• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 33
  • 8
  • 7
  • 5
  • 4
  • 3
  • 2
  • 1
  • 1
  • 1
  • Tagged with
  • 83
  • 27
  • 14
  • 12
  • 11
  • 11
  • 10
  • 10
  • 9
  • 9
  • 8
  • 8
  • 8
  • 8
  • 8
  • 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.
61

Transcriptional control of muscle cell excitation-contraction coupling:the role of activity and mitochondrial function

Hänninen, S. L. (Sandra Lynn) 04 June 2019 (has links)
Abstract Cardiac and skeletal muscle cell contraction is a result of excitation-contraction coupling (ECC), where an electrical signal leads to a rise in intracellular calcium levels and contraction. This process is carefully regulated to meet physiological demand and heavily dependent on an adequate energy supply. Disturbed ECC can have severe consequences on muscle cell function and underlies many cardiac and skeletal muscle pathologies. Cell stress, changing intracellular Ca2+ concentrations, and calcium signal dynamics can all play a role in the transcriptional regulation of genes involved in myocyte Ca2+-handling. In this thesis project, the transcriptional control of ECC was studied in skeletal and cardiac myocytes. Skeletal myocyte calsequestrin (CASQ1) was downregulated in a mouse model of mitochondrial myopathy and it contributed to the decreased SR Ca2+ load and impaired Ca2+ handling in Tfam-/- skeletal myocytes. In cultured neonatal cardiomyocytes, mitochondrial uncoupler FCCP-induced mitochondrial dysfunction led to downregulation of cardiac calsequestrin (CASQ2) and similarly impaired Ca2+ handling. Whereas there was no increase in reactive oxygen species (ROS) levels in Tfam-/- myocytes, cultured cells exposed to FCCP did display increased ROS, an effect that was counteracted by coexposure with the ROS scavenger (NAC). NAC attenuated FCCP-induced CASQ2 downregulation and restored Ca2+ handling. Therefore, mitochondrial dysfunction led to CASQ1/2 downregulation and impaired Ca2+ handling in these two cell types, but by different mechanisms. This project also looked at the role of Ca2+ dynamics on the transcriptional regulation of Ca2+ handling genes. Increased intracellular Ca2+ levels and β-adrenergic stimulation of cardiomyocytes activate Ca2+-calmodulin kinase II (CaMKII) and can trigger hypertrophic remodeling. It was found that CaMKII downregulated expression of the L-type Ca2+ channel α1c-subunit (Cacna1c) in cultured cardiomyocytes. Analysis of the Cacna1c promoter revealed that the transcriptional repressor DREAM bound to a putative downstream regulatory element. The results shed light on the complex interplay between muscle cell energetics and transcriptional regulation of SR Ca2+ handling proteins. A unique pathway for Cacna1c transcriptional regulation by CaMKII and DREAM was also described. / Tiivistelmä Sydän- ja luustolihassolujen supistuminen on seurausta ärsytys-supistuskytkennästä (ECC), jossa sähköinen ärsytys kohottaa solunsisäistä kalsiumpitoisuutta ja aiheuttaa supistuksen. Tätä säädellään tarkasti fysiologisen tarpeen mukaan, ja se riippuu riittävästä energian saannista. Häiriintynyt ECC voi aiheuttaa vakavia seurauksia lihassolujen toiminnalle, ja se on mukana monien sydän- ja luustolihasten sairauksien synnyssä. Tässä tutkimuksessa ECC:n transkriptionaalista säätelyä tutkittiin luustolihasten ja sydämen lihassoluissa. Luustolihassolujen kalsekvestriinin (CASQ1) väheneminen pienensi SR:n Ca2+-määrää mitokondrioiden myopatian hiirimallissa ja heikensi Ca2+-tasapainon ylläpitoa Tfam-/--luustolihassoluissa. Viljellyissä vastasyntyneiden kammio-sydänlihassoluissa mitokondrio-irtikytkijän FCCP:n aiheuttama mitokondrioiden toimintahäiriö johti sydämen kalsekvestriinin (CASQ2) vähenemiseen ja heikensi samalla tavalla Ca2+-tasapainon ylläpitoa. Vaikka Tfam-/--myosyyteissä reaktiivisten happilajien (ROS) tasot eivät olleet koholla, FCCP:lle altistetuissa viljellyissä soluissa ROS kuitenkin lisääntyi. Vaikutusta esti ROS-puhdistaja NAC, joka heikensi FCCP:n aiheuttamaa CASQ2:n laskua ja palautti Ca2+-säätelyn normaaliksi. Mitokondrioiden toimintahäiriö siis johti CASQ1/2:n vähenemiseen ja Ca2+-säätelyn heikentymiseen molemmissa solutyypeissä, mutta eri mekanismeilla. Tässä tutkimuksessa tarkasteltiin myös Ca2+-dynamiikan osuutta Ca2+-tasapainoon osallistuvien geenien transkription säätelyssä. Lisääntynyt solunsisäinen Ca2+-taso ja sydänlihassolujen β-adrenerginen stimulointi aktivoivat Ca2+-kalmoduliinikinaasi II:n (CaMKII), ja ne voivat laukaista sydämen hypertrofisen uudelleenmuovautumisen. Havaittiin, että CaMKII vähensi L-tyypin Ca2+-kanavan a1c-alayksikön (Cacna1c) ilmentymistä viljellyissä sydänlihassoluissa. Promoottorianalyysi osoitti tämän johtuvan transkription repressorin DREAM:n sitoutumisesta oletettuun DRE:hen (alavirrassa sijaitseva säätelyelementti). Nämä tulokset tuovat uutta tietoa lihassolujen energiatalouden ja SR:n Ca2+:n vaikuttavien proteiinien transkription säätelyn vuorovaikutuksesta. Lisäksi havaittiin ainutlaatuinen Cacna1c-transkription säätelyn reitti, johon osallistuvat CaMKII ja DREAM.
62

