Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 14.244
Filtrer
1.
BMJ Open ; 14(7): e082161, 2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-39089721

RÉSUMÉ

INTRODUCTION: Huntington's disease (HD) is an inherited neurodegenerative disease causing progressive cognitive and motor decline, largely due to basal ganglia (BG) atrophy. Rhythmic training offers promise as therapy to counteract BG-regulated deficits. We have developed HD-DRUM, a tablet-based app to enhance movement synchronisation skills and improve cognitive and motor abilities in people with HD. This paper outlines a randomised controlled unblinded trial protocol to determine the feasibility of a larger effectiveness trial for HD-DRUM. Additionally, the trial investigates cognitive and motor function measures, along with brain microstructure, aiming to advance our understanding of the neural mechanisms underlying training effects. METHODS, DESIGN AND ANALYSIS: 50 individuals with HD, confirmed by genetic testing, and a Total Functional Capacity (TFC) score of 9-13, will be recruited into a two-arm randomised controlled feasibility trial. Consenting individuals with HD will be randomised to the intervention group, which entails 8 weeks of at-home usage of HD-DRUM or a usual-activity control group. All participants will undergo cognitive and motor assessments, alongside ultra-strong gradient (300 mT/m) brain microstructural MRI before and after the 8-week period. The feasibility assessment will encompass recruitment, retention, adherence and acceptability of HD-DRUM following prespecified criteria. The study will also evaluate variations in cognitive and motor performance and brain microstructure changes resulting from the intervention to determine effect size estimates for future sample size calculations. ETHICS AND DISSEMINATION: The study has received favourable ethical opinion from the Wales Research Ethics Committee 2 (REC reference: 22/WA/0147) and is sponsored by Cardiff University (SPON1895-22) (Research Integrity, Governance and Ethics Team, Research & Innovation Services, Cardiff University, second Floor, Lakeside Building, University Hospital of Wales, Cardiff, CF14 4XW). Findings will be disseminated to researchers and clinicians in peer-reviewed publications and conference presentations, and to participants, carers and the general public via newsletters and public engagement activities. Data will be shared with the research community via the Enroll-HD platform. TRIAL REGISTRATION NUMBER: ISRCTN11906973.


Sujet(s)
Études de faisabilité , Maladie de Huntington , Humains , Maladie de Huntington/complications , Maladie de Huntington/thérapie , Applications mobiles , Cognition , Essais contrôlés randomisés comme sujet , Adulte , Imagerie par résonance magnétique , Mâle , Traitement par les exercices physiques/méthodes , Femelle
2.
Nat Commun ; 15(1): 6534, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39095390

RÉSUMÉ

Huntington's disease (HD) causes selective degeneration of striatal and cortical neurons, resulting in cell mosaicism of coexisting still functional and dysfunctional cells. The impact of non-cell autonomous mechanisms between these cellular states is poorly understood. Here we generated telencephalic organoids with healthy or HD cells, grown separately or as mosaics of the two genotypes. Single-cell RNA sequencing revealed neurodevelopmental abnormalities in the ventral fate acquisition of HD organoids, confirmed by cytoarchitectural and transcriptional defects leading to fewer GABAergic neurons, while dorsal populations showed milder phenotypes mainly in maturation trajectory. Healthy cells in mosaic organoids restored HD cell identity, trajectories, synaptic density, and communication pathways upon cell-cell contact, while showing no significant alterations when grown with HD cells. These findings highlight cell-type-specific alterations in HD and beneficial non-cell autonomous effects of healthy cells, emphasizing the therapeutic potential of modulating cell-cell communication in disease progression and treatment.


Sujet(s)
Maladie de Huntington , Organoïdes , Phénotype , Télencéphale , Maladie de Huntington/anatomopathologie , Maladie de Huntington/génétique , Maladie de Huntington/métabolisme , Organoïdes/anatomopathologie , Organoïdes/métabolisme , Animaux , Télencéphale/anatomopathologie , Télencéphale/cytologie , Télencéphale/métabolisme , Humains , Souris , Neurones GABAergiques/métabolisme , Neurones GABAergiques/anatomopathologie , Analyse sur cellule unique , Communication cellulaire , Mosaïcisme , Neurones/métabolisme , Neurones/anatomopathologie
3.
Life Sci Alliance ; 7(9)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39054288

