• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 12
  • 6
  • 3
  • 1
  • Tagged with
  • 22
  • 22
  • 22
  • 12
  • 7
  • 6
  • 6
  • 6
  • 5
  • 5
  • 4
  • 4
  • 4
  • 3
  • 3
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

The structure of excitation-contraction coupling in atrial cardiomyocytes

Schulson, Meredith Nicole 05 1900 (has links)
Standard local control theory, which describes Ca²⁺ release during excitation-contraction coupling (ECC), assumes that all Ryanodine Receptor (RyR) complexes are equivalent. Recent data from our laboratory has called this assumption into question. Specifically, we have shown that RyR complexes in ventricular myocytes differ depending on their location within the cell. This, and other data, has led us to hypothesize that similar differences occur within the rat atrial cell. To test this hypothesis, we have triple-labeled enzymatically-isolated, fixed myocytes to examine the distribution and colocalization of RyR, calsequestrin (CSQ), voltage-gated Ca²⁺ channels (Cav1.2), sodium-calcium exchangers (NCX), and caveolin-3 (cav-3). All images were acquired on a wide-field microscope, deconvolved, and subject to extensive analysis, including a novel method of measuring statistical significance of the recorded colocalization values. Overall, eight surface RyR populations were identified, depending on its binding partners. One of these groups, in which RyR, Cav1.2, and NCX colocalize, may provide the structural basis for ‘eager’ sites of Ca²⁺ release in atria, while other groups were defined based on their association with cav-3, and are therefore highly likely to be under the influence of other signaling molecules located within caveolae. Importantly, although a small portion of the surface RyR in atria do colocalize with NCX alone, the majority are tightly linked to Cav1.2 alone or Cav1.2 and NCX together. Therefore, it appears likely that Cav1.2-mediated calcium-induced calcium release (CICR) is the primary method of initiating Ca²⁺ release from the SR during EC coupling.
2

The structure of excitation-contraction coupling in atrial cardiomyocytes

Schulson, Meredith Nicole 05 1900 (has links)
Standard local control theory, which describes Ca²⁺ release during excitation-contraction coupling (ECC), assumes that all Ryanodine Receptor (RyR) complexes are equivalent. Recent data from our laboratory has called this assumption into question. Specifically, we have shown that RyR complexes in ventricular myocytes differ depending on their location within the cell. This, and other data, has led us to hypothesize that similar differences occur within the rat atrial cell. To test this hypothesis, we have triple-labeled enzymatically-isolated, fixed myocytes to examine the distribution and colocalization of RyR, calsequestrin (CSQ), voltage-gated Ca²⁺ channels (Cav1.2), sodium-calcium exchangers (NCX), and caveolin-3 (cav-3). All images were acquired on a wide-field microscope, deconvolved, and subject to extensive analysis, including a novel method of measuring statistical significance of the recorded colocalization values. Overall, eight surface RyR populations were identified, depending on its binding partners. One of these groups, in which RyR, Cav1.2, and NCX colocalize, may provide the structural basis for ‘eager’ sites of Ca²⁺ release in atria, while other groups were defined based on their association with cav-3, and are therefore highly likely to be under the influence of other signaling molecules located within caveolae. Importantly, although a small portion of the surface RyR in atria do colocalize with NCX alone, the majority are tightly linked to Cav1.2 alone or Cav1.2 and NCX together. Therefore, it appears likely that Cav1.2-mediated calcium-induced calcium release (CICR) is the primary method of initiating Ca²⁺ release from the SR during EC coupling.
3

