• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 17
  • 10
  • 2
  • 1
  • 1
  • Tagged with
  • 39
  • 39
  • 21
  • 18
  • 7
  • 7
  • 6
  • 5
  • 4
  • 4
  • 4
  • 4
  • 4
  • 4
  • 4
  • 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.
21

Retinal ciliopathies in Huntington's and SCA7 disorders / Ciliopathies rétiniennes dans la maladie d'Huntington et l'ataxie spinocérébélleuse type7 (SCA7)

Karam, Alice 17 September 2013 (has links)
Les maladies à polyglutamines (polyQ) sont des maladies neurodégénératives héréditaires dominantes causées par une expansion de CAG traduite en longue expansion de polyglutamine dans la protéine correspondante. Ces maladies comprennent l’ataxie spinocérébélleuse 7 (SCA7) et la maladie de Huntington (MH) causées par une expansion de polyQ dans les protéines ataxine-7 (ATXN7) et huntingtine (htt), respectivement. Les souris SCA7 et MH développent des rétinopathies similaires suggérant des pathomécanismes communs toujours inexpliqués. Durant ma thèse, j’ai trouvé qu’en réponse à la toxicité des polyQ, les photorécépteurs (PR) perdent leur différenciation alors que d’autres migrent ou meurent. De plus, cette mortalité cellulaire active la prolifération des cellules gliales de Müller et leur différenciation en PR. Récemment, j’ai trouvé que l’ATXN7 et la htt se trouvent dans le cil primaire et leur mutation mène à une perte des protéines endogènes des cils associée à des défauts du cil. / Polyglutamine (polyQ) disorders are dominantly inherited neurodegenerative disorders caused by the expansion of CAG repeats translated into long polyQ tracts in the corresponding proteins. These diseases include Spinocerebellar ataxia 7(SCA7) and Huntington’s Disease (HD), caused by polyQ expansion ataxin-7 (ATXN7) and huntingtin (htt), respectively. SCA7 and HD mouse models develop similar retinopathies suggesting common pathomechanisms. In my thesis, I found that, in response to polyQ toxicity, SCA7 photoreceptors (PR) undergo several cell fates ranging from their deconstruction, to their migration and their death. Moreover, this cell death activates the proliferation of Müller glial cells and their differentiation into PR like cells. The pathomechanisms underlying HD and SCA7 are still unknown. Recently, I found that ATXN7 and htt are localized to the PR cilia and that the mutant proteins lead to a progressive loss of the wild-type proteins that correlates with defects in the PR cilia.
22

A molecular investigation of a mixed ancestry family displaying dementia and movement disorders

Abrahams-Salaam, Fatima 12 1900 (has links)
Thesis (MScMedSc (Biomedical Sciences. Molecular Biology and Human Genetics))--Stellenbosch University, 2008. / A South African family of Mixed Ancestry presented with a rapidly progressive dementia and a movement disorder which affected a number of individuals across three generations. The initial symptoms included personality changes and tremors that escalated to severe dementia and eventually a completely bedridden state. It was determined that the mean age at onset was in the third decade of life and affected individuals died within 10-15 years after the onset of symptoms. The aim of the present study was to elucidate the genetic cause of the disorder in this family and to further investigate the patho-biology of the disease. Mutations that could possibly cause the observed phenotype in this family were screened for. These included loci implicated in Huntington’s disease, Parkinson’s disease, Dentatorubral-Pallidoluysian Atrophy, Spinocerebellar ataxias (types 1, 2, 3, 6, and 7), Huntington’s disease-like 2 (HDL2) and several mitochondrial disorders. Single-strand Conformation Polymorphism (SSCP) analysis and direct sequencing were used to detect possible mutations while genotyping on an ABI genetic analyser was used to detect disorders caused by repeat expansions. Haplogroup and Short Tandem Repeats (STRs) analyses of the Y-chromosome and mitochondrial DNA of one affected family member was used to determine the family’s genetic ancestry. Reverse transcriptase polymerase chain reaction (RT- PCR) and complementary DNA (cDNA) analyses of the Junctophlin-3 (JPH3) gene was performed to provide information on the expression profile of this gene. After the exclusion of several genetic loci it was shown that this family had HDL2. This is a rare disease caused by a CAG/CTG repeat expansion in an alternatively spliced version of the JPH3 gene. HDL2 occurs almost exclusively in individuals of Black African ancestry. The genetic ancestry data suggested that the family member was most likely of South African Mixed Ancestry making this the first reported family of South African Mixed Ancestry with HDL2. A pilot study investigated the repeat distribution amongst three South African sub-populations in order to determine whether there was a bias in the repeat distribution that possibly predisposes Black Africans to develop the disease. The results showed a statistically significant difference (P= 0.0014) in the distribution of the repeats between the Black African and Caucasian cohorts. However, no conclusions could be drawn as to whether Black Africans harboured larger repeats that predisposes them to developing HDL2. The expanded repeat is located in an alternatively spliced version of the JPH3 mRNA. Interestingly, this repeat is not present in the mouse homologue of the gene although the rest of the genomic sequence is highly conserved across the human, mouse and chimpanzee genomes. Using foetal brain cDNA and PCR primers designed to be specific for different JPH3 isoforms, independent confirmation of the presence of two JPH3 mRNA transcripts (the full length and a shorter alternatively spliced version) was provided. In the absence of brain tissue from an HDL2-affected individual, it was investigated whether both JPH3 mRNA transcripts could be detected in lymphocytes. Using RNA isolated from the transformed lymphocytes of two HDL2-affected family members, real-time PCR was attempted. These experiments produced inconclusive results and required further optimisation. Further RT-PCR experiments for JHP3 expression in different tissues (brain and other) obtained from HDL2-affected individuals would be of interest. The present study identified the first Mixed Ancestry family with HDL2. This family will now be able to request genetic counselling and pre-symptomatic testing for all at-risk family members. Aspects of this study provided independent confirmation of characteristics of the mutated gene. More research on HDL2 will be crucial in understanding the pathogenesis of this disease.
23

