• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 5
  • 2
  • 1
  • 1
  • Tagged with
  • 11
  • 9
  • 8
  • 7
  • 6
  • 6
  • 6
  • 5
  • 5
  • 4
  • 3
  • 3
  • 3
  • 2
  • 2
  • 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

Potent and specific actions of 2-Aminoethoxydiphenyl borate (2-APB) derivatives on Orai channel function

HENDRON, EUNAN January 2013 (has links)
In an effort to dissect the mechanism of SOCe activation, I used two novel 2-APB analogs (DPB162-AE and DPB163-AE) which are ~50-100 times more potent at modifying SOCe than 2-APB. In the presence of STIM1, both compounds (2 µM) differentially affected Orai subtypes, fully blocking endogenous Orai1, but not Orai2 or Orai3 mediated SOCe in DT40 Orai-specific knockout cells. Neither analog directly activated Orai3 over-expressed alone in HEK293 cells. Analysis of constitutively active Orai1 mutant, Orai1V102C, showed an increase in Ca2+ entry after application of DPB162-AE independent of STIM1. When STIM1 was co-expressed with Orai1V102C, there was no inhibitory effect of the analog on the mutant channel complex. DPB162-AE appeared to have a long term effect on the channel complex revealed a lack of SOCe 10 minutes after washout of the analog. STIM1ct-Orai1 Ca2+ entry was moderately increased by DPB162-AE yet constitutively active Stim1ct4EA-Orai1 Ca2+ entry was robustly inhibited. The activation of mutant Orai1V102C indicated the analogs are capable of interacting with Orai1, perhaps to widen the pore, and pointing to a putative mechanism of action for inhibition. FRET analysis indicated no effect on STIM1-Orai1, STIM1ct-Orai1 or SOAR-Orai1 coupling. Thus, the inhibitory effect on STIM1-Orai may be through physical alteration of Orai1 gating. Previously reported as having biphasic effect on SOCe proteins, DPB163-AE appeared to effect its potentiation exclusively via STIM2 with no evident inhibition of STIM2 SOCe. Inhibition by both analogs was mediated by STIM1. DPB162-AE and DPB163-AE had remarkable specificity on Orai1 as opposed to other Ca2+ permeant channels. Neither compound affected Ca2+ entry through TRPC3, TRPC6, or strontium entry through Cav1.2 channels at concentrations (2 µM) that completely inhibited Orai1-mediated SOCe. In summary, DPB162-AE and DPB163-AE are highly specific inhibitors of Orai1 SOCe, with little effect on Orai2 and Orai3, and no effect on other Ca2+ channels. They do not disrupt STIM-Orai coupling but may modify functional Orai1 channel structure to effect their inhibitory action on SOCe. / Biochemistry
2

Structural and Biophysical Studies of the Role of Stromal Interaction Molecules STIM1 and STIM2 in Initiating Store-operated Calcium Entry

Zheng, Le 29 July 2010 (has links)
Store-operated calcium entry (SOCE) is the major Ca2+ entry pathway in most non-excitable cells maintaining prolonged elevation of cytosolic Ca2+ levels required for gene transcription. SOCE is activated by the loss of endoplasmic reticulum (ER) Ca2+ through stromal interaction molecules (STIM), ER-membrane associated Ca2+ sensors. In humans, STIM1 and STIM2 share 65% sequence similarity but differentially regulate SOCE. Biophysical studies on the luminal Ca2+-binding region suggests that STIM2 EF-SAM is more stable than STIM1. The NMR structure of Ca2+-loaded STIM2 EF-SAM determined in this work suggests a more stable SAM and a tighter EF-hand and SAM interaction in STIM2 may be account for its higher stability. Chimeric swapping of the EF-hand and SAM domains generates an unstable ES211. Introducing ES211 into cherryFP-STIM1 shows constitutive puncta which activate SOCE independent of ER depletion. The current work demonstrates that the instability of the EF-SAM plays an important role in regulating SOCE initiation.
3

Structural and Biophysical Studies of the Role of Stromal Interaction Molecules STIM1 and STIM2 in Initiating Store-operated Calcium Entry