The structure of excitation-contraction coupling in atrial cardiomyocytes

Schulson, Meredith Nicole 05 1900 (has links)
Standard local control theory, which describes Ca²⁺ release during excitation-contraction coupling (ECC), assumes that all Ryanodine Receptor (RyR) complexes are equivalent. Recent data from our laboratory has called this assumption into question. Specifically, we have shown that RyR complexes in ventricular myocytes differ depending on their location within the cell. This, and other data, has led us to hypothesize that similar differences occur within the rat atrial cell. To test this hypothesis, we have triple-labeled enzymatically-isolated, fixed myocytes to examine the distribution and colocalization of RyR, calsequestrin (CSQ), voltage-gated Ca²⁺ channels (Cav1.2), sodium-calcium exchangers (NCX), and caveolin-3 (cav-3). All images were acquired on a wide-field microscope, deconvolved, and subject to extensive analysis, including a novel method of measuring statistical significance of the recorded colocalization values. Overall, eight surface RyR populations were identified, depending on its binding partners. One of these groups, in which RyR, Cav1.2, and NCX colocalize, may provide the structural basis for ‘eager’ sites of Ca²⁺ release in atria, while other groups were defined based on their association with cav-3, and are therefore highly likely to be under the influence of other signaling molecules located within caveolae. Importantly, although a small portion of the surface RyR in atria do colocalize with NCX alone, the majority are tightly linked to Cav1.2 alone or Cav1.2 and NCX together. Therefore, it appears likely that Cav1.2-mediated calcium-induced calcium release (CICR) is the primary method of initiating Ca²⁺ release from the SR during EC coupling. / Medicine, Faculty of / Cellular and Physiological Sciences, Department of / Graduate
4

Pathogenesis of Hypertrophic Cardiomyopathy is Mutation Rather Than Disease Specific: A Comparison of the Cardiac Troponin T E163R and R92Q Mouse Models

Ferrantini, Cecilia, Coppini, Raffaele, Pioner, Josè Manuel, Gentile, Francesca, Tosi, Benedetta, Mazzoni, Luca, Scellini, Beatrice, Piroddi, Nicoletta, Laurino, Annunziatina, Santini, Lorenzo, Spinelli, Valentina, Sacconi, Leonardo, De Tombe, Pieter, Moore, Rachel, Tardiff, Jil, Mugelli, Alessandro, Olivotto, Iacopo, Cerbai, Elisabetta, Tesi, Chiara, Poggesi, Corrado 22 July 2017 (has links)
Background-In cardiomyocytes from patients with hypertrophic cardiomyopathy, mechanical dysfunction and arrhythmogenicity are caused by mutation-driven changes in myofilament function combined with excitation-contraction (E-C) coupling abnormalities related to adverse remodeling. Whether myofilament or E-C coupling alterations are more relevant in disease development is unknown. Here, we aim to investigate whether the relative roles of myofilament dysfunction and E-C coupling remodeling in determining the hypertrophic cardiomyopathy phenotype are mutation specific. Methods and Results-Two hypertrophic cardiomyopathy mouse models carrying the R92Q and the E163R TNNT2 mutations were investigated. Echocardiography showed left ventricular hypertrophy, enhanced contractility, and diastolic dysfunction in both models; however, these phenotypes were more pronounced in the R92Q mice. Both E163R and R92Q trabeculae showed prolonged twitch relaxation and increased occurrence of premature beats. In E163R ventricular myofibrils or skinned trabeculae, relaxation following Ca2+ removal was prolonged; resting tension and resting ATPase were higher; and isometric ATPase at maximal Ca2+ activation, the energy cost of tension generation, and myofilament Ca2+ sensitivity were increased compared with that in wildtype mice. No sarcomeric changes were observed in R92Q versus wild-type mice, except for a large increase in myofilament Ca2+ sensitivity. In R92Q myocardium, we found a blunted response to inotropic interventions, slower decay of Ca2+ transients, reduced SERCA function, and increased Ca2+/calmodulin kinase II activity. Contrarily, secondary alterations of E-C coupling and signaling were minimal in E163R myocardium. Conclusions-In E163R models, mutation-driven myofilament abnormalities directly cause myocardial dysfunction. In R92Q, diastolic dysfunction and arrhythmogenicity are mediated by profound cardiomyocytesignaling and E-C coupling changes. Similar hypertrophic cardiomyopathy phenotypes can be generated through different pathways, implying different strategies for a precision medicine approach to treatment.
5

Characterisation of novel cardiac and skeletal ion channels on intracellular Ca2+ stores

