Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Circ Res ; 130(5): 711-724, 2022 03 04.
Article in English | MEDLINE | ID: mdl-35086342

ABSTRACT

BACKGROUND: Oxidative stress in cardiac disease promotes proarrhythmic disturbances in Ca2+ homeostasis, impairing luminal Ca2+ regulation of the sarcoplasmic reticulum (SR) Ca2+ release channel, the RyR2 (ryanodine receptor), and increasing channel activity. However, exact mechanisms underlying redox-mediated increase of RyR2 function in cardiac disease remain elusive. We tested whether the oxidoreductase family of proteins that dynamically regulate the oxidative environment within the SR are involved in this process. METHODS: A rat model of hypertrophy induced by thoracic aortic banding (TAB) was used for ex vivo whole heart optical mapping and for Ca2+ and reactive oxygen species imaging in isolated ventricular myocytes (VMs). RESULTS: The SR-targeted reactive oxygen species biosensor ERroGFP showed increased intra-SR oxidation in TAB VMs that was associated with increased expression of Ero1α (endoplasmic reticulum oxidoreductase 1 alpha). Pharmacological (EN460) or genetic Ero1α inhibition normalized SR redox state, increased Ca2+ transient amplitude and SR Ca2+ content, and reduced proarrhythmic spontaneous Ca2+ waves in TAB VMs under ß-adrenergic stimulation (isoproterenol). Ero1α overexpression in Sham VMs had opposite effects. Ero1α inhibition attenuated Ca2+-dependent ventricular tachyarrhythmias in TAB hearts challenged with isoproterenol. Experiments in TAB VMs and human embryonic kidney 293 cells expressing human RyR2 revealed that an Ero1α-mediated increase in SR Ca2+-channel activity involves dissociation of intraluminal protein ERp44 (endoplasmic reticulum protein 44) from the RyR2 complex. Site-directed mutagenesis and molecular dynamics simulations demonstrated a novel redox-sensitive association of ERp44 with RyR2 mediated by intraluminal cysteine 4806. ERp44-RyR2 association in TAB VMs was restored by Ero1α inhibition, but not by reducing agent dithiothreitol, as hypo-oxidation precludes formation of covalent bond between RyR2 and ERp44. CONCLUSIONS: A novel axis of intraluminal interaction between RyR2, ERp44, and Ero1α has been identified. Ero1α inhibition exhibits promising therapeutic potential by stabilizing RyR2-ERp44 complex, thereby reducing spontaneous Ca2+ release and Ca2+-dependent tachyarrhythmias in hypertrophic hearts, without causing hypo-oxidative stress in the SR.


Subject(s)
Heart Diseases , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Glycoproteins/metabolism , Ryanodine Receptor Calcium Release Channel , Animals , Arrhythmias, Cardiac/metabolism , Calcium/metabolism , Calcium Signaling , Heart Diseases/metabolism , Isoproterenol/pharmacology , Myocytes, Cardiac/metabolism , Oxidoreductases/metabolism , Oxidoreductases/pharmacology , Rats , Reactive Oxygen Species/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism
2.
Biochem Biophys Res Commun ; 661: 89-98, 2023 06 18.
Article in English | MEDLINE | ID: mdl-37087803

ABSTRACT

The ubiquity of wireless electronic-device connectivity has seen microwaves emerge as one of the fastest growing forms of electromagnetic exposure. A growing evidence-base refutes the claim that wireless technologies pose no risk to human health at current safety levels designed to limit thermal (heating) effects. The potential impact of non-thermal effects of microwave exposure, especially in electrically-excitable tissues (e.g., heart), remains controversial. We exposed human embryonic stem-cell derived cardiomyocytes (CM), under baseline and beta-adrenergic receptor (ß-AR)-stimulated conditions, to microwaves at 2.4 GHz, a frequency used extensively in wireless communication (e.g., 4G, Bluetooth™ and WiFi). To control for any effect of sample heating, experiments were done in CM subjected to matched rates of direct heating or CM maintained at 37 °C. Detailed profiling of the temporal and amplitude features of Ca2+ signalling in CM under these experimental conditions was reconciled with the extent and spatial clustering of apoptosis. The data show that exposure of CM to 2.4 GHz EMF eliminated the normal Ca2+ signalling response to ß-AR stimulation and provoked spatially-clustered apoptosis. This is first evidence that non-thermal effects of 2.4 GHz microwaves might have profound effects on human CM function, responsiveness to activation, and survival.