Huntingtine et mitose / Huntingtin and mitosis

Molina-Calavita, Maria 22 October 2012 (has links)
La maladie de Huntington (MH) est une maladie neurodégénérative héréditaire autosomique dominante. Elle résulte d’une expansion anormale de glutamines (polyQ) dans la partie N-terminale de la protéine huntingtine (HTT ; codé par HTT). La MH est caractérisée par la dysfonction et la mort de cellules neuronales dans le cerveau, entraînant l’apparition de symptômes cognitifs, psychiatriques et moteurs, dévastateurs chez les patients. De nombreuses études sur des modèles animaux et cellulaires montrent que l’expansion polyQ dans la protéine mutante conduit à un gain de nouvelles fonctions toxiques, ainsi qu’à la perte de fonctions neuroprotectives de la protéine sauvage. Pendant ma thèse, je me suis intéressée à la description et à la validation fonctionnelle d’un nouvel outil pour étudier la HTT : pARIS-htt. pARIS-htt est un gène synthétique construit pour faciliter le clonage et le marquage de la protéine HTT totale. En utilisant différentes approches cellulaires, nous avons montré que pARIS-htt peut remplacer le rôle de la HTT endogène dans le transport de vésicules du Golgi ainsi que du brain derived neurotrophic factor (BDNF). La version mutante de pARIS-htt ne peut pas restaurer cette fonction. Parallèlement, nous avons généré deux variants de pARIS-htt avec soit une délétion dans la région d’interaction de la HTT avec la dynéine, moteur moléculaire se dirigeant vers l'extrémité négative des microtubules, soit avec la huntingtin associated protein 1 (HAP1), l’un de ses interacteurs. Dans les expériences de remplacement du gène, aucun des deux mutants n’a restauré le transport vésiculaire.Un autre aspect de ma thèse a été d’étudier le rôle de la HTT au cours de la mitose. Nous avons mis en évidence l’importance de la HTT dans le contrôle de l’orientation du fuseau. Cette fonction est perdue lorque la HTT est mutée, mais restaurée lorsque celle-ci est phosphorylée par Akt à la sérine 421. Le contrôle de l’orientation du fuseau est particulièrement important durant la neurogénèse puisque cette orientation ainsi que le mode de division sont impliqués dans la détermination des devenirs cellulaires. Cette fonction de la HTT est conservée chez la D. melanogaster.Cette étude a donc permis de mieux comprendre les fonctions de la HTT, et de proposer de nouvelles cibles thérapeutiques pour traiter la MH. / Huntington disease (HD) is an autosomal-dominant neurodegenerative disorder caused by the pathogenic expansion of the poly-glutamine (polyQ) N-terminal stretch in the huntingtin protein (HTT; encoded by HTT). HD is characterized by the dysfunction and death of neurons in the brain, leading to devastating cognitive, psychiatric, and motor symptoms in patients. Studies in multiple cell and animal model systems support the notion that polyQ expansion in mutant HTT leads to the gain of new toxic functions and loss of the neuroprotective functions of the wild-type HTT. During my thesis, I focused on the description and functional validation of a new tool to study HTT: pARIS-htt. pARIS-htt is a synthetic gene built to facilitate cloning and tagging of full-length HTT. Using different cellular approaches, we showed that pARIS-htt can replace endogenous HTT in the transport of Golgi and brain derived neurotrophic factor (BDNF) containing vesicles. pARIS-htt mutant version could not restore vesicular transport when endogenous HTT was knocked-down. Moreover, we generated pARIS-htt deletion mutants for HTT interaction domain with dynein, a minus-end directed motor protein, and huntingtin associated protein 1 (HAP1), a HTT interactor. Both deletion mutants failed to restore vesicular transport in gene replacement assays. Another aspect of my thesis was the study of HTT during mitosis. We showed that HTT monitors spindle orientation though its interaction with diverse proteins involved in cell division. This function is lost when HTT is mutated and can be reverted by Akt phosphorylation at serine 421. The control of spindle orientation is particularly important during neurogenesis since spindle orientation and the mode of division of apical progenitors are implicated in the determination of cell fate. This function of HTT is conserved in D. melanogaster. This study contributes to the understanding of HTT functions and suggests new therapeutical approaches to treat HD.
24

