Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
J Pers Med ; 12(10)2022 Sep 22.
Article in English | MEDLINE | ID: mdl-36294700

ABSTRACT

SRX246, an orally available CNS penetrant vasopressin (VP) V1a receptor antagonist, was studied in Huntington's disease (HD) patients with irritability and aggressive behavior in the exploratory phase 2 trial, Safety, Tolerability, and Activity of SRX246 in Irritable HD patients (STAIR). This was a dose-escalation study; subjects received final doses of 120 mg BID, 160 mg BID, or placebo. The compound was safe and well tolerated. In this paper, we summarize the results of exploratory analyses of measures of problematic behaviors, including the Cohen-Mansfield Agitation Inventory (CMAI), Aberrant Behavior Checklist (ABC), Problem Behaviors Assessment-short form (PBA-s), Irritability Scale (IS), Clinical Global Impression (CGI), HD Quality of Life (QoL), and Caregiver Burden questionnaires. In addition to these, we asked subjects and caregivers to record answers to short questions about mood, irritability, and aggressive conduct in an eDiary. STAIR was the first rigorously designed study of behavioral endpoints like these in HD. The exploratory analyses showed that SRX246 reduced aggressive acts. Readily observed behaviors should be used as trial endpoints.

2.
Neurol Clin Pract ; 12(2): 131-138, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35747889

ABSTRACT

Background and Objectives: Suicidality is a common concern in the routine care of persons with Huntington disease (HD) and for the many participants in HD clinical trials. In a previous analysis, we identified baseline and time-dependent factors associated with suicidal ideation and attempts from 2CARE, a large, randomized, double-blind clinical trial. Methods: The present analysis extends our prior methodology to 2 other large interventional HD clinical trials, CARE-HD and CREST-E. Results: We observed relationships across studies between suicidality events and prior suicidal ideation at baseline, antidepressant/anxiolytic use, chorea, increasing age, and several domains in the Unified Huntington Disease Rating Scale (UHDRS) Behavioral Assessment (depressed mood, low self-esteem, aggression, and active suicidality). Discussion: These data may form the basis for a subscale of demographic and UHDRS items with the potential for prospectively identifying suicidality risk in HD clinics and clinical trials. Trial Registration Information: 2CARE and CREST are registered at clinicaltrials.gov. 2CARE NCT00608881, registered February 6, 2008; first enrollment March 2008. CREST-E NCT00712426, registered July 10, 2008; first enrollment September 2009. CARE-HD, not registered; first enrollment July 1997.

3.
Front Physiol ; 12: 663898, 2021.
Article in English | MEDLINE | ID: mdl-34366879

ABSTRACT

There is increasing evidence that impairments of cerebrovascular function and/or abnormalities of the cerebral vasculature might contribute to early neuronal cell loss in Huntington's disease (HD). Studies in both healthy individuals as well as in patients with other neurodegenerative disorders have used an exogenous carbon dioxide (CO2) challenge in conjunction with functional magnetic resonance imaging (fMRI) to assess regional cerebrovascular reactivity (CVR). In this study, we explored potential impairments of CVR in HD. Twelve gene expanded HD individuals, including both pre-symptomatic and early symptomatic HD and eleven healthy controls were administered a gas mixture targeting a 4-8 mmHg increase in CO2 relative to the end-tidal partial pressure of CO2 (P ET CO2) at rest. A Hilbert Transform analysis was used to compute the cross-correlation between the time series of regional BOLD signal changes (ΔBOLD) and increased P ET CO2, and to estimate the response delay of ΔBOLD relative to P ET CO2. After correcting for age, we found that the cross-correlation between the time series for regional ΔBOLD and for P ET CO2 was weaker in HD subjects than in controls in several subcortical white matter regions, including the corpus callosum, subcortical white matter adjacent to rostral and caudal anterior cingulate, rostral and caudal middle frontal, insular, middle temporal, and posterior cingulate areas. In addition, greater volume of dilated perivascular space (PVS) was observed to overlap, primarily along the periphery, with the areas that showed greater ΔBOLD response delay. Our preliminary findings support that alterations in cerebrovascular function occur in HD and may be an important, not as yet considered, contributor to early neuropathology in HD.

4.
Neurology ; 96(6): e890-e894, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33106388

ABSTRACT

OBJECTIVE: To quantify the percent volume of dilated perivascular space (PVS) in the subcortical forebrain in patients with early Huntington disease (HD) and to explore the relationship between PVS and disease severity. METHODS: MRI scans were performed on 25 patients with HD and 23 healthy age-matched controls at Massachusetts General Hospital. The imaging data were analyzed with a novel algorithm to determine regional PVS volume. A fractional logistic regression analysis was used to quantify the association between regional percent PVS volume and (1) disease designation (HD or control) and (2) disease severity as assessed by normalized caudate volume. RESULTS: Patients with HD had the greatest percent volume of dilated PVS in the putamen (left putamen: odds ratio 2.06 [95% confidence interval (CI) 1.62-2.62], HD 3.27% [95% CI 2.83-3.78] vs controls 1.62% [95% CI 1.32-1.97], p fdr < 0.001; right putamen: odds ratio 1.66 [95% CI 1.33-2.08], HD 3.43% [95% CI 2.94-4.01] vs controls 2.09% [95% CI 1.79-2.45], p fdr < 0.001) and several subcortical white matter regions compared to controls. Dilated PVS increased with disease severity. CONCLUSIONS: The objective quantification of dilated PVS suggests that PVS burden is high, is associated with disease severity, and may affect the distribution and success of treatments administered either intrathecally such as antisense oligonucleotides or by intraparenchymal administration such as cell and gene therapies. CLASSIFICATION OF EVIDENCE: This study provides Class II evidence that increased dilated PVS is associated with worse HD severity. The study is rated Class II because of the cross-sectional design.


Subject(s)
Glymphatic System/pathology , Huntington Disease/pathology , Huntington Disease/physiopathology , Putamen/pathology , White Matter/pathology , Adult , Cross-Sectional Studies , Female , Glymphatic System/diagnostic imaging , Humans , Huntington Disease/diagnostic imaging , Image Interpretation, Computer-Assisted , Magnetic Resonance Imaging , Male , Middle Aged , Putamen/diagnostic imaging , Severity of Illness Index , White Matter/diagnostic imaging
5.
J Clin Med ; 9(11)2020 Nov 16.
Article in English | MEDLINE | ID: mdl-33207828

ABSTRACT

SRX246 is a vasopressin (AVP) 1a receptor antagonist that crosses the blood-brain barrier. It reduced impulsive aggression, fear, depression and anxiety in animal models, blocked the actions of intranasal AVP on aggression/fear circuits in an experimental medicine fMRI study and demonstrated excellent safety in Phase 1 multiple-ascending dose clinical trials. The present study was a 3-arm, multicenter, randomized, placebo-controlled, double-blind, 12-week, dose escalation study of SRX246 in early symptomatic Huntington's disease (HD) patients with irritability. Our goal was to determine whether SRX246 was safe and well tolerated in these HD patients given its potential use for the treatment of problematic neuropsychiatric symptoms. Participants were randomized to receive placebo or to escalate to 120 mg twice daily or 160 mg twice daily doses of SRX246. Assessments included standard safety tests, the Unified Huntington's Disease Rating Scale (UHDRS), and exploratory measures of problem behaviors. The groups had comparable demographics, features of HD and baseline irritability. Eighty-two out of 106 subjects randomized completed the trial on their assigned dose of drug. One-sided exact-method confidence interval tests were used to reject the null hypothesis of inferior tolerability or safety for each dose group vs. placebo. Apathy and suicidality were not affected by SRX246. Most adverse events in the active arms were considered unlikely to be related to SRX246. The compound was safe and well tolerated in HD patients and can be moved forward as a candidate to treat irritability and aggression.

6.
Mov Disord Clin Pract ; 5(3): 306-311, 2018.
Article in English | MEDLINE | ID: mdl-30363459

ABSTRACT

BACKGROUND: There is increasing evidence that the effects of Huntington's disease (HD) extend beyond the central nervous system. In particular, significant cardiac dysfunction has been described in transgenic mouse models and suggested in symptomatic patients, in whom cardiac involvement could provide an independent risk for sudden cardiac death. METHODS: Standard 12-lead electrocardiograms (ECGs) obtained at screening from 590 early symptomatic (Stage 1 and 2) HD patients participating in a multi-site Phase III study were analyzed. RESULTS: Evaluating only those ECGs in individuals not on medications or with potentially contributing medical conditions, the prevalence of bradycardia was 28.3% (marked in 5.8%), prolonged QRS 4.9%, intraventricular conduction delay 3.4%, right bundle branch block 1.3%, and QTc prolongation 3.7%. CONCLUSION: Significant cardiac abnormalities, characterized primarily by conduction abnormalities, were found in a larger than expected number of patients. Abnormal intraventricular conduction may lead to increased risk for arrhythmia and may be compounded by prescription of QT-prolonging medications.

7.
Neurology ; 89(6): 594-601, 2017 Aug 08.
Article in English | MEDLINE | ID: mdl-28701493

ABSTRACT

OBJECTIVE: To investigate whether creatine administration could slow progressive functional decline in adults with early symptoms of Huntington disease. METHODS: We conducted a multicenter, randomized, double-blind, placebo-controlled study of up to 40 g daily of creatine monohydrate in participants with stage I and II HD treated for up to 48 months. The primary outcome measure was the rate of change in total functional capacity (TFC) between baseline and end of follow-up. Secondary outcome measures included changes in additional clinical scores, tolerability, and quality of life. Safety was assessed by adverse events and laboratory studies. RESULTS: At 46 sites in North America, Australia, and New Zealand, 553 participants were randomized to creatine (275) or placebo (278). The trial was designed to enroll 650 patients, but was halted for futility after the first interim analysis. The estimated rates of decline in the primary outcome measure (TFC) were 0.82 points per year for participants on creatine, 0.70 points per year for participants on placebo, favoring placebo (nominal 95% confidence limits -0.11 to 0.35). Adverse events, mainly gastrointestinal, were significantly more common in participants on creatine. Serious adverse events, including deaths, were more frequent in the placebo group. Subgroup analysis suggested that men and women may respond differently to creatine treatment. CONCLUSIONS: Our data do not support the use of creatine treatment for delaying functional decline in early manifest HD. CLINICALTRIALSGOV IDENTIFIER: NCT00712426. CLASSIFICATION OF EVIDENCE: This study provides Class II evidence that for patients with early symptomatic HD, creatine monohydrate is not beneficial for slowing functional decline.


Subject(s)
Creatine/administration & dosage , Huntington Disease/drug therapy , Neuroprotective Agents/administration & dosage , Australia , Creatine/adverse effects , Disease Progression , Double-Blind Method , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neuroprotective Agents/adverse effects , New Zealand , North America , Quality of Life , Treatment Outcome
8.
Proc Natl Acad Sci U S A ; 114(23): E4676-E4685, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28533375

ABSTRACT

The activity of the transcription factor nuclear factor-erythroid 2 p45-derived factor 2 (NRF2) is orchestrated and amplified through enhanced transcription of antioxidant and antiinflammatory target genes. The present study has characterized a triazole-containing inducer of NRF2 and elucidated the mechanism by which this molecule activates NRF2 signaling. In a highly selective manner, the compound covalently modifies a critical stress-sensor cysteine (C151) of the E3 ligase substrate adaptor protein Kelch-like ECH-associated protein 1 (KEAP1), the primary negative regulator of NRF2. We further used this inducer to probe the functional consequences of selective activation of NRF2 signaling in Huntington's disease (HD) mouse and human model systems. Surprisingly, we discovered a muted NRF2 activation response in human HD neural stem cells, which was restored by genetic correction of the disease-causing mutation. In contrast, selective activation of NRF2 signaling potently repressed the release of the proinflammatory cytokine IL-6 in primary mouse HD and WT microglia and astrocytes. Moreover, in primary monocytes from HD patients and healthy subjects, NRF2 induction repressed expression of the proinflammatory cytokines IL-1, IL-6, IL-8, and TNFα. Together, our results demonstrate a multifaceted protective potential of NRF2 signaling in key cell types relevant to HD pathology.


Subject(s)
Huntington Disease/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Adult , Aged , Animals , Brain/drug effects , Brain/metabolism , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Female , HEK293 Cells , Humans , Huntington Disease/genetics , Kelch-Like ECH-Associated Protein 1/chemistry , MPTP Poisoning/metabolism , MPTP Poisoning/prevention & control , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Middle Aged , NF-E2-Related Factor 2/chemistry , Neural Stem Cells/metabolism , Neuroprotective Agents/pharmacology , Protein Conformation/drug effects , Rats , Signal Transduction
9.
Ann Clin Transl Neurol ; 2(7): 756-68, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26273688

ABSTRACT

OBJECTIVE: Huntington's disease (HD) is a rare neurodegenerative disease caused by the expansion of an N-terminal repeat in the huntingtin protein. The protein is expressed in all cells in the body; hence, peripheral tissues, such as blood, may recapitulate processes in the brain. The plasma metabolome may provide a window into active processes that influence brain health and a unique opportunity to noninvasively identify processes that may contribute to neurodegeneration. Alterations in metabolic pathways in brain have been shown to profoundly impact HD. Therefore, identification and quantification of critical metabolomic perturbations could provide novel biomarkers for disease onset and disease progression. METHODS: We analyzed the plasma metabolomic profiles from 52 premanifest (PHD), 102 early symptomatic HD, and 140 healthy controls (NC) using liquid chromatography coupled with a highly sensitive electrochemical detection platform. RESULTS: Alterations in tryptophan, tyrosine, purine, and antioxidant pathways were identified, including many related to energetic and oxidative stress and derived from the gut microbiome. Multivariate statistical modeling demonstrated mutually distinct metabolomic profiles, suggesting that the processes that determine onset were likely distinct from those that determine progression. Gut microbiome-derived metabolites particularly differentiated the PHD metabolome, while the symptomatic HD metabolome was increasingly influenced by metabolites that may reflect mutant huntingtin toxicity and neurodegeneration. INTERPRETATION: Understanding the complex changes in the delicate balance of the metabolome and the gut microbiome in HD, and how they relate to disease onset, progression, and phenotypic variability in HD are critical questions for future research.

10.
Neurology ; 82(10): 850-7, 2014 Mar 11.
Article in English | MEDLINE | ID: mdl-24510496

ABSTRACT

OBJECTIVE: To assess the safety and tolerability of high-dose creatine, the feasibility of enrolling premanifest and 50% at-risk subjects in a prevention trial, and the potential of cognitive, imaging, and blood markers. METHODS: Sixty-four eligible consenting participants were randomly allocated (1:1) to 15 g twice daily of creatine monohydrate or placebo for a 6-month double-blind phase followed by a 12-month open-label extension. Subjects included premanifest (tested) and at-risk (not tested) individuals without clinical symptoms or signs of Huntington disease (HD). Primary outcomes were safety and tolerability. Exploratory endpoints included fine motor, visuospatial, and memory performance; structural and diffusion MRI; and selected blood markers. RESULTS: Forty-seven HD carriers and 17 non-HD controls were enrolled. Fifteen discontinued treatment (2 assigned to placebo); all were followed for the entire study period. Primary analysis was by intent to treat. The most common adverse events were gastrointestinal. Neuroimaging demonstrated treatment-related slowing of cortical and striatal atrophy at 6 and 18 months. CONCLUSION: We describe a design that preserves the autonomy of subjects not wanting genetic testing while including controls for assessing the specificity of treatment effects. Our results demonstrate the feasibility of prevention trials for HD and the safety of high-dose creatine, provide possible evidence of disease modification, support future studies of creatine, and illustrate the value of prodromal biomarkers. CLASSIFICATION OF EVIDENCE: This study provides Class I evidence that high-dose creatine is safe and tolerable.


Subject(s)
Brain/drug effects , Cognition Disorders/prevention & control , Creatine/pharmacology , Huntington Disease/prevention & control , Adult , Atrophy , Biomarkers/blood , Brain/pathology , Cognition Disorders/blood , Cognition Disorders/pathology , Creatine/administration & dosage , Creatine/adverse effects , Cross-Over Studies , Diffusion Magnetic Resonance Imaging , Double-Blind Method , Feasibility Studies , Genetic Predisposition to Disease , Humans , Huntington Disease/blood , Huntington Disease/pathology , Prodromal Symptoms , Time Factors , Treatment Outcome
11.
Neurology ; 81(13): 1134-40, 2013 Sep 24.
Article in English | MEDLINE | ID: mdl-23966247

ABSTRACT

OBJECTIVE: We measured the levels of mutant huntingtin (mtHtt) and total huntingtin (tHtt) in blood leukocytes from Prospective Huntington At-Risk Observational Study (PHAROS) subjects at 50% risk of carrying the Huntington disease mutation using a homogeneous time-resolved fluorescence (HTRF) assay to assess its potential as a biomarker. METHODS: Peripheral blood mononuclear cells from consenting PHAROS subjects were analyzed by HTRF using antibodies that simultaneously measured mtHtt and tHtt. mtHtt levels were normalized to tHtt, double-stranded DNA, or protein and analyzed according to cytosine-adenine-guanine repeat length (CAGn), demographics, predicted time to clinical onset or known time since clinical onset, and available clinical measures. RESULTS: From 363 assayed samples, 342 met quality control standards. Levels of mtHtt and mt/tHtt were higher in 114 subjects with expanded CAG repeats (CAG ≥ 37) compared with 228 subjects with nonexpanded CAG repeats (CAG <37) (p < 0.0001). Analysis of relationships to predicted time to onset or to phenoconversion suggested that the HTRF signal could mark changes during the Huntington disease prodrome or after clinical onset. CONCLUSIONS: The HTRF assay can effectively measure mtHtt in multicenter sample sets and may be useful in trials of therapies targeting huntingtin.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Huntington Disease/blood , Huntington Disease/pathology , Leukocytes, Mononuclear/metabolism , Nerve Tissue Proteins/metabolism , Observation , Adult , Clinical Trials as Topic , Double-Blind Method , Female , Frontal Lobe/metabolism , Frontal Lobe/pathology , Humans , Huntingtin Protein , Huntington Disease/genetics , Longitudinal Studies , Male , Middle Aged , Mutation/genetics , Nerve Tissue Proteins/genetics , Postmortem Changes , Retrospective Studies , Trinucleotide Repeat Expansion/genetics
12.
Neurogenetics ; 14(3-4): 173-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23644918

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder characterized by motor, cognitive, and behavioral disturbances. It is caused by the expansion of the HTT CAG repeat, which is the major determinant of age at onset (AO) of motor symptoms. Aberrant function of N-methyl-D-aspartate receptors and/or overexposure to dopamine has been suggested to cause significant neurotoxicity, contributing to HD pathogenesis. We used genetic association analysis in 1,628 HD patients to evaluate candidate polymorphisms in N-methyl-D-aspartate receptor subtype genes (GRIN2A rs4998386 and rs2650427, and GRIN2B rs1806201) and functional polymorphisms in genes in the dopamine pathway (DAT1 3' UTR 40-bp variable number tandem repeat (VNTR), DRD4 exon 3 48-bp VNTR, DRD2 rs1800497, and COMT rs4608) as potential modifiers of the disease process. None of the seven polymorphisms tested was found to be associated with significant modification of motor AO, either in a dominant or additive model, after adjusting for ancestry. The results of this candidate-genetic study therefore do not provide strong evidence to support a modulatory role for these variations within glutamatergic and dopaminergic genes in the AO of HD motor manifestations.


Subject(s)
Huntington Disease/genetics , Polymorphism, Genetic , Receptors, Dopamine/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Age of Onset , Catechol O-Methyltransferase/genetics , Dopamine Plasma Membrane Transport Proteins/genetics , Genetic Association Studies , Humans , Huntington Disease/epidemiology , Neural Pathways/metabolism , Receptors, Dopamine D2/genetics , Receptors, Dopamine D4/genetics
13.
Hum Genet ; 131(12): 1833-40, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22825315

ABSTRACT

Huntington's disease (HD) is an inherited neurodegenerative disorder characterized by motor, cognitive and behavioral disturbances, caused by the expansion of a CAG trinucleotide repeat in the HD gene. The CAG allele size is the major determinant of age at onset (AO) of motor symptoms, although the remaining variance in AO is highly heritable. The rs7665116 SNP in PPARGC1A, encoding the mitochondrial regulator PGC-1α, has been reported to be a significant modifier of AO in three European HD cohorts, perhaps due to affected cases from Italy. We attempted to replicate these findings in a large collection of (1,727) HD patient DNA samples of European origin. In the entire cohort, rs7665116 showed a significant effect in the dominant model (p value = 0.008) and the additive model (p value = 0.009). However, when examined by origin, cases of Southern European origin had an increased rs7665116 minor allele frequency (MAF), consistent with this being an ancestry-tagging SNP. The Southern European cases, despite similar mean CAG allele size, had a significantly older mean AO (p < 0.001), suggesting population-dependent phenotype stratification. When the generalized estimating equations models were adjusted for ancestry, the effect of the rs7665116 genotype on AO decreased dramatically. Our results do not support rs7665116 as a modifier of AO of motor symptoms, as we found evidence for a dramatic effect of phenotypic (AO) and genotypic (MAF) stratification among European cohorts that was not considered in previously reported association studies. A significantly older AO in Southern Europe may reflect population differences in genetic or environmental factors that warrant further investigation.


Subject(s)
Heat-Shock Proteins/genetics , Huntington Disease/genetics , Polymorphism, Single Nucleotide , Transcription Factors/genetics , Adult , Age of Onset , Cohort Studies , Europe/epidemiology , Female , Genetics, Population , Humans , Huntingtin Protein , Huntington Disease/epidemiology , Male , Middle Aged , Nerve Tissue Proteins/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Trinucleotide Repeat Expansion
14.
Biochem Biophys Res Commun ; 424(3): 404-8, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22771793

ABSTRACT

Huntington's disease is a neurodegenerative disorder caused by an expanded CAG trinucleotide repeat whose length is the major determinant of age at onset but remaining variation appears to be due in part to the effect of genetic modifiers. GRIK2, which encodes GluR6, a mediator of excitatory neurotransmission in the brain, has been suggested in several studies to be a modifier gene based upon a 3' untranslated region TAA trinucleotide repeat polymorphism. Prior to investing in detailed studies of the functional impact of this polymorphism, we sought to confirm its effect on age at onset in a much larger dataset than in previous investigations. We genotyped the HD CAG repeat and the GRIK2 TAA repeat in DNA samples from 2,911 Huntington's disease subjects with known age at onset, and tested for a potential modifier effect of GRIK2 using a variety of statistical approaches. Unlike previous reports, we detected no evidence of an influence of the GRIK2 TAA repeat polymorphism on age at motor onset. Similarly, the GRIK2 polymorphism did not show significant modifier effect on psychiatric and cognitive age at onset in HD. Comprehensive analytical methods applied to a much larger sample than in previous studies do not support a role for GRIK2 as a genetic modifier of age at onset of clinical symptoms in Huntington's disease.


Subject(s)
Codon, Terminator/genetics , Huntington Disease/genetics , Receptors, Kainic Acid/genetics , Trinucleotide Repeats/genetics , 3' Untranslated Regions/genetics , Adolescent , Adult , Age of Onset , Aged , Aged, 80 and over , Alleles , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Polymorphism, Genetic , Young Adult , GluK2 Kainate Receptor
15.
Am J Hum Genet ; 90(3): 434-44, 2012 Mar 09.
Article in English | MEDLINE | ID: mdl-22387017

ABSTRACT

Age at the onset of motor symptoms in Huntington disease (HD) is determined largely by the length of a CAG repeat expansion in HTT but is also influenced by other genetic factors. We tested whether common genetic variation near the mutation site is associated with differences in the distribution of expanded CAG alleles or age at the onset of motor symptoms. To define disease-associated single-nucleotide polymorphisms (SNPs), we compared 4p16.3 SNPs in HD subjects with population controls in a case:control strategy, which revealed that the strongest signals occurred at a great distance from the HD mutation as a result of "synthetic association" with SNP alleles that are of low frequency in population controls. Detailed analysis delineated a prominent ancestral haplotype that accounted for ∼50% of HD chromosomes and extended to at least 938 kb on about half of these. Together, the seven most abundant haplotypes accounted for ∼83% of HD chromosomes. Neither the extended shared haplotype nor the individual local HTT haplotypes were associated with altered CAG-repeat length distribution or residual age at the onset of motor symptoms, arguing against modification of these disease features by common cis-regulatory elements. Similarly, the 11 most frequent control haplotypes showed no trans-modifier effect on age at the onset of motor symptoms. Our results argue against common local regulatory variation as a factor influencing HD pathogenesis, suggesting that genetic modifiers be sought elsewhere in the genome. They also indicate that genome-wide association analysis with a small number of cases can be effective for regional localization of genetic defects, even when a founder effect accounts for only a fraction of the disorder.


Subject(s)
Chromosomes, Human, Pair 4 , Huntington Disease/genetics , Age of Onset , Alleles , Case-Control Studies , Founder Effect , Genome-Wide Association Study/methods , Haplotypes , Humans , Mutation , Polymorphism, Single Nucleotide , Trinucleotide Repeats
16.
Arch Neurol ; 69(7): 887-93, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22393169

ABSTRACT

BACKGROUND: Aberrant accumulation of transition metals in the brain may have an early and important role in the pathogenesis of several neurodegenerative disorders, including Huntington disease (HD). OBJECTIVE: To comprehensively evaluate and validate the distribution of metal deposition in the brain using advanced magnetic resonance imaging methods from the premanifest through symptomatic stages of HD. DESIGN: Observational study. SETTING: University imaging center. PARTICIPANTS: Twenty-eight HD expanded gene carriers, 34 patients with symptomatic HD, and 56 age- and sex-matched healthy control subjects were included in the study. INTERVENTIONS: Participants underwent magnetic resonance imaging for the quantification of the phase evolution of susceptibility-weighted images. MAIN OUTCOME MEASURES: To verify the identity of the metals responsible for the changes in the phase evolution of the susceptibility signal in the brain and to assess correlations with systemic levels. Inductively coupled plasma mass spectrometry was used to measure transition metal concentrations in postmortem brains. RESULTS: In the basal ganglia, progressive increases in the phase evolution were found in HD, beginning in premanifest individuals who were far from expected onset and increasing with proximity to expected onset and thereafter. Increases in the cerebral cortex were regionally selective and present only in symptomatic HD. Increases were verified by excessive deposition of brain iron, but a complex alteration in other transition metals was found. CONCLUSION: An important and early role of altered metal homeostasis is suggested in the pathogenesis of HD.


Subject(s)
Brain/metabolism , Huntington Disease/metabolism , Huntington Disease/pathology , Iron/metabolism , Adult , Brain Mapping , Case-Control Studies , Disease Progression , Female , Humans , Huntington Disease/genetics , Imaging, Three-Dimensional , Magnetic Resonance Imaging , Male , Middle Aged , Observation , Severity of Illness Index
17.
Proc Natl Acad Sci U S A ; 108(41): 17141-6, 2011 Oct 11.
Article in English | MEDLINE | ID: mdl-21969577

ABSTRACT

Huntington disease (HD) is a progressive neurodegenerative disease that affects 30,000 individuals in North America. Treatments that slow its relentless course are not yet available, and biomarkers that can reliably measure disease activity and therapeutic response are urgently needed to facilitate their development. Here, we interrogated 119 human blood samples for transcripts associated with HD. We found that the dynamic regulator of chromatin plasticity H2A histone family, member Y (H2AFY) is specifically overexpressed in the blood and frontal cortex of patients with HD compared with controls. This association precedes the onset of clinical symptoms, was confirmed in two mouse models, and was independently replicated in cross-sectional and longitudinal clinical studies comprising 142 participants. A histone deacetylase inhibitor that suppresses neurodegeneration in animal models reduces H2AFY levels in a randomized phase II clinical trial. This study identifies the chromatin regulator H2AFY as a potential biomarker associated with disease activity and pharmacodynamic response that may become useful for enabling disease-modifying therapeutics for HD.


Subject(s)
Histones/genetics , Histones/metabolism , Huntington Disease/genetics , Huntington Disease/metabolism , Adult , Aged , Animals , Case-Control Studies , Cross-Sectional Studies , Disease Models, Animal , Double-Blind Method , Female , Frontal Lobe/metabolism , Gene Expression , Histone Deacetylase Inhibitors/pharmacology , Histones/blood , Humans , Huntington Disease/blood , Longitudinal Studies , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Middle Aged , Nerve Degeneration/drug therapy , RNA, Messenger/genetics , RNA, Messenger/metabolism
18.
Hum Mol Genet ; 20(20): 3986-96, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21791548

ABSTRACT

Sirtuin 2 (SIRT2) is one of seven known mammalian protein deacetylases homologous to the yeast master lifespan regulator Sir2. In recent years, the sirtuin protein deacetylases have emerged as candidate therapeutic targets for many human diseases, including metabolic and age-dependent neurological disorders. In non-neuronal cells, SIRT2 has been shown to function as a tubulin deacetylase and a key regulator of cell division and differentiation. However, the distribution and function of the SIRT2 microtubule (MT) deacetylase in differentiated, postmitotic neurons remain largely unknown. Here, we show abundant and preferential expression of specific isoforms of SIRT2 in the mammalian central nervous system and find that a previously uncharacterized form, SIRT2.3, exhibits age-dependent accumulation in the mouse brain and spinal cord. Further, our studies reveal that focal areas of endogenous SIRT2 expression correlate with reduced α-tubulin acetylation in primary mouse cortical neurons and suggest that the brain-enriched species of SIRT2 may function as the predominant MT deacetylases in mature neurons. Recent reports have demonstrated an association between impaired tubulin acetyltransferase activity and neurodegenerative disease; viewed in this light, our results showing age-dependent accumulation of the SIRT2 neuronal MT deacetylase in wild-type mice suggest a functional link between tubulin acetylation patterns and the aging brain.


Subject(s)
Aging/metabolism , Central Nervous System/metabolism , Microtubules/metabolism , Neurons/metabolism , Sirtuin 2/metabolism , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Developmental , Gene Order , Humans , Male , Mice , Mice, Inbred C57BL , Microtubules/genetics , Protein Isoforms/metabolism , Sirtuin 2/genetics
19.
Mov Disord ; 26(9): 1691-7, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21611979

ABSTRACT

Over the past several years, increased attention has been devoted to understanding regionally selective brain changes that occur in Huntington's disease and their relationships to phenotypic variability. Clinical progression is also heterogeneous, and although CAG repeat length influences age of onset, its role, if any, in progression has been less clear. We evaluated progression in Huntington's disease using a novel longitudinal magnetic resonance imaging analysis. Our hypothesis was that the rate of brain atrophy is influenced by the age of onset of Huntington's disease. We scanned 22 patients with Huntington's disease at approximately 1-year intervals; individuals were divided into 1 of 3 groups, determined by the relative age of onset. We found significant differences in the rates of atrophy of cortex, white matter, and subcortical structures; patients who developed symptoms earlier demonstrated the most rapid rates of atrophy compared with those who developed symptoms during middle age or more advanced age. Rates of cortical atrophy were topologically variable, with the most rapid changes occurring in sensorimotor, posterior frontal, and portions of the parietal cortex. There were no significant differences in the rates of atrophy in basal ganglia structures. Although both CAG repeat length and age influenced the rate of change in some regions, there was no significant correlation in many regions. Rates of regional brain atrophy seem to be influenced by the age of onset of Huntington's disease symptoms and are only partially explained by CAG repeat length. These findings suggest that other genetic, epigenetic, and environmental factors play important roles in neurodegeneration in Huntington's disease.


Subject(s)
Cerebral Cortex/pathology , Huntington Disease/pathology , Huntington Disease/physiopathology , Magnetic Resonance Imaging , Age of Onset , Atrophy/pathology , Disease Progression , Female , Humans , Huntington Disease/genetics , Longitudinal Studies , Male , Trinucleotide Repeats/genetics
20.
J Biol Chem ; 286(20): 18320-30, 2011 May 20.
Article in English | MEDLINE | ID: mdl-21454633

ABSTRACT

Huntington disease (HD) is a progressive neurodegenerative disorder caused by expression of polyglutamine-expanded mutant huntingtin protein (mhtt). Most evidence indicates that soluble mhtt species, rather than insoluble aggregates, are the important mediators of HD pathogenesis. However, the differential roles of soluble monomeric and oligomeric mhtt species in HD and the mechanisms of oligomer formation are not yet understood. We have shown previously that copper interacts with and oxidizes the polyglutamine-containing N171 fragment of huntingtin. In this study we report that oxidation-dependent oligomers of huntingtin form spontaneously in cell and mouse HD models. Levels of these species are modulated by copper, hydrogen peroxide, and glutathione. Mutagenesis of all cysteine residues within N171 blocks the formation of these oligomers. In cells, levels of oligomerization-blocked mutant N171 were decreased compared with native N171. We further show that a subset of the oligomerization-blocked form of glutamine-expanded N171 huntingtin is rapidly depleted from the soluble pool compared with "native " mutant N171. Taken together, our data indicate that huntingtin is subject to specific oxidations that are involved in the formation of stable oligomers and that also delay removal from the soluble pool. These findings show that inhibiting formation of oxidation-dependent huntingtin oligomers, or promoting their dissolution, may have protective effects in HD by decreasing the burden of soluble mutant huntingtin.


Subject(s)
Cysteine/metabolism , Huntington Disease/metabolism , Mutation, Missense , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Protein Multimerization , Animals , COS Cells , Chlorocebus aethiops , Cysteine/genetics , Disease Models, Animal , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/pathology , Mice , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Oxidation-Reduction , Protein Structure, Tertiary , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL
...