Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
J Pharmacol Toxicol Methods ; 117: 107193, 2022.
Article in English | MEDLINE | ID: mdl-35792285

ABSTRACT

According to the ICH S7B guideline, drug candidates are screened for hERG block prior to first-in-human testing to predict the likelihood of delayed repolarization associated with a rare, but life-threatening, ventricular tachyarrhythmia. The new ICH E14 Q&As guideline allows hERG results to be used in later clinical development for decision-making (Q&As 5.1 and 6.1). To pursue this path, the hERG assay should be conducted following the new ICH S7B Q&A 2.1 guideline, which calls for best practice considerations of the recording temperature, voltage protocol, stimulation frequency, recording/data quality, and concentration verification. This study investigated hERG block by cisapride, dofetilide, terfenadine, sotalol, and E-4031 - positive controls commonly used to demonstrate assay sensitivity - using the manual whole cell patch clamp method and an action potential-like voltage protocol presented at 0.2 Hz. Recordings were conducted at room and near physiological temperature. Drug concentrations were measured using samples collected during real patch clamp experiments and satellite experiments. Results showed temperature effects for E-4031, terfenadine, and sotalol, but not cisapride and dofetilide. Cisapride and terfenadine showed substantial concentration losses, largely due to nonspecific binding to the perfusion apparatus. Using concentrations measured from the real and satellite experiments to assess block potencies yielded comparable results, indicating that satellite sample collection may be viable for drugs with nonspecific binding concerns only. In summary, this study provides block potencies for 5 hERG positive controls, and serves as a case study for hERG assays conducted, and results illustrated in accordance with the new ICH E14/S7B Q&As.


Subject(s)
Ether-A-Go-Go Potassium Channels , Sotalol , Cisapride , Ether-A-Go-Go Potassium Channels/metabolism , Humans , Phenethylamines , Sotalol/pharmacology , Sulfonamides , Temperature , Terfenadine/pharmacology
2.
ACS Med Chem Lett ; 11(3): 358-364, 2020 Mar 12.
Article in English | MEDLINE | ID: mdl-32184970

ABSTRACT

We describe the discovery of three structurally differentiated potent and selective MTH1 inhibitors and their subsequent use to investigate MTH1 as an oncology target, culminating in target (in)validation. Tetrahydronaphthyridine 5 was rapidly identified as a highly potent MTH1 inhibitor (IC50 = 0.043 nM). Cocrystallization of 5 with MTH1 revealed the ligand in a Φ-cis-N-(pyridin-2-yl)acetamide conformation enabling a key intramolecular hydrogen bond and polar interactions with residues Gly34 and Asp120. Modification of literature compound TH287 with O- and N-linked aryl and alkyl aryl substituents led to the discovery of potent pyrimidine-2,4,6-triamine 25 (IC50 = 0.49 nM). Triazolopyridine 32 emerged as a highly selective lead compound with a suitable in vitro profile and desirable pharmacokinetic properties in rat. Elucidation of the DNA damage response, cell viability, and intracellular concentrations of oxo-NTPs (oxidized nucleoside triphosphates) as a function of MTH1 knockdown and/or small molecule inhibition was studied. Based on our findings, we were unable to provide evidence to further pursue MTH1 as an oncology target.

3.
Am J Physiol Heart Circ Physiol ; 310(3): H426-35, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26637557

ABSTRACT

Pathological enhancement of late Na(+) current (INa) can potentially modify intracellular ion homeostasis and contribute to cardiac dysfunction. We tested the hypothesis that modulation of late INa can be a source of intracellular Na(+) ([Na(+)]i) overload. Late INa was enhanced by exposing rabbit ventricular myocytes to Anemonia sulcata toxin II (ATX-II) and measured using whole cell patch-clamp technique. [Na(+)]i was determined with fluorescent dye Asante NaTRIUM Green-2 AM. Pacing-induced changes in the dye fluorescence measured at 37°C were more pronounced in ATX-II-treated cells than in control (dye washout prevented calibration). At 22-24°C, resting [Na(+)]i was 6.6 ± 0.8 mM. Treatment with 5 nM ATX-II increased late INa 8.7-fold. [Na(+)]i measured after 2 min of electrical stimulation (1 Hz) was 10.8 ± 1.5 mM and 22.1 ± 1.6 mM (P < 0.001) in the absence and presence of 5 nM ATX-II, respectively. Inhibition of late INa with GS-967 (1 µM) prevented Na(+) i accumulation. A strong positive correlation was observed between the late INa and the pacing-induced increase of [Na(+)]i (R(2) = 0.88) and between the rise in [Na(+)]i and the increases in cytosolic Ca(2+) (R(2) = 0.96). ATX-II, tetrodotoxin, or GS-967 did not affect [Na(+)]i in quiescent myocytes suggesting that late INa was solely responsible for triggering the ATX-II effect on [Na(+)]i. Experiments with pinacidil and E4031 indicate that prolongation of the action potential contributes to as much as 50% of the [Na(+)]i overload associated with the increase in late INa caused by ATX-II. Enhancement of late INa can cause intracellular Na(+) overload in ventricular myocytes.


Subject(s)
Calcium/metabolism , Cardiotonic Agents/pharmacology , Cnidarian Venoms/pharmacology , Myocytes, Cardiac/drug effects , Sodium Channels/drug effects , Sodium/metabolism , Animals , Green Fluorescent Proteins , Heart Ventricles/cytology , Indoles , Myocytes, Cardiac/metabolism , Optical Imaging , Patch-Clamp Techniques , Rabbits , Sodium Channels/metabolism
4.
J Mol Cell Cardiol ; 76: 247-56, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25252177

ABSTRACT

An increase of late Na(+) current (INaL) in cardiac myocytes can raise the cytosolic Na(+) concentration and is associated with activation of Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) and alterations of mitochondrial metabolism and Ca(2+) handling by sarcoplasmic reticulum (SR). We tested the hypothesis that augmentation of INaL can increase mitochondrial reactive oxygen species (ROS) production and oxidation of CaMKII, resulting in spontaneous SR Ca(2+) release and increased diastolic Ca(2+) in myocytes. Increases of INaL and/or of the cytosolic Na(+) concentration led to mitochondrial ROS production and oxidation of CaMKII to cause dysregulation of Ca(2+) handling in rabbit cardiac myocytes.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Myocytes, Cardiac/enzymology , Sodium/metabolism , Action Potentials , Animals , Calcium Signaling , Female , Heart Ventricles/cytology , Heart Ventricles/enzymology , Intracellular Space/metabolism , Oxidation-Reduction , Oxidative Stress , Rabbits , Reactive Oxygen Species/metabolism
5.
Am J Physiol Heart Circ Physiol ; 302(1): H253-61, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21984545

ABSTRACT

The role of calsequestrin (CASQ2) in cardiac sarcoplasmic reticulum (SR) calcium (Ca(2+)) transport has gained significant attention since point mutations in CASQ2 were reported to cause ventricular arrhythmia. In the present study, we have critically evaluated the functional consequences of expressing the CASQ2(D307H) mutant protein in the CASQ2 null mouse. We recently reported that the mutant CASQ2(D307H) protein can be stably expressed in CASQ2 null hearts, and it targets appropriately to the junctional SR (Kalyanasundaram A, Bal NC, Franzini-Armstrong C, Knollmann BC, Periasamy M. J Biol Chem 285: 3076-3083, 2010). In this study, we found that introduction of CASQ2(D307H) protein in the CASQ2 null background partially restored triadin 1 levels, which were decreased in the CASQ2 null mice. Despite twofold expression (relative to wild-type CASQ2), the mutant protein failed to increase SR Ca(2+) load. We also found that the Ca(2+) transient decays slower in the CASQ2 null and CASQ2(D307H) cells. CASQ2(D307H) myocytes, when rhythmically paced and challenged with isoproterenol, exhibit spontaneous Ca(2+) waves similar to CASQ2 null myocytes; however, the stability of Ca(2+) cycling was increased in the CASQ2(D307H) myocytes. In the presence of isoproterenol, Ca(2+)-transient amplitude in CASQ2(D307H) myocytes was significantly decreased, possibly indicating an inherent defect in Ca(2+) buffering capacity and release from the mutant CASQ2 at high Ca(2+) concentrations. We also observed polymorphic ventricular tachycardia in the CASQ2(D307H) mice, although lesser than in the CASQ2 null mice. These data suggest that CASQ2(D307H) point mutation may affect Ca(2+) buffering capacity and Ca(2+) release. We propose that poor interaction between CASQ2(D307H) and triadin 1 could affect ryanodine receptor 2 stability, thereby increasing susceptibility to delayed afterdepolarizations and triggered arrhythmic activity.


Subject(s)
Calcium Signaling , Calcium/metabolism , Calsequestrin/metabolism , Myocytes, Cardiac/metabolism , Point Mutation , Sarcoplasmic Reticulum/metabolism , Animals , Calcium Signaling/drug effects , Calsequestrin/genetics , Cardiac Pacing, Artificial , Cardiotonic Agents/pharmacology , Carrier Proteins/metabolism , Disease Models, Animal , Electrocardiography , Genotype , Intracellular Signaling Peptides and Proteins , Isoproterenol/pharmacology , Mice , Mice, Knockout , Mice, Transgenic , Muscle Proteins/metabolism , Myocytes, Cardiac/drug effects , Phenotype , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/drug effects , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/metabolism , Time Factors
6.
PLoS One ; 7(12): e52005, 2012.
Article in English | MEDLINE | ID: mdl-23300588

ABSTRACT

Nitric oxide (NO) and superoxide (O(2) (-)) are important cardiac signaling molecules that regulate myocyte contraction. For appropriate regulation, NO and O(2) (.-) must exist at defined levels. Unfortunately, the NO and O(2) (.-) levels are altered in many cardiomyopathies (heart failure, ischemia, hypertrophy, etc.) leading to contractile dysfunction and adverse remodeling. Hence, rescuing the nitroso-redox levels is a potential therapeutic strategy. Nitrone spin traps have been shown to scavenge O(2) (.-) while releasing NO as a reaction byproduct; and we synthesized a novel, cell permeable nitrone, 2-2-3,4-dihydro-2H-pyrrole 1-oxide (EMEPO). We hypothesized that EMEPO would improve contractile function in myocytes with altered nitroso-redox levels. Ventricular myocytes were isolated from wildtype (C57Bl/6) and NOS1 knockout (NOS1(-/-)) mice, a known model of NO/O(2) (.-) imbalance, and incubated with EMEPO. EMEPO significantly reduced O(2) (.-) (lucigenin-enhanced chemiluminescence) and elevated NO (DAF-FM diacetate) levels in NOS1(-/-) myocytes. Furthermore, EMEPO increased NOS1(-/-) myocyte basal contraction (Ca(2+) transients, Fluo-4AM; shortening, video-edge detection), the force-frequency response and the contractile response to ß-adrenergic stimulation. EMEPO had no effect in wildtype myocytes. EMEPO also increased ryanodine receptor activity (sarcoplasmic reticulum Ca(2+) leak/load relationship) and phospholamban Serine16 phosphorylation (Western blot). We also repeated our functional experiments in a canine post-myocardial infarction model and observed similar results to those seen in NOS1(-/-) myocytes. In conclusion, EMEPO improved contractile function in myocytes experiencing an imbalance of their nitroso-redox levels. The concurrent restoration of NO and O(2) (.-) levels may have therapeutic potential in the treatment of various cardiomyopathies.


Subject(s)
Calcium/metabolism , Myocardial Contraction/drug effects , Myocytes, Cardiac/metabolism , Nitric Oxide Synthase Type I/physiology , Nitric Oxide/metabolism , Nitrogen Oxides/pharmacology , Sarcoplasmic Reticulum/metabolism , Animals , Esterification , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Oxidation-Reduction , Sarcoplasmic Reticulum/drug effects , Spin Labels , Superoxides/metabolism
7.
Am J Physiol Cell Physiol ; 301(3): C577-86, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21677263

ABSTRACT

Late Na(+) current (I(NaL)) and Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) are both increased in the diseased heart. Recently, CaMKII was found to phosphorylate the Na(+) channel 1.5 (Na(v)1.5), resulting in enhanced I(NaL). Conversely, an increase of I(NaL) would be expected to cause elevation of intracellular Ca(2+) and activation of CaMKII. However, a relationship between enhancement of I(NaL) and activation of CaMKII has yet to be demonstrated. We investigated whether Na(+) influx via Na(v)1.5 leads to CaMKII activation and explored the functional significance of this pathway. In neonatal rat ventricular myocytes (NRVM), treatment with the I(NaL) activators anemone toxin II (ATX-II) or veratridine increased CaMKII autophosphorylation and increased phosphorylation of CaMKII substrates phospholamban and ryanodine receptor 2. Knockdown of Na(v)1.5 (but not Na(v)1.1 or Na(v)1.2) prevented ATX-II-induced CaMKII phosphorylation, providing evidence for a specific role of Na(v)1.5 in CaMKII activation. In support of this view, CaMKII activity was also increased in hearts of transgenic mice overexpressing a gain-of-function Na(v)1.5 mutant (N(1325)S). The effects of both ATX-II and the N(1325)S mutation were reversed by either I(NaL) inhibition (with ranolazine or tetrodotoxin) or CaMKII inhibition (with KN93 or autocamtide 2-related inhibitory peptide). Furthermore, ATX-II treatment also induced CaMKII-Na(v)1.5 coimmunoprecipitation. The same association between CaMKII and Na(v)1.5 was also found in N(1325)S mice, suggesting a direct protein-protein interaction. Pharmacological inhibitions of either CaMKII or I(NaL) also prevented ATX-II-induced cell death in NRVM and reduced the incidence of polymorphic ventricular tachycardia induced by ATX-II in rat perfused hearts. Taken together, these results suggest that a Na(v)1.5-dependent increase in Na(+) influx leads to activation of CaMKII, which in turn phosphorylates Na(v)1.5, further promoting Na(+) influx. Pharmacological inhibition of either CaMKII or Na(v)1.5 can ameliorate cardiac dysfunction caused by excessive Na(+) influx.


Subject(s)
Amino Acid Substitution/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Heart Ventricles/metabolism , Myocytes, Cardiac/metabolism , Sodium Channels/metabolism , Sodium/metabolism , Acetanilides/pharmacology , Acetanilides/therapeutic use , Animals , Animals, Newborn , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Calcium-Binding Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Caspase 3/metabolism , Cell Death/drug effects , Cell Survival/drug effects , Cnidarian Venoms/pharmacology , Dose-Response Relationship, Drug , Electrophysiological Phenomena/drug effects , Electrophysiological Phenomena/physiology , Female , Gene Expression/drug effects , Heart Ventricles/cytology , Heart Ventricles/drug effects , Humans , Mice , Mice, Inbred Strains , Mice, Transgenic , Myocytes, Cardiac/drug effects , NAV1.5 Voltage-Gated Sodium Channel , Peptides/pharmacology , Peptides/therapeutic use , Perfusion , Phosphorylation/drug effects , Piperazines/pharmacology , Piperazines/therapeutic use , Protein Binding/drug effects , Protein Binding/physiology , RNA, Small Interfering/genetics , Rabbits , Ranolazine , Rats , Rats, Sprague-Dawley , Ryanodine Receptor Calcium Release Channel/metabolism , Sodium Channels/genetics , Sodium-Calcium Exchanger/antagonists & inhibitors , Sodium-Calcium Exchanger/metabolism , Tachycardia, Ventricular/chemically induced , Tachycardia, Ventricular/prevention & control , Tetrodotoxin/pharmacology , Veratridine/pharmacology
8.
Neoplasia ; 13(5): 428-38, 2011 May.
Article in English | MEDLINE | ID: mdl-21532883

ABSTRACT

The calcium-sensing receptor (CaR) is responsible for the regulation of extracellular calcium (Ca(2+) (o)) homeostasis. CaR activation has been shown to increase proliferation in several cancer cell lines; however, its presence or function has never been documented in lung cancer. We report that Ca(2+) (o)-activated CaR results in MAPK-mediated stimulation of parathyroid hormone-related protein (PTHrP) production in human lung squamous cell carcinoma (SCC) lines and humoral hypercalcemia of malignancy (HHM) in vivo. Furthermore, a single nucleotide polymorphism in CaR identified from a hypercalcemia-inducing lung SCC reduced the receptor's activation threshold leading to increased PTHrP expression and secretion. Increasing the expression of either wild-type CaR or a CaR variant with a single nucleotide polymorphism in the cytoplasmic domain was both necessary and sufficient for lung SCC to induce HHM. Because lung cancer patients who frequently develop HHM and PTHrP expression in lung cancer has been only partially explained, the significance of our findings indicates that CaR variants may provide a positive feedback between PTHrP and calcium and result in the syndrome of HHM.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Hypercalcemia/metabolism , Hypercalcemia/physiopathology , Lung Neoplasms/metabolism , Parathyroid Hormone-Related Protein , Receptors, Calcium-Sensing/metabolism , Animals , Calcium/metabolism , Cell Line , Cell Proliferation , Gene Expression , Gene Knockdown Techniques , Humans , Mice , Mice, Nude , Mitogen-Activated Protein Kinases , Parathyroid Hormone-Related Protein/biosynthesis , Parathyroid Hormone-Related Protein/genetics , Parathyroid Hormone-Related Protein/metabolism , Polymorphism, Single Nucleotide , Receptors, Calcium-Sensing/genetics , Transplantation, Heterologous
9.
J Physiol ; 588(Pt 15): 2905-17, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20530114

ABSTRACT

The sarcoplasmic reticulum (SR) Ca(2+) release channel (ryanodine receptor, RyR2) has been proposed to be an end target of neuronal nitric oxide synthase (NOS1) signalling. The purpose of this study is to investigate the mechanism of NOS1 modulation of RyR2 activity and the corresponding effect on myocyte function. Myocytes were isolated from NOS1 knockout (NOS1(/)) and wild-type mice. NOS1(/) myocytes displayed a decreased fractional SR Ca(2+) release, NOS1 knockout also led to reduced RyR2 S-nitrosylation levels. RyR2 channels from NOS1(/) hearts had decreased RyR2 open probability. Additionally, knockout of NOS1 led to a decrease in [(3)H]ryanodine binding, Ca(2+) spark frequency (CaSpF) and a rightward shift in the SR Ca(2+) leak/load relationship. Similar effects were observed with acute inhibition of NOS1. These data are indicative of decreased RyR2 activity in myocytes with NOS1 knockout or acute inhibition. Interestingly, the NO donor and nitrosylating agent SNAP reversed the depressed RyR2 open probability, the reduced CaSpF, and caused a leftward shift in the leak/load relationship in NOS1(/) myocytes. SNAP also normalized Ca(2+) transient and cell shortening amplitudes and SR fractional release in myocytes with NOS1 knockout or acute inhibition. Furthermore, SNAP was able to normalize the RyR2 S-nitrosylation levels. These data suggest that NOS1 signalling increases RyR2 activity via S-nitrosylation, which contributes to the NOS1-induced positive inotropic effect. Thus, RyR2 is an important end target of NOS1.


Subject(s)
Muscle Contraction/physiology , Myocytes, Cardiac/physiology , Nitric Oxide Synthase Type I/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Cells, Cultured , Feedback, Physiological/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout
10.
Cardiovasc Res ; 84(3): 387-95, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19617226

ABSTRACT

AIMS: Although cardiac alternans is a known predictor of lethal arrhythmias, its underlying causes remain largely undefined in disease settings. The potential role of, and mechanisms responsible for, beat-to-beat alternations in the amplitude of systolic Ca(2+) transients (Ca(2+) alternans) was investigated in a canine post-myocardial infarction (MI) model of sudden cardiac death (SCD). METHODS AND RESULTS: Post-MI dogs had preserved left ventricular (LV) function and susceptibility to ventricular fibrillation (VF) during exercise. LV wedge preparations from VF dogs were more susceptible to action potential (AP) alternans and the frequency-dependence of Ca(2+) alternans was shifted towards slower rates in myocytes isolated from VF dogs relative to controls. In both groups of cells, cytosolic Ca(2+) transients ([Ca(2+)](c)) alternated in phase with changes in diastolic Ca(2+) in sarcoplasmic reticulum ([Ca(2+)](SR)), but the dependence of [Ca(2+)](c) amplitude on [Ca(2+)](SR) was steeper in VF cells. Abnormal ryanodine receptor (RyR) function in VF cells was indicated by increased fractional Ca(2+) release for a given amplitude of Ca(2+) current and elevated diastolic RyR-mediated SR Ca(2+) leak. SR Ca(2+) uptake activity did not differ between VF and control cells. VF myocytes had an increased rate of reactive oxygen species production and increased RyR oxidation. Treatment of VF myocytes with reducing agents normalized parameters of Ca(2+) handling and shifted the threshold of Ca(2+) alternans to higher frequencies. CONCLUSION: Redox modulation of RyRs promotes generation of Ca(2+) alternans by enhancing the steepness of the Ca(2+) release-load relationship and thereby providing a substrate for post-MI arrhythmias.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Death, Sudden, Cardiac/etiology , Disease Models, Animal , Myocardial Infarction/physiopathology , Ryanodine Receptor Calcium Release Channel/physiology , Action Potentials/physiology , Animals , Death, Sudden, Cardiac/epidemiology , Dogs , Female , Male , Myocardial Infarction/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidation-Reduction , Patch-Clamp Techniques , Risk Factors , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sodium-Calcium Exchanger/metabolism , Tachycardia, Ventricular/epidemiology , Tachycardia, Ventricular/pathology , Tachycardia, Ventricular/physiopathology , Ventricular Fibrillation/epidemiology , Ventricular Fibrillation/pathology , Ventricular Fibrillation/physiopathology
11.
Biophys J ; 95(4): 2037-48, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18469084

ABSTRACT

Cardiac calsequestrin (CASQ2) is an intrasarcoplasmic reticulum (SR) low-affinity Ca-binding protein, with mutations that are associated with catecholamine-induced polymorphic ventricular tachycardia (CPVT). To better understand how CASQ2 mutants cause CPVT, we expressed two CPVT-linked CASQ2 mutants, a truncated protein (at G112+5X, CASQ2(DEL)) or CASQ2 containing a point mutation (CASQ2(R33Q)), in canine ventricular myocytes and assessed their effects on Ca handling. We also measured CASQ2-CASQ2 variant interactions using fluorescence resonance transfer in a heterologous expression system, and evaluated CASQ2 interaction with triadin. We found that expression of CASQ2(DEL) or CASQ2(R33Q) altered myocyte Ca signaling through two different mechanisms. Overexpressing CASQ2(DEL) disrupted the CASQ2 polymerization required for high capacity Ca binding, whereas CASQ2(R33Q) compromised the ability of CASQ2 to control ryanodine receptor (RyR2) channel activity. Despite profound differences in SR Ca buffering strengths, local Ca release terminated at the same free luminal [Ca] in control cells, cells overexpressing wild-type CASQ2 and CASQ2(DEL)-expressing myocytes, suggesting that a decline in [Ca](SR) is a signal for RyR2 closure. Importantly, disrupting interactions between the RyR2 channel and CASQ2 by expressing CASQ2(R33Q) markedly lowered the [Ca](SR) threshold for Ca release termination. We conclude that CASQ2 in the SR determines the magnitude and duration of Ca release from each SR terminal by providing both a local source of releasable Ca and by effects on luminal Ca-dependent RyR2 gating. Furthermore, two CPVT-inducing CASQ2 mutations, which cause mechanistically different defects in CASQ2 and RyR2 function, lead to increased diastolic SR Ca release events and exhibit a similar CPVT disease phenotype.


Subject(s)
Calcium Signaling , Calcium/metabolism , Calsequestrin/metabolism , Death, Sudden, Cardiac , Myocytes, Cardiac/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Calsequestrin/genetics , Cells, Cultured , Dogs , Humans
12.
Blood ; 111(6): 3190-9, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18192508

ABSTRACT

Effective administration of flavopiridol in advanced-stage chronic lymphocytic leukemia (CLL) is often associated with early biochemical evidence of tumor cell lysis. Previous work using other cell types showed that flavopiridol impacts mitochondria, and in CLL cells flavopiridol down-regulates the mitochondrial protein Mcl-1. We therefore investigated mitochondrial structure and function in flavopiridol-treated CLL patient cells and in the lymphoblastic cell line 697 using concentrations and times at which tumor lysis is observed in treated patients. Mitochondrial membrane depolarization was detected in flavopiridol-treated CLL cells by 6 hours, well before the onset of cell death. Flavopiridol-induced mitochondrial depolarization was not blocked by caspase inhibitors or by the calcium chelator EGTA, but was reduced by Bcl-2 overexpression. Intracellular calcium mobilization was noted at early time points using fluorescence microscopy. Furthermore, electron paramagnetic resonance oximetry showed a gradual but significant reduction in cellular oxygen consumption rate by 6 hours, corresponding with ultrastructural mitochondrial damage detected by electron microscopy. These observations suggest that in CLL and 697 cells, flavopiridol mediates its cytotoxic effects via induction of the mitochondrial permeability transition and changes in intracellular calcium.


Subject(s)
Calcium/metabolism , Flavonoids/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Mitochondria/drug effects , Mitochondria/pathology , Oxygen/metabolism , Piperidines/pharmacology , Apoptosis/drug effects , Biological Transport , Caspase Inhibitors , Caspases/metabolism , Cell Membrane Permeability/drug effects , Cell Shape/drug effects , Flow Cytometry , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Microscopy, Electron, Transmission , Mitochondria/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Time Factors
13.
Life Sci ; 81(14): 1152-9, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17884106

ABSTRACT

While cardiac resynchronization therapy (CRT) has been shown to reduce morbidity and mortality in heart failure (HF) patients, the fundamental mechanisms for the efficacy of CRT are poorly understood. The lack of understanding of these basic mechanisms represents a significant barrier to our understanding of the pathogenesis of HF and potential recovery mechanisms. Our purpose was to determine cellular mechanisms for the observed improvement in chronic HF after CRT. We used a canine model of chronic nonischemic cardiomyopathy. After 15 months, dogs were randomized to continued RV tachypacing (untreated HF) or CRT for an additional 9 months. Six minute walk tests, echocardiograms, and electrocardiograms were done to assess the functional response to therapy. Left ventricular (LV) midmyocardial myocytes were isolated to study electrophysiology and intracellular calcium regulation. Compared to untreated HF, CRT improved HF-induced increases in LV volumes, diameters and mass (p<0.05). CRT reversed HF-induced prolongations in LV myocyte repolarization (p<0.05) and normalized HF-induced depolarization (p<0.03) of the resting membrane potential. CRT improved HF-induced reductions in calcium (p<0.05). CRT did not attenuate the HF-induced increases in LV interstitial fibrosis. Using a translational approach in a chronic HF model, CRT significantly improved LV structure; this was accompanied by improved LV myocyte electrophysiology and calcium regulation. The beneficial effects of CRT may be attributable, in part, to improved LV myocyte function.


Subject(s)
Cardiac Pacing, Artificial , Cardiomyopathies/physiopathology , Cardiomyopathies/therapy , Ventricular Remodeling , Animals , Calcium/metabolism , Chronic Disease , Defibrillators, Implantable , Disease Models, Animal , Dogs , Echocardiography , Electrocardiography , Electrophysiology , Heart Failure/physiopathology , Heart Failure/therapy , Heart Ventricles/metabolism , Heart Ventricles/pathology , In Vitro Techniques , Myocytes, Cardiac/physiology , Pacemaker, Artificial
14.
Am J Physiol Regul Integr Comp Physiol ; 293(5): R1787-97, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17761517

ABSTRACT

Isolated diastolic dysfunction is found in almost half of asymptomatic patients with well-controlled diabetes and may precede diastolic heart failure. However, mechanisms that underlie diastolic dysfunction during diabetes are not well understood. We tested the hypothesis that isolated diastolic dysfunction is associated with impaired myocardial Ca(2+) handling during type 1 diabetes. Streptozotocin-induced diabetic rats were compared with age-matched placebo-treated rats. Global left ventricular myocardial performance and systolic function were preserved in diabetic animals. Diabetes-induced diastolic dysfunction was evident on Doppler flow imaging, based on the altered patterns of mitral inflow and pulmonary venous flows. In isolated ventricular myocytes, diabetes resulted in significant prolongation of action potential duration compared with controls, with afterdepolarizations occurring in diabetic myocytes (P < 0.05). Sustained outward K(+) current and peak outward component of the inward rectifier were reduced in diabetic myocytes, while transient outward current was increased. There was no significant change in L-type Ca(2+) current; however, Ca(2+) transient amplitude was reduced and transient decay was prolonged by 38% in diabetic compared with control myocytes (P < 0.05). Sarcoplasmic reticulum Ca(2+) load (estimated by measuring the integral of caffeine-evoked Na(+)-Ca(2+) exchanger current and Ca(2+) transient amplitudes) was reduced by approximately 50% in diabetic myocytes (P < 0.05). In permeabilized myocytes, Ca(2+) spark amplitude and frequency were reduced by 34 and 20%, respectively, in diabetic compared with control myocytes (P < 0.05). Sarco(endo)plasmic reticulum Ca(2+)-ATPase-2a protein levels were decreased during diabetes. These data suggest that in vitro impairment of Ca(2+) reuptake during myocyte relaxation contributes to in vivo diastolic dysfunction, with preserved global systolic function, during diabetes.


Subject(s)
Calcium/metabolism , Diabetic Angiopathies/metabolism , Muscle Cells/metabolism , Action Potentials/physiology , Animals , Blotting, Western , Calcium/physiology , Calcium Channels, L-Type/metabolism , Calcium Signaling/drug effects , Diabetes Mellitus, Experimental/metabolism , Diabetic Angiopathies/physiopathology , Diastole/physiology , Electrocardiography , Heart Ventricles/cytology , Heart Ventricles/metabolism , Male , Potassium Channels/drug effects , Potassium Channels/metabolism , Rats , Rats, Wistar , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
15.
J Physiol ; 583(Pt 1): 71-80, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17569730

ABSTRACT

In cardiac muscle, intracellular Ca2+ release is controlled by a number of proteins including the ryanodine receptor (RyR2), calsequestrin (CASQ2), triadin-1 (Trd) and junctin (Jn) which form a complex in the junctional sarcoplasmic reticulum (SR) membrane. Within this complex, Trd appears to link CASQ2 to RyR2 although the functional significance of this interaction is unclear. In this study, we explored the functional importance of Trd-CASQ2 interactions for intracellular Ca2+ handling in rat ventricular myocytes. A peptide encompassing the homologous CASQ2 binding domain of Trd (residues 206-230 in the rat; TrdPt) was expressed in the lumen of the SR to disrupt Trd-CASQ2 interactions. Myocytes expressing TrdPt exhibited increased responsiveness of SR Ca2+ release to activation by ICa as manifested by flattened and broadened voltage dependency of the amplitude of cytosolic Ca2+ transients. Rhythmically paced, TrdPt-expressing myocytes exhibited spontaneous arrhythmogenic oscillations of intracellular Ca2+ and membrane potential that was not seen in control cells. In addition, the frequency of spontaneous Ca2+ sparks and Ca2+ waves was significantly increased in TrdPt-expressing, permeabilized myocytes. These alterations in SR Ca2+ release were accompanied by a significant decrease in basal free intra-SR[Ca2+] and total SR Ca2+ content in TrdPt-expressing cells. At the same time a synthetic peptide corresponding to the CASQ2 binding domain of Trd produced no direct effects on the activity of single RyR2 channels incorporated into lipid bilayers while interfering with the ability of CASQ2 to inhibit the RyR2 channel. These results suggest that CASQ2 stabilizes SR Ca2+ release by inhibiting the RyR2 channel through interaction with Trd. They also show that intracellular Ca2+ cycling in the heart relies on coordinated interactions between proteins of the RyR2 channel complex and that disruption of these interactions may represent a molecular mechanism for cardiac disease.


Subject(s)
Calcium/metabolism , Calsequestrin/metabolism , Carrier Proteins/metabolism , Muscle Proteins/metabolism , Myocytes, Cardiac/metabolism , Sarcoplasmic Reticulum/metabolism , Action Potentials/physiology , Animals , Electrophysiology , Intracellular Signaling Peptides and Proteins , Male , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Ryanodine Receptor Calcium Release Channel/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/physiology
16.
Cardiovasc Res ; 75(1): 69-78, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17449018

ABSTRACT

OBJECTIVE: A naturally-occurring mutation in cardiac calsequestrin (CASQ2) at amino acid 307 was discovered in a highly inbred family and hypothesized to cause Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT). The goal of this study was to establish a causal link between CASQ2(D307H) and the CPVT phenotype using an in vivo model. METHODS AND RESULTS: Cardiac-specific expression of the CASQ2(D307H) transgene was achieved using the alpha-MHC promoter. Multiple transgenic (TG) mouse lines expressing CASQ2(D307H) from 2- to 6-fold possess structurally normal hearts without any sign of hypertrophy. The hearts displayed normal ventricular function. Myocytes isolated from TG mice had diminished I(Ca)-induced Ca2+ transient amplitude and duration, as well as increased Ca2+ spark frequency. These myocytes, when exposed to isoproterenol and caffeine, displayed disturbances in their rhythmic Ca2+ oscillations and membrane potential, and delayed afterdepolarizations. ECG monitoring revealed that TG mice challenged with isoproterenol and caffeine developed complex ventricular arrhythmias, including non-sustained polymorphic ventricular tachycardia. CONCLUSIONS: The findings of the present study demonstrate that expression of mutant CASQ2(D307H) in the mouse heart results in abnormal myocyte Ca2+ handling and predisposes to complex ventricular arrhythmias similar to the CPVT phenotype observed in human patients.


Subject(s)
Calcium/metabolism , Calsequestrin/genetics , Death, Sudden, Cardiac/etiology , Mutation, Missense , Sarcoplasmic Reticulum/metabolism , Tachycardia, Ventricular/genetics , Animals , Caffeine/pharmacology , Calcium Signaling , Cardiotonic Agents/pharmacology , Electrocardiography , Isoproterenol/pharmacology , Mice , Mice, Transgenic , Microscopy, Confocal , Models, Animal , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/pathology
17.
Circulation ; 114(10): 1012-9, 2006 Sep 05.
Article in English | MEDLINE | ID: mdl-16908766

ABSTRACT

BACKGROUND: Four distinct mutations in the human cardiac calsequestrin gene (CASQ2) have been linked to catecholaminergic polymorphic ventricular tachycardia (CPVT). The mechanisms leading to the clinical phenotype are still poorly understood because only 1 CASQ2 mutation has been characterized in vitro. METHODS AND RESULTS: We identified a homozygous 16-bp deletion at position 339 to 354 leading to a frame shift and a stop codon after 5aa (CASQ2(G112+5X)) in a child with stress-induced ventricular tachycardia and cardiac arrest. The same deletion was also identified in association with a novel point mutation (CASQ2(L167H)) in a highly symptomatic CPVT child who is the first CPVT patient carrier of compound heterozygous CASQ2 mutations. We characterized in vitro the properties of CASQ2 mutants: CASQ2(G112+5X) did not bind Ca2+, whereas CASQ2(L167H) had normal calcium-binding properties. When expressed in rat myocytes, both mutants decreased the sarcoplasmic reticulum Ca2+-storing capacity and reduced the amplitude of I(Ca)-induced Ca2+ transients and of spontaneous Ca2+ sparks in permeabilized myocytes. Exposure of myocytes to isoproterenol caused the development of delayed afterdepolarizations in CASQ2(G112+5X). CONCLUSIONS: CASQ2(L167H) and CASQ2(G112+5X) alter CASQ2 function in cardiac myocytes, which leads to reduction of active sarcoplasmic reticulum Ca2+ release and calcium content. In addition, CASQ2(G112+5X) displays altered calcium-binding properties and leads to delayed afterdepolarizations. We conclude that the 2 CASQ2 mutations identified in CPVT create distinct abnormalities that lead to abnormal intracellular calcium regulation, thus facilitating the development of tachyarrhythmias.


Subject(s)
Calsequestrin/genetics , Syncope/genetics , Tachycardia, Ventricular/genetics , Amino Acid Substitution , Animals , Child , Female , Gene Transfer Techniques , Genetic Carrier Screening , Humans , Male , Muscle Cells/physiology , Mutagenesis, Site-Directed , Mutation , Pedigree , Point Mutation , Rats , Tachycardia, Ventricular/physiopathology , Transfection
18.
Circ Res ; 98(9): 1151-8, 2006 May 12.
Article in English | MEDLINE | ID: mdl-16601229

ABSTRACT

Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a familial arrhythmogenic disorder associated with mutations in the cardiac ryanodine receptor (RyR2) and cardiac calsequestrin (CASQ2) genes. Previous in vitro studies suggested that RyR2 and CASQ2 interact as parts of a multimolecular Ca(2+)-signaling complex; however, direct evidence for such interactions and their potential significance to myocardial function remain to be determined. We identified a novel CASQ2 mutation in a young female with a structurally normal heart and unexplained syncopal episodes. This mutation results in the nonconservative substitution of glutamine for arginine at amino acid 33 of CASQ2 (R33Q). Adenoviral-mediated expression of CASQ2(R33Q) in adult rat myocytes led to an increase in excitation-contraction coupling gain and to more frequent occurrences of spontaneous propagating (Ca2+ waves) and local Ca2+ signals (sparks) with respect to control cells expressing wild-type CASQ2 (CASQ2WT). As revealed by a Ca2+ indicator entrapped inside the sarcoplasmic reticulum (SR) of permeabilized myocytes, the increased occurrence of spontaneous Ca2+ sparks and waves was associated with a dramatic decrease in intra-SR [Ca2+]. Recombinant CASQ2WT and CASQ2R33Q exhibited similar Ca(2+)-binding capacities in vitro; however, the mutant protein lacked the ability of its WT counterpart to inhibit RyR2 activity at low luminal [Ca2+] in planar lipid bilayers. We conclude that the R33Q mutation disrupts interactions of CASQ2 with the RyR2 channel complex and impairs regulation of RyR2 by luminal Ca2+. These results show that intracellular Ca2+ cycling in normal heart relies on an intricate interplay of CASQ2 with the proteins of the RyR2 channel complex and that disruption of these interactions can lead to cardiac arrhythmia.


Subject(s)
Calsequestrin/metabolism , Death, Sudden, Cardiac/etiology , Exercise , Ryanodine Receptor Calcium Release Channel/metabolism , Tachycardia, Ventricular/genetics , Amino Acid Substitution , Animals , Arginine , Binding, Competitive , Calcium/metabolism , Calsequestrin/genetics , Cardiac Pacing, Artificial/methods , Catecholamines/metabolism , Female , Glutamine , Humans , Intracellular Membranes/metabolism , Mutation , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Rats , Recombinant Proteins/metabolism , Sarcoplasmic Reticulum/metabolism , Syncope/genetics , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/physiopathology
19.
Proc Natl Acad Sci U S A ; 102(39): 14104-9, 2005 Sep 27.
Article in English | MEDLINE | ID: mdl-16172392

ABSTRACT

Diminished Ca release from the sarcoplasmic reticulum (SR) is an important contributor to the impaired contractility of the failing heart. Despite extensive effort, the underlying causes of abnormal SR Ca release in heart failure (HF) remain unknown. We used a combination of simultaneous imaging of cytosolic and SR intraluminal [Ca] in isolated cardiomyocytes and recordings from single-ryanodine receptor (RyR) channels reconstituted into lipid bilayers to investigate alterations in intracellular Ca handling in an experimental model of chronic HF. We found that diastolic free [Ca] inside the SR was dramatically reduced because of a Ca leak across the SR membrane, mediated by spontaneous local release events (Ca sparks), in HF myocytes. Additionally, the magnitudes of intrastore Ca depletion signals during global and focal Ca release events were blunted, and [Ca]SR recovery was slowed after global but not focal Ca release in HF myocytes. At the single-RyR level, the sensitivity of RyRs to activation by luminal Ca was greatly enhanced, providing a molecular mechanism for the maintained potentiation of Ca sparks (and increased Ca leak) at reduced intra-SR [Ca] in HF. This work shows that the diminished SR Ca release characteristic of failing myocardium could be explained by increased sensitivity of RyRs to luminal Ca, leading to enhanced spark-mediated SR Ca leak and reduced intra-SR [Ca].


Subject(s)
Calcium Signaling , Calcium/metabolism , Cardiac Output, Low/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Calcium/analysis , Cardiac Output, Low/pathology , Cardiac Output, Low/physiopathology , Chronic Disease , Dogs , Heart/physiopathology , Muscle Proteins/analysis , Muscle Proteins/metabolism , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Sarcoplasmic Reticulum/chemistry
20.
J Physiol ; 567(Pt 2): 493-504, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-15975978

ABSTRACT

We used confocal Ca2+ imaging and the patch-clamp technique to investigate the interplay between Ca2+ entries through L-type Ca2+ channels (LCCs) and reverse-mode Na+-Ca2+ exchange (NCX) in activating Ca2+-induced Ca2+ release (CICR) from the sarcoplasmic reticulum (SR) in cardiac myocytes from normal and failing rat hearts. In normal myocytes exposed to N(6),2'-O-dibutyryl adenosine-3',5'-cyclic monophosphate (db-cAMP, membrane-permeable form of cAMP), the bell-shaped voltage dependence of cytosolic Ca2+ transients was dramatically broadened due to activation of SR Ca2+ release at high membrane potentials (30-120 mV). This broadening of Ca2+-transient voltage dependence could be prevented by KB-R7943, an inhibitor of the reverse-mode NCX. Trans-sarcolemmal Ca2+ entries were measured fluorometrically in myocytes during depolarizing steps to high membrane potentials. The total Ca2+ entry (deltaF(Tot)) was separated into two Ca2+ entry components, LCC-mediated (deltaF(LCC)) and NCX-mediated (deltaF(NCX)), by exposing the cells to the specific inhibitors of LCCs and reverse-mode NCX, nifedipine and KB-R7943, respectively. In the absence of protein kinase A (PKA) stimulation the amplitude of the Ca2+-inflow signal (deltaF(Tot)) corresponded to the arithmetic sum of the amplitudes of the KB-R7943- and nifedipine-resistant components (deltaF(Tot)=deltaF(LCC)+deltaF(NCX)). PKA activation resulted in significant increases in deltaF(Tot) and deltaF(LCC). Paradoxically, deltaF(Tot) became approximately threefold larger than the sum of the deltaF(NCX) and deltaF(LCC) components. In myocytes from failing hearts, stimulation of PKA failed to induce a shift in Ca2+ release voltage dependence toward more positive membrane potentials. Although the total and NCX-mediated Ca2+ entries were increased again, deltaF(Tot) did not significantly exceed the sum of deltaF(LCC) and deltaF(NCX). We conclude that the LCC and NCX Ca2+-entry pathways interact synergistically to trigger SR Ca2+ release on depolarization to positive membrane potentials in PKA-stimulated cardiac muscle. In heart failure, this new form of Ca2+ release is diminished and may potentially account for the compromised contractile performance and reduced functional reserve in failing hearts.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Signaling , Calcium/metabolism , Cardiomyopathies/metabolism , Myocytes, Cardiac/metabolism , Sodium-Calcium Exchanger/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...