Mechanism of spreading of prion and polyglutamine aggregates and role of the cellular prion protein in Huntington’s disease / Mécanisme de dissémination du prion ainsi que des agrégats polyglutaminiques et rôle de la protéine cellulaire prion dans la maladie de Huntington

Costanzo, Maddalena 28 September 2012 (has links)
La pathogénèse de la plupart des maladies neurodégénératives incluant les maladies transmissibles comme les encéphalopathies à prion, les maladies génétiques de type maladie de Huntington et les maladies sporadiques comme les maladies d’Alzheimer et de Parkinson est directement liée à la formation d’agrégats protéiques fibrillaires. Pendant de nombreuses années, le concept de dissémination et d’infectivité de ces agrégats a été réservé aux maladies à prion. Cependant, de récents résultats montrent que ces protéines amyloidiques extracellulaires (β-amyloïde) comme intracellulaires (α-synucléine, tau, huntingtin) sont capables de bouger (et possiblement de se répliquer) d’une zone à l’autre du cerveau à la façon des prions (Brundin et al., 2010; Jucker and Walker, 2011; Aguzzi and Rajendran, 2009). Récemment une nouveau lien a été établie entre prions et différentes protéinopathies à agrégats. Il a été suggéré que le prion cellulaire, PrPC, dont la forme pathologique (PrPSc) est responsable des maladies à prion, pourrait servir de médiateur dans la toxicité de la protéine β-amyloïde impliquée dans la maladie d’Alzheimer comme dans d’autres conformations-β, indépendamment de la propagation des prions infectieux (revue de Biasini et al., 2012). Malgré une intense recherche sur les maladies neurodégénératives à prion ou non, de nombreuses questions restent ouvertes à la fois au niveau du mécanisme de dissémination des agrégats protéiques que du mécanisme de toxicité. Dans la première partie de ma thèse, j’ai contribué à étudier le rôle de cellules dendritiques (DCs) dans la dissémination de l’infection à prion aux neurones. J’ai démontré que le transfert de PrPSc des cellules dendritiques infectées par un homogénat de cerveau infecté par du prion vers les neurones était dû à contact direct entre ces cellules et a pour résultat la transmission de l’infectivité aux neurones en co-culture. Ces résultats confirment le possible rôle des cellules dendritiques dans la propagation du prion de la périphérie vers le système nerveux central. J’ai aussi trouvé un potentiel mécanisme de transfert de PrPSc des cellules dendritiques aux neurones via des nanotubes (TNTs) et exclu l’implication de la sécrétion de PrPSc dans notre système. Dans la seconde partie de ma thèse, j’ai étudié les mécanismes de dissémination et de toxicité des agrégats protéiques huntingtin et le possible rôle de PrPC dans ces évènements. J’ai démontré que les agrégats Htt sont transférés entre les lignées de cellules neuronales et les neurones primaires et qu’un contact direct cellule à cellule est requis. De même, j’ai montré l’implication des TNTs dans ce transfert et l’agrégation des Htt sauvages endogènes dans les neurones primaires, probablement en suivant le transfert des agrégats Htt. La dernière partie de mes résultats montre que PrPC est impliqué dans la propagation de la toxicité induite par les Htt mutants dans des neurones primaires en culture. / The pathogenesis of most neurodegenerative diseases, including transmissible diseases like prion encephalopathies, inherited disorders like Huntington’s disease, and sporadic diseases like Alzheimer’s and Parkinson’s diseases, appear to be directly linked to the formation of fibrillar protein aggregates. For many years, the concept of aggregate spreading and infectivity has been confined to prion diseases. However, recent evidence indicate that both extracellular (e.g. amyloid-β) and intracellular (α- synuclein, tau, huntingtin) amyloidogenic protein are able to move (and possibly replicate) within the brains of affected individuals, thereby contributing to the spread of pathology in a prion-like manner (Brundin et al., 2010; Jucker and Walker, 2011; Aguzzi and Rajendran, 2009). Recently another intriguing connection has been made between prions and other aggregation proteinopathies, as it was suggested that the cellular prion protein, PrPC, whose pathological counterpart is responsible for prion diseases, possibly mediates the toxicity of Aβ, the pathogenic protein in Alzheimer’s disease, and of other β- conformers independently of the propagation of infectious prions (reviewed in Biasini et al., 2012). However, despite the intense research, many questions in prion and non-prion neurodegenerative diseases are still open regarding both the mechanism of protein aggregate spreading and the mechanism of toxicity. In the first part of my thesis, I contributed to investigate the role of DCs (dendritic cells) in the spreading of prion infection to neuronal cells. I demonstrated that the transfer of PrPSc from DCs (loaded with prion infected brain homogenate) to primary neurons was triggered by direct cell–cell contact and resulted in transmission of infectivity to the co-cultured neurons. These data confirm the possible role of DCs in prion spreading from the periphery to the nervous system. I also provided a plausible transfer mechanism of PrPSc through tunneling nanotubes (TNTs) shown to connect DCs to primary neurons and excluded the involvement of PrPSc secretion in our system. In the second part of my thesis, I investigated the mechanisms of the spreading and toxicity of Htt aggregates and the possible role of PrPC in these events. I demonstrated that Htt aggregates transfer between neuronal cells and primary neurons and that cell-cell contact is required. I also showed the involvement of TNTs in the transfer and reported the aggregation of endogenous wild-type Htt in primary neurons, possibly following the transfer of Htt aggregates. Finally, the last part of my results provides evidences that PrPC is involved in the spreading of the toxicity mediated by mutant Htt in primary neuronal cultures.
25

