• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 10
  • 4
  • 2
  • 1
  • 1
  • Tagged with
  • 22
  • 13
  • 8
  • 7
  • 7
  • 7
  • 6
  • 6
  • 5
  • 4
  • 3
  • 3
  • 3
  • 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.
11

Structural and Functional Studies on Human Mitochondrial Iron-Sulfur Cluster Biosynthesis

Tsai, Chi-Lin 2011 May 1900 (has links)
Iron-sulfur (Fe-S) clusters are critical protein cofactors found in all life forms. In eukaryotes, a well-conserved biosynthetic pathway located in the mitochondria is used to assemble Fe-S clusters. Although proteins required for Fe-S cluster biosynthesis have been identified, their precise function and mechanism remain elusive. In this study, biochemical and biophysical methods are applied to understand molecular details for the core components of the human Fe-S cluster biosynthesis: Nfs1, Isd11, Isu2, and frataxin (Fxn). Nfs1 is a cysteine desulfurase that converts cysteine into alanine and transfers the sulfur to a scaffold protein Isu2 for Fe-S clusters. Fxn depletion is associated with the neurodegenerative disease Friedreich’s ataxia (FRDA), and results in a complicated phenotype that includes loss of Fe-S clusters. The results presented here provide the first in vitro evidence for a stable protein complex that exists in at least two forms: an inactive complex with Nfs1, Isd11, and Isu2 (SDU) components and an active form that also includes Fxn (SDUF). Fxn binding dramatically changes the catalytic efficiency (kcat/KM) of Nfs1 from 25 to 10,100 M-1s-1 and enhances the rate of Fe-S cluster biosynthesis 25 fold. Oxidizing conditions diminish the levels of both complex formation and Fxn-based activation, whereas Fe2 further stimulates Nfs1 activity. Mutagenesis coupled to enzyme kinetics indicate that one of the three conserved cysteines (C104) on Isu2 accepts the sulfane sulfur from Nfs1 and that this transfer event likely requires prior binding of Fxn. In vitro interrogation of FRDA I154F and W155R and related Fxn variants revealed the binding affinity to SDU followed the trend Fxn ~ I154F > W155F > W155A ~ W155R. The Fxn variants also have diminished ability to facilitate both sulfur transfer and Fe-S cluster assembly. Fxn crystallographic structures reveal specific rearrangements associated with the loss of function. Importantly, the weaker binding and lower activity of the W155R variant compared to I154F explains the earlier onset and more severe disease progression. Finally, these experimental results coupled with computational docking studies suggest a model for how human Fxn functions as an allosteric activator and triggers sulfur transfer and Fe-S cluster assembly.
12

Genotype and phenotype characterisation of Friedreich ataxia mouse models and cells