Rôle de PGC-1α dans le système cardiovasculaire : Recherche d'activateurs cœur-spécifiques et étude de ses mécnismes de régulation dans le muscle lisse aortique

Ruiz, Matthieu 14 September 2012 (has links) (PDF)
L'insuffisance cardiaque (IC) reste la cause majeure de morbimortalité dans les pays industrialisés justifiant ainsi la recherche de traitements plus ciblés. Caractérisée par des désordres métaboliques importants qui impliquent notamment une dysfonction mitochondriale, le métabolisme énergétique apparait comme une composante majeure du développement de l'IC. Ces dernières années, le co-activateur transcriptionnel PGC-1α a été proposé comme un acteur central du contrôle de la fonction mitochondriale et constitue ainsi une cible thérapeutique d'intérêt. Ainsi, l'objectif principal de ce travail est de développer un test cellulaire robotisé permettant la recherche d'activateurs de PGC-1α dans un contexte cardiaque.La mise en place de ce test cellulaire de criblage dans des cellules H9c2 différenciées en cellules pseudo-cardiaques a permis l'identification de trois familles majeures : les hormones stéroïdiennes, les vitamines B et les acides gras, capables d'activer l'expression de PGC-1α et par ce biais d'induire une biogenèse mitochondriale ainsi qu'une augmentation de la respiration mitochondriale. La validation de ces effets dans des cardiomyocytes de rat adulte a permis d'une part de valider la pertinence du test et du choix du modèle cellulaire et d'autre part de vérifier qu'une induction de l'expression de PGC-1α se répercute bien sur la cascade transcriptionnelle de la biogenèse mitochondriale. Ce test constitue donc un atout majeur dans le recherche de nouveaux activateurs de PGC-1α pour mieux comprendre ses mécanismes de régulation dans le cœur, mais offre aussi des perspectives intéressantes pour la recherche de composés pharmacologiques à visée thérapeutique.Par ailleurs, peu de connaissances sont disponibles dans la littérature concernant le contrôle de la biogenèse mitochondriale dans le muscle lisse vasculaire et plus particulièrement dans l'hypertension artérielle. Ainsi, la deuxième partie de ce travail a été de caractériser la biogenèse mitochondriale dans un contexte d'hypertension. A travers l'utilisation d'un modèle expérimental d'hypertension et après confirmation dans des cellules musculaires lisses en culture, nous avons montré une induction importante de la biogenèse mitochondriale dans l'hypertension par un mécanisme stress oxydant-dépendant. De plus, cette induction est corrélée à une forte activation de la CaMKII, totalement bloquée par la présence d'un anti-oxydant : le resvératrol. Ces résultats suggèrent donc un contrôle de la biogenèse mitochondriale dépendante de la balance pro/anti-oxydante via l'activation de la CaMKII dans le muscle lisse vasculaire.
63

Study On The Molecular Basis Of Individual Variation In Spatial Memory In Rats

Gokcek Sarac, Cigdem 01 June 2012 (has links) (PDF)
Despite very extensive studies related to molecular processes underlying memory formation, still little known about the potential differences in the brain biochemistry between &ldquo / good&rdquo / and &ldquo / poor&rdquo / learners belonging to a random population of young animals. In the present study, an attempt was taken to correlate the individual variation in short- and long-term spatial memory in three different lines of young, healthy rats: inbred Wistar (W), outcrossed Wistar/Spraque Dawley (W/S) and pigmented Long-Evans rats, with hippocampal levels of selected enzymes known as &ldquo / memory molecules&rdquo / including neuronal (n), endothelial (e) and inducible (i) NOS, CaMKII&alpha / , PKA and ChAT. Additionally, in order to indirectly estimate the activity of CaMKII&alpha / and PKA, hippocampal levels of their phosphorylated forms (pCaMKII&alpha / and pPKA) were assessed. Rats were classified as &ldquo / good&rdquo / and &ldquo / poor&rdquo / learners on the basis of their performance in a partially baited 12-arm radial maze. The hippocampal protein levels were measured using Western Blot technique. In addition to individual variation in animals&rsquo / learning capacity, strain-depended differences have also been observed. Deficient performance recorded in inbred W rats compared to outcrossed W/S rats, and &ldquo / poor&rdquo / learners from both rat groups had predominantly related to the higher frequency of reference memory errors. The results of biochemical assays showed strain-depended differences in the NOS expression. The overall NOS levels were significantly higher in outcrossed W/S rats compared to inbred W rats. In both rat lines, the rate of learning positively correlated with hippocampal levels of nNOS and negatively correlated with iNOS levels. Hippocampal eNOS levels correlated negatively with animals&rsquo / performance but only in the W rats. These results suggested that all 3 NOS isoforms are implemented in the learning process playing, however, different roles in neural signaling. Experiments carried out on Long-Evans rats did not reveal a significant difference in the basal hippocampal levels of the CaMKII&alpha / , however, the level of the pCaMKII&alpha / , was significantly higher in &ldquo / good&rdquo / learners. Also, hippocampal levels of both PKA and pPKA, as well as that of ChAT were significantly higher in &ldquo / good&rdquo / as compared to &ldquo / poor&rdquo / learners. Taken together, the latter findings indicate that low hippocampal expression of PKA and ChAT as well as low CaMKII&alpha / or PKA activation may cause learning deficits in random population of young rats, and thus, these enzymes can be considered target molecules when looking for cognitive enhancers to treat memory deficits in young subjects.
64

Identification de nouveaux mécanismes régulateurs des pulsars calciques endothéliaux d’artères mésentériques de souris

Toussaint, Fanny 06 1900 (has links)
No description available.
65

Des Polycystines au centrosome, une enzyme clef : la calcium/calmoduline dependent kinase 2 / From polycystins to centrosomes, a key enzyme : the calcium/calmodulin dependant kinase 2

Ribe-Pinachyan, Emilie 16 December 2010 (has links)
La polykystose rénale autosomique dominante (ADPKD) est la maladie monogéniquehumaine la plus fréquente (prévalence 1/800). Les gènes responsables de cette maladie sont PKD1(codant pour PC1) ou PKD2 (codant pour PC2). La maladie évolue vers l’insuffisance rénale terminale.Aujourd’hui, seul un traitement symptomatique est proposé aux malades. Les mécanismes à l’originede l’ADPKD sont mal connus. Les modèles animaux permettent de mieux comprendre laphysiopathologie d’une maladie. Il n’existe pas de bon modèle de polykystose (même causemoléculaire, même mode de transmission, même signes cliniques). En utilisant la transgénose degrands fragments, nous avons créé un modèle de surexpression de PKD2 humain. Le transgène estsous le contrôle de son promoteur naturel humain. Cette souris exprime deux fois plus de PC2 queles sauvages. Elle ne présente que quelques microkystes mais une tubulopathie associant défaut deconcentration des urines et protéinurie tubulaire. La surexpression de PC2 inhibe l’expression degènes codant pour des protéines de la matrice extracellulaire. Le phénotype cellulaire de cesanimaux est remarquable : un tiers des cellules présentent un nombre élevé de centrosomes. Cephénotype cellulaire a été retrouvé chez des souris sous exprimant Pkd2 et chez des souris sousexprimant Pkd1. Ce caractère multicentrosomique est corrigé en incubant les cellules avec uninhibiteur de CaMKII ou en croisant nos souris transgéniques avec une souris KO de Camk2. Nousavons réussi à lier CaMKII, la duplication des centrosomes et les polycystines, in vitro et in vivo. Ceciamène un éclairage nouveau sur la duplication du centrosome et la physiopathologie de l’ADPKD. / Autosomal Dominant Polycystic Kidney Disease (ADPKD) is the most common monogenic human disease (prevalence 1/800). Genes responsible for this disease are PKD1 (encoding PC1) or PKD2 (encoding PC2). The disease progresses to end stage renal disease. Today, only symptomatic treatment is offered to patients. The mechanisms underlying the ADPKD are unknown. Animals models allow better understand the disease’s pathophysiology. There is no good model of ADPKD (same molecular cause, same clinical signs). We created a mice model of human PKD2 overexpression. The transgène is under the control of its human natural promoter. This mouse expresses PC2 twice as much as the wild. It shows only few microcysts but tubulopathy involving lack of urine concentration and tubular proteinuria. PC2 overexpression inhibits the expression of genes encoding proteins of the extracellular matrix. The cellular phenotype of these animals is special : one third of the cells have a high number of centrosomes. This cellular phenotype was found in Pkd2 Knockout mice and in Pkd1 knockout mice. This multicentrosomic character is corrected by incubating the cells with a CaMKII inhibitor or by crossing our transgenic mice with Camk2 knockout mice. We propose a link between CaMKII, Centrosome duplication and polycystin in vitro and in vivo. This brings a new light on centrosome duplication and pathophysiology of ADPKD.
66

Mechanisms of microRNA-mediated regulation of the rapid delayed rectifier potassium current, IKr, during sustained beta-adrenergic receptor stimulation

Enoch Amarh (17598138) 12 December 2023 (has links)
<p dir="ltr"><b>Background</b></p><p dir="ltr">Heart failure (HF) is a chronic clinical syndrome characterized by symptoms including breathlessness, fatigue, swelling of the ankles, and signs such as edema pulmonary crackles etc. During HF, pathogenic mechanisms including hemodynamic overload, ventricular remodeling, aberrant calcium handling, excessive neurohormonal stimulation contribute to the worsening and progression of the condition. Ventricular arrhythmias are the common cause of sudden cardiac death (SCD) in HF patients.</p><p dir="ltr">Hyperactivation of the sympathetic nervous system (SNS), a characteristic of HF, causes an increase in circulating catecholamines which becomes detrimental to-adrenergic receptors (-AR) leading to signaling dysfunction, and decrease in contractility and the ionotropic reserve. Expression of calcium/calmodulin-dependent protein kinase II (CaMKII), a downstream effector of-AR and a key regulator of calcium homeostasis, has been shown to be enhanced in HF. CaMKII-mediated mechanisms have been demonstrated to contribute to cardiac remodeling, arrhythmias by pathological regulation of ion channels, and contractile dysfunction.</p><p dir="ltr">The human ether-a-go-go related gene (hERG) encodes the pore-forming subunit of the voltage-gated potassium channel that conduct the rapid component of the delayed rectifier potassium current, <i>I</i><sub>Kr</sub>. The gating kinetics of <i>I</i><sub>Kr </sub>makes it a crucial determinant of the duration of the plateau phase of atrial and ventricular action potential (AP). Reduced <i>I</i><sub>Kr</sub> density due to loss-of-function mutations or pharmacological blockage of hERG channels precipitate arrhythmias. Downregulation of <i>I</i><sub>Kr</sub> density and protein have been reported in HF. Recent studies suggest that microRNAs (miRNAs) are involved in pathological downregulation of hERG.</p><p dir="ltr">miRNA are small non-coding RNAs of approximately 22 nucleotides in length that function as gene expression regulatory elements by repression translation. Aberrant miRNA expression has associated with cancer, cardiovascular, autoimmune, and inflammatory disorders.</p><p dir="ltr"><b>Objective</b></p><p dir="ltr">The overarching objective of this study is to investigate the mechanisms of CaMKII-mediated regulation of hERG function, including assessment of an interplay with miR-362-3p during sustained β-AR stimulation. In Specific Aim 1, the effect of CaMKII activation through sustained β-AR stimulation on hERG function and miR-362-3p expression will be assessed. The mechanism of miR-362-3p upregulation will be evaluated in Specific Aim 2, and in Specific Aim 3, the interactome of miR-362-3p and binding sites will be characterized and predicted, respectively.</p><p dir="ltr"><b>Methods</b></p><p dir="ltr">Whole-cell, voltage clamp electrophysiology experiments were performed in HEK 293 cells stably expressing hERG (hERG-HEK) and both hERG and wild-type CaMKIIδ<br>(hERG/CaMKII-HEK) following treatment with isoproterenol for 48 hours, and after transfection with miR-362-3p. The effect of CaMKII activation on miR-362-3p was assessed using real-time quantitative polymerase chain reaction (RT-qPCR). Total RNA was isolated 48 hours after isoproterenol treatment and the TaqMan assay was used to reverse transcribe and analyze miR-362-3p expression. Cells were transfected with cJun siRNA and precursor miR-362-3p to assess the role of cJun miR-362-3p upregulation during sustained β-AR stimulation with isoproterenol. The interactome of miR-362-3p was assessed in both cell lines using enhanced crosslinking immunoprecipitation (eCLIP) assay. miR-362-3p binding sites were predicted using RNAStructure Duplexfold after identification of miR-362-3p chimeric molecules from eCLIP experiment. Interaction analysis was performed using GeneMania in Cytoscape to identify genes that were potentially downregulated by miR-362-3p and been reported to interact with hERG.</p><p dir="ltr"><b>Results</b></p><p dir="ltr">In Specific Aim 1, the effect of sustained β-AR stimulation on hERG currents and endogenous miR-362-3p was assessed in hERG-HEK and hERG/CaMKII-HEK cells. Using whole-cell voltage clamp electrophysiology, we demonstrated that 48 hours treatment with 100 nM isoproterenol reduced hERG currents in hERG/CaMKII-HEK cells (p = 0.032) but had no effect on the voltage dependence of activation (p = 0.61) relative to control vehicle. Isoproterenol treatment for 48 hours, however, had no effect on hERG currents (p = 0.58) and the voltage dependence of activation (p = 0.99) in hERG-HEK cells. The effect of sustained isoproterenol treatment on miR-362-3p was also assessed using RT-qPCR. In hERG/CaMKII cells, 48 hours isoproterenol treatment increased miR-362-3p expression (2.3 folds; p = 0.038) relative to control vehicle. hERG/CaMKII-HEK cells were also treated with 500 nM KN-93 or its inactive analogue, KN-92, in an attempt to reverse CaMKII effect on miR-362-3p expression. Treatment with KN-93 decreased miR-362-3p expression (0.5-fold; p = 0.002) relative KN-92 treatment. Isoproterenol treatment had no effect on miR-362-3p expression in hERG-HEK cells (p = 0.38).</p><p dir="ltr">The regulatory mechanism of miR-362-3p expression was evaluated in Specific Aim 2. The role of an activator protein-1 (AP-1)-like sequence located at 98 base pairs upstream of miR-362-3p transcription start site was probed using siRNA inhibition of cJun, a central protein of the AP-1 complex, and deletion of the site sequence. The effect of exogenous miR-362-3p on hERG currents were first assessed. Precursor miR-362-3p decreased hERG currents (p = 0.003) compared to control plasmid. The effect of CaMKII overexpression was also assessed on exogenous miR-363-3p expression. Isoproterenol treatment in hERG/CaMKII-HEK cells transfected with precursor miR-362-3p increased mature miR-362-3p expression (0.029) compared to control vehicle treatment. Inhibition of cJun inhibition with cJun-specific siRNA decreased mature miR-362-3p expression (0.5-fold; p = 0.027) compared to scramble siRNA in hERG-HEK cells. In hERG-HEK cells transfected with mutated precursor miR-362-3p (AP-1-like site deleted), cJun inhibition with siRNA had no effect on miR-362-3p expression (p = 0.40).</p><p dir="ltr">The focus of Specific Aim 3 was to characterize the interactome of miR-362-3p as well as predict the miRNA response element (MRE) of its target mRNAs using enhanced crosslinking immunoprecipitation. A network analysis was also performed to identify miR-362-3p targets that have been reported to interact with hERG. Approximately 23% of miR-362-3p mRNA targets from the eCLIP assay have also been catalogued in miRNA database, TargetScanHuman, as miR-362-3p targets. miR-362-3p chimeric molecules with 853 unique targets, of which 75 were identified to interact with hERG through the network analysis. Four unique chimeric molecules between miR-362-3p and hERG mRNA were identified, but the interactions were non-canonical (located in the coding sequence of hERG and outside the seed region of miR-362-3p). Thirty five of the 75 miR-362-3p targets that were identified to interact had a chimeric read ≥ 3, a cutoff number indicating non-random chimeric formation. Using RNAStructure DuplexFold, miR-362-3p was predicted to form canonical binding with 12 of 35 mRNA targets. HSPA4, a heat shock protein involved in the maturation and trafficking of hERG, was identified in a canonical interaction (8-mer) with miR-362-3p.</p><p dir="ltr"><b>Conclusion</b>:</p><p dir="ltr">Sustained β-AR stimulation increases miR-362-3p expression and decreases hERG currents in CaMKII overexpressing cells. cJun mediates miR-362-3p upregulation by interacting with an AP-1-like sequence upstream of miR-362-3p transcription start site. Pathological regulation of <i>I</i><sub>Kr</sub> by CaMKII mediated by miR-362-3p during sustained-AR may contribute to increased risk of arrhythmias in states of increase catecholaminergic activity, such as HF.</p>
67

The Transient Receptor Potential Canonical 3 (TRPC3) Channel: Novel Role in Endothelial Cell Apoptosis and its Impact on Atherosclerosis

Ampem, Prince Tuffour 03 October 2017 (has links)
No description available.
68

Molecular physiology of ankyrin-G in the heart:Critical regulator of cardiac cellular excitability and architecture.

Makara, Michael A. 12 August 2016 (has links)
No description available.
69

Molecular Regulation of Inducible Nitric Oxide Synthase

Wang, Tingting 18 December 2012 (has links)
No description available.
70

Rôle de PGC-1α dans le système cardiovasculaire : recherche d’activateurs cœur-spécifiques et étude de ses mécanismes de régulation dans le muscle lisse aortique / PGC-1 alpha role in the cardiovascular system : search for inducers of its expression in heart and study of signaling pathways controlling PGC-1 alpha in aortic smooth muscle

Ruiz, Matthieu 14 September 2012 (has links)
L’insuffisance cardiaque (IC) reste la cause majeure de morbimortalité dans les pays industrialisés justifiant ainsi la recherche de traitements plus ciblés. Caractérisée par des désordres métaboliques importants qui impliquent notamment une dysfonction mitochondriale, le métabolisme énergétique apparait comme une composante majeure du développement de l’IC. Ces dernières années, le co-activateur transcriptionnel PGC-1α a été proposé comme un acteur central du contrôle de la fonction mitochondriale et constitue ainsi une cible thérapeutique d’intérêt. Ainsi, l’objectif principal de ce travail est de développer un test cellulaire robotisé permettant la recherche d’activateurs de PGC-1α dans un contexte cardiaque.La mise en place de ce test cellulaire de criblage dans des cellules H9c2 différenciées en cellules pseudo-cardiaques a permis l’identification de trois familles majeures : les hormones stéroïdiennes, les vitamines B et les acides gras, capables d’activer l’expression de PGC-1α et par ce biais d’induire une biogenèse mitochondriale ainsi qu’une augmentation de la respiration mitochondriale. La validation de ces effets dans des cardiomyocytes de rat adulte a permis d’une part de valider la pertinence du test et du choix du modèle cellulaire et d’autre part de vérifier qu’une induction de l’expression de PGC-1α se répercute bien sur la cascade transcriptionnelle de la biogenèse mitochondriale. Ce test constitue donc un atout majeur dans le recherche de nouveaux activateurs de PGC-1α pour mieux comprendre ses mécanismes de régulation dans le cœur, mais offre aussi des perspectives intéressantes pour la recherche de composés pharmacologiques à visée thérapeutique.Par ailleurs, peu de connaissances sont disponibles dans la littérature concernant le contrôle de la biogenèse mitochondriale dans le muscle lisse vasculaire et plus particulièrement dans l’hypertension artérielle. Ainsi, la deuxième partie de ce travail a été de caractériser la biogenèse mitochondriale dans un contexte d’hypertension. A travers l’utilisation d’un modèle expérimental d’hypertension et après confirmation dans des cellules musculaires lisses en culture, nous avons montré une induction importante de la biogenèse mitochondriale dans l’hypertension par un mécanisme stress oxydant-dépendant. De plus, cette induction est corrélée à une forte activation de la CaMKII, totalement bloquée par la présence d’un anti-oxydant : le resvératrol. Ces résultats suggèrent donc un contrôle de la biogenèse mitochondriale dépendante de la balance pro/anti-oxydante via l’activation de la CaMKII dans le muscle lisse vasculaire. / Heart failure (HF) is still the major cause of morbimortality in industrialized countries that justify the research of new treatments. Characterized in part by metabolic disorders including mitochondrial dysfunction, energetic metabolism appears as an essential component in HF development. These last years, PGC-1α has been proposed as a central actor of mitochondrial function control and thus as a therapeutic target of interest.The development of a cellular robotized assay in cardiac-like differentiated H9c2 cells allowed identification of three families: steroid hormones, B vitamins and fatty acids, able to induce the expression of PGC-1α and thus up-regulate mitochondrial biogenesis and mitochondrial respiration. The validation of these effects in adult rat cardiomyocytes lets in the one hand to validate the suitability of the assay and in the other hand to confirm that PGC-1α induction leads to mitochondrial biogenesis activation. Consequently, this assay constitutes a major asset to find new activators of PGC-1α to better understand its regulation in heart and provides interesting perspectives for the research of therapeutic pharmacologic compounds.Mechanisms controlling mitochondrial biogenesis in response to hypertension in vascular smooth muscle remain unclear. In this context, the second part of this work was to identify how mitochondrial biogenesis is modulated in arterial hypertension. Using an experimental model of hypertension and after validation in cultivated smooth muscle cells, we show a mitochondrial biogenesis induction in response to hypertension in relation with an increase in oxidative stress. Moreover, this induction is associated with a significant increase in CaMKII activity which was totally blocked by an antioxidant: resveratrol. These results suggest a regulation of mitochondrial biogenesis by oxidative stress via a CaMKII mechanism in vascular smooth muscle.

Page generated in 0.0302 seconds