RÉSUMÉ

Huntington's disease (HD) is a fatal neurodegenerative disorder caused by an expanded CAG tract in the huntingtin (HTT) gene, leading to toxic gains of function. HTT-lowering treatments are in clinical trials, but the risks imposed are unclear. Recent studies have reported on the consequences of widespread HTT loss in mice, where one group described early HTT loss leading to fatal pancreatitis, but later loss as benign. Another group reported no pancreatitis but found widespread neurological phenotypes including subcortical calcification. To better understand the liabilities of widespread HTT loss, we knocked out Htt with two separate tamoxifen-inducible Cre lines. We find that loss of HTT at 2 mo of age leads to progressive tremors and severe subcortical calcification at examination at 14 mo of age but does not result in acute pancreatitis or histological changes in the pancreas. We, in addition, report that HTT loss is followed by sustained induction of circulating neurofilament light chain. These results confirm that global loss of HTT in mice is associated with pronounced risks, including progressive subcortical calcification and neurodegeneration.


Sujet(s)
Modèles animaux de maladie humaine , Protéine huntingtine , Maladie de Huntington , Souris knockout , Pancréas , Animaux , Protéine huntingtine/génétique , Protéine huntingtine/métabolisme , Souris , Pancréas/anatomopathologie , Pancréas/métabolisme , Maladie de Huntington/génétique , Maladie de Huntington/anatomopathologie , Maladie de Huntington/métabolisme , Maladies neurodégénératives/génétique , Maladies neurodégénératives/métabolisme , Maladies neurodégénératives/étiologie , Maladies neurodégénératives/anatomopathologie , Mâle , Calcinose/génétique , Calcinose/anatomopathologie , Phénotype , Femelle
4.
Clin Transl Sci ; 17(7): e13852, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38988035

RÉSUMÉ

N-methyl-D-aspartate receptor (NMDAR)-positive allosteric modulators (PAMs) represent a potential therapeutic strategy for cognitive impairment in disorders associated with NMDAR hypofunction, including Huntington's disease (HD) and Alzheimer's disease. Dalzanemdor (SAGE-718) is a novel, investigational NMDAR PAM being evaluated for the potential treatment of cognitive impairment in these disorders. We report first-in-human, phase I, double-blind, dose-finding studies to assess the safety, tolerability, and clinical pharmacology of dalzanemdor. A single-ascending dose study (dalzanemdor 0.35, 0.75, 1.5, or 3.0 mg vs. placebo) was conducted in healthy participants and included food effects. A multiple-ascending dose study (14 days) was conducted in healthy participants (dalzanemdor 0.5 or 1.0 mg vs. placebo) and HD participants (open-label dalzanemdor 1.0 mg) and included exploratory pharmacodynamics on cognitive performance. Dalzanemdor was generally well tolerated with no adverse events leading to discontinuation. Dalzanemdor exhibited pharmacokinetic parameters appropriate for once-daily dosing. Following single and multiple doses in healthy participants, median terminal half-life was 8-118 h, and the median time to reach maximum plasma concentration was 4-7 h. Exposures were dose-proportional after single dose (6-46 ng/mL) and more than dose-proportional after multiple doses (6-41 ng/mL). With multiple dosing, a steady state was achieved after 11 days in healthy participants and 13 days in HD participants. Dalzanemdor exposure decreased slightly with food. In HD participants, results suggest that dalzanemdor may improve cognitive performance on tests of executive function. These results support continued clinical development of dalzanemdor for the potential treatment of cognitive impairment in disorders of NMDAR hypofunction.


Sujet(s)
Relation dose-effet des médicaments , Maladie de Huntington , Récepteurs du N-méthyl-D-aspartate , Humains , Mâle , Adulte , Maladie de Huntington/traitement médicamenteux , Femelle , Adulte d'âge moyen , Méthode en double aveugle , Récepteurs du N-méthyl-D-aspartate/antagonistes et inhibiteurs , Régulation allostérique/effets des médicaments et des substances chimiques , Jeune adulte , Volontaires sains , Adolescent , Cognition/effets des médicaments et des substances chimiques , Sujet âgé
5.
Cells ; 13(13)2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38994995

RÉSUMÉ

Huntington's disease (HD) is a rare but progressive and devastating neurodegenerative disease characterized by involuntary movements, cognitive decline, executive dysfunction, and neuropsychiatric conditions such as anxiety and depression. It follows an autosomal dominant inheritance pattern. Thus, a child who has a parent with the mutated huntingtin (mHTT) gene has a 50% chance of developing the disease. Since the HTT protein is involved in many critical cellular processes, including neurogenesis, brain development, energy metabolism, transcriptional regulation, synaptic activity, vesicle trafficking, cell signaling, and autophagy, its aberrant aggregates lead to the disruption of numerous cellular pathways and neurodegeneration. Essential heavy metals are vital at low concentrations; however, at higher concentrations, they can exacerbate HD by disrupting glial-neuronal communication and/or causing dysbiosis (disturbance in the gut microbiota, GM), both of which can lead to neuroinflammation and further neurodegeneration. Here, we discuss in detail the interactions of iron, manganese, and copper with glial-neuron communication and GM and indicate how this knowledge may pave the way for the development of a new generation of disease-modifying therapies in HD.