Eberhardt, David Richard January 2018 (has links)
Excitation-contraction (EC) coupling is the process by which Ca<sup>2+</sup> is released from the sarcoplasmic reticulum (SR) and is fundamental to cardiac and skeletal muscle function. The SR contains many uncharacterised ion channels and proteins which may influence EC coupling and in this thesis I have investigated the biophysical properties of some of these channels. I have demonstrated that the single-channel gating and conducting properties of SR K<sup>+</sup> channels from various mammalian species (rabbit, sheep and mouse) are very similar. I investigated the actions of possible physiological regulators of these channels and demonstrated that luminal Ca<sup>2+</sup> and Mg<sup>2+</sup> can block K<sup>+</sup> flux in a voltage-dependent manner, while luminal Ca<sup>2+</sup>, Ni<sup>2+</sup>, and alkaline pH can reduce Po by additional mechanisms. I also characterised the single-channel properties of the various SR anion channels that are observed after incorporating mammalian SR vesicles into artificial membranes. The trimeric intracellular cation channels (TRIC-A and TRIC-B) and Mitsugumin 23 (MG23) are suggested to be SR cation channels. I have therefore utilised Tric-a KO and Mg23 KO mice to study SR membranes devoid of TRIC-A and MG23. Additionally, I have begun to investigate the single-channel properties of purified c. elegans TRIC-B1 and human TRIC-A. I found that SR K<sup>+</sup> channel function was altered in SR from Tric-a KO or Mg23 KO tissue, however the underlying mechanisms for the observed changes appear to be complex. My initial studies of the purified TRIC-A and TRIC-B proteins show that they are permeable to K<sup>+</sup>, Ca<sup>2+</sup>, choline, and Cl<sup>-</sup>, properties which deviate from those of SR K<sup>+</sup> channels from rabbit, mouse and sheep. This may reflect species differences or alterations to protein function caused during the purification process or that SR K<sup>+</sup> channels remain an unidentified class of ion channel.
6

Régulations monoaminergiques AMPc-dépendantes du coeur sain et pathologique / cAMP-dependent monoaminergic regulations of the healthy and failing heart