Anjomani Virmouni, Sara January 2013 (has links)
Friedreich ataxia (FRDA) is an autosomal recessive neurodegenerative disorder, caused by a GAA repeat expansion mutation within intron 1 of the FXN gene, resulting in reduced level of frataxin protein. Normal individuals have 5 to 40 GAA repeat sequences, whereas affected individuals have approximately 70 to more than 1000 GAA triplets. Frataxin is a mitochondrial protein involved in iron-sulphur cluster and heme biosynthesis. The reduction in frataxin expression leads to oxidative stress, mitochondrial iron accumulation and consequential cell death with the primary sites of neurons of the dorsal root ganglia and the dentate nucleus of the cerebellum. FRDA, which is the most common inherited ataxia, affecting 1:50,000 Caucasians, is characterised by neurodegeneration, cardiomyopathy, diabetes mellitus and skeletal deformities. To investigate FRDA molecular disease mechanisms and therapy, several human FXN YAC transgenic mouse models have been established: Y47R, containing normal-sized (GAA)9 repeats; YG8R and YG22R, which initially contained expanded GAA repeats of 90-190 units and 190 units, respectively, but which have subsequently been bred to now contain expanded GAA repeats of 120-220 units and 170-260 units, respectively, and YG8sR (YG8R with a small GAA band) that was recently generated from YG8R breeding. To determine the FXN transgene copy number in the enhanced GAA repeat expansion-based FRDA mouse lines, a TaqMan qPCR assay was developed. The results demonstrated that the YG22R and Y47R lines had a single copy of the FXN transgene while the YG8R line had two copies. The YG8s lines showed less than one copy of the target gene, suggesting potential deletion of the FXN gene. Single integration sites of all transgenes were confirmed by fluorescence in situ hybridisation (FISH) analysis of metaphase and interphase chromosomes. However, in the YG8s line, at least 25% of the YG8s cells had no signals, while the remaining cells showed one signal corresponding to the transgenic FXN gene. In addition, the analysis of FXN exons in YG8s rescue mice by PCR confirmed the presence of all FXN exons in these lines, suggesting the incidence of somatic mosaicism in these lines. Extended functional analysis was carried out on these mice from 4 to 12 months of age. Coordination ability of YG8R, YG8sR and YG22R ‘FRDA-like’ mice, together with Y47R and C57BL6/J wild-type control mice, was assessed using accelerating rotarod analysis. The results indicated a progressive decrease in the motor coordination of YG8R, YG22R and YG8sR mice compared to Y47R or C57BL6/J controls. Locomotor activity was also assessed using an open field beam-breaker apparatus followed by four additional functional analyses including beam-walk, hang wire, grip strength and foot print tests. The results indicated significant functional deficits in the FRDA mouse models. Glucose and insulin tolerance tests were also conducted in the FRDA mouse models, indicating glucose intolerance and insulin hypersensitivity in the aforementioned lines. To investigate the correlation between the FRDA-like pathological phenotype and frataxin deficiency in the FRDA mouse models, frataxin mRNA and protein levels as well as somatic GAA repeat instability were examined. The results indicated that somatic GAA repeats increased in the cerebellum and brain of YG22R, YG8R and YG8sR mice, together with significantly reduced levels of FXN mRNA and protein in the liver of YG8R and YG22R compared to Y47R. However, YG8sR lines showed a significant decrease in FXN mRNA in all of the examined tissues compared to Y47R human FXN and C57BL6/J mouse Fxn mRNA. Protein expression levels were also considerably reduced in all the tissues of YG8sR mice compared to Y47R. Subsequently, the telomere length of human and mouse FRDA and control fibroblasts was assessed using qPCR and Q-FISH. The results indicated that the FRDA cells had chromosomes with relatively longer telomeric repeats in comparison to the controls. FRDA cells were screened for expression of telomerase activity using the TRAP assay and a quantitative assay for hTERT mRNA expression using TaqMan qRT-PCR. The results indicated that telomerase activity was not present in the FRDA cells. To investigate whether FRDA cells maintained their telomeres by ALT associated PML bodies (APBs), co-localisation of PML bodies with telomeres was assessed in these cells using combined immunofluorescence to PML and Q-FISH for telomere detection. The results demonstrated that the FRDA cells had significantly higher co-localised PML foci with telomeric DNA compared to the normal cells. Moreover, telomere sister chromatid exchange (T-SCE) frequencies were analysed in the human FRDA cell lines using chromosome orientation FISH (CO-FISH). The results indicated a significant increase in T-SCE levels of the FRDA cell lines relative to the controls. Furthermore, growth curve and population doubling analysis of the human FRDA and control fibroblasts was carried out. The results showed that the FRDA fibroblast cell cultures underwent growth arrest with higher cumulative population doubling compared to the controls. Though, further analysis of telomere length at different passage numbers revealed that the FRDA cells lost telomeres faster than the controls. Finally, the telomere dysfunction-induced foci (TIF) assay was performed to detect DNA damage in the human FRDA fibroblast cells using an antibody against DNA damage marker γ-H2AX and a synthetic PNA probe for telomeres. The frequency of γ-H2AX foci was significantly higher in the FRDA cells compared to the controls. Similarly, the FRDA cells had greater frequencies of TIFs in comparison to the controls, suggesting induced telomere dysfunction in the FRDA cells.
13