Sujet(s)
Microbiome gastro-intestinal , Maladie de Huntington , Métaux lourds , Névroglie , Maladie de Huntington/microbiologie , Maladie de Huntington/métabolisme , Maladie de Huntington/anatomopathologie , Humains , Névroglie/métabolisme , Névroglie/anatomopathologie , Métaux lourds/métabolisme , Métaux lourds/toxicité , Animaux
6.
PLoS One ; 19(7): e0305358, 2024.
Article de Anglais | MEDLINE | ID: mdl-39008492

RÉSUMÉ

BACKGROUND: Huntington's disease (HD) is an extremely harmful autosomal inherited neurodegenerative disease. Motor dysfunction, mental disorder, and cognitive deficits are the characteristic features of this disease. The current study examined whether 6-shogaol has a protective effect against 3-Nitropropionic Acid (3-NPA)-induced HD in rats. METHODS: A total of thirty male Wistar rats received 6-shogaol (10 and 20 mg/kg, per oral) an hour before injection of 3-NPA (10 mg/kg i.p.) for 15 days. Behavioral tests were performed, including narrow beam walk, rotarod test, and grip strength test. Biochemical tests promoting oxidative stress were evaluated [superoxide dismutase (SOD), reduced glutathione (GSH), catalase (CAT) and malondialdehyde (MDA)], including changes to neurotransmitters serotonin (5-HT), dopamine (DA), norepinephrine (NE), homovanillic acid (HVA), (3,4-dihydroxyphenylacetic acid (DOPAC), γ-aminobutyric acid (GABA), and 5-hydroxy indole acetic acid (5-HIAA), nuclear factor kappa-B (NF-κB), tumor necrosis factor-α (TNF-α), interleukins-1ß (IL-1ß), IL-6, brain-derived neurotrophic factor (BDNF), and nuclear factor erythroid 2-related factor 2 (Nrf2). The 6-shogaol was docked to the active site of TNF-α (2AZ5), NF-κB (1SVC), BDNF) [1B8M], and Nrf2 [5FZN] proteins using AutoDock tools. RESULTS: The 6-shogaol group significantly improved behavioral activity over the 3-NPA-injected control rats. Moreover, 3-NPA-induced significantly altered neurotransmitters, biochemical and neuroinflammatory indices, which could efficiently be reversed by 6-shogaol. The 6-shogaol showed favorable negative binding energies at -9.271 (BDNF) kcal/mol. CONCLUSIONS: The present investigation demonstrated the neuroprotective effects of 6-shogaol in an experimental animal paradigm against 3-NPA-induced HD in rats. The suggested mechanism is supported by immunohistochemical analysis and western blots, although more research is necessary for definite confirmation.


Sujet(s)
Facteur neurotrophique dérivé du cerveau , Catéchols , Cytokines , Maladie de Huntington , Simulation de docking moléculaire , Facteur-2 apparenté à NF-E2 , Facteur de transcription NF-kappa B , Composés nitrés , Propionates , Rat Wistar , Animaux , Maladie de Huntington/métabolisme , Maladie de Huntington/induit chimiquement , Maladie de Huntington/traitement médicamenteux , Propionates/pharmacologie , Mâle , Facteur neurotrophique dérivé du cerveau/métabolisme , Rats , Facteur de transcription NF-kappa B/métabolisme , Facteur-2 apparenté à NF-E2/métabolisme , Catéchols/pharmacologie , Catéchols/composition chimique , Cytokines/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques , Comportement animal/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie
7.
J Huntingtons Dis ; 13(2): 119-131, 2024.
Article de Anglais | MEDLINE | ID: mdl-38968054

RÉSUMÉ

The field of Huntington's disease research covers many different scientific disciplines, from molecular biology all the way through to clinical practice, and as our understanding of the disease has progressed over the decades, a great deal of different terminology has accrued. The field is also renowned for its collaborative spirit and use of standardized reagents, assays, datasets, models, and clinical measures, so the use of standardized terms is especially important. We have set out to determine, through a consensus exercise involving basic and clinical scientists working in the field, the most appropriate language to use across disciplines. Nominally, this article will serve as the style guide for the Journal of Huntington's Disease (JHD), the only journal devoted exclusively to HD, and we lay out the preferred and standardized terminology and nomenclature for use in JHD publications. However, we hope that this article will also serve as a useful resource to the HD research community at large and that these recommended naming conventions will be adopted widely.