Meschin, Pierre 01 December 2014 (has links)
La fonction cardiaque est finement régulée par des hormones de type monoamines qui constituent des régulateurs cruciaux de l’activité cardiaque (chronotropie et inotropie). Ces hormones dérivées d’acides aminés aromatiques comprenant les catécholamines et la sérotonine maintiennent l’activité du myocarde dans un cadre physiologique tout en lui permettant de s’adapter aux contraintes environnementales. Les récepteurs cellulaires des monoamines sont couplés à des voies de signalisation qui impliquent un nucléotide cyclique, l’AMPc, et modulent la contractilité des cardiomyocytes par l’intermédiaire de multiples phosphorylations des protéines régulatrices du cycle du calcium (canal calcique de type L, RyR2 ou phospholamban) par la protéine kinase A AMPc-dépendante. Lorsque les monoamines voient leurs activités dérégulées en contextes pathologiques tels que l’insuffisance cardiaque (IC) ou un lors d'un traitement antidépresseur, elles conduisent à une hyperstimulation de leurs récepteurs spécifiques. Cette dernière altère alors les voies impliquant l’AMPc et les flux calciques engendrant des évènements ectopiques proarythmogéniques nommés post-dépolarisations. Ces dysfonctions de la contractilité cellulaire et de l'homéostasie calcique peuvent être à l’origine d’arythmies tissulaires et de morts subites cardiaques. Les altérations de l’homéostasie calcique subsistent en dépit des approches thérapeutiques actuelles (!-bloquants, inhibiteurs de l’enzyme de conversion de l’angiotensine) qui vise à freiner le remodelage myocardique post-ischémique et constituent donc une cible active de la recherche cardiovasculaire. Les Rycals, stabilisateurs pharmacologiques du RyR2, représentent une nouvelle approche visant à remédier à ces altérations. Au sein de ces travaux de recherche, nous avons axé nos études sur les deux voies monoaminergiques AMPc cardiaques majeures, les voies adrénergiques et sérotoninergiques. Un premier axe d’étude a consisté en l’évaluation des bénéfices potentiels d’un nouveau Rycal, le S44121, sur la survenue d’arythmies cellulaires et tissulaires en comparaison d’un !-bloquant de référence, le métoprolol, dans un contexte d’IC post-infarctus chez le rat. L’étude n’a cependant pas mise en évidence de bénéfices du S44121 mais a confirmé la cardioprotection exercée par le métoprolol. Un deuxième axe d’étude a évalué l’implication potentielle au niveau cardiaque de la protéine S100A10 dans la modulation de la voie du récepteur à la sérotonine de type 4 (5-HT4R) en conditions physiologiques ou en contexte d’IC. Cette étude originale a mis en avant pour la première fois dans le coeur sain un rôle de la S100A10 dans l’apparition d’une voie 5-HT4R proarythmogène lorsque son expression est induite par une neurotrophine (Brain-derived neurotrophic factor) ou un antidépresseur (imipramine). En revanche, le rôle de la S100A10 dans la modulation de la voie 5-HT4R en contexte d’IC n’a pas été déterminé de façon certaine. / Cardiac function is tightly regulated by hormones such as monoamines which are substantial modulators of cardiac activity (chronotropy and inotropy). These hormones, derived from aromatic amino acids, maintain myocardial activity in a physiological range and allow the cardiac adaptation to environmental conditions. The cellular receptors to monoamines are coupled to signaling pathways involving a cyclic nucleotide, cAMP, and modulate cardiac activity by phosphorylating several key proteins of calcium handling (L-type calcium channel, RyR2 or phospholamban) by the cAMP-dependent protein kinase A. Deregulation of monoamines in pathological conditions such as heart failure (HF) or during antidepressanttreatment leads to a hyperstimulation of their specific receptors. It therefore induces alterations of the cAMP signaling pathway and calcium handling leading to the occurrence of proarrhythmogenic ectopic cellular events known as afterdepolarizations. These dysfunctions in cellular contractility and calcium handling may cause tissue arrhythmias andeven sudden cardiac death. Calcium handling alterations leading to cardiac arrhythmias remain a clinically relevant issue despite the current therapeutical approaches (!-blockers, angiotensin-converting-enzyme inhibitors) which slow the post-ischemic myocardial remodeling and thus represent an active target in the cardiovascular research field. Rycals, RyR2 pharmacological stabilisers, are a new approach to prevent these alterations. In this work, we focused on the two major monoaminergic cAMP-dependent pathways in the heart, the adrenergic and serotoninergic pathways. In the first part of this work, we aimed to evaluate the potential benefits of a new Rycal, S44121, on cellular and tissue arrhythmias occurrence in post-myocardial infarction rat model. These effects were compared to those of the well-known !-blocker, metoprolol. This study failed to show any strong benefit of S44121 but confirmed the cardioprotection associated with the metoprolol use. In a second part of the work presented here, we aimed to evaluate the potential involvement of the S100A10 protein in the modulation of the cardiac serotonin receptor 4 pathway (5-HT4R) in physiological conditions or during HF. This original study unraveled for the first time a new role for S100A10 in the healthy heart by revealing a functional 5-HT4R pathway when S100A10 expression is induced by neurotrophins such as brain-derived neurotrophic factor or by antidepressant drugs such as imipramine. However, we failed to conclude on a direct evidence for a role of S100A10 in the modulation of the 5-HT4R pathway in the failingheart.
7

Caractérisation des voies de mort cellulaire lors du remodelage cardiaque dans les cardiopathies d'origine ischémique / Characterization of cell death pathway during myocardial ischemia reperfusion

