Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Alzheimers Dement (Amst) ; 14(1): e12324, 2022.
Article in English | MEDLINE | ID: mdl-35634535

ABSTRACT

Research suggests a link between Alzheimer's Disease in Down Syndrome (DS) and the overproduction of amyloid plaques. Using Positron Emission Tomography (PET) we can assess the in-vivo regional amyloid load using several available ligands. To measure amyloid distributions in specific brain regions, a brain atlas is used. A popular method of creating a brain atlas is to segment a participant's structural Magnetic Resonance Imaging (MRI) scan. Acquiring an MRI is often challenging in intellectually-imparied populations because of contraindications or data exclusion due to significant motion artifacts or incomplete sequences related to general discomfort. When an MRI cannot be acquired, it is typically replaced with a standardized brain atlas derived from neurotypical populations (i.e. healthy individuals without DS) which may be inappropriate for use in DS. In this project, we create a series of disease and diagnosis-specific (cognitively stable (CS-DS), mild cognitive impairment (MCI-DS), and dementia (DEM-DS)) probabilistic group atlases of participants with DS and evaluate their accuracy of quantifying regional amyloid load compared to the individually-based MRI segmentations. Further, we compare the diagnostic-specific atlases with a probabilistic atlas constructed from similar-aged cognitively-stable neurotypical participants. We hypothesized that regional PET signals will best match the individually-based MRI segmentations by using DS group atlases that aligns with a participant's disorder and disease status (e.g. DS and MCI-DS). Our results vary by brain region but generally show that using a disorder-specific atlas in DS better matches the individually-based MRI segmentations than using an atlas constructed from cognitively-stable neurotypical participants. We found no additional benefit of using diagnose-specific atlases matching disease status. All atlases are made publicly available for the research community. Highlight: Down syndrome (DS) joint-label-fusion atlases provide accurate positron emission tomography (PET) amyloid measurements.A disorder-specific DS atlas is better than a neurotypical atlas for PET quantification.It is not necessary to use a disease-state-specific atlas for quantification in aged DS.Dorsal striatum results vary, possibly due to this region and dementia progression.

2.
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.

3.
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
4.
J Huntingtons Dis ; 8(3): 339-356, 2019.
Article in English | MEDLINE | ID: mdl-31306138

ABSTRACT

BACKGROUND: Huntington's disease (HD) is a genetic disorder known for affecting motor control. Despite evidence for the impact of HD on visual cortico-striatal loops, evidence for impaired visual perception in early symptomatic HD patients is limited; much less is known about what happens during the HD prodrome. OBJECTIVE: The goals of this study were to evaluate perceptual processing in motor pre-manifest HD gene-carriers (Pre-HDs) during a visual mental rotation task. METHODS: To achieve this goal, 79 participants including 24 Pre-HD participants and 55 healthy matched controls were scanned using functional MRI as they performed a mental rotation task. Another group of 36 subjects including 15 pre-HDs and 21 healthy age/gender matched controls participated in a control behavioral test of judgment of line orientation outside the scanner. RESULTS: We found that, although Pre-HDs (in this stage of disease) did not demonstrate slower response times, their response accuracy was lower than controls. On the fMRI task, controls showed a significant decrease in activity in the occipito-temporal (OT) visual network and increase in activity in the caudo-fronto-parietal (CFP) network with mental rotation load. Interestingly, the amount of mental rotation-related activity decrease in the OT network was reduced in Pre-HDs compared to controls while, the level of CFP response remained unchanged between the two groups. Comparing the link between the evoked BOLD activity within these networks and response accuracy (i.e., behavior), we found that the models fit to data from controls were less accurate in predicting response accuracy of Pre-HDs. CONCLUSION: These findings provide some of the earliest functional evidence of impaired visual processing and altered neural processing underlying visual perceptual decision making during the HD prodrome.


Subject(s)
Brain/physiopathology , Huntington Disease/physiopathology , Huntington Disease/psychology , Pattern Recognition, Visual/physiology , Adult , Brain Mapping , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Motion Perception/physiology , Rotation , Young Adult
5.
J Neurosci ; 36(40): 10456-10471, 2016 10 05.
Article in English | MEDLINE | ID: mdl-27707978

ABSTRACT

The caudate nucleus is a part of the visual corticostriatal loop (VCSL), receiving input from different visual areas and projecting back to the same cortical areas via globus pallidus, substantia nigra, and thalamus. Despite perceptual and navigation impairments in patients with VCSL disruption due to caudate atrophy (e.g., Huntington's disease, HD), the relevance of the caudate nucleus and VCSL on cortical visual processing is not fully understood. In a series of fMRI experiments, we found that the caudate showed a stronger functional connection to parahippocampal place area (PPA) compared with adjacent regions (e.g., fusiform face area, FFA) within the temporal visual cortex. Consistent with this functional link, the caudate showed a higher response to scenes compared with faces, similar to the PPA. Testing the impact of VCSL disruption on neural processes within PPA, HD patients showed reduced scene-selective activity within PPA compared with healthy matched controls. In contrast, the level of selective activity in adjacent cortical and subcortical face-selective areas (i.e., FFA and amygdala) remained intact. These results show some of the first evidence for the direct impact and potential clinical significance of VCSL on the generation of "selective" activity within PPA. SIGNIFICANCE STATEMENT: Visual perception is often considered the product of a multistage feedforward neural processing between visual cortical areas, ignoring the likely impact of corticosubcortical loops on this process. Here, we provide evidence for the contribution of visual corticostriatal loop and the caudate nucleus on generating selective response within parahippocampal place area (PPA). Our results show that disruption of this loop in Huntington's disease patients reduces the level of selective activity within PPA, which may lead to related perceptual impairments in these patients.


Subject(s)
Neostriatum/physiology , Parahippocampal Gyrus/physiology , Visual Cortex/physiology , Visual Pathways/physiology , Adult , Aged , Amygdala/physiology , Brain Mapping , Discrimination, Psychological/physiology , Face , Female , Form Perception/physiology , Humans , Huntington Disease/physiopathology , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Middle Aged , Recognition, Psychology/physiology , Visual Perception/physiology , Young Adult
6.
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.

7.
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
8.
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
9.
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
10.
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
11.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...