Zheng, Le 29 July 2010 (has links)
Store-operated calcium entry (SOCE) is the major Ca2+ entry pathway in most non-excitable cells maintaining prolonged elevation of cytosolic Ca2+ levels required for gene transcription. SOCE is activated by the loss of endoplasmic reticulum (ER) Ca2+ through stromal interaction molecules (STIM), ER-membrane associated Ca2+ sensors. In humans, STIM1 and STIM2 share 65% sequence similarity but differentially regulate SOCE. Biophysical studies on the luminal Ca2+-binding region suggests that STIM2 EF-SAM is more stable than STIM1. The NMR structure of Ca2+-loaded STIM2 EF-SAM determined in this work suggests a more stable SAM and a tighter EF-hand and SAM interaction in STIM2 may be account for its higher stability. Chimeric swapping of the EF-hand and SAM domains generates an unstable ES211. Introducing ES211 into cherryFP-STIM1 shows constitutive puncta which activate SOCE independent of ER depletion. The current work demonstrates that the instability of the EF-SAM plays an important role in regulating SOCE initiation.
4

Dissecting the mechanism of STIM coupling to Orai

Deng, Xiaoxiang January 2011 (has links)
Store-operated Ca2+ entry (SOCE) triggered by the depletion of endoplasmic reticulum (ER) luminal Ca2+ stores is a major Ca2+ entry pathway in non-excitable cells and is essential in physiological Ca2+ signaling and homeostasis. STIM proteins are sensors of ER luminal Ca2+, which translocate to ER-plasma membrane (PM) junctional regions to activate the family of Orai channels mediating Ca2+ entry. This study is focused on dissecting the mechanism of STIM interacting with Orai. A powerful modifier of SOCE, 2-aminoethoxydiphenyl borate (2-APB) is utilized. First, the action of 2-APB on the mammalian Orai homologues are characterized using the DT40 STIM knockout cells. 50 ìM 2-APB directly activates Orai3 but not Orai1 or Orai2. Second, while it stimulates the STIM2-mediated constitutive Ca2+ entry through Orai, 2-APB also induces the cytosolic STIM C-terminus fragments to translocate to the PM and activate Orai1. These data reveal 50 ìM 2-APB enhances STIM-Orai coupling. Further, this enhanced binding of STIM and Orai leads to a conformational change within the STIM-Orai complex, which is possibly the underlying mechanism for the 50 ìM 2-APB inhibitory effect on SOCE. Finally, six residues (344-349) at the N-terminus of the STIM-Orai activation region (SOAR) prove to be critical for this inhibitory action. These same six amino acid region also constitutes an ancillary Orai binding site within SOAR, in addition to the main polybasic region. The deletion of this ancillary site abolishes the ability of SOAR to bind to and activate Orai1, but can be compensated for by the STIM-Orai binding enhancing effect of 50 ìM 2-APB. The majority of STIM1 is located on the ER membrane, while a small proportion of STIM1 is on the PM. Using an extracellularly applied STIM1 antibody, the PM STIM1 can be aggregated to exert an influence on the ER STIM1. Although the PM STIM1 is not obligatory for STIM1-mediated Orai activation, it nevertheless may have a functional presence in the PM. Lastly, a regulatory link between voltage-gated Ca2+ channels (Cav channels) and the STIM proteins is established. After activation by store depletion, STIM strongly suppresses the Cav1.2 channels. There is a biochemical interaction between STIM1 and the Cav1.2 pore subunit á1C. This inhibitory effect is independent of Orai1 activation. Hence, STIM1 interacts with and reciprocally controls two major Ca2+ channels. / Biochemistry
5

