Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Pharmacol Toxicol Methods ; 81: 263-73, 2016.
Article in English | MEDLINE | ID: mdl-27095424

ABSTRACT

INTRODUCTION: A priority in the development and approval of new drugs is assessment of cardiovascular risk. Current methodologies for screening compounds (e.g. HERG testing) for proarrhythmic risk lead to many false positive and false negative results, resulting in the attrition of potentially therapeutic compounds in early development, and the advancement of other candidates that cause adverse effects. With improvements in the technologies of high content imaging and human stem cell differentiation, it is now possible to directly screen compounds for arrhythmogenic tendencies in human stem cell derived cardiomyocytes (hSC-CMs). METHODS: A training panel of 90 compounds consisting of roughly equal numbers of QT-prolonging and negative control (non-QT-prolonging) compounds, and a follow-up blinded study of 35 compounds including 16 from the 90 compound panel and 2 duplicates, were evaluated for prolongation of the calcium transient in hSC-CMs using kinetic image cytometry (KIC), a specialized form of high content analysis. RESULTS: The KIC-hSC-CM assay identified training compounds that prolong the calcium transient with 98% specificity, 97% precision, 80% sensitivity, and 89% accuracy in predicting clinical QT prolongation by these compounds. The follow-up study of 35 blinded compounds confirmed the reproducibility and strong diagnostic accuracy of the assay. DISCUSSION: The correlation of the KIC-hSC-CM results to clinical observations met or surpassed traditional preclinical assessment of cardiac risk utilizing animal models. Thus, the KIC-hSC-CM assay, which can be accomplished in high throughput and at relatively low cost, is an effective new model system for testing chemicals for cardiovascular risk.


Subject(s)
Induced Pluripotent Stem Cells/drug effects , Long QT Syndrome/chemically induced , Myocytes, Cardiac/drug effects , Action Potentials/drug effects , Animals , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/physiopathology , Calcium Signaling/drug effects , Cells, Cultured , Humans , Image Cytometry , Long QT Syndrome/physiopathology , Predictive Value of Tests , Quality Control , Reproducibility of Results
2.
Cardiovasc Res ; 111(3): 274-86, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27097650

ABSTRACT

AIMS: Current mechanisms driving cardiac pacemaker function have focused on ion channel and gap junction channel function, which are essential for action potential generation and propagation between pacemaker cells. However, pacemaker cells also harbour desmosomes that structurally anchor pacemaker cells to each other in tissue, but their role in pacemaker function remains unknown. METHODS AND RESULTS: To determine the role of desmosomes in pacemaker function, we generated a novel mouse model harbouring cardiac conduction-specific ablation (csKO) of the central desmosomal protein, desmoplakin (DSP) using the Hcn4-Cre-ERT2 mouse line. Hcn4-Cre targets cells of the adult mouse sinoatrial node (SAN) and can ablate DSP expression in the adult DSP csKO SAN resulting in specific loss of desmosomal proteins and structures. Dysregulation of DSP via loss-of-function (adult DSP csKO mice) and mutation (clinical case of a patient harbouring a pathogenic DSP variant) in mice and man, respectively, revealed that desmosomal dysregulation is associated with a primary phenotype of increased sinus pauses/dysfunction in the absence of cardiomyopathy. Underlying defects in beat-to-beat regulation were also observed in DSP csKO mice in vivo and intact atria ex vivo. DSP csKO SAN exhibited migrating lead pacemaker sites associated with connexin 45 loss. In vitro studies exploiting ventricular cardiomyocytes that harbour DSP loss and concurrent early connexin loss phenocopied the loss of beat-to-beat regulation observed in DSP csKO mice and atria, extending the importance of DSP-associated mechanisms in driving beat-to-beat regulation of working cardiomyocytes. CONCLUSION: We provide evidence of a mechanism that implicates an essential role for desmosomes in cardiac pacemaker function, which has broad implications in better understanding mechanisms underlying beat-to-beat regulation as well as sinus node disease and dysfunction.