Mechanisms of Dopaminergic Neurodegeneration in Parkinson's Disease

Verma, Aditi January 2018 (has links) (PDF)
Parkinson’s disease (PD) is a debilitating movement disorder. The cardinal symptoms of PD are bradykinesia, resting tremors and rigidity. PD is characterized by degeneration of dopaminergic neurons of A9 region, substantia nigra pars compacta (SNpc) and loss of dopaminergic terminals in striatum while the dopaminergic neurons of A10 region, ventral tegmental area (VTA) are relatively protected. Putative mechanisms, such as mitochondrial dysfunction, dysregulation of the ubiquitin proteasome system and increased oxidative stress have been hypothesized to mediate PD pathology. However, precise mechanisms that underlie selective vulnerability of SNpc dopaminergic neurons to degeneration are unknown. The aim of this thesis was to evaluate the pathological mechanisms that may contribute to degeneration of SNpc dopaminergic neurons in PD. Dopaminergic neurons of SNpc are pacemakers and constant calcium entry through L-type calcium channel, Cav1.3 has been reported in these neurons during pacemaking. In addition, these neurons have poor calcium buffering capacity. Together, this leads to dysregulation of calcium homeostasis in the SNpc dopaminergic neurons leading to increased oxidative stress. Gene expression of the full length channel and the variant was investigated in the mouse midbrain and further their presence was verified in mouse SNpc and VTA and also in SNpc and VTA in the MPTP mouse model of PD. Gene expression of Cav1.3 -42 and its variant was also studied in SNpc from autopsy tissue from PD patients and age matched controls. Having studied differential expression of the calcium channels, global changes in gene expression in SNpc from the MPTP mouse model of PD and PD autopsy tissues were next examined. This is the first report of transcriptome profile alterations from SNpc in mouse model and PD tissue performed using RNA-seq. Gene expression profiles were examined from SNpc 1 day post single exposure to MPTP, in which case there is no neuronal death and 14 days after daily MPTP treatment where SNpc has undergone ~50% cell death. Further, RNA- seq was performed to study gene expression alterations in SNpc from human PD patients and age- matched controls. The RNA-seq data was taken through extensive analyses; analysed for differential gene expression, gene-set enrichment analysis, pathway analysis and network analysis. Glutaredoxin 1 (Grx1) is a thiol disulfide oxidoreductase that catalyses the deglutathionylation of proteins and is important for regulation of cellular protein thiol redox homeostasis. Down-regulation of Grx1 has been established to exacerbate neurodegeneration through impairment of cell survival signalling. Previous work from our laboratory has demonstrated that perturbation of protein thiol redox homeostasis through diamide injection into SNpc leads to development of PD pathology and motor deficits. It was therefore investigated if Grx1 down-regulation in vivo, leading to increased glutathionylation and protein thiol oxidation, could result in PD pathology. This work is thus the first study of RNA-seq based transcriptomic profile alterations in SNpc from human PD patients. This work also highlights several differences between mouse model and human PD tissue indicating that the underlying mechanisms of PD pathogenesis differ from mouse to humans in addition to developing a novel model for PD.
26

Modification des activités de réseaux in vivo chez un modèle murin de la maladie de Huntington / In vivo circuit activity changes in a mouse model of Huntington’s disease