Store-Operated Calcium Channels in the Function of Intracardiac Neurons

Bonds, Timetria 01 January 2012 (has links)
Proper autonomic regulation of mammalian cardiac function is dependent upon very complex and precise communication among the intracardiac ganglia and individual neurons within the ganglia. An array of neuromodulators is found within the ganglia that direct neuronal activity by modulating the movement of calcium. The current study determines that opioidergic agonists, which have been found to contribute to severe cardiac disease states and intracellular calcium mobilization, are also responsible for changes in the function of the intracardiac neuron via their effects on store-operated calcium channels (SOCs). Previous studies suggest that phosphorylation plays a role in SOC regulation. Using Fura-2 calcium fluorometry, we determined that protein kinase A (PKA), protein kinase C (PKC), and cyclic adenosine monophosphate (cAMP) had no effect on store-operated calcium entry in the presence of antagonists, phorbol 12, 13 dibutyrate (PDBu), forskolin, and 8-Br cAMP, respectively. We also found pharmacologically that using both electrophysiology and calcium imaging that μ-opioid agonists, met-enkephalin (ME) and endomorphin (EM) depress SOC activity in intracardiac neurons. Arachidonic acid (AA), which has been found to depress SOC function in rat liver cells and μ-opioid receptor activation (MOR), blocked both store-operated calcium entry (SOCE) and the calcium release-activated current (ICRAC) significantly. Contrastingly, AA metabolites, prostaglandin E2)(PGE2) and prostaglandin D2 (PGD2), do not significantly influence SOCE which suggests that the effects of AA may be direct. The block elicited by EM was partially reversed by pertussis toxin (PTX), indicative of activation of a PTX-sensitive G-protein following MOR activation. Similarly, PLA2 inhibitors, OBAA and AACOCF3, decreased the percent block of SOCE due to opioid agonist-induced inhibition. Using the perforated-patch method of I-clamp electrophysiology, we demonstrated that gadolinium, at low micromolar concentrations, reversibly reduced action potential firing. Importantly, these results suggest that SOCs may influence action potential firing in mammalian intracardiac neurons. Similarly, AA and EM depressed action potential firing. Taken together, these experiments suggest that a pathway involving EM and AA influences repetitive firing through SOC inhibition. The importance of SOCs in the maintenance of action potential firing and more specifically, the expression and biophysical functionality of the individual pore-forming subunits (Orai1, 2, and 3) in any neuronal cell type has previously not been explored. Quantitative RT-PCR along with I-clamp electrophysiology revealed that Orai3 was exclusive to repetitively firing neurons. As a result, we hypothesize that robust Ca2+-dependent fast inactivation, also associated Orai3, is a factor in the maintenance of repetitive action potential firing. Using Fura-2 calcium fluorometry and patch-clamp electrophysiology, we determined pharmacologically that μ-opioid receptor activation precedes an intracellular cascade that is dependent on a PTX-sensitive G-protein and AA but independent of prostaglandin and protein kinase activity. Finally, we used RT-PCR to determine the Orai subunits expressed in the intracardiac neurons and their influence on neuronal firing patterns. This study is the first to determine the role expressed subunits has in the maintenance of the electrical activity of the neuron.
6

Expressing human Orai3 in insect cells for pharmacological studies

Bennett, Orville R. 21 March 2012 (has links)
No description available.
7

Characterisation of store-operated calcium entry in a vascular endothelial cell line and impact on the production of nitric oxide

Batchelor, Helen R. January 2014 (has links)
Store-operated calcium entry (SOCE) is a principal mechanism for extracellular calcium entry in non-excitable cell types, and is primarily facilitated by the calcium- release activated calcium (CRAC) channel; itself comprised of the pore-forming Orai-1 and calcium-sensing Stromal interaction molecule (STIM)-1 proteins. Depletion of endoplasmic reticulum (ER) calcium stores initiates STIM-1 translocation to defined ER-plasma membrane puncta, and subsequent Orai-STIM interaction and opening of Orai. The importance of this mechanism in calcium signalling in diverse tissue types is becoming increasingly clear. The vascular endothelium is a dynamic tissue, involved in the maintenance of vascular homeostasis and haemostasis. Many endothelium-derived bioactive agents, such as endothelin-1, prostaglandins, and the potent vasodilator nitric oxide (NO), are known to be produced via calcium- dependent mechanisms. However, the role of the CRAC channel in the vascular endothelium is poorly defined with little known about downstream targets of calcium influx through CRAC channels. The dysregulation of NO production by endothelial nitric oxide synthase (eNOS) is a major contributory factor in many vascular disease states, yet the calcium channel responsible for eNOS activation has yet to be identified. Within this thesis, I establish the endothelial cell line sEnd.1 as a new model system for studying CRAC channel signalling in the vascular endothelium, defining sEnd.1 SOCE as being CRAC channel-dependent. Inhibition of CRAC channels with an array of inhibitors, and knock-down of STIM-1, both reduced ATP- and TG-induced SOCE. The sEnd.1 model system was subsequently used to identify calcium entry through the CRAC channel as the elusive activation mechanism for eNOS. Through real-time imaging with the fluorescent NO dye DAF-2-DA, we established that NO production is non-linear, with a slow initial increase preceding a faster NO production phase. These kinetics, with a characteristic delay before fast production have, to our knowledge, not previously been reported. The time taken to reach the fast phase of NO production could be manipulated through changes in both local and bulk calcium rises, which indicated roles for both elements of calcium signalling in eNOS activation. eNOS regulation by calcium is complex, occurring not only through direct binding of calcium-calmodulin, but additionally through changing post-translational modifications, which in turn regulate the calcium-dependency of eNOS, such as phosphorylation of Ser1177. We propose that the delay in fast production of NO is due to the time taken to alter eNOS post-translational modifications, which thus remove inhibition on eNOS. Activation of CRAC channels increased phosphorylation of residue Ser1177 via calcium-calmodulin kinase II (CaMKII), with a similar time course to that required to reach the fast phase of NO production. Inhibition of CaMKII increased the time taken to reach fast activation. In conclusion this thesis presents a new model system for investigation of CRAC channel signalling in the endothelium. Furthermore, we clearly identify a critical endothelial pathway as being regulated by CRAC channels, by demonstrating the production of NO in response to both ATP and TG, which stimulate calcium entry through CRAC channels.
8