Subject(s)
Biological Clocks , Desmosomes , Heart Rate , Sick Sinus Syndrome/physiopathology , Sinoatrial Node/physiopathology , Action Potentials , Age Factors , Animals , Atrial Function , Cells, Cultured , Connexins/metabolism , Desmoplakins/deficiency , Desmoplakins/genetics , Desmosomes/metabolism , Desmosomes/ultrastructure , Genetic Predisposition to Disease , Humans , Mice, Knockout , Mutation , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Phenotype , Sick Sinus Syndrome/genetics , Sick Sinus Syndrome/metabolism , Sick Sinus Syndrome/pathology , Sinoatrial Node/metabolism , Sinoatrial Node/ultrastructure , Time Factors
3.
Toxicol Sci ; 148(2): 503-16, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26358003

ABSTRACT

Human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) are emerging as a powerful in vitro model for cardiac safety assessment which may allow for better identification of compounds with poor arrhythmogenic liability profiles early in the drug discovery process. Here, we describe our examination of the Kinetic Image Cytometer (KIC) system's ability to predict adverse compound effects using hiPS-CMs and a library of 53 compounds, the majority of which are known to be cardioactive compounds, and several negative controls. The KIC provides a high throughput method for analyzing intracellular calcium transients. In the cardiomyocyte, intracellular calcium transients integrate the electrochemical signals of the action potential (AP) with the molecular signaling pathways regulating contraction. Drug-induced alterations in the shape and duration of AP result in changes to the shape and duration of the intracellular calcium transient. By examining calcium transient dynamics in hiPS-CMs, KIC can be used as a phenotypic screen to assess compound effects across multiple ion channel types (MITs), detecting MITs, calcium handling and signaling effects. The results of this blinded study indicate that using hiPS-CMs, KIC is able to accurately detect drug-induced changes in Ca(2+) transient dynamics (ie, duration and beat rate) and therefore, may be useful in predicting drug-induced arrhythmogenic liabilities in early de-risking within the drug discovery phase.


Subject(s)
Calcium Signaling/drug effects , Heart Diseases/chemically induced , High-Throughput Screening Assays , Image Cytometry , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Toxicity Tests/methods , Action Potentials , Animal Testing Alternatives , Cardiotoxicity , Cell Line , Dose-Response Relationship, Drug , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/physiopathology , Heart Rate/drug effects , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Kinetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phenotype , Reproducibility of Results , Risk Assessment
4.
Cell Mol Bioeng ; 8(2): 237-246, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-27087858

ABSTRACT

Volume loading of the cardiac ventricles is known to slow electrical conduction in the rabbit heart, but the mechanisms remain unclear. Previous experimental and modeling studies have investigated some of these mechanisms, including stretch-activated membrane currents, reduced gap junctional conductance, and altered cell membrane capacitance. In order to quantify the relative contributions of these mechanisms, we combined a monomain model of rabbit ventricular electrophysiology with a hyperelastic model of passive ventricular mechanics. First, a simplified geometric model with prescribed homogeneous deformation was used to fit model parameters and characterize individual MEF mechanisms, and showed good qualitative agreement with experimentally measured strain-CV relations. A 3D model of the rabbit left and right ventricles was then compared with experimental measurements from optical electrical mapping studies in the isolated rabbit heart. The model was inflated to an end-diastolic pressure of 30 mmHg, resulting in epicardial strains comparable to those measured in the anterior left ventricular free wall. While the effects of stretch activated channels did alter epicardial conduction velocity, an increase in cellular capacitance was required to explain previously reported experimental results. The new results suggest that for large strains, various mechanisms can combine and produce a biphasic relationship between strain and conduction velocity. However, at the moderate strains generated by high end-diastolic pressure, a stretch-induced increase in myocyte membrane capacitance is the dominant driver of conduction slowing during ventricular volume loading.