Cabanas, Magali 14 December 2016 (has links)
La maladie de Huntington est une pathologie héréditaire qui se caractérise par une dégénérescence sélective des neurones striataux de la voie indirecte des ganglions de la base. Chez les patients ainsi que chez les souris modèles de la pathologie, en plus des symptômes moteurs, cognitifs et psychiatriques, des troubles du sommeil peuvent aussi apparaitre dès la phase pré-symptomatique. L’étude électrophysiologique in vivo des souris transgéniques R6/1a, en outre, révélé en début de phase symptomatique l’apparition du rythme pathologique β observé principalement durant le sommeil. Ces travaux de thèses ont donc eut pour but d’étudier le lien entre les modifications d’activités de réseaux cérébraux, les troubles du sommeil et l’émergence du rythme β ainsi que l’implication de ces anomalies dans les perturbations comportementales observées chez les souris R6/1. Notre étude de l’imagerie c-Fos a montré une hyperactivation de la voie frontostriatale chez ces souris, et ceci uniquement au stade pré-symptomatique sans aucune modification d’activation de la voie indirecte. Notre étude pharmacogénétique a démontré que la modification d’activité de ces neurones de projection striataux pouvait modifier l’alternance veille/sommeil mais ne pouvaient générer le rythme β. Enfin, notre étude pharmacologique a établit le lien entre le dysfonctionnement du système orexinergique et l’émergence du rythme β chez les souris R6/1. Ces travaux ont permis de mieux décrire des modifications d’activités de réseaux associées aux différents stades de la pathologie, en particulier au stade présymptomatique, et leurs contributions aux troubles du sommeil et l’émergence du rythme β. / Huntington’s disease (HD) is an inherited pathology that causes selective degeneration ofindirect striatal pathway neurons of the basal ganglia. In addition to the classic motor,cognitive and psychiatric symptoms, patients and mouse models of HD develop sleepdisorders, which can appear at as early as pre-symptomatic stage. Furthermore, in vivoelectrophysiological study of R6/1 transgenic mice revealed a unique and pathological βrhythm that appear at early symptomatic stage and which is mainly observed during sleep.The aim of this thesis work was to examine the link between changes in cerebral networkactivities, sleep disturbances and β rhythm, and to determine the contribution of theseabnormalities to the behavioral disturbances observed in R6/1 mice. Our neuroimaging study of the marker of neuronal activity c-Fos showed a hyperactivation of frontostriatal pathway at pre-symptomatic stage without any activity changes of the vulnerable indirect pathway neurons. Our pharmacogenetic study demonstrated that changes of striatal projection neuronal activity can modify sleep/wake behaviors, without inducing the pathological β rhythm. Finally, our pharmacological study established a link between orexinergic system dysfunction and β rhythm emergence in R6/1 mice. Our data, therefore, described further the natures of altered neural circuit activity associated with different disease stages, in particular pre-motor symptomatic period, and the importance of these alterations for sleep disturbances as well as β rhythm appearance in transgenic HD mice.
27

Troubles hormonaux et leur implication dans la progression de la maladie de Huntington

Saleh, Nadine 29 September 2009 (has links)
Les processus physiopathologiques qui mènent à la dégénérescence neuronale ainsi qu’aux symptômes de la maladie de Huntington (MH) demeurent non identifiés et les hypothèses actuelles ne permettent pas d’expliquer l’hétérogénéité intra et interindividuelle de l’évolution de ces symptômes. Ainsi, la progression de la maladie reste donc difficile voire impossible à prédire. Dans ce contexte, il est important d’explorer d’autres facteurs qui semblent être impliqués dans le processus pathogène de la maladie mais qui pourraient également influencer l’évolution de ces symptômes et ainsi prédire la progression de la maladie. Plusieurs éléments de preuve renforcent l’hypothèse de l’existence de troubles hormonaux dans la MH tels que l’atteinte de l’hypothalamus et la perte de poids. Cependant, en raison du peu d’études, de leur qualité et de la discordance de leurs résultats, l’existence des modifications hormonales dans la maladie de Huntington et plus particulièrement leur lien avec la progression de la maladie reste controversée. L’objectif de ce travail est de décrire le profil hormonal de l’axe hypothalamohypophysaire dans la MH afin de mieux comprendre le rôle de ces hormones sur la progression et éventuellement sur la physiopathologie de la maladie. Dans notre étude transversale, nous avons mis en évidence une activation de l’axe somatotrope (Growth Hormone/Insulin Growth Factor 1), une inhibition en fonction de la sévérité de la maladie de deux axes : gonadotrope (Testostérone) et thyréotrope (Thyroid Stimulating Hormone et triiodothyronine) mais aucune modification des hormones de l’axe corticotrope ni de la prolactine. De plus, la modification hormonale de l’axe somatotrope était non pathologique et précoce alors qu’elle était tardive pour les deux autres axes. Pour expliquer le lien entre ces modifications et la progression de la maladie une étude longitudinale a été mise en place. Les résultats de cette étude montre que seule l’élévation plasmatique d’IGF1 était prédictive de la détérioration cognitive. L’ensemble de nos résultats apporte une meilleure description et compréhension du profil de l’axe hypothalamo-hypophysaire dans la maladie de Huntington. Les axes pituitaires ne sont pas tous atteints et leur atteinte n’est pas dans le même sens. La relation inverse entre l’activation de l’axe somatotrope et la détérioration cognitive renforce l’hypothèse d’une résistance à l’effet de l’IGF1 dans la maladie de Huntington comme pour la maladie d’alzheimer. En conclusion, compte tenu de l’implication de l’IGF1 dans la prédiction de la progression cognitive dans la maladie de Huntington, il serait intéressant de détecter si les modifications biologiques de l’IGF1 existent dès la phase asymptomatique cognitive afin d’envisager d’utiliser l’IGF1 comme biomarqueur de l’apparition ou de l’évolution des symptômes cognitives. D’un autre côté, il serait important d’étendre les recherches sur les mécanismes responsables des modifications hormonales dans la maladie de Huntington afin de mieux comprendre l’effet de cause à effet s’il existe entre ces modifications et les symptômes de la maladie / The pathophysiological processes leading to neurodegeneration and the symptoms of Huntington's disease (HD) remain unidentified and current hypothesis do not explain the intra and interindividual heterogeneity of the evolution of these symptoms. Thus, the progression of the disease remains difficult or impossible to predict. In this context, it is important to explore other factors that appear to be involved in the pathogenic process of the disease but could also influence the evolution of these symptoms and predict disease progression. Several evidences reinforce the hypothesis of the existence of hormonal disorders in HD such as the atrophy of the hypothalamus and weight loss. Because of few studies, their quality and the discrepancies of their results, the existence of hormonal changes in Huntington's disease and particularly their relationship to disease progression remains controversial. The objective of this work is to describe the hormonal profile of the hypothalamicpituitary axis in HD in order to better understand the role of these hormones on the progression and on the pathophysiology of the disease. In our cross-sectional study, we identified an activation of the somatotropic axis (Growth Hormone / Insulin Growth Factor 1), an inhibition according to the severity of the disease in two axes: gonadotrope (Testosterone) and thyréotrope (Thyroid Stimulating hormone and triiodothyronine) but no change in hormones of corticotropic axis and prolactin. In addition, the somatotropic axis is overactive even in patients with early disease. To explain the link between these changes and the progression of the disease, a longitudinal study was done. The results of this study showed that only the elevated plasma IGF1 was predictive of cognitive impairment. All of our results provide a better description and understanding of the profile of the hypothalamic-pituitary axis in Huntington's disease. Pituitary axes are not all disturbed. The inverse relationship between activation of the somatotropic axis and cognitive impairment strengthens the hypothesis of a resistance to the effect of IGF1 in Huntington's disease like in Alzheimer's disease. In conclusion, given the involvement of IGF1 in the prediction of cognitive progression in Huntington's disease, it would be interesting to detect whether the biological changes of IGF1 are already present at the asymptomatic cognitive stage in order to use IGF1 as a biomarker of the onset or changes in cognitive symptoms. On the other hand, , it would be important to extend research on the mechanisms responsible for hormonal changes in Huntington's disease to better understand the link between these changes and symptoms of the disease
28