Regulation of the Early Growth Response Protein-1 in vascular smooth muscle cells

Simo Cheyou, Estelle Rolande 12 1900 (has links)
Une hyperactivation de la prolifération des cellules musculaires lisses vasculaires (CMLV) contribue à la pathogenèse des maladies des vaisseaux. Des travaux antérieurs suggèrent que l’augmentation de l'adénosine monophosphate cyclique (AMPc) inhibe la prolifération des CMLV. Provoquer une augmentation d’AMPc préviendrait aussi certaines maladies vasculaires qui sont associées à des altérations dans sa signalisation impliquant l'activité de la protéine kinase A (PKA). Des études ont démontré la contribution du facteur de transcription « early growth response protein-1» (Egr-1) dans la pathogenèse des maladies vasculaires et une surexpression d’Egr-1 a été rapportée dans des modèles d'athérosclérose et d'hyperplasie intimale. Divers agents vasoactifs contrôlent l'expression d’Egr-1 suivant des mécanismes qui ont fait l’objet de plusieurs études mais demeurent incomplètement élucidés. L'angiotensine-II (Ang-II) est l'un des principaux peptides vasoactifs impliqués dans la pathogenèse des maladies vasculaires. Une des voies de signalisation induite par l’Ang-II implique l’augmentation du calcium (Ca2+) intracellulaire. Celle-ci se produit par l’activation de l'entrée de calcium opérée par la relâche des réserves (SOCE) de Ca2+ réticulaire suite à l’activation du récepteur à l’inositol-3-phosphate (IP3R) et le recrutement ultérieur du complexe conducteur formé par la molécule d'interaction stromale 1 (STIM-1) et le canal Orai-1. Bien qu’il ait déjà été démontré que l’expression de l'Egr-1 est régulée par la signalisation calcique en réponse à plusieurs stimuli, l'implication du complexe STIM-1/Orai-1 dans l'expression d'Egr-1 dans la CMLV n’a jamais été étudiée. De même, la question de savoir si la signalisation induite par l'Ang-II conduisant à l'expression d'Egr-1 est modulée par l'AMPc n’a jamais été explorée. Par conséquent, les travaux menés dans cette thèse ont consisté à examiner le rôle de la signalisation du Ca2+ dans l'expression d'Egr-1 induite par l’Ang-II dans la CMLV avec une attention particulière portée sur le rôle joué par STIM-1 et Orai-1. En outre, nous avons examiné l'effet de l’augmentation de l’AMPc sur l'expression d'Egr-1 induite par l’Ang-II et étudié les voies de signalisation associées. Nos données montrent que l’inhibition du récepteur IP3R et du SOCE par le 2-aminoéthoxydiphénylborate atténue la libération de Ca2+ induite par l’Ang-II et ceci s’accompagne d’une baisse des niveaux d’expression de protéine et d’ARN messager de l’Egr-1. La stimulation de l’expression de l'Egr-1 a également été supprimée à la suite du blocage de la calmoduline et de la protéine kinase CaMKII. De plus, le blocage par interférence d’ARN de l’expression de STIM-1 et Orai-1 a atténué l'expression d'Egr-1 induite par l’Ang-II ainsi que la phosphorylation des protéines ERK et CREB. Par ailleurs, l'isoproterenol (ISO) et la forskoline (FSK), deux activateurs de l'adénylate cyclase ont atténué de manière dose-dépendante l'expression d'Egr-1 induite par l’Ang-II. Des réponses similaires ont été observées en utilisant des analogues non spécifique (dibutyryl-cAMP) et PKA-spécifique (Benzoyl-cAMP) de l’AMPc, ainsi qu'un inhibiteur à large spectre de l'activité phosphodiesterase intracellualaire (isobutylméthylxanthine). L'inhibition de l'expression d'Egr-1 induite par l’Ang-II s’accompagne d'une augmentation de l’activité de la PKA mesurée par la phosphorylation de la « phosphoprotéine activée par les vasodilatateurs (VASP) », et d’une diminution concomitante de la phosphorylation de la protéine ERK. Le blocage pharmacologique de la PKA a réduit la phosphorylation de VASP et restauré la phosphorylation de la protéine ERK ainsi que l'expression d'Egr-1 en présence de l’Ang-II. En résumé, nos données démontrent que la voie STIM-1/Orai-1 /Ca2+ médie l'expression de l'Egr-1 induite par l'Ang-II dans la CMLV et suggèrent que la suppression de la réponse à l’Ang-II menant à l’expression de l'Egr-1 peut expliquer les effets vasoprotecteurs de l’AMPc. En outre, ces travaux montrent que les mécanismes moléculaires de régulation de l’expression d’Egr-1 en réponse aux signaux externes culminent vers la modulation des cascades de signalisation en aval de la protéine ERK dans les CMLV. / Aberrant vascular smooth muscle cell (VSMC) proliferative responses contribute to the development of neointimal lesions. Cyclic adenosine monophosphate (cAMP) is believed to inhibit VSMC proliferation, and vascular diseases are associated with impairments in cAMP-induced signalling responses involving protein kinase A (PKA) signaling. An enhanced expression of the early growth response protein-1 (Egr-1), a zinc finger transcription factor, has been reported in models of vascular diseases and, a crucial role of Egr-1 in regulating the expression of genes implicated in neointimal formation leading to atherogenesis has been suggested. Various vasoactive factors have been shown to modulate Egr-1 expression in VSMC via mechanisms which remain to be completely understood. Angiotensin-II (Ang-II) is one of the key vasoactive peptides implicated in the pathogenesis of vascular diseases. Ang-II elevates intracellular calcium (Ca2+) through activation of voltage-gated calcium channels as well as store-operated calcium channels. The store-operated calcium entry (SOCE) involves an inositol-3-phosphate receptor (IP3R)-coupled depletion of endoplasmic reticular Ca2+ and a subsequent activation of the stromal interaction molecule 1 (STIM-1) /Orai-1 complex. Although Egr-1 has been demonstrated to be upregulated in a Ca2+-dependent fashion in response to several stimuli, the involvement of STIM-1/Orai-1-dependent signaling in Egr-1 expression in VSMC has never been addressed. Besides, whether Ang-II-induced signaling leading to Egr-1 expression is modulated by cAMP-dependent signaling pathway remains unexplored. Therefore, in the present studies, we have examined the role of Ca2+ signaling in Ang-II-induced Egr-1 expression in VSMC and investigated the contribution of STIM-1 or Orai-1. Additionnaly, we have examined the effect of cAMP on Ang-II-induced expression of Egr-1 and have investigated the associated signalling pathways. Pharmacological blockade of IP3R and SOCE by 2-aminoethoxydiphenylborate (2-APB) decreased Ang-II-induced Ca2+ release and attenuated Ang-II-induced enhanced expression of Egr-1 protein and mRNA levels. Egr-1 upregulation was also suppressed following blockade of calmodulin and CaMKII. Furthermore, RNA interference-mediated depletion of STIM-1 or Orai-1 attenuated Ang-II-induced Egr-1 expression, as well as Ang-II-induced phosphorylation of ERK1/2 and CREB. Moreover, isoproterenol (ISO) and forskolin (FSK), two respective receptor and non-receptor activators of adenylate cyclase, attenuated Ang-II-induced Egr-1 expression in a dose-dependent fashion. Similar responses were observed using non-specific (dibutyryl-cAMP) and PKA-specific (Benzoyl-cAMP) analogs of cAMP, as well as a broad spectrum inhibitor of intracellular phosphodiesterase activity (isobutylmethylxanthine). The inhibition of Ang-II-induced Egr-1 expression was accompanied by an increase in serine 157 phosphorylation of the vasodilator-activated phosphoprotein (VASP), a marker of PKA activity, and this was associated with a concomitant decrease in ERK phosphorylation. Pharmacological blockade of PKA using H89 decreased VASP phosphorylation, restored Ang-II-induced ERK phosphorylation and abolished ISO- and FSK-mediated inhibition of Ang-II-induced Egr-1 expression. In summary, our data demonstrate that STIM-1/Orai-1/Ca2+-dependent signaling pathways mediate Ang-II-induced Egr-1 expression in A-10 VSMC and suggest that PKA-mediated suppression of Ang-II-induced Egr-1 expression and phosphorylation of ERK may be among the mechanisms by which cAMP exerts its vasoprotective effects. In addition, our data supports the notion that stimuli-induced regulation of Egr-1 expression involves the participation of signaling cascades downstream of ERK in VSMC.
9

Cellular mechanisms underlying the regulation of calcium signaling in brain pericytes

Phillips, Braxton 06 1900 (has links)
Les cellules murales du cerveau sont un groupe de cellules neurovasculaires qui présentent une hétérogénéité moléculaire, morphologique et fonctionnelle exceptionnelle. Celles en contact avec les plus petis vaisseaux du cerveau, les péricytes du lit capillaire moyen sont connues pour être essentielles à l'homéostasie cérébrale, bien que leur capacité contractile ait longtemps été débattue. Cependant, nombre de leurs propriétés physiologiques, telles que leurs mécanismes de signalisation calcique, n'ont pas encore été élucidées. Cette thèse vise donc à identifier les mécanismes cellulaires de la signalisation calcique des péricytes des capillaires cérébraux. Dans le chapitre 2, nous utilisons la pharmacologie et l'imagerie des péricytes cérébraux exprimant l'indicateur de calcium GCaMP6f (provenant de souris transgéniques PDGFRβ-Cre::GCaMP6f) pour découvrir ces mécanismes. Contrairement aux péricytes engainants dont la signalisation du calcique dépend des canaux calcique voltage-dépendants, nous constatons que les signaux calcique des péricytes capillaire moyen sont indépendants des canaux calcique voltage-dépendants. Au contraire, nous constatons que les signaux calciques transitoires des pericytes du lit capillaire moyen sont inhibés par l'élimination du Ca2+ extracellulaire, l'inhibition des canaux Orai opérés par les réserves, le blocage du remplissage des réserves du réticulum endoplasmique, ainsi que l'inhibition des récepteurs de la ryanodine (RyRs) et des récepteurs de l'inositol trisphosphate (IP3Rs). Nous constatons également que l'entrée de Ca2+ opérée par les réserves peut être induite par la déplétion des réserves du réticulum endoplasmique et inhibée par les bloqueurs d'Orai dans les pericytes du lit capillaire moyen, et que l'influx basal de Ca2+ est largement dépendant de la déplétion des réserves. Enfin, nous montrons que l'entrée de Ca2+ opérée par les réserves d'Orai amplifie les élévations de Ca2+ cytosolique en réponse au vasoconstricteur endothéline-1. Nous concluons que la signalisation calcique dans les pericytes du lit capillaire moyen, qu'elle soit spontanée ou induite de façon agoniste, est régulée par le couplage entre la libération des réserves du réticulum endoplasmique et les voies d'influx opérées par les réserves. / Brain mural cells are a grouping of neurovascular cells that display exceptional molecular, morphological, and functional heterogeneity. Mid-capillary pericytes, the mural cells which contact the smallest vessels of the brain, are known to be critical to brain homeostasis, and their contractile ability has long been debated. However, many of their physiological properties, such as their Ca2+ signaling mechanisms, have not been elucidated. This thesis aims to uncover the cellular mechanisms of brain mid-capillary pericyte Ca2+ signaling. In chapter 2, we harness pharmacology and imaging of brain pericytes expressing the calcium indicator GCaMP6f (from transgenic PDGFRβ-Cre::GCaMP6f mice) to uncover these mechanisms. In contrast to ensheathing pericytes whose Ca2+ signaling is dependent on voltage-gated Ca2+ channels (VGCCs), we find that mid-capillary pericyte Ca2+ signals are independent of VGCCs. Instead, we find that mid-capillary pericyte Ca2+ transients are inhibited by removal of extracellular Ca2+, inhibition of store-operated Orai channels, blockade of endoplasmic reticulum store filling, as well as inhibition of ryanodine receptors (RyRs) and inositol triphosphate receptors (IP3Rs). We further find that store-operated Ca2+ entry can be induced by endoplasmic reticulum store depletion and inhibited by Orai blockers in mid-capillary pericytes, and that basal Ca2+ influx is largely dependent on store depletion. Finally, we show that Orai store-operated Ca2+ entry amplifies cytosolic Ca2+ elevations in response to the vasoconstrictor endothelin-1. We conclude that both spontaneous and Gq-coupled protein receptor agonist-induced Ca2+ signaling in mid-capillary pericytes is regulated by coupling between endoplasmic reticulum store release and store-operated influx pathways.
10