Sujet(s)
Maladie de Huntington , Terminologie comme sujet , Maladie de Huntington/classification , Maladie de Huntington/diagnostic , Humains , Recherche biomédicale/normes
8.
Sci Adv ; 10(29): eado5264, 2024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39028820

RÉSUMÉ

Huntingtin protein, mutated in Huntington's disease, is implicated in nucleic acid-mediated processes, yet the evidence for direct huntingtin-nucleic acid interaction is limited. Here, we show wild-type and mutant huntingtin copurify with nucleic acids, primarily RNA, and interact directly with G-rich RNAs in in vitro assays. Huntingtin RNA-immunoprecipitation sequencing from patient-derived fibroblasts and neuronal progenitor cells expressing wild-type and mutant huntingtin revealed long noncoding RNA NEAT1 as a significantly enriched transcript. Altered NEAT1 levels were evident in Huntington's disease cells and postmortem brain tissues, and huntingtin knockdown decreased NEAT1 levels. Huntingtin colocalized with NEAT1 in paraspeckles, and we identified a high-affinity RNA motif preferred by huntingtin. This study highlights NEAT1 as a huntingtin interactor, demonstrating huntingtin's involvement in RNA-mediated functions and paraspeckle regulation.


Sujet(s)
Protéine huntingtine , Maladie de Huntington , ARN long non codant , Protéines de liaison à l'ARN , Humains , Protéine huntingtine/métabolisme , Protéine huntingtine/génétique , ARN long non codant/métabolisme , ARN long non codant/génétique , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Maladie de Huntington/métabolisme , Maladie de Huntington/génétique , Maladie de Huntington/anatomopathologie , Liaison aux protéines , Fibroblastes/métabolisme , Mutation
9.
Sci Rep ; 14(1): 15729, 2024 07 08.
Article de Anglais | MEDLINE | ID: mdl-38977715

RÉSUMÉ

Huntington's disease (HD) is a hereditary neurodegenerative disorder caused by the extension of the CAG repeats in exon 1 of the HTT gene and is transmitted in a dominant manner. The present study aimed to assess whether patients' sex, in the context of mutated and normal allele length, contributes to age on onset (AO) of HD. The study population comprised a large cohort of 3723 HD patients from the European Huntington's Disease Network's REGISTRY database collected at 160 sites across 17 European countries and in one location outside Europe. The data were analyzed using regression models and factorial analysis of variance (ANOVA) considering both mutated allele length and sex as predictors of patients' AO. AO, as described by the rater's estimate, was found to be later in affected women than in men across the whole population. This difference was most pronounced in a subgroup of 1273 patients with relatively short variants of the mutated allele (40-45 CAG repeats) and normal alleles in a higher half of length distribution-namely, more than 17 CAG repeats; however, it was also observed in each group. Our results presented in this observational study point to sex-related differences in AO, most pronounced in the presence of the short mutated and long normal allele, which may add to understanding the dynamics of AO in Huntington's Disease.Trial registration: ClinicalTrials.gov identifier NCT01590589.


Sujet(s)
Âge de début , Protéine huntingtine , Maladie de Huntington , Humains , Maladie de Huntington/génétique , Mâle , Femelle , Adulte d'âge moyen , Adulte , Protéine huntingtine/génétique , Allèles , Répétitions de trinucléotides/génétique , Expansion de trinucléotide répété/génétique , Facteurs sexuels , Sujet âgé , Mutation , Europe/épidémiologie
10.
Parkinsonism Relat Disord ; 125: 107048, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38959686