Roberge, Stéphanie 09 December 2013 (has links)
L'ischémie se caractérise par l'obstruction d'une artère coronaire qui prive le tissu d'un apport en oxygène et nutriments. Bien que nécessaire, la reperfusion, c'est-à-dire la réouverture de l'artère, s'accompagne de lésions tissulaires, appelées lésions de reperfusion. Au cours de l'I/R, le TNF-α, cytokine pro-inflammatoire, augmente. Sa liaison sur son récepteur TNFR1 induit le recrutement des protéines FADD et procaspase-8 formant le complexe DISC qui permet l'activation de la caspase-8. La caspase-8 clive une protéine pro-apoptotique, Bid, qui induit une perméabilisation de la membrane mitochondriale entrainant une production excessive de radicaux libres et une libération de cytochrome c. Cette dernière associée à Apaf-1 et procaspase-9 sert de plateforme d'activation à la caspase-9, qui, une fois activée, clive et active la caspase-3. La caspase-2 est une caspase initiatrice, tout comme la caspase-8. Pourtant, son rôle dans l'I/R cardiaque est peu connu. La production de ROS via la voie TNF-α/caspase-8 provoque des dommages à l'ADN. Ceci entraine l'activation de PARP-1, une enzyme impliquée dans la réparation de l'ADN. En fonctionnant, PARP-1 produit de l'ADP-ribose qui peut se fixer sur le canal TRPM2 et ainsi l'activer. L'ouverture de ce canal cationique provoque une entrée de Ca2+ qui contribue à la mort cellulaire et aux lésions de reperfusion. L'objectif de ce travail est de déterminer les mécanismes de mort cellulaire faisant intervenir la caspase-8, la caspase-2 et TRPM2 et d'évaluer les effets d'une inhibition de ces protéines sur les lésions de reperfusion. Un modèle de rat I/R met en évidence une augmentation de TNF-α après seulement 1h de reperfusion suivie d'une activation de la caspase-8. Cette activation entraine une production de ROS qui altère la structure et la fonction du canal RyR2, favorisant la fuite de Ca2+ du reticulum sarcoplasmique vers le cytosol. La caspase-2, exprimée dans le ventricule gauche, est activée avant la caspase-8 et induit une voie apoptotique de type intrinsèque. L'inhibition de la caspase-8 ou de la caspase-2 diminue les lésions de reperfusion. Parallèlement, le TNF-α induit un courant de type TRPM2 via l'activation de la caspase-8 et la production de ROS. In vivo, l'inhibition de TRPM2 par le clotrimazole diminue les lésions de reperfusion chez un modèle de souris I/R. La caspase-8, la caspase-2 et TRPM2 contribuent aux lésions de reperfusion et apparaissent comme de bonnes cibles dans la cardioprotection. / Myocardial ischemia and reperfusion (I/R) lead to repefusion injury. TNF-α, a pro-inflammatory cytokine, increases during reperfusion and contributes to this injury. The binding TNF/TNFR1 leads to the recruitment of FADD, TRADD and procaspase-8 and form a complexe named DISC. This complexe activates caspase-8, which cleaves Bid, a pro-apoptotic member of Bcl-2 family. tBid disrupts the mitochondrial membrane and induces a ROS production and a release of cytochome c, localized in intermembrane space. In cytosol, a complexe named apoptosome is formed with cytochrome c, Apaf-1 and procaspase-9 to activate caspase-9, which cleaves and activates caspase-3. Like caspase-8, caspase-2 is an initiator caspase. But little data exists on the role of this caspase in myocardial I/R.The disruption of mitochondria induces a ROS production which causes DNA damage. The enzyme PARP-1, involved in DNA repair, is then activated. By operating, PARP-1 produces ADP-ribose which can bind on TRPM2, a non selective cationic channel of TRP family. The opening of TRPM2 causes an increase of cytosolic calcium promoting cell death and reperfusion injury. The goal of this study was to determine the mechanisms of cell death after I/R involving caspase-8, caspase-2 and TRPM2 and to test an inhibitor of each protein on reperfusion injury. With a model of rat I/R, we demonstrated that TNF-α increases after only 1h of reperfusion following by a caspase-8 activation and a ROS production. Oxidative stress causes a modification of RyR2 with a leak of calcium in cytosol. Caspase-2, also expressed in ventricles, is activated before caspase-8 and induces an intrinsic apoptotic pathway until caspase-3 activation. An inhibition of caspase-8 or caspase-2 decreases the reperfusion injury.In mouse cardiomyocytes, TNF-α induces a TRPM2-like current through caspase-8 activation and ROS production. TRPM2 inhibition by clotrimazole decreases cell death and reperfusion injury in vivo.In conclusion, caspase-2, caspase-8 and TRPM2 play an important role in cell death pathway ans should be good therapeutical tools.
8