Caractérisation des canaux calciques dans les polynucléaires neutrophiles : rôle dans la phagocytose et la production des radicaux libres oxygénés

Djillani, Alaeddine 26 September 2013 (has links) (PDF)
Les polynucléaires neutrophiles représentent 50-70% des leucocytes sanguins et possèdent un rôle majeur dans la défense de l'organisme contre les pathogènes. Le Ca2+ est un second messager qui joue un rôle primordial dans le chimiotactisme, la phagocytose, la dégranulation et la production de formes réactives de l'oxygène (FRO) afin de neutraliser l'agent pathogène. Dans ces cellules, l'influx calcique de type SOCE est essentiel pour l'homéostasie calcique. Il est peu étudié en raison du manque d'outils pharmacologiques spécifiques d'où l'importance dans un premier temps de chercher de nouvelles molécules. Les cellules T Jurkat dont le SOCE est largement caractérisé servent de modèle pour la caractérisation initiale de ces molécules. Le 2-APB est parmi les molécules les plus largement utilisées dans la caractérisation du SOCE en raison de sa double activité sur le SOCE avec une potentialisation à [1-10 μM] et une inhibition à [> 20 μM]. En revanche, ce produit manque de spécificité et agit sur d'autres cibles cellulaires comme les récepteurs à l'inositol (1,4,5)-trisphosphate (InsP3Rs). La 1ère étape est de sélectionner à partir d'analogues commerciaux du 2-APB (Methoxy-APB, Dimethoxy-APB, Cyclic-APB, Benzothienyl-APB, Thienyl-APB et MDEB), des composés plus spécifiques et également plus efficaces que la molécule mère. Deux molécules se sont distinguées : le MDEB comme uniquement potentialisant du SOCE et le Benzothienyl-APB comme un puissant inhibiteur. En revanche, tous les analogues du 2-APB inhibent les InsP3Rs à l'exception du MDEB qui semble plus spécifique du SOCE. L'effet du MDEB sur le courant calcique, ICRAC, a été étudié grâce à la technique du patch-clamp. Il augmente d'environ 4 fois l'amplitude de ICRAC par rapport à celle enregistrée dans les cellules contrôle. Par ailleurs, le MDEB ralentie l'inactivation rapide de ICRAC due au Ca2+. Sur le plan physiologique, le MDEB à des concentrations croissantes inhibe la synthèse de l'IL-2 dans les cellules Jurkat stimulées et ceci malgré son effet potentialisant du SOCE. Cette activité est liée à son effet pro-apoptotique dans les cellules Jurkat stimulées. Le MDEB et le Benzothienyl-APB caractérisés dans la 1ère partie nous ont servi d'outils potentiels afin d'étudier le SOCE des cellules PLB-985 différenciées en cellules proches de neutrophiles. Le SOCE a été induit soit par un traitement des cellules avec la thapsigargine (Tg) soit de manière physiologique avec les peptides fMLF et le WKYMVm deux chimioattractants, ligands des récepteurs aux peptides formylés FPR et FPRL1 respectivement. En plus, le SOCE induit par la Tg est modulable par le 2-APB, potentialisé par le MDEB et inhibé par le Benzothienyl-APB. La phagocytose des levures par les cellules PLB-985 différenciées ainsi que la production de FRO intraphagosomales ont été inhibées par le MDEB et le Benzothienyl-APB. Les FRO extracellulaires ont été également inhibées par Benzothienyl-APB en revanche à cause de la forte interférence du MDEB avec la technique de mesure nous n'avons pas pu étudier ses activités. En conclusion, le MDEB et le Benzothienyl-APB sont de nouveaux outils pharmacologiques potentialisant ou inhibant le SOCE des leucocytes, qui nous permettront dans l'avenir une meilleure compréhension de l'entrée calcique et ses rôles dans ces cellules.

Page generated in 0.4254 seconds