• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 26
  • 9
  • 5
  • 3
  • Tagged with
  • 54
  • 38
  • 25
  • 18
  • 14
  • 9
  • 9
  • 9
  • 7
  • 7
  • 7
  • 7
  • 6
  • 5
  • 5
  • 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

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.
22

Caractérisation de P42, région cruciale pour la fonction de la Huntingtine et peptide capable d’inhiber la toxicité associée à la Chorée de Huntington / Characterisation of P42 : a crucial region of Huntingtin and a therapeutic peptide for the treatment of Huntington's Disease

Arribat, Yoan 24 October 2012 (has links)
La Maladie de Huntington (MH) reste à ce jour une pathologie neurodégénérative dévastatrice pour laquelle aucun traitement n'est disponible. L'agrégation de la Huntingtine Mutante (Htt PolyQ) joue un rôle majeur dans les processus pathologiques. Dans ce contexte, des études récentes ont démontré que la partie N-terminale de la Huntingtine Humaine (Htt wt) ou de son Homologue drosophile (dHtt) sont toutes deux capables de réduire l'agrégation et la toxicité de Htt PolyQ. En se basant sur cette observation, le travail de thèse décrit dans ce manuscrit a mis au point un découpage du fragment N-terminal de Htt wt de manière à isoler en son sein, une courte séquence de 23 acides aminés (nommée P42), capable d'inhiber spécifiquement l'agrégation de Htt PolyQ en modèle cellulaire. L'effet protecteur de ce peptide a été confirmé in vivo, sur un modèle drosophile de la MH. Le potentiel thérapeutique que représente P42 a servi de point de départ à une étude menée sur le modèle murin R6/2 de la MH. L'effet de P42 a été potentialisé par l'ajout du peptide de transduction TAT de manière à faciliter son entrée dans les cellules cibles. Puis, la protéine fusion P42-TAT a été vectorisée sous forme de microémulsion de manière à assurer à la fois une administration simple de la molécule, et un accès au système nerveux central. Ce protocole original a permis d'observer des bénéfices sans précédent sur les phénotypes comportementaux, histologiques et moléculaires que présentent les souris R6/2.Au-delà de son aspect thérapeutique, P42 est avant tout une séquence méconnue située dans une région cruciale de la Huntingtine. L'étude du rôle physiologique de ce site, a mené à une meilleure compréhension de la fonction sauvage de la protéine toute entière. En outre, une analyse biochimique a montré la capacité du fragment N-terminal de Htt wt à se lier aux microtubules. Cette interaction avec le cytosquelette dépend de plusieurs processus (clivages, dimérisation) et semble affilier la Huntingtine à la grande famille des MAP.L'identification de P42 ouvre donc une voie nouvelle vers la compréhension du rôle physiologique de la Huntingtine, mais représente surtout un espoir thérapeutique captivant. / Huntington's disease (HD) is a devastating and incurable neurodegenerative disorder. Aggregation processes of mutant Huntingtin (Htt PolyQ) play a central part in the pathology onset. In this context, recent studies pointed out the capacities of wild-type Huntingtin N-terminus to reduce both aggregation and toxicity associated with Htt PolyQ. The drosophila Homologue shares the sames properties. Basing on these observations, the present work realised a cut of human Huntingtin N-terminus in order to identify the region responsible for therapeutic benefits. This screen highlighted a 23 amino-acid sequence (noted P42), that inhibits Htt PolyQ aggregation in a HeLa cells model. Then, the protective effect of this peptide was confirmed in vivo, in a HD drosophila model.P42 therapeutic potential was explored in the R6/2 HD mouse model. The entry of the peptide into cells, was potentialised by grafting to P42, the transduction sequence of TAT. Then, the fusion protein P42-TAT was vectorised in microemulsion, in order to enhance the delivery of the peptide to the brain by resorting to a non-invasive administration way. This original protocol exhibited highly-significant rescues on behavioural, histological and molecular R6/2 phenotypes..Over the therapeutic aspect, P42 also represents an important region of Huntingtin. The study of this site led to a better understanding of Huntingtin physiological function. Biochemestrial experiments underlined the binding of Htt N-terminus on microtubules networks. This interaction depends on a range of complex processes (dimerization, cleavage) and suggests that the Huntingtin belongs to the family of Structual MAPs.In summary, the identification of P42 enhances the knowledge about Huntingtin function, and opens a new promising therapeutical avenue for HD.
23

Implication de la huntingtine dans les troubles de l'humeur : approche comportementale et neurogénique / Implication of huntingtin in mood disorders : A behavioural and neurogenic approach