Investigating the existence of a link between mitochondria and microRNAs / Étude d'un lien entre la mitochondrie et les microARNs

Bandiera, Simonetta 05 November 2012 (has links)
La mitochondrie est une organite ayant un rôle central dans le métabolisme énergétique de la cellule. Bien que la mitochondrie exprime son propre génome, plusieurs protéines et ARN non-codants issus du génome nucléaire sont nécessaires à la biogenèse et aux fonctions mitochondriales. Les microARNs (miRNAs) sont de petits ARN non-codants qui s'associent à la protéine Argonaute 2 (Ago2) pour moduler l'expression génique au niveau post-transcriptionnelle par ARN interférence. Classiquement, les miRNAs s’apparient à des sites de liaison complémentaires situés dans le 3’-UTR de l’ARNm cible. Nous faisons l'hypothèse que les miRNAs seraient impliqués dans la communication entre le noyau et la mitochondrie. Notre travail a donc porté sur l’étude du rôle des miRNAs dans le contexte de maladies génétiques caractérisées par une dysfonction mitochondriale. Nous avons choisi d’étudier l’ataxie de Friedreich, la plus fréquente des ataxies héréditaires, qui est causée par un déficit d’expression de la protéine mitochondriale frataxine (FXN). Nous avons montré qu'environ 90% de patients étaient homozygotes pour un haplotype spécifique des variants génétiques du 3'-UTR du gène FXN. Ce résultat a été retrouvé dans deux cohortes de patients indépendantes. Par une combinaison d’approche bioinformatique et d’expériences de co-transfections, nous avons montré que les miRNAs, et en particulier miR-124, ciblent les variants du 3’-UTR. En parallèle, nous avons évalué la possibilité que les miRNAs ciblent directement la mitochondrie. Pour cela, nous avons analysé l’expression des miRNAs dans des fractions d'ARN mitochondriale et cytosolique isolées à partir de mêmes cellules HeLa. Nous avons identifié une signature de 13 miRNAs spécifiquement enrichis dans la fraction d'ARN mitochondriale que nous avons appelé «mitomiRs». Nos prédictions ont révèle des fonctions spécifiques de ces mitomiRs à la mitochondrie, y compris la modulation de l'activité de la chaîne respiratoire. Nous avons également montré une localisation de la protéine Ago2 à l’espace inter-membranaire mitochondriale.Notre travail définit ainsi les miRNAs et Ago2 comme un nouveau niveau de communication entre le noyau et les mitochondries. Nous discutons de la possibilité que la mitochondrie agisse comme acteur du ARN interférence ou plutôt comme organite cible. Notre travail ouvre la voie à un nouveau domaine de recherche, qui pourrait avoir une utilité thérapeutique pour palier les dysfonctions mitochondriales. / Mitochondria are organelles that have a central role in the energetic metabolism of the cell. Although mitochondria express their own genome, they rely on the expression of the nuclear genome for their biogenesis and function. microRNAs (miRNAs) are small non-coding RNAs that associate with Argonaute 2 (Ago2) protein to regulate gene expression post-transcriptionallythrough RNA interference. The ‘classic’ view of RNA interference describes the pairing of miRNAs with complementary binding sites within the 3’untranslated region (3’-UTR) of the target mRNA. We hypothesized that miRNAs might be instrumental to the cross-talk between the nucleus and the mitochondria. In the first part, we assessed the role of miRNAs in the context of a rare genetic disease involving mitochondrial dysfunction. We focused on Friedreich's ataxia, the most frequent of inherited ataxia in Europe, which is caused by reduced expression of the mitochondrial protein frataxin (FXN). Intwo independent cohorts of patients, we discovered that about 90% of patients were homozygous forone specific haplotype of genetic variants of the FXN3'-UTR. By a combination of computational target prediction analysis and co-transfection experiments, we showed that miRNAs, and specifically miR-124, are involved in the regulation of the FXN.We then challenged further the relationship between the miRNAs and mitochondria through questioning their localization at mitochondria. To this end, we studied miRNAs from mitochondrial and cytosolic RNA fractions isolated from the same HeLa cells. We identified a signature of 13 miRNAs specifically enriched in the mitochondrial RNA fraction that we termed ‘mitomiRs’. Through pathway-enrichement analysis, we observed a specific mitochondrial role for mitomiRs, including regulation of ATP synthesis coupled electron trasport. We also provided the evidence of Ago2 protein location inside human mitochondria at the inter-membrane space. Our work sketches miRNAs and Ago2 as a novel layer of the interplay between the nucleus and the mitochondria. We discuss whether mitochondria may be instrumental to RNA interference or a target per se. Our work paves the way to a new field of research, which may unravel therapeutic outcomes to rescue mitochondrial dysfunction.
14