Huntingtin gene profiling, towards allele-specific treatment

Håkansson, Mimmi January 2020 (has links)
Huntington diseases(HD) is a fatal autosomal neurodegenerative genetic disorder, caused by a CAG trinucleotide repeat expansion in the huntingtin (HTT) gene, resulting in a toxic gain-of-function in the mutant huntingtin protein(mHTT). To date, there is no approved treatment to either cure or halt the course of HD. It has been established that wild-type(wt) HTT protein is essential for development and has a critical role for maintaining neuronal health, thus, a preferable approach for treatment is an mHTT specific lowering maintaining the wild type HTT expression. The achievement of an allele specific therapies depends on targetable allele variation, hence in this project, was the allele frequency in the Swedish population investigated and compared with both the total population and the European population selective. The data demonstrated that there is significant differences between populations. Additionally, the gene expression in five human fibroblast from HD patients with CAG repeats varying from 40 up to180, was analyzed as well as the gene variation across tissue , where the human HD brain and two animal brains; a nonhuman primate and a transgenic minipig, was compared. The result demonstrated that there is similarity in the gene expression between the two models and the human brain, where the highest expression was seen in the prefrontal cortex. The results from the gene expression analyze in the cell lines of fibroblast demonstrated that there is difference in expression between CAG repeats. Furthermore could it be seen that there were only two cell lines, HD180 and HD70, that was heterozygous for dACTT, rs362307, and for the SNP, rs7223906, in exon 67. There are various therapeutic approaches in the pipeline for HD as shown in this thesis, and hopefully a treatment for the disease in the not too distant future. / Huntingtons sjukdom är en dödlig autosomal neurodegenerativ genetisk avvikelse, orsakad av en specifik DNA-sekvens, CAG, upprepning i arvsanlaget som kodar för proteinet huntingtin (HTT). Det muterade HTT skadar nervcellerna i hjärnan och leder till att cellerna bryts ner. Idag finns ännu inga godkända terapier för att bota eller stoppa förloppet av Huntingtons sjukdom. Det har konstaterats att det friska HTT protein är betydelsefullt för utvecklingen och att den har en kritisk roll för att upprätthålla hjärnans nervceller. Därför skulle det vara fördelaktigt att som behandling sänka nivåerna av det muterade HTT och samtidigt behålla nivåerna av det friska HTT i en så kallad allel-specifik strategi. Utförandet av en allel-specifik behandling är beroende allel variationen mellan den friska genen och den muterade. Därför undersöktes allel-frekvensen i den svenska populationen och jämfördes mellan den europiska populationens frekvens. Resultatet från denna undersökning påvisade att det finns tydliga skillnader mellan förekomst av allel-variationer mellan olika populationer. Utöver detta undersöktes även genuttrycket i fem mänskliga friboblaster från patienter med Huntingtons med varierande CAG längd, från 40 repetitioner upp till 180 repetitioner, samt genvariationen mellan vävnader i hjärnan. För den sistnämnda användes data från en mänsklig hjärnan med Huntingtons sjukdom och två djurhjärnor; en ifrån en icke-mänsklig primat och ifrån en transgen minigris. Resultatet påvisade likheter mellan genuttrycket mellan den mänskliga hjärnan och djurhjärnorna, och det högsta uttrycket återfanns i prefrontala cortex. Resultat från fibroblastproverna visade att det finns skillnader i genuttryck mellan patienter som innehar olika längd på CAG-sekvensen. D, dessutom var det endast två cellinjerna, HD180 och HD70, som var heterozygoter för dACTT, rs 362307, var det enda somoch variationen i exon 67, rs7223906. Det finns varierande en multitud av tillvägagångssätt som anges i denna uppsats för att behandla utvecklandet av Huntingtons sjukdom i utveckling, , som anges i denna uppsats, och förhoppningsvis är finns ett botemedel inte i en inte alltför avlägsen framtid.
29

Biology and characterisation of polyalanine as an emerging pathological marker

Stochmanski, Shawn Joseph 12 1900 (has links)
Dix-huit maladies humaines graves ont jusqu'ici été associées avec des expansions de trinucléotides répétés (TNR) codant soit pour des polyalanines (codées par des codons GCN répétés) soit pour des polyglutamines (codées par des codons CAG répétés) dans des protéines spécifiques. Parmi eux, la dystrophie musculaire oculopharyngée (DMOP), l’Ataxie spinocérébelleuse de type 3 (SCA3) et la maladie de Huntington (MH) sont des troubles à transmission autosomale dominante et à apparition tardive, caractérisés par la présence d'inclusions intranucléaires (IIN). Nous avons déjà identifié la mutation responsable de la DMOP comme étant une petite expansion (2 à 7 répétitions supplémentaires) du codon GCG répété du gène PABPN1. En outre, nous-mêmes ainsi que d’autres chercheurs avons identifié la présence d’événements de décalage du cadre de lecture ribosomique de -1 au niveau des codons répétés CAG des gènes ATXN3 (SCA3) et HTT (MH), entraînant ainsi la traduction de codons répétés hybrides CAG/GCA et la production d'un peptide contenant des polyalanines. Or, les données observées dans la DMOP suggèrent que la toxicité induite par les polyalanines est très sensible à leur quantité et leur longueur. Pour valider notre hypothèse de décalage du cadre de lecture dans le gène ATXN3 dans des modèles animaux, nous avons essayé de reproduire nos constatations chez la drosophile et dans des neurones de mammifères. Nos résultats montrent que l'expression transgénique de codons répétés CAG élargis dans l’ADNc de ATXN3 conduit aux événements de décalage du cadre de lecture -1, et que ces événements sont néfastes. À l'inverse, l'expression transgénique de codons répétés CAA (codant pour les polyglutamines) élargis dans l’ADNc de ATXN3 ne conduit pas aux événements de décalage du cadre de lecture -1, et n’est pas toxique. Par ailleurs, l’ARNm des codons répétés CAG élargis dans ATXN3 ne contribue pas à la toxicité observée dans nos modèles. Ces observations indiquent que l’expansion de polyglutamines dans nos modèles drosophile et de neurones de mammifères pour SCA3 ne suffit pas au développement d'un phénotype. Par conséquent, nous proposons que le décalage du cadre de lecture ribosomique -1 contribue à la toxicité associée aux répétitions CAG dans le gène ATXN3. Pour étudier le décalage du cadre de lecture -1 dans les maladies à expansion de trinucléotides CAG en général, nous avons voulu créer un anticorps capable de détecter le produit présentant ce décalage. Nous rapportons ici la caractérisation d’un anticorps polyclonal qui reconnaît sélectivement les expansions pathologiques de polyalanines dans la protéine PABPN1 impliquée dans la DMOP. En outre, notre anticorps détecte également la présence de protéines contenant des alanines dans les inclusions intranucléaires (IIN) des échantillons de patients SCA3 et MD. / Eighteen severe human diseases have thus far been associated with trinucleotide repeat (TNR) expansions coding for either polyalanine (encoded by a GCN repeat tract) or polyglutamine (encoded by a CAG repeat tract) in specific proteins. Among them, oculopharyngeal muscular dystrophy (OPMD), spinocerebellar ataxia type-3 (SCA3), and Huntington’s disease (HD) are late-onset autosomal-dominant disorders characterised by the presence of intranuclear inclusions (INIs). We have previously identified the OPMD causative mutation as a small expansion (2 to 7) of a GCG repeat tract in the PABPN1 gene. In addition, we and others have reported the occurrence of -1 ribosomal frameshifting events in expanded CAG repeat tracts in the ATXN3 (SCA3) and HTT (HD) genes, which result in the translation of a hybrid CAG/GCA repeat tract and the production of a polyalanine-containing peptide. Data from OPMD suggests that polyalanine-induced toxicity is very sensitive to the dosage and length of the alanine stretch. To validate our ATXN3 -1 frameshifting hypothesis in animal models, we set out to reproduce our findings in Drosophila and mammalian neurons. Our results show that the transgenic expression of expanded CAG repeat tract ATXN3 cDNA led to -1 frameshifting events, and that these events are deleterious. Conversely, the expression of polyglutamine-encoding expanded CAA repeat tract ATXN3 cDNA was neither frameshifted nor toxic. Furthermore, expanded CAG repeat tract ATXN3 mRNA does not contribute to the toxicity observed in our models. These observations indicate that expanded polyglutamine repeats in Drosophila and mammalian neuron models of SCA3 are insufficient for the development of a phenotype. Hence, we propose that -1 ribosomal frameshifting contributes to the toxicity associated with CAG repeat tract expansions in the ATXN3 gene. To further investigate ribosomal frameshifting in expanded CAG repeat tract diseases, we sought to create an antibody capable of detecting the frameshifted product. Here we report the characterization of a polyclonal antibody that selectively recognizes pathological expansions of polyalanine in the protein implicated in OPMD, PABPN1. Furthermore, our antibody also detects the presence of alanine proteins in the intranuclear inclusions (INIs) of SCA3 and HD patient samples.
30

How the manipulation of the Ras homolog enriched in striatum alters the behavioral and molecular progression of Huntington’s disease

Lee, Franklin A 18 December 2015 (has links)
Huntington’s disease is an incurable, progressive neurological disorder characterized by loss of motor control, psychiatric dysfunction, and eventual dystonia leading to death. Despite the fact that this disorder is caused by a mutation in one single gene, there is no cure. The mutant Huntingtin (mHtt) protein is expressed ubiquitously throughout the brain but frank cell death is limited to the striatum. Recent work has suggested that Rhes, Ras homolog enriched in striatum, which is selectively expressed in the striatum, may play a role in Huntington’s disease neuropathology. In vitro studies have shown Rhes to be an E3 ligase for the post-translational modification protein SUMO. Rhes increases binding of SUMO to mHtt which competes for the same binding site as Ubiquitin. SUMOylation of mHtt leads to disaggregation and cellular death, whereas ubiquitination leads to aggregation and cellular protection. In a previous study we showed that deletion of Rhes caused a decrease in the Huntington’s disease phenotype in mice. We hypothesized that mice lacking Rhes would also show increased aggregation in the striatum and this increased aggregation would correlate in a rescue of behavioral symptoms. Despite the prior in vitro and in vivo evidence, deletion of Rhes in vivo did not alter the aggregation of mHtt in the striatum of mice however deletion of Rhes still showed a rescue from the diseased phenotype. This result would indicate that deletion of Rhes alters the neurobehavioral phenotype of Huntington’s disease through a different pathway than promoting aggregation in striatal cells.

Page generated in 0.0849 seconds