Orvoen, Sophie 18 September 2012 (has links)
La maladie de Huntington (HD) est une maladie génétique neurodégénérative qui touche environ 6000 personnes en France. Les manifestations psychiatriques sont une des composantes majeures des symptômes précoces de la pathologie. Ainsi, des épisodes dépressifs parfois associés à de l’anxiété généralisée sont communément observés au cours des stades pré-symptomatiques de la maladie. On connaît mal à l’heure actuelle les raisons de cette prévalence élevée. L'allèle responsable de la maladie code une protéine appelée huntingtine (HTT) dont l'expansion polyglutaminique (polyQ) en N-terminal est plus longue que dans la HTT non pathogénique. La huntingtine est impliquée dans diverses fonctions cellulaires et notamment dans le transport et l’expression d’un facteur neurotrophique, le Brain-Derived Neurotrophic Factor (BDNF). Celui-ci est d’ailleurs connu pour son rôle dans la régulation des troubles de l’humeur, de la neurogénèse hippocampique chez l’adulte, ainsi que dans la réponse thérapeutique aux antidépresseurs. Nous avons émis l'hypothèse que la huntingtine, en plus de ses rôles connus dans le cortex et le striatum, puisse jouer également un rôle dans l'hippocampe. Ainsi, une altération du transport de BDNF dans l’hippocampe pourrait en partie expliquer les troubles de l’humeur observés chez les patients HD.Par une approche in vivo, en utilisant différents modèles de souris, nous avons ainsi démontré que la huntingtine stimule le trafic vésiculaire et la sécrétion de BDNF dans les neurones hippocampiques et que cette action peut être modulée par la mutation polyQ ou par le statut de phosphorylation de la protéine sur les sérines 1181 et 1201. Cela aboutit à des modifications des voies de signalisation (Akt, ERK, CREB) activées par le BDNF. Nous mettons également en évidence que la huntingtine sauvage est impliquée dans le soutien exercé par les neurones matures sur les nouveaux neurones, nécessaire à leur survie à long terme et à la formation d’une arborisation dendritique complexe. Le BDNF est l’intermédiaire idéal grâce à ses effets sur la neurogenèse hippocampique. Enfin, la huntingtine sauvage et ses formes mutées (polyQ et phosphorylation sur les sérines 1181 et 1201) sont impliquées dans le comportement anxio-dépressif des souris. / Huntington disease (HD) is a genetic neurodegenerative disorder that affects about 6,000 people in France. Psychiatric manifestations are an important component of the early symptoms of the disease. Indeed, depressive episodes sometimes associated with generalized anxiety are commonly observed during the pre-symptomatic stages of disease. Few information is available about the reasons for this high prevalence.The allele responsible for the disease encodes a protein called huntingtin (HTT) whose polyglutamine expansion (polyQ) in the N-terminal region is longer than in the non-pathogenic HTT. Huntingtin is involved in various cellular functions including the transport and the expression of a neurotrophic factor, the Brain-Derived Neurotrophic Factor (BDNF). This factor is also known for its role in the regulation of mood, adult hippocampal neurogenesis, and in the therapeutic response to antidepressants.We hypothesized that huntingtin, in addition to its known roles in the cortex and striatum, may play a role in the hippocampus. Thus, an impaired transport of BDNF in the hippocampus could partly explain the mood disorders observed in HD patients.By an in vivo approach using different mouse models, we demonstrated that huntingtin stimulates vesicular trafficking and secretion of BDNF in hippocampal neurons and that this action may be modulated by the polyQ mutation or by the phosphorylation status of the protein on serines 1181 and 1201. These lead to changes in signaling pathways (Akt, ERK, CREB) activated by BDNF.We also demonstrate that normal huntingtin is involved in the support provided by mature neurons to new neurons for their long-term survival and the formation of a complex dendritic arborization. BDNF is the ideal candidate to mediate these effects on hippocampal neurogenesis. Finally, normal huntingtin and its mutated forms (polyQ and phosphorylated on serines 1181 and 1201) are involved in anxiety and depressive-like phenotype in mice.
24

Huntington disease and breast cancer / maladie de Huntington et cancer du sein

Sousa, Cristovao 11 July 2013 (has links)
La maladie de Huntington (MH) est une maladie neurodégénérative autosomale dominante causée par une expansion anormale de CAG dans le gène codant la huntingtine (HTT) qui se traduit dans la protéine HTT par une répétition de polyglutamine, entrainant la mort neuronale. Néanmoins, la MH entraine aussi le développement de symptômes périphériques comme la HTT est une protéine exprimée de façon ubiquitaire. Notamment, la MH a été associé à une plus faible incidence des cancers, mais les mécanismes sous-jacents ne sont pas décrits. Nous avons étudié le rôle de HTT mutée et sauvage dans le cancer du sein, où la protéine est fortement exprimée. Des modèles murins de cancer du sein (MMTV-PyVT et MMTV-ErbB2) exprimant la HTT mutée (souris knock-in transportant 111 GAC) développent des tumeurs mammaires agressives par rapport aux souris exprimant la HTT sauvage. La transition épithéliale-mésenchymateuse est accélérée avec une augmentation de la motilité cellulaire ainsi que de la formation de métastases. Ces tumeurs accumulent le récepteur tyrosine-kinase HER2 à la membrane, en raison d'un défaut d'endocytose dynamine-dépendante en présence de la HTT mutée. La signalisation accrue de HER2 est responsable de l'agressivité des tumeurs exprimant la HTT mutée, comme en témoigne le traitement trastuzumab, un anticorps dirigé contre HER2 qui restaure la motilité et l'invasion des cellules tumorales porteuses de la mutation responsable de la MH. La HTT sauvage a elle-même un rôle protecteur dans le cancer, retardant l’apparition des métastases en raison d'un potentiel rôle dans l’adhésion intercellulaire. Ainsi, notre travail met en évidence des rôles clés de la HTT mutée et sauvage au cours de la progression du cancer du sein. / Huntington disease (HD) is an autosomal dominant neurodegenerative disorder caused by an abnormal CAG expansion in the huntingtin (HTT) gene. The corresponding polyglutamine expansion in the HTT protein causes specific neuronal death, but the consequences of HTT mutation in other tissues are less well understood. Nevertheless, HD mutation causes peripheral symptoms as HTT is an ubiquitous protein. HD was associated to lower cancer incidence, however, the mechanisms behind this effect were not described. Here we have studied the role of wild-type and mutant HTT in breast cancer, where we found the protein to be highly expressed. We demonstrate that mouse breast cancer models (MMTV-PyVT and MMTV-ErbB2) expressing mutant HTT (knock-in mice carrying 111 CAGs) develop aggressive mammary tumors as compared to control mice. Epithelial-to-mesenchymal transition is enhanced with subsequent increased cell motility and metastasis. These tumors accumulate tyrosine-kinase receptor HER2 at the membrane, due to a dynamin-dependent endocytosis defect in the presence of mutant HTT. HER2 enhanced signaling is responsible for the aggressiveness of the mutant HTT expressing tumors, as demonstrated by Trastuzumab treatment, an antibody against HER2 that restores motility and invasion in tumor cells carrying HD mutation. The wild-type HTT has itself a protective role in cancer, inhibiting metastasis due to a possible role in cellular junction maintenance. Thus, our work unravels a key role of HTT in breast cancer progression, with the mutant HTT triggering the development of aggressive and metastatic tumors.
25

Evolutionarily Conserved Function of Huntingtin in Cellular Dynamics Related to Cell Adhesion and the Cytoskeleton

Thompson, Morgan Nicole 15 March 2013 (has links)
Huntington's disease (HD) is a rare, dominantly inherited neurodegenerative disorder characterized by progressive chorea, emotional and behavioral disturbances, and cognitive decline. The single, causative mutation is an expanded trinucleotide repeat of cytosine, adenosine, and guanine (CAG) of more than 37 residues in the HD gene (currently referred to as HTT). Genetic evidence suggests that the CAG repeat expansion results in a gain of huntingtin function. While huntingtin and its numerous interactors have been implicated in a variety of essential cellular processes, the role of the full-length, endogenous protein remains unclear. Multiple studies have implicated huntingtin in processes related to cytoskeletal structure and dynamics in HD patients and model organisms. However, alterations in cellular dynamics related to the cytoskeleton — including cell adhesion — have not been characterized in a comprehensive, rigorous manner. Using Mus musculus genetic models of the HD mutation and/or deficiency and a Dictyostelium discoideum genetic deficiency model, I have undertaken an investigation of evolutionarily conserved huntingtin function in the cytoskeleton and cell adhesion. The results of these studies support a role for huntingtin in cell-cell and cell-substrate adhesion, as well as maintaining actin cytoskeletal structure. Furthermore, my thesis research sets the stage for future work to elucidate the molecular mechanism by which huntingtin is acting and determine the effect of the CAG repeat expansion on huntingtin function. Evolutionary conservation affords an invaluable tool to identify crucial function(s) of the huntingtin molecule and the effect of the pathogenic HD mutation on function, enabling therapeutic development while providing novel insights into cytoskeletal biology and cell adhesion.
26

The effects of chronic simvastatin treatment on the expression of behavioral symptoms in a transgenic mouse model of Huntington’s disease

Whitmarsh, Ashley 20 December 2013 (has links)
Huntington’s disease (HD) is a heritable, neurodegenerative disorder characterized by motor, cognitive, and psychiatric disturbances. An unstable CAG expansion within the gene normally encoding for the Huntingtin protein is responsible. The expanded mutant form of Huntingtin and the putative protein co-factor Rhes interact and cause cell death within the striatum. We hypothesized chronic treatment with simvastatin, a cholesterol lowering drug, would disrupt the biosynthetical pathway which gives both Rhes and its target cells binding sites and render Rhes inactive. Healthy and HD mice were treated with simvastatin or a vehicle. Animals’ motor behavior was assessed with three separate tests over the first four months of life. No significant differences were found between the HD groups; however, the HD treated animals’ performance on the rotarod test, at month 4, was intermediate between healthy mice and HD vehicle treated mice. The results hint at simvastatin’s therapeutic potential, but are interpreted cautiously.
27

Charakterizace imunitních buněk a sledování změn zánětlivých proteinů u miniprasečího modelu Huntingtonovy choroby / Characterization of immune cells and monitoring changes of inflammatory proteins in minipig model of Huntington's disease

Butalová, Nikola January 2017 (has links)
The Huntington disease (HD) is a hereditary neuro-degenerative disorder caused by a mutation of the huntigtin gene that codes a protein of the same name. The mutated form of the huntigtin gene plays its part in many pathological interactions and influences a number of cellular mechanisms, including the immune system that could serve as a modifier of the neuropathology of the disease. The cells of the monocyte-macrophage system express cytokines whose production changes in relation to the activation of the cell. The presence of the mutated huntingtin protein in these cells renders them hyper-responsive to immunity incentives leading to changes in the production of cytokines. These differences are discernible a few years prior to the appearance of the symptoms. Therefore, the changes in the levels of certain cytokines could serve as appropriate biomarkers for monitoring of the onset of the disease and its progression. The HD pathogenesis includes an inflammation of the central neutral system. Inflammatory changes in peripheral tissues could reflect inflammatory processes in the central neural system. A miniature TgHD pig could represent an appropriate model organism for studying of the impact of the mHtt on the immune system. This model enables to observe a slow progression of the disease. Changes in...
28

Alterations in mRNA 3′UTR Isoform Abundance Accompany Gene Expression Changes in Huntington's Disease

Romo, Lindsay S. 10 July 2017 (has links)
Huntington’s disease is a neurodegenerative disorder caused by expansion of the CAG repeat in huntingtin exon 1. Early studies demonstrated the huntingtin gene is transcribed into two 3′UTR isoforms in normal human tissue. Decades later, researchers identified a truncated huntingtin mRNA isoform in disease but not control human brain. We speculated the amount of huntingtin 3′UTR isoforms might also vary between control and Huntington’s disease brains. We provide evidence that the abundance of huntingtin 3′UTR isoforms, including a novel mid-3′UTR isoform, differs between patient and control neural stem cells, fibroblasts, motor cortex, and cerebellum. Both alleles of huntingtin contribute to isoform changes. We show huntingtin 3′UTR isoforms are metabolized differently. The long and mid isoforms have shorter half-lives, shorter polyA tails, and more microRNA and RNA binding protein sites than the short isoform. 3′UTR Isoform changes are not limited to huntingtin. Isoforms from 11% of genes change abundance in Huntington’s motor cortex. Only 17% of genes with isoform alterations are differentially expressed in disease tissue. However, gene ontology analysis suggests they share common pathways with differentially expressed genes. We demonstrate knockdown of the RNA binding protein CNOT6 in control fibroblasts results in huntingtin isoform changes similar to those in disease fibroblasts. This study further characterizes Huntington’s disease molecular pathology and suggests RNA binding protein expression may influence mRNA isoform expression in the Huntington’s disease brain.
29

Towards Trans-Splicing Gene Therapy for HD : Intronic Targets Identification in the Huntingtin Gene / Vers la mise au point d’une thérapie génique par trans-épissage pour la maladie de Huntington : identification de cibles introniques dans le gène Huntingtine

Maire, Séverine 09 March 2018 (has links)
La maladie de Huntington (MH) est une maladie autosomale dominante causée par une expansion de la répétition CAG codant pour une expansion de la polyglutamine dans le premier exon du gène Huntingtine (HTT). Ce gène code pour une protéine ubiquitaire dont la mutation entraine de graves symptômes moteurs, psychiatriques et cognitifs, dus à la dégénérescence spécifique des neurones GABAergique épineux moyens du striatum. Nous proposons d'utiliser le trans-épissage pour développer un vecteur de thérapie génique qui réduira significativement voir éliminera l'expression de la protéine mutée tout en restaurant un niveau physiologique de HTT normale dans les cellules affectées par la mutation du gène Huntingtine. Cette technologie est basée sur le remplacement de l'exon muté par un exon sans mutation pendant l'étape de maturation de l'ARNm. Du fait du caractère dominant de la mutation,l'efficacité thérapeutique nécessitera une réaction de trans-épissage très efficace capable de convertir une portion significative de pre-ARNm HTT mutés en en ARNm HTT normaux. Nous avons donc développé un système rapporteur fluorescent permettant la détection des évènements de trans-épissage afin d’identifier les séquences les plus performantes parmi une centaine de molécules candidates. Nous avons validé notre stratégie de criblage basée sur la fluorescence et réalisé le criblage sur plusieurs introns HTT (3, 9 et 20) qui ont démontré des zones favorables au trans-épissage. Une méthode de quantification directe et absolue du taux de trans-épissage a également été validée pour déterminer très précisément le taux de correction. L’ensemble de ce travail a permis de contribuer à la mise en évidence de la faisabilité du trans-épissage dans le contexte de la MH. / Huntington’s disease (HD) is an autosomal dominant genetic disorder caused by the expansion of a CAG repeat encoding a polyglutamine tract in the first exon of the Huntingtin gene (HTT). This gene encode a ubiquitous protein in which mutation lead to severe motor, psychiatric and cognitive deficits and causes degeneration of specific neuronal populations, in particular the GABAergic medium spiny neurons of the striatum. We propose to use trans-splicing to develop a gene therapy vector that will significantly reduce or eliminate the expression of the mutant protein while restoring a physiological level of normal HTT in cells affected by the HD mutation. This technology is based on replacement of the mutated exon by a normal version during the mRNA maturation process. HTT mutation being dominant, therapeutic benefits necessitates a highly efficient trans-splicing reaction that would convert a significant proportion of mutant-HTT pre-mRNA into normal HTT mRNA. For this purpose, we developed a fluorescent reporter system enabling the detection of trans-splicing events in high content screening in order to identify the most potent trans-splicing sequences among hundreds of molecules. We validated our fluorescent screening strategy and implement trans-splicing screening on 3 HTT introns (3, 9 and 20), in which we demonstrated the presence of hotspot promoting trans-splicing reactions. A direct and absolute quantification method was also validated to accurately assess the correction rate. Overall, this work generated additional evidences of trans-splicing feasibility in HD.
30

Analysis of the role of arginine methylation in the pathogenesis of Huntington’s disease

Migazzi, Alice 25 October 2019 (has links)
Huntington's disease (HD) is a fatal neurodegenerative disorder characterized by progressive loss of striatal and cortical neurons. HD is caused by an abnormal polyglutamine (polyQ) expansion in Huntingtin protein (HTT). HTT controls vesicular trafficking along axons in neurons through interaction with components of the molecular motor machinery. Arginine methylation is one of the most abundant post-translational modifications (PTMs) and is catalyzed by protein arginine methyltransferases (PRMTs). Recent evidence supports a key role for arginine methylation in neurodegeneration and particularly in polyglutamine diseases. However, whether HTT is methylated at arginine residues has not been investigated yet and the role of arginine methylation in HD pathogenesis remains to be fully elucidated. In this thesis, I show that vesicle-associated HTT is methylated in vivo at two evolutionarily conserved arginine residues, namely R101 and R118. Methylation of HTT at R118 is catalyzed by Protein Arginine Methyltransferase 6 (PRMT6), which localizes on vesicles together with HTT, whereas further analyses are required to identify the enzyme(s) responsible for R101 methylation. Interestingly, loss of PRMT6-mediated R118 methylation reduces the association of HTT with vesicles, impairs anterograde axonal transport and exacerbates polyQ-expanded HTT toxicity. Conversely, PRMT6 overexpression improves the global efficiency of anterograde axonal transport and rescues cell death in neurons expressing polyQ-expanded HTT. These findings establish a crucial role of arginine methylation as a modulator of both normal HTT function and polyQ-expanded HTT toxicity and identify PRMT6 as a novel modifier of HD pathogenesis. Importantly, defects in HTT methylation may contribute to neurodegeneration in HD and promoting arginine methylation of HTT might represent a new therapeutic strategy for HD.

Page generated in 0.0637 seconds