RÉSUMÉ

INTRODUCTION: Huntington's disease (HD) is a hereditary condition caused by the expansion of the CAG trinucleotide in the huntingtin gene on chromosome 4, resulting in motor, cognitive, and psychiatric disorders that significantly impact patients' quality of life. Despite the lack of effective treatments for the disease, various surgical strategies have been explored to alleviate symptoms and slow its progression. METHODOLOGY: A comprehensive systematic literature review was conducted, including MeSH terms, yielding only 38 articles that were categorized based on the surgical procedure. The study aimed to describe the types of surgeries performed and their efficacy in HD patients. RESULTS: Deep brain stimulation (DBS) involved 41 predominantly male patients with bilateral implantation in the globus pallidus, showing a preoperative Unified Huntington's Disease Rating Scale (UHDRS) score of 60.25 ± 16.13 and a marked postoperative value of 48.54 ± 13.93 with a p < 0.018 at one year and p < 0.040 at three years. Patients experienced improvement in hyperkinesia but worsening of bradykinesia. Additionally, cell transplantation in 119 patients resulted in a lower preoperative UHDRS score of 34.61 ± 14.61 and a significant postoperative difference of 32.93 ± 15.87 (p < 0.016), respectively, in the first to third years of following. Some now, less used procedures were crucial for understanding brain function, such as pallidotomies in 3 patients, showing only a 25 % difference from their baseline. CONCLUSION: Despite advancements in technology, there is still no curative treatment, only palliative options. Promising treatments like trophic factor implantation offer new prospects for the future.


Sujet(s)
Stimulation cérébrale profonde , Maladie de Huntington , Maladie de Huntington/chirurgie , Maladie de Huntington/thérapie , Humains , Procédures de neurochirurgie/méthodes
11.
Int J Mol Sci ; 25(14)2024 Jul 14.
Article de Anglais | MEDLINE | ID: mdl-39062952

RÉSUMÉ

Gastrodin (GAS) is the main chemical component of the traditional Chinese herb Gastrodia elata (called "Tianma" in Chinese), which has been used to treat neurological conditions, including headaches, epilepsy, stroke, and memory loss. To our knowledge, it is unclear whether GAS has a therapeutic effect on Huntington's disease (HD). In the present study, we evaluated the effect of GAS on the degradation of mutant huntingtin protein (mHtt) by using PC12 cells transfected with N-terminal mHtt Q74. We found that 0.1-100 µM GAS had no effect on the survival rate of Q23 and Q74 PC12 cells after 24-48 h of incubation. The ubiquitin-proteasome system (UPS) is the main system that clears misfolded proteins in eukaryotic cells. Mutated Htt significantly upregulated total ubiquitinated protein (Ub) expression, decreased chymotrypsin-like, trypsin-like and caspase-like peptidase activity, and reduced the colocalization of the 20S proteasome with mHtt. GAS (25 µM) attenuated all of the abovementioned pathological changes, and the regulatory effect of GAS on mHtt was found to be abolished by MG132, a proteasome inhibitor. The autophagy-lysosome pathway (ALP) is another system for misfolded protein degradation. Although GAS downregulated the expression of autophagy markers (LC3II and P62), it increased the colocalization of LC3II with lysosomal associated membrane protein 1 (LAMP1), which indicates that ALP was activated. Moreover, GAS prevented mHtt-induced neuronal damage in PC12 cells. GAS has a selective effect on mHtt in Q74 PC12 cells and has no effect on Q23 and proteins encoded by other genes containing long CAGs, such as Rbm33 (10 CAG repeats) and Hcn1 (>30 CAG repeats). Furthermore, oral administration of 100 mg/kg GAS increased grip strength and attenuated mHtt aggregates in B6-hHTT130-N transgenic mice. This is a high dose (100 mg/kg GAS) when compared with experiments on HD mice with other small molecules. We will design more doses to evaluate the dose-response relationship of the inhibition effect of GAS on mHtt in our next study. In summary, GAS can promote the degradation of mHtt by activating the UPS and ALP, making it a potential therapeutic agent for HD.


Sujet(s)
Autophagie , Alcools benzyliques , Glucosides , Protéine huntingtine , Lysosomes , Proteasome endopeptidase complex , Ubiquitine , Animaux , Protéine huntingtine/génétique , Protéine huntingtine/métabolisme , Rats , Proteasome endopeptidase complex/métabolisme , Cellules PC12 , Autophagie/effets des médicaments et des substances chimiques , Lysosomes/métabolisme , Lysosomes/effets des médicaments et des substances chimiques , Ubiquitine/métabolisme , Alcools benzyliques/pharmacologie , Glucosides/pharmacologie , Souris , Maladie de Huntington/métabolisme , Maladie de Huntington/traitement médicamenteux , Maladie de Huntington/génétique , Protéolyse/effets des médicaments et des substances chimiques , Mutation
12.
J Neurosci Res ; 102(7): e25366, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38953592