Subject(s)
Microwaves , Receptors, Adrenergic, beta , Humans , Myocytes, Cardiac , Signal Transduction , Electromagnetic Fields
3.
Int J Mol Sci ; 22(23)2021 Dec 02.
Article in English | MEDLINE | ID: mdl-34884838

ABSTRACT

Alterations to amino acid residues G4946 and I4790, associated with resistance to diamide insecticides, suggests a location of diamide interaction within the pVSD voltage sensor-like domain of the insect ryanodine receptor (RyR). To further delineate the interaction site(s), targeted alterations were made within the same pVSD region on the diamondback moth (Plutella xylostella) RyR channel. The editing of five amino acid positions to match those found in the diamide insensitive skeletal RyR1 of humans (hRyR1) in order to generate a human-Plutella chimeric construct showed that these alterations strongly reduce diamide efficacy when introduced in combination but cause only minor reductions when introduced individually. It is concluded that the sites of diamide interaction on insect RyRs lie proximal to the voltage sensor-like domain of the RyR and that the main site of interaction is at residues K4700, Y4701, I4790 and S4919 in the S1 to S4 transmembrane domains.


Subject(s)
Diamide/chemistry , Insect Proteins/chemistry , Ryanodine Receptor Calcium Release Channel/chemistry , Animals , Binding Sites , Caffeine/pharmacology , Calcium Signaling/drug effects , Diamide/metabolism , Diamide/pharmacology , Humans , Insect Proteins/genetics , Insect Proteins/metabolism , Insecticide Resistance/drug effects , Insecticides/chemistry , Insecticides/metabolism , Insecticides/pharmacology , Moths/metabolism , Mutagenesis, Site-Directed , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , ortho-Aminobenzoates/chemistry , ortho-Aminobenzoates/metabolism , ortho-Aminobenzoates/pharmacology
4.
J Cell Sci ; 131(15)2018 08 03.
Article in English | MEDLINE | ID: mdl-29930088

ABSTRACT

The cardiac muscle ryanodine receptor-Ca2+ release channel (RyR2) constitutes the sarcoplasmic reticulum (SR) Ca2+ efflux mechanism that initiates myocyte contraction, while cardiac myosin-binding protein-C (cMyBP-C; also known as MYBPC3) mediates regulation of acto-myosin cross-bridge cycling. In this paper, we provide the first evidence for the presence of direct interaction between these two proteins, forming a RyR2-cMyBP-C complex. The C-terminus of cMyBP-C binds with the RyR2 N-terminus in mammalian cells and the interaction is not mediated by a fibronectin-like domain. Notably, we detected complex formation between both recombinant cMyBP-C and RyR2, as well as between the native proteins in cardiac tissue. Cellular Ca2+ dynamics in HEK293 cells is altered upon co-expression of cMyBP-C and RyR2, with lowered frequency of RyR2-mediated spontaneous Ca2+ oscillations, suggesting that cMyBP-C exerts a potential inhibitory effect on RyR2-dependent Ca2+ release. Discovery of a functional RyR2 association with cMyBP-C provides direct evidence for a putative mechanistic link between cytosolic soluble cMyBP-C and SR-mediated Ca2+ release, via RyR2. Importantly, this interaction may have clinical relevance to the observed cMyBP-C and RyR2 dysfunction in cardiac pathologies, such as hypertrophic cardiomyopathy.


Subject(s)
Carrier Proteins/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Calcium/metabolism , Calcium Signaling/physiology , Cytosol/metabolism , HEK293 Cells , Humans , Protein Binding , Sarcoplasmic Reticulum/metabolism
5.
Circ Res ; 116(8): 1324-35, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25648700

ABSTRACT

RATIONALE: Flecainide, a class 1c antiarrhythmic, has emerged as an effective therapy in preventing arrhythmias in patients with catecholaminergic polymorphic ventricular tachycardia (CPVT) refractory to ß-adrenergic receptor blockade. It has been proposed that the clinical efficacy of flecainide in CPVT is because of the combined actions of direct blockade of ryanodine receptors (RyR2) and Na(+) channel inhibition. However, there is presently no direct evidence to support the notion that flecainide blocks RyR2 Ca(2+) flux in the physiologically relevant (luminal-to-cytoplasmic) direction. The mechanism of flecainide action remains controversial. OBJECTIVE: To examine, in detail, the effect of flecainide on the human RyR2 channel and to establish whether the direct blockade of physiologically relevant RyR2 ion flow by the drug contributes to its therapeutic efficacy in the clinical management of CPVT. METHODS AND RESULTS: Using single-channel analysis, we show that, even at supraphysiological concentrations, flecainide did not inhibit the physiologically relevant, luminal-to-cytosolic flux of cations through the channel. Moreover, flecainide did not alter RyR2 channel gating and had negligible effect on the mechanisms responsible for the sarcoplasmic reticulum charge-compensating counter current. Using permeabilized cardiac myocytes to eliminate any contribution of plasmalemmal Na(+) channels to the observed actions of the drug at the cellular level, flecainide did not inhibit RyR2-dependent sarcoplasmic reticulum Ca(2+) release. CONCLUSIONS: The principal action of flecainide in CPVT is not via a direct interaction with RyR2. Our data support a model of flecainide action in which Na(+)-dependent modulation of intracellular Ca(2+) handling attenuates RyR2 dysfunction in CPVT.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Flecainide/pharmacology , Myocytes, Cardiac/drug effects , Ryanodine Receptor Calcium Release Channel/drug effects , Tachycardia, Ventricular/drug therapy , Voltage-Gated Sodium Channel Blockers/pharmacology , Animals , Calcium Signaling/drug effects , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Male , Membrane Potentials , Myocytes, Cardiac/metabolism , Potassium Channels/drug effects , Potassium Channels/metabolism , Rats, Sprague-Dawley , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/physiopathology , Time Factors , Transfection
6.
Br J Pharmacol ; 180(3): 255-263, 2023 02.
Article in English | MEDLINE | ID: mdl-36529953

ABSTRACT

Scientists who plan to publish in the British Journal of Pharmacology (BJP) should read this article before undertaking studies utilising anaesthetics in mammalian animals. This editorial identifies certain gaps in the reporting of details on the use of anaesthetics in animal research studies published in the BJP. The editorial also provides guidance, based upon current best practices, for performing in vivo experiments that require anaesthesia. In addition, mechanisms of action and physiological impact of specific anaesthetic agents are discussed. Our goal is to identify best practices and to provide guidance on the information required for manuscripts submitted to the BJP that involve the use of anaesthetic agents in studies with experimental animals.


Subject(s)
Anesthesia , Anesthetics , Animal Experimentation , Animals , Anesthetics/pharmacology , Mammals
7.
Am J Physiol Cell Physiol ; 303(9): C897-910, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22843795

ABSTRACT

The normal contractile, electrical, and energetic function of the heart depends on the synchronization of biological oscillators and signal integrators that make up cellular signaling networks. In this review we interpret experimental data from molecular, cellular, and transgenic models of cardiac signaling behavior in the context of established concepts in cell network architecture and organization. Focusing on the cellular Ca(2+) handling machinery, we describe how the plasticity and adaptability of normal Ca(2+) signaling is dependent on dynamic network configurations that operate across a wide range of functional states. We consider how (mal)adaptive changes in signaling pathways restrict the dynamic range of the network such that it cannot respond appropriately to physiologic stimuli or perturbation. Based on these concepts, a model is proposed in which pathologic abnormalities in cardiac rhythm and contractility (e.g., arrhythmias and heart failure) arise as a consequence of progressive desynchronization and reduction in the dynamic range of the Ca(2+) signaling network. We discuss how a systems-level understanding of the network organization, cellular noise, and chaotic behavior may inform the design of new therapeutic modalities that prevent or reverse the disease-linked unraveling of the Ca(2+) signaling network.


Subject(s)
Calcium Signaling/physiology , Heart/physiology , Adaptation, Physiological , Animals , Heart Diseases/physiopathology , Humans , Mice , Models, Cardiovascular , Rats
8.
J Lipid Res ; 53(2): 282-91, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22058426

ABSTRACT

Synthesis inhibition is the basis for the treatment of type 1 Gaucher disease by the glucosylceramide synthase (GCS) inhibitor eliglustat tartrate. However, the extended use of eliglustat and related compounds for the treatment of glycosphingolipid storage diseases with CNS manifestations is limited by the lack of brain penetration of this drug. Property modeling around the D-threo-1-phenyl-2-decanoylamino-3-morpholino-propanol (PDMP) pharmacophore was employed in a search for compounds of comparable activity against the GCS but lacking P-glycoprotein (MDR1) recognition. Modifications of the carboxamide N-acyl group were made to lower total polar surface area and rotatable bond number. Compounds were screened for inhibition of GCS in crude enzyme and whole cell assays and for MDR1 substrate recognition. One analog, 2-(2,3-dihydro-1H-inden-2-yl)-N-((1R,2R)-1-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-1-hydroxy-3-(pyrrolidin-1-yl)propan-2-yl)acetamide (CCG-203586), was identified that inhibited GCS at low nanomolar concentrations with little to no apparent recognition by MDR1. Intraperitoneal administration of this compound to mice for 3 days resulted in a significant dose dependent decrease in brain glucosylceramide content, an effect not seen in mice dosed in parallel with eliglustat tartrate.


Subject(s)
Brain/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Glucosylceramides/metabolism , Glucosyltransferases/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Brain/metabolism , Cells, Cultured , Dioxanes/chemical synthesis , Dioxanes/pharmacology , Dose-Response Relationship, Drug , Drug Design , Drug Evaluation, Preclinical/methods , Indans/chemical synthesis , Indans/pharmacology , Injections, Intraperitoneal , Mice , Mice, Inbred C57BL , Morpholines/chemistry , Vinblastine/pharmacokinetics
9.
Biochem J ; 438(3): 545-53, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21692749

ABSTRACT

Sperm PLCζ (phospholipase Cζ) is a distinct phosphoinositide-specific PLC isoform that is proposed to be the physiological trigger of egg activation and embryo development at mammalian fertilization. Recombinant PLCζ has the ability to trigger Ca²âº oscillations when expressed in eggs, but it is not known how PLCζ activity is regulated in sperm or eggs. In the present study, we have transfected CHO (Chinese-hamster ovary) cells with PLCζ fused with either YFP (yellow fluorescent protein) or luciferase and found that PLCζ-transfected cells did not display cytoplasmic Ca²âº oscillations any differently from control cells. PLCζ expression was not associated with changes in CHO cell resting Ca²âº levels, nor with a significantly changed Ca²âº response to extracellular ATP compared with control cells transfected with either YFP alone, a catalytically inactive PLCζ or luciferase alone. Sperm extracts containing PLCζ also failed to cause Ca²âº oscillations in CHO cells. Despite these findings, PLCζ-transfected CHO cell extracts exhibited high recombinant protein expression and PLC activity. Furthermore, either PLCζ-transfected CHO cells or derived cell extracts could specifically cause cytoplasmic Ca²âº oscillations when microinjected into mouse eggs. These data suggest that PLCζ-mediated Ca²âº oscillations may require specific factors that are only present within the egg cytoplasm or be inhibited by factors present only in somatic cell lines.


Subject(s)
Calcium/metabolism , Oocytes/cytology , Phosphoinositide Phospholipase C/metabolism , Adenosine Triphosphate/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cricetinae , Cytoplasm/genetics , Cytoplasm/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mammals/genetics , Mammals/metabolism , Mice , Oocytes/metabolism , Phosphoinositide Phospholipase C/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
10.
Adv Exp Med Biol ; 740: 183-215, 2012.
Article in English | MEDLINE | ID: mdl-22453943

ABSTRACT

In excitable tissues, the ryanodine receptor Ca(2+) release channel (RyR) protein complex regulates excitation-contraction coupling, exocytosis, gene expression and apoptosis. Defects in RyR function, in genetic or acquired pathologies, lead to massive disruptions of Ca(2+) release that can be lethal. Therefore, RyR has emerged as a putative therapeutic target and an increasing number of RyR-targeting drugs are currently being tested.Nonetheless this large-size channel is still a mystery in terms of structure, which hinders full characterization of the properties of this central protein. This chapter is dedicated to the methods available to examine RyR structure and function. The aim of the article is to concentrate on contemporary methodologies rather than focusing overtly on the progress that has been achieved using these techniques. Here we review a series of reliable approaches that are routinely employed to investigate this channel. Technical limitations are discussed, and technological developments are presented. This work is not a handbook, but it can be used as a resource and a starting point for the investigation of RyR at different levels of resolution.


Subject(s)
Ryanodine Receptor Calcium Release Channel/physiology , Animals , Binding Sites , Calcium/metabolism , Humans , Lipid Bilayers/chemistry , Protein Structure, Tertiary , Ryanodine Receptor Calcium Release Channel/analysis , Ryanodine Receptor Calcium Release Channel/chemistry
11.
Br J Pharmacol ; 179(11): 2558-2563, 2022 06.
Article in English | MEDLINE | ID: mdl-34698387

ABSTRACT

Flecainide is used to treat catecholaminergic polymorphic ventricular tachycardia (CPVT), an arrhythmia caused by disrupted cellular Ca2+ handling following ß-adrenergic stimulation. The clinical efficacy of flecainide in this context involves complex effects on multiple ion channels that may be influenced by the disease state. A compelling narrative has been constructed around flecainide's nonselective block of sarcoplasmic reticulum (SR) lumen-to-cytoplasm Ca2+ release through intracellular calcium release channels (RyR2). However, ion fluxes across the SR membrane during heart contraction are bidirectional, and here, we review experimental evidence that flecainide's principal action on RyR2 involves the partial block of ion flow in the cytoplasm-to-lumen direction (i.e., flecainide inhibits RyR2-mediated SR 'countercurrent'). Experimental approaches that could advance new knowledge on the mechanism of RyR2 block by flecainide are proposed. Some impediments to progress in this area, that must be overcome to enable the development of superior drugs to treat CPVT, are also considered.


Subject(s)
Flecainide , Tachycardia, Ventricular , Anti-Arrhythmia Agents/pharmacology , Anti-Arrhythmia Agents/therapeutic use , Calcium/metabolism , Flecainide/pharmacology , Flecainide/therapeutic use , Humans , Mutation , Myocytes, Cardiac , Ryanodine Receptor Calcium Release Channel , Sarcoplasmic Reticulum , Tachycardia, Ventricular/drug therapy
12.
Pest Manag Sci ; 78(3): 869-880, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34821007

ABSTRACT

BACKGROUND: Resistance to diamide insecticides in Lepidoptera is known to be caused primarily by amino acid changes on the ryanodine receptor (RyR). Recently, two new target site mutations, G4946V and I4790M, have emerged in populations of diamondback moth, Plutella xylostella, as well as in other lepidopteran species, and both mutations have been shown empirically to decrease diamide efficacy. Here, we quantify the impact of the I4790M mutation on diamide activation of the receptor, as compared to alterations at the G4946 locus. RESULTS: I4790M when introduced into P. xylostella RyR expressed in an insect-derived Sf9 cell line was found to mediate just a ten-fold reduction in chlorantraniliprole efficacy (compared to 104- and 146-fold reductions for the G4946E and G4946V variants, respectively), whilst in the field its presence is associated with a ≥150-fold reduction. I4790M-mediated resistance to flubendiamide was estimated to be >24-fold. When the entire coding sequence of P. xylostella RyR was integrated into Drosophila melanogaster, the I4790M variant conferred ~4.4-fold resistance to chlorantraniliprole and 22-fold resistance to flubendiamide in the 3rd instar larvae, confirming that it imparts only a moderate level of resistance to diamide insecticides. Although the I4790M substitution appears to bear no fitness costs in terms of the flies' reproductive capacity, when assessed in a noncompetitive environment, it does, however, have potentially major impacts on mobility at both the larval and adult stages. CONCLUSIONS: I4790M imparts only a moderate level of resistance to diamide insecticides and potentially confers significant fitness costs to the insect.


Subject(s)
Insecticide Resistance , Moths , Ryanodine Receptor Calcium Release Channel , Animals , Animals, Genetically Modified , Cell Line , Diamide/pharmacology , Drosophila melanogaster/genetics , Insecticide Resistance/genetics , Moths/genetics , Mutation , Ryanodine Receptor Calcium Release Channel/genetics
13.
Br J Pharmacol ; 179(15): 3907-3913, 2022 08.
Article in English | MEDLINE | ID: mdl-35673806

ABSTRACT

Scientists who plan to publish in British Journal of Pharmacology (BJP) must read this article before undertaking a study. This editorial provides guidance for the design of experiments. We have published previously two guidance documents on experimental design and analysis (Curtis et al., 2015; Curtis et al., 2018). This update clarifies and simplifies the requirements on design and analysis for BJP manuscripts. This editorial also details updated requirements following an audit and discussion on best practice by the BJP editorial board. Explanations for the requirements are provided in the previous articles. Here, we address new issues that have arisen in the course of handling manuscripts and emphasise three aspects of design that continue to present the greatest challenge to authors: randomisation, blinded analysis and balance of group sizes.


Subject(s)
Research Design
15.
J Immunol ; 183(1): 506-17, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19542461

ABSTRACT

TLR overactivation may lead to end organ damage and serious acute and chronic inflammatory conditions. TLR responses must therefore be tightly regulated to control disease outcomes. We show in this study the ability of the soluble form of TLR2 (sTLR2) to regulate proinflammatory responses, and demonstrate the mechanisms underlying sTLR2 regulatory capacity. Cells overexpressing sTLR2, or stimulated in the presence of the sTLR2 protein, are hyporesponsive to TLR2 ligands. Regulation was TLR2 specific, and affected NF-kappaB activation, phagocytosis, and superoxide production. Natural sTLR2-depleted serum rendered leukocytes hypersensitive to TLR2-mediated stimulation. Mice administered sTLR2 together with Gram-positive bacteria-derived components showed lower peritoneal levels of the neutrophil (PMN) chemoattractant, keratinocyte-derived chemokine; lower PMN numbers; and a reduction in late apoptotic PMN. Mononuclear cell recruitment remained unaffected, and endogenous peritoneal sTLR2 levels increased. Notably, the capacity of sTLR2 to modulate acute inflammatory parameters did not compromise the ability of mice to clear live Gram-positive bacteria-induced infection. Mechanistically, sTLR2 interfered with TLR2 mobilization to lipid rafts for signaling, acted as a decoy microbial receptor, and disrupted the interaction of TLR2 with its coreceptor, CD14, by associating with CD14. These findings establish sTLR2 as a regulator of TLR2-mediated inflammatory responses, capable of blunting immune responses without abrogating microbial recognition and may inform the design of novel therapeutics against acute and chronic inflammatory conditions.


Subject(s)
Inflammation Mediators/physiology , Peritonitis/immunology , Peritonitis/microbiology , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Toll-Like Receptor 2/physiology , Acute Disease , Amino Acid Sequence , Animals , Bacterial Adhesion/genetics , Bacterial Adhesion/immunology , CHO Cells , Cell Line , Cricetinae , Cricetulus , Humans , Immunity, Innate/genetics , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/blood , Ligands , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharide Receptors/physiology , Membrane Microdomains/genetics , Membrane Microdomains/immunology , Membrane Microdomains/microbiology , Membrane Microdomains/pathology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Peritonitis/pathology , Peritonitis/prevention & control , Signal Transduction/genetics , Signal Transduction/immunology , Staphylococcal Infections/pathology , Staphylococcal Infections/prevention & control , Staphylococcus epidermidis/immunology , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 2/blood , Toll-Like Receptor 2/genetics
16.
Circ Res ; 100(6): 874-83, 2007 Mar 30.
Article in English | MEDLINE | ID: mdl-17322175

ABSTRACT

Ca(2+) release via type 2 ryanodine receptors (RyR2) regulates cardiac function. Molecular cloning of human RyR2 identified 2 alternatively spliced variants, comprising 30- and 24-bp sequence insertions; yet their role in shaping cardiomyocyte Ca(2+) signaling and cell phenotype is unknown. We profiled the developmental regulation and the tissue and species specificity of these variants and showed that their recombinant expression in HL-1 cardiomyocytes profoundly modulated nuclear and cytoplasmic Ca(2+) release. All splice variants localized to the sarcoplasmic reticulum, perinuclear Golgi apparatus, and to finger-like invaginations of the nuclear envelope (nucleoplasmic reticulum). Strikingly, the 24-bp splice insertion that was present at low levels in embryonic and adult hearts was essential for targeting RyR2 to an intranuclear Golgi apparatus and promoted the intracellular segregation of this variant. The amplitude variability of nuclear and cytoplasmic Ca(2+) fluxes were reduced in nonstimulated cardiomyocytes expressing both 30- and 24-bp splice variants and were associated with lower basal levels of apoptosis. Expression of RyR2 containing the 24-bp insertion also suppressed intracellular Ca(2+) fluxes following prolonged caffeine exposure (1 mmol/L, 16 hours) that protected cells from apoptosis. The antiapoptotic effects of this variant were linked to increased levels of Bcl-2 phosphorylation. In contrast, RyR2 containing the 30-bp insertion, which was abundant in human embryonic heart but was decreased during cardiac development, did not protect cardiomyocytes from caffeine-evoked apoptosis. Thus, we provide the first evidence that RyR2 splice variants exquisitely modulate intracellular Ca(2+) signaling and are key determinants of cardiomyocyte apoptotic susceptibility.


Subject(s)
Alternative Splicing/genetics , Apoptosis/genetics , Calcium Signaling/physiology , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Apoptosis/drug effects , Caffeine/pharmacology , Cell Nucleus/metabolism , Cells, Cultured , Cytoplasm/metabolism , Gene Expression Regulation, Developmental , Gene Transfer Techniques , Humans , Mice , Molecular Sequence Data , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/metabolism , Species Specificity
17.
Cardiovasc Res ; 77(2): 302-14, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18006486

ABSTRACT

Heart failure (HF) is a chronic multi-factorial disease characterized by sarcoplasmic reticulum (SR) dysfunction that manifests as severely reduced contractility and increased risk of arrhythmia. Several lines of evidence have revealed the existence of defective ryanodine receptor (RyR2)-mediated Ca(2+) leak in HF, although its relevance as a causative factor rather than a phenotypic consequence of the disease is questioned. This review will consider the relative contribution of RyR2-mediated Ca(2+) leak to the profound cellular, transcriptional and electrical remodelling associated with HF. In particular, it will focus on our current understanding of the role of defective phosphorylation of RyR2 as a both a chronic mediator of excitation-contraction coupling (ECC) dysfunction and as a potent catalyst of RyR2-dependent arrhythmogenesis. A hypothetical concept that SR Ca(2+) leak fundamentally underlies the increased arrhythmogenic susceptibility in HF, but that it may not directly contribute to contractile dysfunction, which may involve maladaptive perturbations in metabolism and energy utilization, is also discussed.


Subject(s)
Calcium/metabolism , Heart Failure/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Arrhythmias, Cardiac/etiology , Humans , Myocardial Contraction , Myocardium/metabolism , Phosphorylation , Ryanodine Receptor Calcium Release Channel/metabolism
18.
Front Physiol ; 10: 340, 2019.
Article in English | MEDLINE | ID: mdl-30984025

ABSTRACT

The connectedness of signaling components in network structures is a universal feature of biologic information processing. Such organization enables the transduction of complex input stimuli into coherent outputs and is essential in modulating activities as diverse as the cooperation of bacteria within populations and the dynamic organization of mitochondria within cells. Here, we highlight some common principles that underpin collectivization in bacteria and mitochondrial populations and the advantages conferred by such behavior. We discuss the concept that bacteria and mitochondria act as signal transducers of their localized metabolic environments to bring about energy-dependent clustering to modulate higher-order function across multiple scales.

19.
Circ Res ; 98(1): 88-97, 2006 Jan 06.
Article in English | MEDLINE | ID: mdl-16339485

ABSTRACT

Arrhythmogenic cardiac ryanodine receptor (RyR2) mutations are associated with stress-induced malignant tachycardia, frequently leading to sudden cardiac death (SCD). The causative mechanisms of RyR2 Ca2+ release dysregulation are complex and remain controversial. We investigated the functional impact of clinically-severe RyR2 mutations occurring in the central domain, and the C-terminal I domain, a key locus of RyR2 autoregulation, on interdomain interactions and Ca2+ release in living cells. Using high-resolution confocal microscopy and fluorescence resonance energy transfer (FRET) analysis of interaction between fusion proteins corresponding to amino- (N-) and carboxyl- (C-) terminal RyR2 domains, we determined that in resting cells, RyR2 interdomain interaction remained unaltered after introduction of SCD-linked mutations and normal Ca2+ regulation was maintained. In contrast, after channel activation, the abnormal Ca2+ release via mutant RyR2 was intrinsically linked to altered interdomain interaction that was equivalent with all mutations and exhibited threshold characteristics (caffeine >2.5 mmol/L; Ca2+ >150 nmol/L). Noise analysis revealed that I domain mutations introduced a distinct pattern of conformational instability in Ca2+ handling and interdomain interaction after channel activation that was absent in signals obtained from the central domain mutation. I domain-linked channel instability also occurred in intact RyR2 expressed in CHO cells and in HL-1 cardiomyocytes. These new insights highlight a critical role for mutation-linked defects in channel autoregulation, and may contribute to a molecular explanation for the augmented Ca2+ release following RyR2 channel activation. Our findings also suggest that the mutational locus may be an important mechanistic determinant of Ca2+ release channel dysfunction in arrhythmia and SCD.


Subject(s)
Arrhythmias, Cardiac/etiology , Mutation , Ryanodine Receptor Calcium Release Channel/genetics , Animals , CHO Cells , Caffeine/pharmacology , Calcium/metabolism , Cricetinae , Death, Sudden, Cardiac/etiology , Fluorescence Resonance Energy Transfer , Homeostasis , Humans , Protein Structure, Tertiary , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/physiology , Tachycardia, Ventricular/etiology
SELECTION OF CITATIONS
SEARCH DETAIL