Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Front Pharmacol ; 12: 794620, 2021.
Article in English | MEDLINE | ID: mdl-35115940

ABSTRACT

The congenital short QT syndrome (SQTS) is a cardiac condition that leads to abbreviated ventricular repolarization and an increased susceptibility to arrhythmia and sudden death. The SQT3 form of the syndrome is due to mutations to the KCNJ2 gene that encodes Kir2.1, a critical component of channels underlying cardiac inwardly rectifying K+ current, IK1. The first reported SQT3 KCNJ2 mutation gives rise to the D172N Kir2.1 mutation, the consequences of which have been studied on recombinant channels in vitro and in ventricular cell and tissue simulations. The aim of this study was to establish the effects of the D172N mutation on ventricular repolarization through real-time replacement of IK1 using the dynamic clamp technique. Whole-cell patch-clamp recordings were made from adult guinea-pig left ventricular myocytes at physiological temperature. Action potentials (APs) were elicited at 1 Hz. Intrinsic IK1 was inhibited with a low concentration (50 µM) of Ba2+ ions, which led to AP prolongation and triangulation, accompanied by a ∼6 mV depolarization of resting membrane potential. Application of synthetic IK1 through dynamic clamp restored AP duration, shape and resting potential. Replacement of wild-type (WT) IK1 with heterozygotic (WT-D172N) or homozygotic (D172N) mutant formulations under dynamic clamp significantly abbreviated AP duration (APD90) and accelerated maximal AP repolarization velocity, with no significant hyperpolarization of resting potential. Across stimulation frequencies from 0.5 to 3 Hz, the relationship between APD90 and cycle length was downward shifted, reflecting AP abbreviation at all stimulation frequencies tested. In further AP measurements at 1 Hz from hiPSC cardiomyocytes, the D172N mutation produced similar effects on APD and repolarization velocity; however, resting potential was moderately hyperpolarized by application of mutant IK1 to these cells. Overall, the results of this study support the major changes in ventricular cell AP repolarization with the D172N predicted from prior AP modelling and highlight the potential utility of using adult ventricular cardiomyocytes for dynamic clamp exploration of functional consequences of Kir2.1 mutations.

2.
Clin Pharmacol Ther ; 107(1): 102-111, 2020 01.
Article in English | MEDLINE | ID: mdl-31709525

ABSTRACT

This white paper presents principles for validating proarrhythmia risk prediction models for regulatory use as discussed at the In Silico Breakout Session of a Cardiac Safety Research Consortium/Health and Environmental Sciences Institute/US Food and Drug Administration-sponsored Think Tank Meeting on May 22, 2018. The meeting was convened to evaluate the progress in the development of a new cardiac safety paradigm, the Comprehensive in Vitro Proarrhythmia Assay (CiPA). The opinions regarding these principles reflect the collective views of those who participated in the discussion of this topic both at and after the breakout session. Although primarily discussed in the context of in silico models, these principles describe the interface between experimental input and model-based interpretation and are intended to be general enough to be applied to other types of nonclinical models for proarrhythmia assessment. This document was developed with the intention of providing a foundation for more consistency and harmonization in developing and validating different models for proarrhythmia risk prediction using the example of the CiPA paradigm.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Computer Simulation , Drug-Related Side Effects and Adverse Reactions/etiology , Risk Assessment/methods , Arrhythmias, Cardiac/prevention & control , Drug-Related Side Effects and Adverse Reactions/prevention & control , Humans , Models, Theoretical , Validation Studies as Topic
3.
J Physiol ; 597(6): 1705-1733, 2019 03.
Article in English | MEDLINE | ID: mdl-30629744

ABSTRACT

KEY POINTS: Tymothy syndrome (TS) is a multisystem disorder featuring cardiac arrhythmias, autism and adrenal gland dysfunction that originates from a de novo point mutation in the gene encoding the Cav1.2 (CACNA1C) L-type channel. To study the role of Cav1.2 channel signals in autism, the autistic TS2-neo mouse has been generated bearing the G406R point-mutation associated with TS type-2. Using heterozygous TS2-neo mice, we report that the G406R mutation reduces the rate of inactivation and shifts leftward the activation and inactivation of L-type channels, causing marked increase of resting Ca2+ influx ('window' Ca2+ current). The increased 'window current' causes marked reduction of NaV channel density, switches normal tonic firing to abnormal burst firing, reduces mitochondrial metabolism, induces cell swelling and decreases catecholamine release. Overnight incubations with nifedipine rescue NaV channel density, normal firing and the quantity of catecholamine released. We provide evidence that chromaffin cell malfunction derives from altered Cav1.2 channel gating. ABSTRACT: L-type voltage-gated calcium (Cav1) channels have a key role in long-term synaptic plasticity, sensory transduction, muscle contraction and hormone release. A point mutation in the gene encoding Cav1.2 (CACNA1C) causes Tymothy syndrome (TS), a multisystem disorder featuring cardiac arrhythmias, autism spectrum disorder (ASD) and adrenal gland dysfunction. In the more severe type-2 form (TS2), the missense mutation G406R is on exon 8 coding for the IS6-helix of the Cav1.2 channel. The mutation causes reduced inactivation and induces autism. How this occurs and how Cav1.2 gating-changes alter cell excitability, neuronal firing and hormone release on a molecular basis is still largely unknown. Here, using the TS2-neo mouse model of TS we show that the G406R mutation altered excitability and reduced secretory activity in adrenal chromaffin cells (CCs). Specifically, the TS2 mutation reduced the rate of voltage-dependent inactivation and shifted leftward the activation and steady-state inactivation of L-type channels. This markedly increased the resting 'window' Ca2+ current that caused an increased percentage of CCs undergoing abnormal action potential (AP) burst firing, cell swelling, reduced mitochondrial metabolism and decreased catecholamine release. The increased 'window' Ca2+ current caused also decreased NaV channel density and increased steady-state inactivation, which contributed to the increased abnormal burst firing. Overnight incubation with the L-type channel blocker nifedipine rescued the normal AP firing of CCs, the density of functioning NaV channels and their steady-state inactivation. We provide evidence that CC malfunction derives from the altered Cav1.2 channel gating and that dihydropyridines are potential therapeutics for ASD.


Subject(s)
Action Potentials , Autistic Disorder/genetics , Calcium Channels, L-Type/genetics , Chromaffin Cells/metabolism , Exocytosis , Long QT Syndrome/genetics , Syndactyly/genetics , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Catecholamines/metabolism , Cells, Cultured , Chromaffin Cells/drug effects , Chromaffin Cells/physiology , Ion Channel Gating , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Nifedipine/pharmacology , Point Mutation , Sodium Channels/metabolism
4.
Glia ; 66(11): 2324-2339, 2018 11.
Article in English | MEDLINE | ID: mdl-30151840

ABSTRACT

To study the role of L-type voltage-gated Ca++ channels in oligodendrocyte development, we used a mouse model of Timothy syndrome (TS) in which a gain-of-function mutation in the α1 subunit of the L-type Ca++ channel Cav1.2 gives rise to an autism spectrum disorder (ASD). Oligodendrocyte progenitor cells (OPCs) isolated from the cortex of TS mice showed greater L-type Ca++ influx and displayed characteristics suggestive of advanced maturation compared to control OPCs, including a more complex morphology and higher levels of myelin protein expression. Consistent with this, expression of Cav1.2 channels bearing the TS mutation in wild-type OPCs triggered process formation and promoted oligodendrocyte-neuron interaction via the activation of Ca++ /calmodulin-dependent protein kinase II. To ascertain whether accelerated OPC maturation correlated with functional enhancements, we examined myelination in the TS brain at different postnatal time points. The expression of myelin proteins was significantly higher in the corpus callosum, cortex and striatum of TS animals, and immunohistochemical analysis for oligodendrocyte stage-specific markers revealed an increase in the density of myelinating oligodendrocytes in several areas of the TS brain. Along the same line, electron microscopy studies in the corpus callosum of TS animals showed significant increases both in the percentage of myelinated axons and in the thickness of myelin sheaths. In summary, these data indicate that OPC development and oligodendrocyte myelination is enhanced in the brain of TS mice, and suggest that this mouse model of a syndromic ASD is a useful tool to explore the role of L-type Ca++ channels in myelination.


Subject(s)
Autistic Disorder/complications , Autistic Disorder/pathology , Cell Differentiation/genetics , Gene Expression Regulation, Developmental/genetics , Long QT Syndrome/complications , Long QT Syndrome/pathology , Myelin Proteins/metabolism , Oligodendroglia/physiology , Syndactyly/complications , Syndactyly/pathology , Animals , Animals, Newborn , Autistic Disorder/genetics , Autophagy-Related Proteins , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Calcium/metabolism , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Coculture Techniques , Disease Models, Animal , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Long QT Syndrome/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neurons/ultrastructure , Oligodendrocyte Precursor Cells/drug effects , Oligodendrocyte Precursor Cells/metabolism , Oligodendrocyte Precursor Cells/pathology , Oligodendroglia/pathology , Oligodendroglia/ultrastructure , Potassium/pharmacology , Syndactyly/genetics
5.
Sci Rep ; 8(1): 11246, 2018 07 26.
Article in English | MEDLINE | ID: mdl-30050137

ABSTRACT

Brugada syndrome (BrS) is an inherited cardiac arrhythmia commonly associated with SCN5A mutations, yet its ionic mechanisms remain unclear due to a lack of cellular models. Here, we used human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) from a BrS patient (BrS1) to evaluate the roles of Na+ currents (INa) and transient outward K+ currents (Ito) in BrS induced action potential (AP) changes. To understand the role of these current changes in repolarization we employed dynamic clamp to "electronically express" IK1 and restore normal resting membrane potentials and allow normal recovery of the inactivating currents, INa, ICa and Ito. HiPSC-CMs were generated from BrS1 with a compound SCN5A mutation (p. A226V & p. R1629X) and a healthy sibling control (CON1). Genome edited hiPSC-CMs (BrS2) with a milder p. T1620M mutation and a commercial control (CON2) were also studied. CON1, CON2 and BrS2, had unaltered peak INa amplitudes, and normal APs whereas BrS1, with over 75% loss of INa, displayed a loss-of-INa basal AP morphology (at 1.0 Hz) manifested by a reduced maximum upstroke velocity (by ~80%, p < 0.001) and AP amplitude (p < 0.001), and an increased phase-1 repolarization pro-arrhythmic AP morphology (at 0.1 Hz) in ~25% of cells characterized by marked APD shortening (~65% shortening, p < 0.001). Moreover, Ito densities of BrS1 and CON1 were comparable and increased from 1.0 Hz to 0.1 Hz by ~ 100%. These data indicate that a repolarization deficit could be a mechanism underlying BrS.


Subject(s)
Brugada Syndrome/physiopathology , Membrane Potentials , Myocytes, Cardiac/pathology , Potassium/metabolism , Sodium/metabolism , Cell Differentiation , Humans , Mutation , NAV1.5 Voltage-Gated Sodium Channel/genetics , Patch-Clamp Techniques , Pluripotent Stem Cells/physiology
6.
Metabolomics ; 13(11)2017 Nov.
Article in English | MEDLINE | ID: mdl-29147105

ABSTRACT

INTRODUCTION: Autism spectrum disorders (ASD) are a group of neurodevelopmental disorders lacking a clinical biomarker for diagnosis. Emerging evidence shows that intestinal microflora from ASD subjects can be distinguished from controls, suggesting metabolite differences due to the action of intestinal microbes may provide a means for identifying potential biomarkers for ASD. OBJECTIVES: The aim of this study was to determine if quantitative differences in levels of stercobilin and stercobilinogen, metabolites produced by biological action of intestinal microflora, exist in the fecal matter between an ASD mouse model population and controls. METHODS: Pairs of fecal samples were collected from two mouse groups, an ASD model group with Timothy syndrome 2 (TS2-NEO) and a gender-matched control group. After centrifugation, supernatant was spiked with an 18O-labeled stercobilin isotopomer and subjected to solid phase extraction for processing. Extracted samples were spotted on a stainless steel plate and subjected to matrix-assisted laser desorption and ionization mass spectrometry using dihydroxybenzoic acid as the matrix (n = 5). Peak areas for bilins and 18O-stercobilin isotopomers were determined in each fecal sample. RESULTS: A 40-45% depletion in stercobilin in TS2-NEO fecal samples compared with controls was observed with p < 0.05; a less dramatic depletion was observed for stercobilinogen. CONCLUSIONS: The results show that stercobilin depletion in feces is observed for an ASD mouse model vs. controls. This may help to explain recent observations of a less diverse microbiome in humans with ASD and may prove helpful in developing a clinical ASD biomarker.

7.
Biol Sex Differ ; 8(1): 26, 2017 08 14.
Article in English | MEDLINE | ID: mdl-28807015

ABSTRACT

BACKGROUND: Women have a higher risk of lethal arrhythmias than men in long QT syndrome type 2 (LQTS2), but the mechanisms remain uncertain due to the limited availability of healthy control human tissue. We have previously reported that in female rabbits, estrogen increases arrhythmia risk in drug-induced LQTS2 by upregulating L-type Ca2+ (ICa,L) and sodium-calcium exchange (INCX) currents at the base of the epicardium by a genomic mechanism. This study investigates if the effects of estrogen on rabbit ICa,L and INCX apply to human hearts. METHODS: Postmortem human left ventricular tissue samples were probed with selective antibodies for regional heterogeneities of ion channel protein expression and compared to rabbit myocardium. Functionally, ICa,L and INCX were measured from female and male cardiomyocytes derived from human induced pluripotent stem cells (iPS-CMs) with the voltage-clamp technique from control and estrogen-treated iPS-CMs. RESULTS: In women (n = 12), Cav1.2α (primary subunit of the L-type calcium channel protein 1) and NCX1 (sodium-calcium exchange protein) levels were higher at the base than apex of the epicardium (40 ± 14 and 81 ± 30%, respectively, P < 0.05), but not in men (n = 6) or postmenopausal women (n = 6). Similarly, in cardiomyocytes derived from female human iPS-CMs, estrogen (1 nM, 1-2 days) increased ICa,L (31%, P < 0.05) and INCX (7.5-fold, - 90 mV, P < 0.01) and their mRNA levels (P < 0.05). Moreover, in male human iPS-CMs, estrogen failed to alter ICa,L and INCX. CONCLUSIONS: The results show that estrogen upregulates cardiac ICa,L and INCX in women through genomic mechanisms that account for sex differences in Ca2+ handling and spatial heterogeneities of repolarization due to base-apex heterogeneities of Cav1.2α and NCX1. By analogy with rabbit studies, these effects account for human sex-difference in arrhythmia risk.


Subject(s)
Calcium Channels, L-Type/metabolism , Estrogens/metabolism , Myocytes, Cardiac/metabolism , Sex Characteristics , Sodium-Calcium Exchanger/metabolism , Up-Regulation/physiology , Adolescent , Adult , Aged , Animals , Cells, Cultured , Estrogens/administration & dosage , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Membrane Potentials/physiology , Middle Aged , Myocardium/metabolism , RNA, Messenger/metabolism , Rabbits , Young Adult
9.
Cell Host Microbe ; 17(5): 642-52, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25974303

ABSTRACT

Toxoplasma gondii is a protozoan pathogen in the phylum Apicomplexa that resides within an intracellular parasitophorous vacuole (PV) that is selectively permeable to small molecules through unidentified mechanisms. We have identified GRA17 as a Toxoplasma-secreted protein that localizes to the parasitophorous vacuole membrane (PVM) and mediates passive transport of small molecules across the PVM. GRA17 is related to the putative Plasmodium translocon protein EXP2 and conserved across PV-residing Apicomplexa. The PVs of GRA17-deficient parasites have aberrant morphology, reduced permeability to small molecules, and structural instability. GRA17-deficient parasites proliferate slowly and are avirulent in mice. These GRA17-deficient phenotypes are rescued by complementation with Plasmodium EXP2. GRA17 functions synergistically with a related protein, GRA23. Exogenous expression of GRA17 or GRA23 alters the membrane conductance properties of Xenopus oocytes in a manner consistent with a large non-selective pore. Thus, GRA17 and GRA23 provide a molecular basis for PVM permeability and nutrient access.


Subject(s)
Antigens, Protozoan/metabolism , Membrane Transport Proteins/metabolism , Toxoplasma/physiology , Vacuoles/parasitology , Virulence Factors/metabolism , Animals , Antigens, Protozoan/genetics , Biological Transport , Gene Deletion , Genetic Complementation Test , Membrane Transport Proteins/genetics , Mice , Toxoplasma/growth & development , Toxoplasma/metabolism , Toxoplasmosis, Animal/parasitology , Toxoplasmosis, Animal/pathology , Virulence , Virulence Factors/genetics , Xenopus
10.
J Mol Cell Cardiol ; 84: 52-60, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25889894

ABSTRACT

The inward rectifier potassium current, IK1, contributes to the terminal phase of repolarization of the action potential (AP), as well as the value and stability of the resting membrane potential. Regional variation in IK1 has been noted in the canine heart, but the biophysical properties have not been directly compared. We examined the properties and functional contribution of IK1 in isolated myocytes from ventricular, atrial and Purkinje tissue. APs were recorded from canine left ventricular midmyocardium, left atrial and Purkinje tissue. The terminal rate of repolarization of the AP in ventricle, but not in Purkinje, depended on changes in external K(+) ([K(+)]o). Isolated ventricular myocytes had the greatest density of IK1 while atrial myocytes had the lowest. Furthermore, the outward component of IK1 in ventricular cells exhibited a prominent outward component and steep negative slope conductance, which was also enhanced in 10 mM [K(+)]o. In contrast, both Purkinje and atrial cells exhibited little outward IK1, even in the presence of 10 mM [K(+)]o, and both cell types showed more persistent current at positive potentials. Expression of Kir2.1 in the ventricle was 76.9-fold higher than that of atria and 5.8-fold higher than that of Purkinje, whereas the expression of Kir2.2 and Kir2.3 subunits was more evenly distributed in Purkinje and atria. Finally, AP clamp data showed distinct contributions of IK1 for each cell type. IK1 and Kir2 subunit expression varies dramatically in regions of the canine heart and these regional differences in Kir2 expression likely underlie regional distinctions in IK1 characteristics, contributing to variations in repolarization in response to in [K(+)]o changes.


Subject(s)
Action Potentials/physiology , Heart/physiology , Potassium Channels, Inwardly Rectifying/metabolism , Animals , Dogs , Female , Heart Atria/metabolism , Heart Ventricles/metabolism , Ion Channel Gating , Kinetics , Male , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Polyamines/metabolism , Potassium/metabolism , Potassium Channels, Inwardly Rectifying/genetics , Protein Subunits/genetics , Protein Subunits/metabolism , Purkinje Cells/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
Dis Model Mech ; 8(5): 457-66, 2015 May.
Article in English | MEDLINE | ID: mdl-25791035

ABSTRACT

Duchenne muscular dystrophy (DMD) is caused by mutations in the dystrophin gene (DMD), and is characterized by progressive weakness in skeletal and cardiac muscles. Currently, dilated cardiomyopathy due to cardiac muscle loss is one of the major causes of lethality in late-stage DMD patients. To study the molecular mechanisms underlying dilated cardiomyopathy in DMD heart, we generated cardiomyocytes (CMs) from DMD and healthy control induced pluripotent stem cells (iPSCs). DMD iPSC-derived CMs (iPSC-CMs) displayed dystrophin deficiency, as well as the elevated levels of resting Ca(2+), mitochondrial damage and cell apoptosis. Additionally, we found an activated mitochondria-mediated signaling network underlying the enhanced apoptosis in DMD iPSC-CMs. Furthermore, when we treated DMD iPSC-CMs with the membrane sealant Poloxamer 188, it significantly decreased the resting cytosolic Ca(2+) level, repressed caspase-3 (CASP3) activation and consequently suppressed apoptosis in DMD iPSC-CMs. Taken together, using DMD patient-derived iPSC-CMs, we established an in vitro model that manifests the major phenotypes of dilated cardiomyopathy in DMD patients, and uncovered a potential new disease mechanism. Our model could be used for the mechanistic study of human muscular dystrophy, as well as future preclinical testing of novel therapeutic compounds for dilated cardiomyopathy in DMD patients.


Subject(s)
Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/therapy , Induced Pluripotent Stem Cells/cytology , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/therapy , Muscular Dystrophy, Duchenne/therapy , Adolescent , Animals , Apoptosis/drug effects , Calcium/metabolism , Caspase 3/metabolism , Cell Differentiation/drug effects , Disease Models, Animal , Humans , Induced Pluripotent Stem Cells/ultrastructure , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Muscular Dystrophy, Duchenne/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Poloxamer/pharmacology , Sequence Analysis, RNA , Signal Transduction/drug effects , Transcriptome/genetics
13.
J Physiol ; 593(6): 1347-60, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25772290

ABSTRACT

This paper is the second of a series of three reviews published in this issue resulting from the University of California Davis Cardiovascular Symposium 2014: Systems approach to understanding cardiac excitation-contraction coupling and arrhythmias: Na(+) channel and Na(+) transport. The goal of the symposium was to bring together experts in the field to discuss points of consensus and controversy on the topic of sodium in the heart. The present review focuses on Na(+) channel function and regulation, Na(+) channel structure and function, and Na(+) channel trafficking, sequestration and complexing.


Subject(s)
Myocytes, Cardiac/metabolism , Sodium Channels/metabolism , Action Potentials , Amino Acid Sequence , Animals , Congresses as Topic , Humans , Molecular Sequence Data , Myocytes, Cardiac/drug effects , Protein Transport , Sodium Channel Blockers/pharmacology , Sodium Channels/chemistry
14.
J Mol Cell Cardiol ; 81: 81-93, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25644533

ABSTRACT

BACKGROUND AND OBJECTIVES: The creation of cardiomyocytes derived from human induced pluripotent stem cells (hiPS-CMs) has spawned broad excitement borne out of the prospects to diagnose and treat cardiovascular diseases based on personalized medicine. A common feature of hiPS-CMs is their spontaneous contractions but the mechanism(s) remain uncertain. METHODS: Intrinsic activity was investigated by the voltage-clamp technique, optical mapping of action potentials (APs) and intracellular Ca(2+) (Cai) transients (CaiT) at subcellular-resolution and pharmacological interventions. RESULTS: The frequency of spontaneous CaiT (sCaiT) in monolayers of hiPS-CMs was not altered by ivabradine, an inhibitor of the pacemaker current, If despite high levels of HCN transcripts (1-4). HiPS-CMs had negligible If and IK1 (inwardly-rectifying K(+)-current) and a minimum diastolic potential of -59.1±3.3mV (n=18). APs upstrokes were preceded by a depolarizing-foot coincident with a rise of Cai. Subcellular Cai wavelets varied in amplitude, propagated and died-off; larger Cai-waves triggered cellular sCaTs and APs. SCaiTs increased in frequency with [Ca(2+)]out (0.05-to-1.8mM), isoproterenol (1µM) or caffeine (100µM) (n≥5, p<0.05). HiPS-CMs became quiescent with ryanodine receptor stabilizers (K201=2µM); tetracaine; Na-Ca exchange (NCX) inhibition (SEA0400=2µM); higher [K(+)]out (5→8mM), and thiol-reducing agents but could still be electrically stimulated to elicit CaiTs. Cell-cell coupling of hiPS-CM in monolayers was evident from connexin-43 expression and CaiT propagation. SCaiTs from an ensemble of dispersed hiPS-CMs were out-of-phase but became synchronous through the outgrowth of inter-connecting microtubules. CONCLUSIONS: Automaticity in hiPS-CMs originates from a Ca(2+)-clock mechanism involving Ca(2+) cycling across the sarcoplasmic reticulum linked to NCX to trigger APs.


Subject(s)
Calcium/metabolism , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Sarcoplasmic Reticulum/metabolism , Sodium-Calcium Exchanger/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Aniline Compounds/pharmacology , Animals , Benzazepines/pharmacology , Caffeine/pharmacology , Cardiovascular Agents/pharmacology , Cell Differentiation , Cell Line , Cellular Reprogramming , Connexin 43/metabolism , Dependovirus/genetics , Genetic Vectors , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Isoproterenol/pharmacology , Ivabradine , Microtubules/drug effects , Microtubules/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques , Phenyl Ethers/pharmacology , Sarcoplasmic Reticulum/drug effects , Sodium-Calcium Exchanger/antagonists & inhibitors , Tetracaine/pharmacology , Transfection
16.
Cardiovasc Res ; 104(2): 258-69, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25209314

ABSTRACT

AIMS: Familial hypertrophic cardiomyopathy (HCM) is one the most common heart disorders, with gene mutations in the cardiac sarcomere. Studying HCM with patient-specific induced pluripotent stem-cell (iPSC)-derived cardiomyocytes (CMs) would benefit the understanding of HCM mechanism, as well as the development of personalized therapeutic strategies. METHODS AND RESULTS: To investigate the molecular mechanism underlying the abnormal CM functions in HCM, we derived iPSCs from an HCM patient with a single missense mutation (Arginine442Glycine) in the MYH7 gene. CMs were next enriched from HCM and healthy iPSCs, followed with whole transcriptome sequencing and pathway enrichment analysis. A widespread increase of genes responsible for 'Cell Proliferation' was observed in HCM iPSC-CMs when compared with control iPSC-CMs. Additionally, HCM iPSC-CMs exhibited disorganized sarcomeres and electrophysiological irregularities. Furthermore, disease phenotypes of HCM iPSC-CMs were attenuated with pharmaceutical treatments. CONCLUSION: Overall, this study explored the possible patient-specific and mutation-specific disease mechanism of HCM, and demonstrates the potential of using HCM iPSC-CMs for future development of therapeutic strategies. Additionally, the whole methodology established in this study could be utilized to study mechanisms of other human-inherited heart diseases.


Subject(s)
Cardiomyopathy, Hypertrophic, Familial/genetics , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Action Potentials , Adult , Animals , Calcium Signaling/genetics , Cardiac Myosins/genetics , Cardiomyopathy, Hypertrophic, Familial/metabolism , Cardiomyopathy, Hypertrophic, Familial/pathology , Case-Control Studies , Cell Proliferation/genetics , Cell Separation/methods , Cells, Cultured , Cellular Reprogramming , Cellular Reprogramming Techniques , Female , Gene Expression Profiling/methods , Gene Expression Regulation , Gene Regulatory Networks , Genetic Predisposition to Disease , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/ultrastructure , Mice, Inbred NOD , Mice, SCID , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/ultrastructure , Myosin Heavy Chains/genetics , Phenotype , Sarcomeres/metabolism , Sarcomeres/ultrastructure , Transcriptome
18.
Heart Rhythm ; 10(12): 1903-10, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24055949

ABSTRACT

BACKGROUND: Human-induced pluripotent stem cell (h-iPSC)-derived cardiac myocytes are a unique model in which human myocyte function and dysfunction are studied, especially those from patients with genetic disorders. They are also considered a major advance for drug safety testing. However, these cells have considerable unexplored potential limitations when applied to quantitative action potential (AP) analysis. One major factor is spontaneous activity and resulting variability and potentially anomalous behavior of AP parameters. OBJECTIVE: To demonstrate the effect of using an in silico interface on electronically expressed I(K1), a major component lacking in h-iPSC-derived cardiac myocytes. METHODS: An in silico interface was developed to express synthetic I(K1) in cells under whole-cell voltage clamp. RESULTS: Electronic I(K1) expression established a physiological resting potential, eliminated spontaneous activity, reduced spontaneous early and delayed afterdepolarizations, and decreased AP variability. The initiated APs had the classic rapid upstroke and spike and dome morphology consistent with data obtained with freshly isolated human myocytes as well as the readily recognizable repolarization attributes of ventricular and atrial cells. The application of 1 µM of BayK-8644 resulted in anomalous AP shortening in h-iPSC-derived cardiac myocytes. When I(K1) was electronically expressed, BayK-8644 prolonged the AP, which is consistent with the existing results on native cardiac myocytes. CONCLUSIONS: The electronic expression of I(K1) is a simple and robust method to significantly improve the physiological behavior of the AP and electrical profile of h-iPSC-derived cardiac myocytes. Increased stability enables the use of this preparation for a controlled quantitative analysis of AP parameters, for example, drug responsiveness, genetic disorders, and dynamic behavior restitution profiles.


Subject(s)
Arrhythmias, Cardiac/metabolism , Calcium Channels, L-Type/biosynthesis , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/cytology , Action Potentials , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques
19.
Circ Res ; 113(3): 313-21, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23748429

ABSTRACT

RATIONALE: Atrial fibrillation (AF) contributes significantly to morbidity and mortality in elderly and hypertensive patients and has been correlated to enhanced atrial fibrosis. Despite a lack of direct evidence that fibrosis causes AF, reversal of fibrosis is considered a plausible therapy. OBJECTIVE: To evaluate the efficacy of the antifibrotic hormone relaxin (RLX) in suppressing AF in spontaneously hypertensive rats (SHR). METHODS AND RESULTS: Normotensive Wistar-Kyoto (WKY) and SHR were treated for 2 weeks with vehicle (WKY+V and SHR+V) or RLX (0.4 mg/kg per day, SHR+RLX) using implantable mini-pumps. Hearts were perfused, mapped optically to analyze action potential durations, intracellular Ca²âº transients, and restitution kinetics, and tested for AF vulnerability. SHR hearts had slower conduction velocity (CV; P<0.01 versus WKY), steeper CV restitution kinetics, greater collagen deposition, higher levels of transcripts for transforming growth factor-ß, metalloproteinase-2, metalloproteinase-9, collagen I/III, and reduced connexin 43 phosphorylation (P<0.05 versus WKY). Programmed stimulation triggered sustained AF in SHR (n=5/5) and SHR+V (n=4/4), but not in WKY (n=0/5) and SHR+RLX (n=1/8; P<0.01). RLX treatment reversed the transcripts for fibrosis, flattened CV restitution kinetics, reduced action potential duration at 90% recovery to baseline, increased CV (P<0.01), and reversed atrial hypertrophy (P<0.05). Independent of antifibrotic actions, RLX (0.1 µmol/L) increased Na⁺ current density, INa (≈2-fold in 48 hours) in human cardiomyocytes derived from inducible pluripotent stem cells (n=18/18; P<0.01). CONCLUSIONS: RLX treatment suppressed AF in SHR hearts by increasing CV from a combination of reversal of fibrosis and hypertrophy and by increasing INa. The study provides compelling evidence that RLX may provide a novel therapy to manage AF in humans by reversing fibrosis and hypertrophy and by modulating cardiac ionic currents.


Subject(s)
Atrial Fibrillation/drug therapy , Cardiomyopathies/drug therapy , Myocytes, Cardiac/physiology , Relaxin/physiology , Relaxin/therapeutic use , Animals , Atrial Fibrillation/physiopathology , Cardiomyopathies/physiopathology , Fibrosis/physiopathology , Fibrosis/prevention & control , Heart Conduction System/drug effects , Heart Conduction System/physiology , Humans , Hypertrophy/drug therapy , Hypertrophy/physiopathology , Male , Myocytes, Cardiac/pathology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use
20.
Nat Neurosci ; 16(2): 201-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23313911

ABSTRACT

L-type voltage gated calcium channels have an important role in neuronal development by promoting dendritic growth and arborization. A point mutation in the gene encoding Ca(V)1.2 causes Timothy syndrome, a neurodevelopmental disorder associated with autism spectrum disorders (ASDs). We report that channels with the Timothy syndrome alteration cause activity-dependent dendrite retraction in rat and mouse neurons and in induced pluripotent stem cell (iPSC)-derived neurons from individuals with Timothy syndrome. Dendrite retraction was independent of calcium permeation through the mutant channel, was associated with ectopic activation of RhoA and was inhibited by overexpression of the channel-associated GTPase Gem. These results suggest that Ca(V)1.2 can activate RhoA signaling independently of Ca(2+) and provide insights into the cellular basis of Timothy syndrome and other ASDs.


Subject(s)
Dendrites/pathology , Long QT Syndrome/pathology , Neurons/pathology , Syndactyly/pathology , Animals , Autistic Disorder , Bacterial Proteins/genetics , Calcium/metabolism , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Cerebral Cortex/cytology , Dendrites/drug effects , Dendrites/ultrastructure , Disease Models, Animal , Embryo, Mammalian , Humans , Long QT Syndrome/genetics , Luminescent Proteins/genetics , Mice , Myosin Light Chains/metabolism , Neurons/drug effects , Photic Stimulation , RNA, Small Interfering/genetics , Rats , Silver Staining , Syndactyly/genetics , Transfection , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism , Red Fluorescent Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...