RÉSUMÉ

Increasing neuroimaging studies have attempted to identify biomarkers of Huntington's disease (HD) progression. Here, we conducted voxel-based meta-analyses of voxel-based morphometry (VBM) studies on HD to investigate the evolution of gray matter volume (GMV) alterations and explore the effects of genetic and clinical features on GMV changes. A systematic review was performed to identify the relevant studies. Meta-analyses of whole-brain VBM studies were performed to assess the regional GMV changes in all HD mutation carriers, in presymptomatic HD (pre-HD), and in symptomatic HD (sym-HD). A quantitative comparison was performed between pre-HD and sym-HD. Meta-regression analyses were used to explore the effects of genetic and clinical features on GMV changes. Twenty-eight studies were included, comparing a total of 1811 HD mutation carriers [including 1150 pre-HD and 560 sym-HD] and 969 healthy controls (HCs). Pre-HD showed decreased GMV in the bilateral caudate nuclei, putamen, insula, anterior cingulate/paracingulate gyri, middle temporal gyri, and left dorsolateral superior frontal gyrus compared with HCs. Compared with pre-HD, GMV decrease in sym-HD extended to the bilateral median cingulate/paracingulate gyri, Rolandic operculum and middle occipital gyri, left amygdala, and superior temporal gyrus. Meta-regression analyses found that age, mean lengths of CAG repeats, and disease burden were negatively associated with GMV atrophy of the bilateral caudate and right insula in all HD mutation carriers. This meta-analysis revealed the pattern of GMV changes from pre-HD to sym-HD, prompting the understanding of HD progression. The pattern of GMV changes may be biomarkers for disease progression in HD.


Sujet(s)
Substance grise , Maladie de Huntington , Neuroimagerie , Maladie de Huntington/imagerie diagnostique , Maladie de Huntington/anatomopathologie , Maladie de Huntington/génétique , Humains , Substance grise/imagerie diagnostique , Substance grise/anatomopathologie , Neuroimagerie/méthodes , Encéphale/anatomopathologie , Encéphale/imagerie diagnostique , Imagerie par résonance magnétique/méthodes
14.
Proc Natl Acad Sci U S A ; 121(32): e2319091121, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39074279

RÉSUMÉ

Understanding the normal function of the Huntingtin (HTT) protein is of significance in the design and implementation of therapeutic strategies for Huntington's disease (HD). Expansion of the CAG repeat in the HTT gene, encoding an expanded polyglutamine (polyQ) repeat within the HTT protein, causes HD and may compromise HTT's normal activity contributing to HD pathology. Here, we investigated the previously defined role of HTT in autophagy specifically through studying HTT's association with ubiquitin. We find that HTT interacts directly with ubiquitin in vitro. Tandem affinity purification was used to identify ubiquitinated and ubiquitin-associated proteins that copurify with a HTT N-terminal fragment under basal conditions. Copurification is enhanced by HTT polyQ expansion and reduced by mimicking HTT serine 421 phosphorylation. The identified HTT-interacting proteins include RNA-binding proteins (RBPs) involved in mRNA translation, proteins enriched in stress granules, the nuclear proteome, the defective ribosomal products (DRiPs) proteome and the brain-derived autophagosomal proteome. To determine whether the proteins interacting with HTT are autophagic targets, HTT knockout (KO) cells and immunoprecipitation of lysosomes were used to investigate autophagy in the absence of HTT. HTT KO was associated with reduced abundance of mitochondrial proteins in the lysosome, indicating a potential compromise in basal mitophagy, and increased lysosomal abundance of RBPs which may result from compensatory up-regulation of starvation-induced macroautophagy. We suggest HTT is critical for appropriate basal clearance of mitochondrial proteins and RBPs, hence reduced HTT proteostatic function with mutation may contribute to the neuropathology of HD.


Sujet(s)
Protéine huntingtine , Lysosomes , Mitochondries , Protéines de liaison à l'ARN , Ubiquitine , Protéine huntingtine/métabolisme , Protéine huntingtine/génétique , Lysosomes/métabolisme , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Humains , Ubiquitine/métabolisme , Mitochondries/métabolisme , Autophagie , Animaux , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Souris , Liaison aux protéines , Maladie de Huntington/métabolisme , Maladie de Huntington/génétique , Maladie de Huntington/anatomopathologie , Peptides/métabolisme
15.
Pharmacol Rep ; 76(4): 693-713, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38982016