Functionaland structural studies of human frataxin: An iron chaperone protein for mitochondrial iron-sulfur cluster and heme biosyntheses

Yoon, Taejin 24 August 2005 (has links)
No description available.
15

Thermodynamic studies on iron-sulfur cluster assembly proteins

Ding, Shu 21 October 2011 (has links)
No description available.
16

Functional Characterization and Surface Mapping of Frataxin (FXN) Interactions with the Fe-S Cluster Assembly Complex

Thorstad, Melissa 16 December 2013 (has links)
In 1996, scientists discovered a connection between the gene for the human protein frataxin (FXN) and the neurodegenerative disease Friedreich’s ataxia (FRDA). Decreased FXN levels result in a variety of aberrant phenotypes including loss of activity for iron-sulfur containing enzymes, mitochondrial iron accumulation, and susceptibility to oxidative stress. These symptoms are the primary focus of current therapeutic efforts. In contrast our group is pursuing an alternate strategy of first defining FXN function at a molecular level then using this information to identify small molecule functional replacements. Recently, our group has discovered that FXN functions as an allosteric activator for the human Fe-S cluster assembly complex. The work presented here helps to further define molecular details of FXN activation and explain how FRDA missense mutants are functionally compromised. First, the FRDA missense mutants L182H and L182F were investigated. Unlike other characterized FRDA missense mutants, the L182F variant was not compromised in its ability to bind and activate the Fe-S assembly complex. The L182H variant exhibited an altered circular dichroism signature; suggesting a change in secondary structure relative to native FXN, and rapidly degraded. Together these studies suggest that L182 variants are less stable than native FXN and are likely prone to degradation in FRDA patients. Second, as a regulatory role of FXN suggests that its function is likely controlled by environmental stimuli, different maturation forms of FXN as well as post-translational modification mimics were tested as mechanisms to control FXN regulation. Here experiments were designed to test if a larger polypeptide form of FXN represents a functional form of the protein. Kinetic and analytical ultracentrifugation studies revealed a complex heterogeneous mixture of species some of which can activate the Fe-S assembly complex. A previously identified acetylation site was also tested using mutants that mimic acetylation. These mutants had little effect on the ability of FXN to bind and activate the assembly complex. Third, mutagenesis experiments were designed in which the FXN surface residues were replaced with alanine and the resulting variants were tested in binding and activity assays. These experiments revealed a localized “hot-spot” on the surface of FXN that suggests small cyclic peptide mimics might be able to replace FXN and function as FRDA therapeutics. Unexpectedly, one of the FXN variants exhibited significantly tighter binding and could have relevance for therapeutic development.
17

Alterations of mitochondrial biogenesis and alterations of mitochondrial antioxidant defense in Friedreich's ataxia

Marmolino, Daniele 25 January 2011 (has links)
Friedreich’s ataxia (FRDA) is an autosomal recessive inherited disorder affecting approximately 1 every 40,000 individuals in Western Europe, is characterized by progressive gait and limb ataxia, dysarthria, areflexia, loss of vibratory and position sense, and a progressive weakness of central origin. Additional features particularly include an hypertrophic cardiomyopathy that can cause premature death. A large GAA repeat expansion in the first intron of the FXN gene is the most common mutation underlying FRDA. Patients show severely reduced levels of the FXN-encoded mitochondrial protein frataxin.<p>Frataxin function is not yet completely elucidated. In frataxin deficiency conditions abnormalities of iron metabolism occur: decreased activities of iron-sulfur cluster (ISC) containing proteins, accumulation of iron in mitochondria and depletion in the cytosol, enhanced cellular iron uptake, and, in some models, reduced heme synthesis. <p>Evidence of oxidative stress has also been found in most though not all models of frataxin deficiency. Accordingly, yfh1-deficient yeast and cells from FRDA patients are highly sensitive to oxidants. Respiratory chain dysfunction further aggravate oxidative stress by increasing leakage of electrons and the formation of superoxide. Frataxin deficient cells not only generate more free radicals, but, they also show a reduced ability to mobilize antioxidant defenses, in particular to induce superoxide dismutase 2 (SOD2).<p>Peroxisome proliferator-activated receptor (PPAR) isoform-gamma play a key role in numerous cellular functions and is a key regulator of mitochondrial biogenesis and of the ROS metabolism. Recruitment of the PPAR coactivator-1a (PGC-1a) mediates many effects of the PPAR-γ activation.<p>In a first work we assessed the potential beneficial effects of a potent PPAR-gamma agonist on frataxin expression in primary fibroblasts from healthy controls and FRDA patients, and Neuroblastoma cells. We used the APAF molecule (1-0-hexadecyl-2-azelaoyl-sn-glycero-3-phosphocoline; C33H66NO9P). Our results show that this compound is able to increase frataxin amount both at transcriptional and post-transcriptional level. At a dose of 20µM frataxin mRNA significantly increases in both controls (p=0.03) and FRDA patients (p=0.002) fibroblasts (1). The finding was confirmed in Neuroblastoma cells (p=0.042). According to previous publications APAF, as others PPAR-gamma agonists is able to up-regulate PGC-1a transcription.<p>In a second part of the study we investigate the role of the PPAR-gamma/PGC-1a pathway in the pathogenesis of FRDA. We performed a microarray analysis of heart and skeletal muscle in a mouse model of frataxin deficiency and we found molecular evidence of increased lipogenesis in skeletal muscle and alteration of fiber-type composition in heart, consistent with insulin resistance and cardiomyopathy, respectively. Since the PPAR-gamma pathway is known to regulate both processes, we hypothesized that dysregulation of this pathway could play a key role in frataxin deficiency. We confirmed this by showing a coordinate dysregulation of Pgc1a and the transcription factor Srebp1 in cellular and animal models of frataxin deficiency, and in cells from FRDA patients, who have marked insulin resistance. Particularly, PGC-1a was found significantly reduced (2) in primary fibroblasts and lymphocytes from FRDA patients (p<0.05). Furthermore, PGC-1a mRNA levels strongly correlate with frataxin relative mRNA levels (r2=0.9, p<0.001). According to this observation, in C2C12 myoblasts, PGC-1a and a reporter gene under the control of the PGC-1a promoter are rapidly down-regulated (p<0.05) when frataxin expression is inhibited by an shRNA in vitro. To further investigate this relation, we then generate PGC-1a deficient fibroblasts cells using a specific siRNA; at 72 hours of transfection frataxin was found down-regulate (p<0.05) in control cells. <p>Taken together those data indicate that some mechanism directly links an early effect of frataxin deficiency with reduced PGC-1a transcription in this cell type, and presumably in other cells that also down-regulate PGC-1α when frataxin levels are low.<p>Finally, since PGC-1a has also emerged as a key factor in the induction of many antioxidant programs in response to oxidative stress, both in vivo and in vitro, in particular in neurons, we tested whether the PGC-1a down-regulation occurring in FRDA cells could be in part responsible for the blunted antioxidant response observed in frataxin deficiency.<p>Using primary fibroblasts from FRDA patients we found reduced SOD2 levels (p<0.05), according to PGC1& / Doctorat en Sciences biomédicales et pharmaceutiques / info:eu-repo/semantics/nonPublished
18

Développement d'un nouveau modèle cellulaire de l'ataxie de Friedreich : différenciation de cellules pluripotentes induites de patients en cardiomyocytes / Induced pluripotent stem cells-derived cardiomyocytes : a new model for Friedreich’s ataxia

Hick, Aurore 04 April 2014 (has links)
L’ataxie de Friedreich (AF) est une maladie neurodégénérative récessive due à un déficit en frataxine, une protéine mitochondriale très conservée. Elle est souvent associée à une atteinte cardiaque. Le déficit en frataxine est responsable d’une diminution de l’activité des enzymes Fe-Set d’une accumulation mitochondriale de fer. Les cellules pluripotentes induites (iPS) générées par reprogrammation de cellules somatiques constituent un outil puissant pour le développement de modèles de maladies monogéniques. Les cardiomyocytes obtenus par différenciation des iPS de patients AF développent une atteinte mitochondriale après 1 mois en culture. Celle-ci se complète par l’apparition secondaire de dépôts de fer, visibles après 4 mois en culture, indiquant une progression dans la physiopathologie de la maladie. Notre étude montre la capacité de ces cardiomyocytes à modéliser le phénotype cardiaque de l’AF, offrant ainsi l’opportunité d’approfondir leur caractérisation physiopathologique. / Friedreich’s ataxia (FA) is a recessive neurodegenerative disorder due to a deficit of frataxin, a highly conserved mitochondrial protein. It is commonly associated with a hypertrophic cardiomyopathy. The main pathophysiological consequences are a decrease of Fe-S enzyme activities and mitochondrial iron accumulation. The recent technical advances in the generation of induced pluripotent stem cells (iPS) from somatic cells provide a powerful tool to create disease specific cellular models. Cardiomyocytes derived from FA-iPS present altered mitochondria after 1 month in culture. After four months in culture, iron deposits can be found in degenerating mitochondria, indicating a progression in the pathophysiology of the disease. Our study illustrates the ability of iPS-derived cardiomyocytes to model the cardiac phenotype associated with FA, and offers new opportunities to further investigate pathological mechanisms linked to frataxin deficiency.
19

Overcoming frataxin gene silencing in Friedreich's ataxia with small molecules: studies on cellular and animal models

Rai, Myriam 05 January 2010 (has links)
Friedreich’s ataxia (FRDA) is an inherited recessive disorder characterized by progressive neurological disability and heart disease. It is caused by a pathological intronic hyperexpansion of a GAA repeat in the FXN gene, encoding the essential mitochondrial protein frataxin. At the homozygous state, the GAA expansion induces a heterochromatin state with decreased histone acetylation and increased methylation, resulting in a partial deficiency of frataxin expression. This was established in cells from FRDA patients. We showed that the same chromatin changes exist in a GAA based mouse model, KIKI, generated in our laboratory. Furthermore, treatment of KIKI mice with a novel Histone Deacetylase Inhibitor (HDACi), 106, a pimelic diphenylamide that increases frataxin levels in FRDA cell culture, restored frataxin levels in the nervous system and heart of KIKI mice and induced histone hyperacetylation near the GAA repeat. As shown by microarrays, most of the differentially expressed genes in KIKI were corrected towards wild type. In an effort to improve the pharmacological profile of compound 106, we synthesized more compounds based on its structure and specificity. We characterized two of these compounds in FRDA patients’ peripheral blood lymphocytes and in the KIKI mouse model. We observed a sustained frataxin upregulation in both systems, and, by following the time course of the events, we concluded that the effects of these compounds last longer than the time of direct exposure to HDACi. Our results support the pre-clinical development of a therapeutic approach based on pimelic diphenylamide HDACis for FRDA. Laboratory tools to follow disease progression and assess drug efficacy are needed in a slowly progressive neurodegenerative disease such as FRDA. We used microarrays to characterize the gene expression profile in peripheral lymphocytes from FRDA patients, carriers and controls. We identified gene expression changes in heterozygous, clinically unaffected GAA expansion carriers, suggesting that they present a biochemical phenotype, consistent with data from animal models of frataxin deficiency. We identified a subset of genes changing in patients as a result of pathological frataxin deficiency establishing robust gene expression changes in peripheral lymphocytes. These changes can be used as a biomarker to monitor disease progression and potentially assess drug efficacy. To this end, we used he same methodology to characterize the gene expression profiles in peripheral lymphocytes after treatment with pimelic diphenylamide HDACi. This treatment had relevant effects on gene expression on peripheral patients’ blood lymphocytes. It increased frataxin levels in a dose-dependent manner, and partially rescued the gene expression phenotype associated with frataxin deficiency in the tested cell model, thus providing the first application of a biomarker gene set in FRDA. / Doctorat en sciences biomédicales / info:eu-repo/semantics/nonPublished
20

Etude du rôle de la frataxine bactérienne CyaY chez Escherichia coli / Study of bacterial frataxin CyaY in Escherichia coli

Roche, Béatrice 01 December 2015 (has links)
Les protéines à centre Fe-S sont impliquées dans de nombreux processus cellulaires. In vivo, la formation des centres Fe-S est réalisée par des machineries multi-protéiques dont ISC et SUF, conservées chez les eucaryotes et les procaryotes. D’autres composants participent à la formation des centres Fe-S chez les eucaryotes, comme la frataxine (FXN). La FXN est une protéine présente chez l’homme, les plantes, la levure ou encore les bactéries à Gram négatif. Chez les eucaryotes, l’absence de FXN conduit à des phénotypes drastiques comme une accumulation de fer dans la mitochondrie, une diminution drastique de l’activité d’enzymes à centre Fe-S ou encore des dommages oxydatifs. Chez l’homme, un déficit en FXN est responsable d’une maladie neurodégénérative, l’ataxie de Friedreich. A la différence des eucaryotes, chez les procaryotes comme Escherichia coli, l’absence de CyaY, homologue bactérien de la FXN, ne conduit à aucun des phénotypes évoqués ci-dessus.Durant ma thèse, je me suis intéressée au rôle de CyaY chez E. coli. J’ai montré que, in vivo, CyaY favorise la formation des centres Fe-S via la machinerie ISC. Un lien génétique entre CyaY et IscX a également pu être établi, montrant que ces deux protéines participent à la formation des centres Fe-S in vivo. Je me suis ensuite intéressée aux bases moléculaires pouvant expliquer la différence entre les phénotypes liés à l’absence de FXN chez les eucaryotes et les procaryotes. J’ai montré que le résidu 108 de IscU joue un rôle clé pour la dépendance de CyaY. Enfin, pour mieux comprendre le rôle de CyaY chez E. coli, j’ai réalisé une approche globale en caractérisant le transcriptome du mutant ∆cyaY. / Fe-S cluster containing proteins are involved in many cellular processes such as respiration, DNA repair or gene regulation. In vivo, Fe-S cluster biogenesis is catalysed by specific protein machineries, ISC and SUF, conserved in both eukaryotes and prokaryotes. Frataxin (FXN) is a small protein found in humans, plants, yeast and Gram negative bacteria. In eukaryotes, a defect in FXN leads to drastic phenotypes such as mitochondrial iron accumulation, drastic decrease of Fe-S cluster protein activity, sensitivity to oxidants. In humans, FXN deficiency is responsible for the neurodegenerative disease, Friedreich’s ataxia. In prokaryotes like E. coli, a defect in CyaY, the bacterial FXN homolog, does not lead to significant phenotypes compared to the wild-type strain. During my thesis, I investigated the role of the bacterial FXN CyaY in E. coli. I showed that, in vivo, CyaY assisted the ISC-catalyzed Fe-S cluster biogenesis. A genetic link was also observed between cyaY and iscX, demonstrating that these proteins participate in Fe-S cluster biogenesis. In a second part, I investigated the differences between the impact of the eukaryotic versus prokaryotic FXN. I showed that the IscU 108th residue is crucial for the CyaY-dependency. Finally, I used a transcriptomic approach to test whether CyaY has a global role in E. coli.

Page generated in 0.0303 seconds