5.
PLoS Genet ; 10(2): e1004114, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24586179

ABSTRACT

Recent interest has focused on the importance of the nucleus and associated nucleoskeleton in regulating changes in cardiac gene expression in response to biomechanical load. Mutations in genes encoding proteins of the inner nuclear membrane and nucleoskeleton, which cause cardiomyopathy, also disrupt expression of a biomechanically responsive gene program. Furthermore, mutations in the outer nuclear membrane protein Nesprin 1 and 2 have been implicated in cardiomyopathy. Here, we identify for the first time a role for the outer nuclear membrane proteins, Nesprin 1 and Nesprin 2, in regulating gene expression in response to biomechanical load. Ablation of both Nesprin 1 and 2 in cardiomyocytes, but neither alone, resulted in early onset cardiomyopathy. Mutant cardiomyocytes exhibited altered nuclear positioning, shape, and chromatin positioning. Loss of Nesprin 1 or 2, or both, led to impairment of gene expression changes in response to biomechanical stimuli. These data suggest a model whereby biomechanical signals are communicated from proteins of the outer nuclear membrane, to the inner nuclear membrane and nucleoskeleton, to result in changes in gene expression required for adaptation of the cardiomyocyte to changes in biomechanical load, and give insights into etiologies underlying cardiomyopathy consequent to mutations in Nesprin 1 and 2.


Subject(s)
Cardiomyopathies/genetics , Myocardium/metabolism , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Animals , Biomechanical Phenomena , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cell Nucleus/metabolism , Cytoskeletal Proteins , Gene Expression Regulation , Humans , Mice , Mutation , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Nuclear Matrix/metabolism , Nuclear Proteins/metabolism
6.
Hum Mol Genet ; 23(5): 1134-50, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24108106

ABSTRACT

Arrhythmogenic right ventricular cardiomyopathy (ARVC) termed a 'disease of the desmosome' is an inherited cardiomyopathy that recently underwent reclassification owing to the identification of left-dominant and biventricular disease forms. Homozygous loss-of-function mutations in the desmosomal component, desmoplakin, are found in patients exhibiting a biventricular form of ARVC; however, no models recapitulate the postnatal hallmarks of the disease as seen in these patients. To gain insights into the homozygous loss-of-function effects of desmoplakin in the heart, we generated cardiomyocyte-specific desmoplakin-deficient mice (DSP-cKO) using ventricular myosin light chain-2-Cre mice. Homozygous DSP-cKO mice are viable but display early ultrastructural defects in desmosomal integrity leading to a cardiomyopathy reminiscent of a biventricular form of ARVC, which includes cell death and fibro-fatty replacement within the ventricle leading to biventricular dysfunction, failure and premature death. DSP-cKO mice also exhibited ventricular arrhythmias that are exacerbated with exercise and catecholamine stimulation. Furthermore, DSP-cKO hearts exhibited right ventricular conduction defects associated with loss of connexin 40 expression and electrical wavefront propagation defects associated with loss of connexin 43 expression. Dose-dependent assessment of the effects of loss of desmoplakin in neonatal ventricular cardiomyocytes revealed primary loss of connexin 43 levels, phosphorylation and function independent of the molecular dissociation of the mechanical junction complex and fibro-fatty manifestation associated with ARVC, suggesting a role for desmoplakin as a primary stabilizer of connexin integrity. In summary, we provide evidence for a novel mouse model, which is reminiscent of the postnatal onset of ARVC while highlighting mechanisms underlying a biventricular form of human ARVC.


Subject(s)
Arrhythmogenic Right Ventricular Dysplasia/genetics , Connexins/deficiency , Animals , Animals, Newborn , Arrhythmias, Cardiac/genetics , Arrhythmogenic Right Ventricular Dysplasia/diagnosis , Arrhythmogenic Right Ventricular Dysplasia/mortality , Brugada Syndrome , Cardiac Conduction System Disease , Catecholamines/pharmacology , Connexin 43/deficiency , Connexin 43/genetics , Connexin 43/metabolism , Connexins/genetics , Desmoplakins/deficiency , Disease Models, Animal , Electrocardiography , Gene Expression , Heart/drug effects , Heart Conduction System/abnormalities , Magnetic Resonance Imaging , Mice , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Myocardium/ultrastructure , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/ultrastructure , Phosphorylation , Physical Conditioning, Animal/adverse effects , Gap Junction alpha-5 Protein
7.
J Biomech Eng ; 136(2): 021007, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24337452

ABSTRACT

Cardiac mechanical contraction is triggered by electrical activation via an intracellular calcium-dependent process known as excitation-contraction coupling. Dysregulation of cardiac myocyte intracellular calcium handling is a common feature of heart failure. At the organ scale, electrical dyssynchrony leads to mechanical alterations and exacerbates pump dysfunction in heart failure. A reverse coupling between cardiac mechanics and electrophysiology is also well established. It is commonly referred as cardiac mechanoelectric feedback and thought to be an important contributor to the increased risk of arrhythmia during pathological conditions that alter regional cardiac wall mechanics, including heart failure. At the cellular scale, most investigations of myocyte mechanoelectric feedback have focused on the roles of stretch-activated ion channels, though mechanisms that are independent of ionic currents have also been described. Here we review excitation-contraction coupling and mechanoelectric feedback at the cellular and organ scales, and we identify the need for new multicellular tissue-scale model systems and experiments that can help us to obtain a better understanding of how interactions between electrophysiological and mechanical processes at the cell scale affect ventricular electromechanical interactions at the organ scale in the normal and diseased heart.


Subject(s)
Excitation Contraction Coupling/physiology , Feedback, Physiological/physiology , Heart Conduction System/physiology , Models, Cardiovascular , Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Ventricular Function/physiology , Animals , Humans
8.
Mol Biol Cell ; 22(22): 4324-34, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21937718

ABSTRACT

Changes in blood flow regulate gene expression and protein synthesis in vascular endothelial cells, and this regulation is involved in the development of atherosclerosis. How mechanical stimuli are transmitted from the endothelial luminal surface to the nucleus is incompletely understood. The linker of nucleus and cytoskeleton (LINC) complexes have been proposed as part of a continuous physical link between the plasma membrane and subnuclear structures. LINC proteins nesprin-1, -2, and -4 have been shown to mediate nuclear positioning via microtubule motors and actin. Although nesprin-3 connects intermediate filaments to the nucleus, no functional consequences of nesprin-3 mutations on cellular processes have been described. Here we show that nesprin-3 is robustly expressed in human aortic endothelial cells (HAECs) and localizes to the nuclear envelope. Nesprin-3 regulates HAEC morpho-logy, with nesprin-3 knockdown inducing prominent cellular elongation. Nesprin-3 also organizes perinuclear cytoskeletal organization and is required to attach the centrosome to the nuclear envelope. Finally, nesprin-3 is required for flow-induced polarization of the centrosome and flow-induced migration in HAECs. These results represent the most complete description to date of nesprin-3 function and suggest that nesprin-3 regulates vascular endothelial cell shape, perinuclear cytoskeletal architecture, and important aspects of flow-mediated mechanotransduction.


Subject(s)
Cell Polarity , Cytoskeleton/metabolism , Endothelial Cells/cytology , Microfilament Proteins/metabolism , Nuclear Envelope/metabolism , Aorta/metabolism , Cell Movement , Cell Nucleus/metabolism , Cell Shape , Cells, Cultured , Centrosome/metabolism , Centrosome/ultrastructure , Cytoskeleton/genetics , Cytoskeleton/ultrastructure , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Humans , Intermediate Filaments/metabolism , Intermediate Filaments/ultrastructure , Mechanotransduction, Cellular , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microfilament Proteins/biosynthesis , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Envelope/genetics , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Interference , RNA, Small Interfering
9.
Biophys J ; 97(4): 1125-9, 2009 Aug 19.
Article in English | MEDLINE | ID: mdl-19686660

ABSTRACT

Calcium is essential for many biological processes involved in cellular motility. However, the pathway by which calcium influences motility, in processes such as muscle contraction and neuronal growth, is often indirect and complex. We establish a simple and direct mechanochemical link that shows how calcium quantitatively regulates the dynamics of a primitive motile system, the actin-based acrosomal bundle of horseshoe crab sperm. The extension of this bundle requires the continuous presence of external calcium. Furthermore, the extension rate increases with calcium concentration, but at a given concentration, we find that the volumetric rate of extension is constant. Our experiments and theory suggest that calcium sequentially binds to calmodulin molecules decorating the actin filaments. This binding leads to a collective wave of untwisting of the actin filaments that drives bundle extension.


Subject(s)
Actins/physiology , Calcium/physiology , Models, Biological , Models, Chemical , Molecular Motor Proteins/physiology , Sperm Motility/physiology , Spermatozoa/physiology , Actins/chemistry , Animals , Calcium/chemistry , Cells, Cultured , Elastic Modulus , Horseshoe Crabs , Male , Molecular Motor Proteins/chemistry , Spermatozoa/chemistry , Stress, Mechanical
10.
Antimicrob Agents Chemother ; 51(9): 3410-2, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17606687

ABSTRACT

We used an assay to measure quinolone sensitivity as a shift in the position of the cleavage-religation equilibrium. This assay was found to be useful in identifying the primary target of a quinolone drug and assessing the effect of quinolone resistance-conferring mutations.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/genetics , Mutation/genetics , Quinolones/pharmacology , DNA Gyrase/genetics , DNA Topoisomerase IV/genetics , Escherichia coli/drug effects , Escherichia coli/genetics , Microbial Sensitivity Tests , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics
11.
Antimicrob Agents Chemother ; 48(2): 608-11, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14742217

ABSTRACT

Replacement of the alpha4 helix of ParC with that of GyrA increased the stability of topoisomerase IV-quinolone-DNA ternary complexes. This mutant topoisomerase IV-mediated cell killing was more efficient than topoisomerase IV-mediated cell killing in Escherichia coli. Thus, the alpha4 helix plays critical roles in determining the stability and the cytotoxicity of ternary complexes.


Subject(s)
DNA Gyrase/genetics , DNA Topoisomerase IV/chemistry , DNA Topoisomerase IV/genetics , DNA, Bacterial/genetics , Escherichia coli/genetics , Quinolones/chemistry , Anti-Infective Agents/pharmacology , Cloning, Molecular , DNA Gyrase/chemistry , DNA, Bacterial/chemistry , Drug Resistance, Bacterial , Escherichia coli/drug effects , Mutation/genetics , Norfloxacin/pharmacology
12.
J Biol Chem ; 278(48): 48154-61, 2003 Nov 28.
Article in English | MEDLINE | ID: mdl-12972433

ABSTRACT

Endothelin-1 (ET-1) is an autocrine factor in the mammalian heart important in enhancing cardiac performance, protecting against myocardial ischemia, and initiating the development of cardiac hypertrophy. The ETA receptor is a seven-transmembrane G-protein-coupled receptor whose precise subcellular localization in cardiac muscle is unknown. Here we used fluorescein ET-1 and 125I-ET-1 to provide evidence for ET-1 receptors in cardiac transverse tubules (T-tubules). Moreover, the ETA receptor and downstream effector phospholipase C-beta 1 were co-localized within T-tubules using standard immunofluorescence techniques, and protein kinase C (PKC)-epsilon-enhanced green fluorescent protein bound reversibly to T-tubules upon activation. Localized photorelease of diacylglycerol further suggested compartmentation of PKC signaling, with release at the myocyte "surface" mimicking the negative inotropic effects of bath-applied PKC activators and "deep" release mimicking the positive inotropic effect of ET-1. The functional significance of T-tubular ET-1 receptors was further tested by rendering the T-tubule lumen inaccessible to bath-applied ET-1. Such "detubulated" cardiac myocytes showed no positive inotropic response to 20 nM ET-1, despite retaining both a nearly normal twitch response to field stimulation and a robust positive inotropic response to 20 nm isoproterenol. We propose that ET-1 enhances myocyte contractility by activating ETA receptor-phospholipase C-beta 1-PKC-epsilon signaling complexes preferentially localized in cardiac T-tubules. Compartmentation of ET-1 signaling complexes may explain the discordant effects of ET-1 versus bath applied PKC activators and may contribute to both the specificity and diversity of the cardiac actions of ET-1.


Subject(s)
Myocardium/metabolism , Myocytes, Cardiac/physiology , Receptors, Endothelin/metabolism , Animals , Blotting, Western , Dogs , Dose-Response Relationship, Drug , Endothelin-1/metabolism , Fluorescein/pharmacology , Green Fluorescent Proteins , Heart Ventricles/metabolism , Isoenzymes/metabolism , Isoproterenol/pharmacology , Kinetics , Luminescent Proteins/metabolism , Male , Microscopy, Fluorescence , Models, Biological , Models, Chemical , Myocardium/cytology , Myocytes, Cardiac/metabolism , Phospholipase C beta , Photons , Protein Binding , Protein Kinase C/metabolism , Protein Kinase C-epsilon , Rats , Rats, Sprague-Dawley , Signal Transduction , Type C Phospholipases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...