RÉSUMÉ

Huntington's disease (HD) is an inherited neurodegenerative disorder caused by an expansion in CAG repeat on huntington (Htt) gene, leading to a degeneration of GABAergic medium spiny neurons (MSNs) in the striatum, resulting in the generation of reactive oxygen species, and decrease antioxidant activity. These pathophysiological alterations impair mitochondrial functions, leading to an increase in involuntary hyperkinetic movement. However, researchers investigated the neuroprotective effect of antioxidants using various animal models. Still, their impact is strictly limited to curtailing oxidative stress and increasing the antioxidant enzyme in the brain, which is less effective in HD. Meanwhile, researchers discovered Mitochondria-targeted antioxidants (MTAXs) that can improve mitochondrial functions and antioxidant activity through the modulation of mitochondrial signaling pathways, including peroxisome proliferator-activated receptor (PPAR)-coactivator 1 (PGC-1α), dynamin-related protein 1 (Drp1), mitochondrial fission protein 1 (Fis1), and Silent mating type information regulation 2 homolog 1 (SIRT-1), showing neuroprotective effects in HD. The present review discusses the clinical and preclinical studies that investigate the neuroprotective effect of MTAXs (SS31, XJB-5-131, MitoQ, bezafibrate, rosiglitazone, meldonium, coenzyme Q10, etc.) in HD. This brief literature review will help to understand the relevance of MTAXs in HD and enlighten the importance of MTAXs in future drug discovery and development.


Sujet(s)
Antioxydants , Maladie de Huntington , Mitochondries , Neuroprotecteurs , Maladie de Huntington/traitement médicamenteux , Maladie de Huntington/métabolisme , Humains , Antioxydants/pharmacologie , Antioxydants/usage thérapeutique , Animaux , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Stress oxydatif/effets des médicaments et des substances chimiques
16.
Int J Mol Sci ; 25(11)2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38892250

RÉSUMÉ

Neurodegenerative diseases are progressive disorders that affect the central nervous system (CNS) and represent the major cause of premature death in the elderly. One of the possible determinants of neurodegeneration is the change in mitochondrial function and content. Altered levels of mitochondrial DNA copy number (mtDNA-CN) in biological fluids have been reported during both the early stages and progression of the diseases. In patients affected by neurodegenerative diseases, changes in mtDNA-CN levels appear to correlate with mitochondrial dysfunction, cognitive decline, disease progression, and ultimately therapeutic interventions. In this review, we report the main results published up to April 2024, regarding the evaluation of mtDNA-CN levels in blood samples from patients affected by Alzheimer's (AD), Parkinson's (PD), and Huntington's diseases (HD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS). The aim is to show a probable link between mtDNA-CN changes and neurodegenerative disorders. Understanding the causes underlying this association could provide useful information on the molecular mechanisms involved in neurodegeneration and offer the development of new diagnostic approaches and therapeutic interventions.


Sujet(s)
Variations de nombre de copies de segment d'ADN , ADN mitochondrial , Mitochondries , Maladies neurodégénératives , Humains , Maladies neurodégénératives/génétique , ADN mitochondrial/génétique , Mitochondries/génétique , Mitochondries/métabolisme , Maladie de Huntington/génétique , Maladie de Huntington/anatomopathologie , Animaux
17.
Acta Neuropathol Commun ; 12(1): 88, 2024 06 05.
Article de Anglais | MEDLINE | ID: mdl-38840253

RÉSUMÉ

Huntington's disease (HD) is an inherited neurodegenerative disorder caused by an expanded CAG repeat in the coding sequence of huntingtin protein. Initially, it predominantly affects medium-sized spiny neurons (MSSNs) of the corpus striatum. No effective treatment is still available, thus urging the identification of potential therapeutic targets. While evidence of mitochondrial structural alterations in HD exists, previous studies mainly employed 2D approaches and were performed outside the strictly native brain context. In this study, we adopted a novel multiscale approach to conduct a comprehensive 3D in situ structural analysis of mitochondrial disturbances in a mouse model of HD. We investigated MSSNs within brain tissue under optimal structural conditions utilizing state-of-the-art 3D imaging technologies, specifically FIB/SEM for the complete imaging of neuronal somas and Electron Tomography for detailed morphological examination, and image processing-based quantitative analysis. Our findings suggest a disruption of the mitochondrial network towards fragmentation in HD. The network of interlaced, slim and long mitochondria observed in healthy conditions transforms into isolated, swollen and short entities, with internal cristae disorganization, cavities and abnormally large matrix granules.