Die Wirkung ionisierender Strahlung auf die elektromechanische Kopplung und den intrazellulären Ca2+-Haushalt in isolierten Herzmuskelzellen / The influence of ionizing radiation on excitation-contraction coupling and Ca2+ cycling of isolated cardiac myocytes

Neumann, Kay 10 July 2012 (has links)
No description available.
9

Die Effekte der Ca2+-Calmodulin-abhängigen Proteinkinase II (CaMKII) auf die Aktionspotential-morphologie bei mechanischer Last / The effects of Calcium2+/Calmodulin-dependent protein kinase II (CaMKII) on action potential morphology under mechanical load

Gupta, Shamindra Nath 29 October 2013 (has links)
No description available.
10

Re-Expression of T-Type Calcium Channels Minimally Affects Cardiac Contractility and Activates Pro-Survival Signaling Pathways in the Myocardium

Jaleel, Naser January 2010 (has links)
The role of T-type calcium channels (TTCCs) in the heart is unclear. TTCCs are transiently expressed throughout the neonatal heart during a period of rapid cardiac development. A few weeks postnatally, TTCCs are no longer found in ventricular myocytes (VMs) and calcium influx via TTCCs (ICa,T) is only detected in the SA node and Purkinje system. However, pathologic cardiac stress is associated with re-expression of TTCCs in VMs. Whether ICa,T in this setting promotes cardiac growth or exacerbates cardiac function is a topic of debate. The focus of this thesis work was to examine the effect of TTCC re-expression in the normal and diseased myocardium. Our experiments were performed in a transgenic mouse model with inducible, cardiac-specific expression of α1G TTCCs. While both the α1G and α1H TTCC subtypes re-appear during cardiac disease, we specifically evaluated the effects of α1G TTCCs since mRNA levels of this TTCC subtype are markedly elevated during cardiac pathology. We found that transgenic mice with α1G overexpression had robust ICa,T with biophysical properties similar to those published in previous studies. α1G mice had a small increase in cardiac function and showed no evidence of cardiac histopathology or increased mortality. These findings were in contrast to the phenotype of transgenic mice with augmented L-type calcium channel (LTCC) activity secondary to overexpression of the β2a regulatory subunit. While the magnitude of calcium influx in α1G and β2a VMs was similar, we found that cardiac contractility of β2a mice was significantly greater than α1G mice. Also, β2a mice had significant cardiac fibrosis, myocyte death, and premature lethality compared to the benign phenotype of α1G mice. We showed that the phenotypic differences are likely related to the differential spatial localization of T- and LTCCs. Whereas α1G TTCCs were principally localized to the surface sarcolemma, LTCCs were primarily found in the transverse tubules in close proximity to the sites of sarcoplasmic reticulum calcium release. We evaluated the effect of TTCC expression during cardiac disease by inducing myocardial infarction (MI) in α1G mice. Acutely (1-week post MI), α1G mice showed similar worsening of cardiac function and mortality rates compared to control post-infarct mice. However, α1G hearts had smaller infarct sizes which correlated with increased Akt and NFAT activation in α1G than control hearts. After chronic heart failure, i.e. 7- weeks post-infarction, α1G hearts had significant hypertrophic response as determined by increased HW/BW ratio, myocyte cross-sectional area, as well as NFAT and Akt activity. Finally, α1G mice had a small survival benefit than control mice, which while statistically non-significant, suggests that TTCC re-expression does not exacerbate cardiac function as hypothesized by some investigators. We conclude that TTCCs play a minimal role in cardiac function and activate pro-survival signaling pathways in the myocardium. / Physiology

Page generated in 0.1914 seconds