Sujet(s)
Modèles animaux de maladie humaine , Maladie de Huntington , Imagerie tridimensionnelle , Mitochondries , Animaux , Maladie de Huntington/anatomopathologie , Maladie de Huntington/génétique , Maladie de Huntington/métabolisme , Mitochondries/ultrastructure , Mitochondries/anatomopathologie , Mitochondries/métabolisme , Imagerie tridimensionnelle/méthodes , Souris , Souris transgéniques , Encéphale/anatomopathologie , Encéphale/ultrastructure , Encéphale/métabolisme , Microscopie électronique/méthodes , Mâle , Neurones/anatomopathologie , Neurones/ultrastructure , Neurones/métabolisme
18.
J Cell Biol ; 223(8)2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-38913026

RÉSUMÉ

The double-stranded RNA-binding protein Staufen1 (STAU1) regulates a variety of physiological and pathological events via mediating RNA metabolism. STAU1 overabundance was observed in tissues from mouse models and fibroblasts from patients with neurodegenerative diseases, accompanied by enhanced mTOR signaling and impaired autophagic flux, while the underlying mechanism remains elusive. Here, we find that endogenous STAU1 forms dynamic cytoplasmic condensate in normal and tumor cell lines, as well as in mouse Huntington's disease knockin striatal cells. STAU1 condensate recruits target mRNA MTOR at its 5'UTR and promotes its translation both in vitro and in vivo, and thus enhanced formation of STAU1 condensate leads to mTOR hyperactivation and autophagy-lysosome dysfunction. Interference of STAU1 condensate normalizes mTOR levels, ameliorates autophagy-lysosome function, and reduces aggregation of pathological proteins in cellular models of neurodegenerative diseases. These findings highlight the importance of balanced phase separation in physiological processes, suggesting that modulating STAU1 condensate may be a strategy to mitigate the progression of neurodegenerative diseases with STAU1 overabundance.


Sujet(s)
Autophagie , Biosynthèse des protéines , Protéines de liaison à l'ARN , Sérine-thréonine kinases TOR , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/génétique , Animaux , Humains , Autophagie/génétique , Souris , Protéines du cytosquelette/métabolisme , Protéines du cytosquelette/génétique , Maladies neurodégénératives/métabolisme , Maladies neurodégénératives/anatomopathologie , Maladies neurodégénératives/génétique , Lysosomes/métabolisme , Lysosomes/génétique , Transduction du signal , Maladie de Huntington/métabolisme , Maladie de Huntington/anatomopathologie , Maladie de Huntington/génétique
19.
BMC Genomics ; 25(1): 633, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38918688

RÉSUMÉ

Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder featured by abnormal movements, arising from the extensive neuronal loss and glial dysfunction in the striatum. Although the causes and pathogenetic mechanisms of HD are well established, the development of disease-modifying pharmacological therapies for HD remains a formidable challenge. Laduviglusib has demonstrated neuroprotective effects through the enhancement of mitochondrial function in the striatum of HD animal models. Ferroptosis is a nonapoptotic form of cell death that occurs as a consequence of lethal iron-dependent lipid peroxidation and mitochondrial dysfunction. However, the ferroptosis-related mechanisms underlying the neuroprotective effects of laduviglusib in the striatum of HD patients remain largely uncharted. In this study, we leveraged single-nucleus RNA sequencing data obtained from the striatum of HD patients in stages 2-4 to identify differentially expressed genes within distinct cell-type. We subsequently integrated these differentially expressed genes of HD, laduviglusib target genes and ferroptosis-related genes to predict the ferroptosis-related mechanisms underpinning the neuroprotective effects of laduviglusib in HD patients. The Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analyses unveiled that the effects of laduviglusib on direct pathway striatal projection neurons (dSPNs) is mainly associated with Th17 cell differentiation pathways. Conversely, its impact on indirect pathway striatal projection neurons (iSPNs) extends to the Neurotrophin signaling pathway, FoxO signaling pathway, and reactive oxygen species pathway. In microglia, laduviglusib appears to contribute to HD pathology via mechanisms related to Th17 cell differentiation and the FoxO signaling pathway. Further, molecular docking results indicated favorable binding of laduviglusib with PARP1 (associated with dSPNs and iSPNs), SCD (associated with astrocytes), ALOX5 (associated with microglia), and HIF1A (associated with dSPNs, iSPNs, and microglia). In addition, the KEGG results suggest that laduviglusib may enhance mitochondrial function and protect against neuronal loss by targeting ferroptosis-related signaling pathways, particularly mediated by ALOX5 in microglia. These findings provide valuable insights into the potential mechanisms through which laduviglusib exerts its effects on distinct cell-types within the HD striatum.


Sujet(s)
Corps strié , Ferroptose , Maladie de Huntington , Ferroptose/effets des médicaments et des substances chimiques , Ferroptose/génétique , Maladie de Huntington/métabolisme , Maladie de Huntington/génétique , Maladie de Huntington/anatomopathologie , Humains , Corps strié/métabolisme , Corps strié/anatomopathologie , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE