Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Mov Disord ; 39(1): 197-203, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38148511

ABSTRACT

BACKGROUND: Huntington's disease (HD) is a genetically determined disease with motor, cognitive, and neuropsychiatric disorders. However, the links between clinical progression and disruptions to dynamics in motor and cognitive large-scale networks are not well established. OBJECTIVE: To investigate changes in dynamic and static large-scale networks using an established tool of disease progression in Huntington's disease, the composite Unified Huntington's Disease Rating Scale (cUHDRS). METHODS: Sixty-four mutation carriers were included. Static and dynamic baseline functional connectivity as well as topological features were correlated to 2-year follow-up clinical assessments using the cUHDRS. RESULTS: Decline in cUHDRS scores was associated with higher connectivity between frontal default-mode and motor networks, whereas higher connectivity in posterior, mainly visuospatial regions was associated with a smaller decline in cUHDRS scores. CONCLUSIONS: Structural disruptions in HD were evident both in posterior parietal/occipital and frontal motor regions, with reciprocal increases in functional connectivity. However, although higher visuospatial network connectivity was tied to a smaller cUHDRS decline, increased motor and frontal default-mode connections were linked to a larger cUHDRS decreases. Therefore, divergent functional compensation mechanisms might be at play in the clinical evolution of HD.


Subject(s)
Huntington Disease , Humans , Huntington Disease/diagnostic imaging , Huntington Disease/genetics , Brain/diagnostic imaging , Magnetic Resonance Imaging , Disease Progression , Frontal Lobe
2.
Mov Disord ; 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39192729

ABSTRACT

BACKGROUND: Emerging research implicates tau protein dysregulation in the pathophysiology of Huntington's disease. OBJECTIVE: This study investigated skin tau quantification as a potential biomarker for Huntington's disease and its correlation with disease burden outcomes. METHODS: In this cross-sectional study, we measured skin tau levels using enzyme-linked immunosorbent assay in 23 Huntington's disease mutations carriers and eight control subjects, examining group discrimination, correlations with genetic markers, clinical assessments, and neuroimaging data. Brain atrophy was quantified by both volumetric measurements from brain segmentation and a voxel-based morphometry approach. RESULTS: Our findings showed elevated skin tau levels in manifest Huntington's disease compared with premanifest and healthy controls. These levels correlated with CAG repeat length, CAG-Age-Product score, composite Unified Huntington's Disease Rating Scale Total Motor Score, cognitive assessments, and disease-related cortical and subcortical volumes, all independent of age and gender. Using skin tau levels in cluster analysis along with genetic and clinical measures led to improved subject stratification, providing enhanced distinction and validity of clusters. CONCLUSIONS: This study not only confirms the feasibility of skin tau quantification in Huntington's disease but also establishes its potential as a biomarker for enhancing group classification and assessing disease severity across the Huntington's disease spectrum, opening new directions in biomarker research. © 2024 International Parkinson and Movement Disorder Society.

3.
Neurol Sci ; 45(9): 4635-4637, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38896185

ABSTRACT

Chorea is a hyperkinetic movement disorder associated with various underlyingconditions, including autoimmune diseases such as antiphospholipid syndrome (APS). APS can manifest with a wide range of neurological symptoms, including chorea. We present a case of a 77-year-old man with subacute generalized chorea secondary to primary APS. Notably, the patient exhibited a left patellar crossed-reflex, a phenomenon rarely documented in chorea cases, the pathophysiology of which has not yet been elucidated. In summary, this case challenges the traditional demographics of antiphospholipid syndrome (APS) by suggesting a potential link between APS and late-age patients. It emphasizes the importance of considering APS in late-onset chorea cases.


Subject(s)
Antiphospholipid Syndrome , Chorea , Humans , Aged , Male , Chorea/etiology , Chorea/physiopathology , Antiphospholipid Syndrome/complications , Reflex/physiology
4.
J Neurosci Res ; 101(6): 990-999, 2023 06.
Article in English | MEDLINE | ID: mdl-36807154

ABSTRACT

Individuals with pre-manifest and early symptomatic Huntington's disease (HD) have shown deficits in solving arithmetic word-problems. However, the neural correlates of these deficits in HD are poorly understood. We explored the structural (gray-matter volume; GMV) and metabolic (18F-FDG PET; SUVr) brain correlates of arithmetic performance using the recently developed HD-word problem arithmetic task (HD-WPA) in seventeen preHD and sixteen HD individuals. Symptomatic participants showed significantly lower scores in the HD-WPA than preHD participants. Lower performance in the HD-WPA was associated with reduced GMV in subcortical, medial frontal, and several posterior-cortical clusters in HD participants. No significant GMV loss was found in preHD participants. 18F-FDG data revealed a widespread pattern of hypometabolism in association with lower arithmetic performance in all participants. In preHD participants, this pattern was restricted to the ventrolateral and orbital prefrontal cortex, the insula, and the precentral gyrus. In HD participants, the pattern extended to several parietal-temporal regions. Word-problem solving arithmetic deficits in HD is subserved by a pattern of asynchronous metabolic and structural compromise across the cerebral cortex as a function of disease stage. In preHD individuals, arithmetic deficits were associated with prefrontal alterations, whereas in symptomatic HD patients, more severe arithmetic deficits are associated with the compromise of several frontal-subcortical and temporo-parietal regions. Our results support the hypothesis that cognitive deficits in HD are not exclusively dominated by frontal-striatal dysfunctions but also involve fronto-temporal and parieto-occipital damage.


Subject(s)
Cognition Disorders , Huntington Disease , Humans , Huntington Disease/metabolism , Fluorodeoxyglucose F18/metabolism , Brain/metabolism , Cognition Disorders/complications , Problem Solving , Magnetic Resonance Imaging
5.
Eur J Neurol ; 30(7): 1871-1879, 2023 07.
Article in English | MEDLINE | ID: mdl-36994811

ABSTRACT

BACKGROUND AND PURPOSE: Cognitive impairment is a central feature of Huntington's disease (HD), but it is unclear to what extent more aggressive cognitive phenotypes exist in HD among individuals with the same genetic load and equivalence in other clinical and sociodemographic variables. METHODS: We included Enroll-HD study participants in early and early-mid stages of HD at baseline and with three consecutive yearly follow-ups for whom several clinical and sociodemographic as well as cognitive measures were recorded. We excluded participants with low and large CAG repeat length (CAG < 39 & > 55), with juvenile or late onset HD, and with dementia at baseline. We explored the existence of different groups according to the profile of cognitive progression using a two-step k-means cluster analysis model based on the combination of different cognitive outcomes. RESULTS: We identified a slow cognitive progression group of 293 participants and an aggressive progression group (F-CogHD) of 235 for which there were no differences at the baseline visit in any of the measures explored, with the exception of a slightly higher motor score in the F-CogHD group. This group showed a more pronounced annual loss of functionality and a more marked motor and psychiatric deterioration. CONCLUSIONS: The rate of progression of cognitive deterioration in HD is strongly variable even between patients sharing, among other variables, equivalent CAG repeat length, age, and disease duration. We can recognize at least two phenotypes that differ in terms of rate of progression. Our findings open new avenues to study additional mechanisms contributing to HD heterogeneity.


Subject(s)
Cognition Disorders , Huntington Disease , Humans , Huntington Disease/complications , Huntington Disease/genetics , Huntington Disease/psychology , Longitudinal Studies , Disease Progression , Cognition
6.
Mov Disord ; 37(2): 343-353, 2022 02.
Article in English | MEDLINE | ID: mdl-34752656

ABSTRACT

BACKGROUND: Huntington's disease is a neurodegenerative disorder characterized by clinical alterations in the motor, behavioral, and cognitive domains. However, the structure and disruptions to large-scale brain cognitive networks have not yet been established. OBJECTIVE: We aimed to profile changes in large-scale cognitive networks in premanifest and symptomatic patients with Huntington's disease. METHODS: We prospectively recruited premanifest and symptomatic Huntington's disease mutation carriers as well as healthy controls. Clinical and sociodemographic data were obtained from all participants, and resting-state functional connectivity data, using both time-averaged and dynamic functional connectivity, was acquired from whole-brain and cognitively oriented brain parcellations. RESULTS: A total of 64 gene mutation carriers and 23 healthy controls were included; 21 patients with Huntington's disease were classified as premanifest and 43 as symptomatic Huntington's disease. Compared with healthy controls, patients with Huntington's disease showed decreased network connectivity within the posterior hubs of the default-mode network and the medial prefrontal cortex, changes that correlated with cognitive (t = 2.25, P = 0.01) and disease burden scores (t = -2.42, P = 0.009). The salience network showed decreased functional connectivity between insular and supramarginal cortices and also correlated with cognitive (t = 2.11, P = 0.02) and disease burden scores (t = -2.35, P = 0.01). Dynamic analyses showed that network variability was decreased for default-central executive networks, a feature already present in premanifest mutation carriers (dynamic factor 8, P = 0.02). CONCLUSIONS: Huntington's disease shows an early and widespread disruption of large-scale cognitive networks. Importantly, these changes are related to cognitive and disease burden scores, and novel dynamic functional analyses uncovered subtler network changes even in the premanifest stages.


Subject(s)
Huntington Disease , Brain/diagnostic imaging , Brain Mapping , Cognition , Humans , Huntington Disease/diagnostic imaging , Huntington Disease/genetics , Magnetic Resonance Imaging
7.
Mov Disord ; 36(9): 2162-2172, 2021 09.
Article in English | MEDLINE | ID: mdl-33998063

ABSTRACT

BACKGROUND: Apathy, a common neuropsychiatric disturbance in Huntington's disease (HD), is subserved by a complex neurobiological network. However, no study has yet employed a whole-brain approach to examine underlying regional vulnerabilities that may precipitate apathy changes over time. OBJECTIVES: To identify whole-brain gray matter volume (GMV) vulnerabilities that may predict longitudinal apathy development in HD. METHODS: Forty-five HD individuals (31 female) were scanned and evaluated for apathy and other neuropsychiatric features using the short-Problem Behavior Assessment for a maximum total of six longitudinal visits (including baseline). In order to identify regions where changes in GMV may describe changes in apathy, we performed longitudinal voxel-based morphometry (VBM) on those 33 participants with a magnetic resonance imaging (MRI) scan on their second visit at 18 ± 6 months follow-up (78 MRI datasets). We next employed a generalized linear mixed-effects model (N = 45) to elucidate whether initial and specific GMV may predict apathy development over time. RESULTS: Utilizing longitudinal VBM, we revealed a relationship between increases in apathy and specific GMV atrophy in the right middle cingulate cortex (MCC). Furthermore, vulnerability in the right MCC volume at baseline successfully predicted the severity and progression of apathy over time. CONCLUSIONS: This study highlights that individual differences in apathy in HD may be explained by variability in atrophy and initial vulnerabilities in the right MCC, a region implicated in action-initiation. These findings thus serve to facilitate the prediction of an apathetic profile, permitting targeted, time-sensitive interventions in neurodegenerative disease with potential implications in otherwise healthy populations. © 2021 International Parkinson and Movement Disorder Society.


Subject(s)
Apathy , Huntington Disease , Neurodegenerative Diseases , Brain/diagnostic imaging , Female , Gray Matter/diagnostic imaging , Humans , Huntington Disease/diagnostic imaging , Magnetic Resonance Imaging
8.
Eur J Nucl Med Mol Imaging ; 47(9): 2165-2174, 2020 08.
Article in English | MEDLINE | ID: mdl-31897582

ABSTRACT

BACKGROUND: Impulse control disorders (ICD) are a common and disrupting complication of Parkinson's disease (PD) treatment. Although their relationship with dopaminergic activity is well studied, their brain metabolic correlates are mostly unknown. METHODS: In this work we studied brain metabolism using brain 18F-FDG-PET. We performed a case-control study nested within a cohort of PD patients free of ICD at baseline to compare ICD patients right after ICD diagnosis and prior to any treatment modification with matched ICD-free patients. We also compared both PD groups with healthy controls. RESULTS: When compared with ICD-free PD patients, PD patients with recently diagnosed ICD showed higher glucose metabolism in widespread areas comprising prefrontal cortices, both amygdalae and default mode network hubs (p < 0.05, corrected). When compared to healthy controls, they did not show hypermetabolism, and the only hypometabolic region was the right caudate. In turn, ICD-free patients showed diffuse hypometabolism when compared to healthy controls. CONCLUSION: Our results suggest brain metabolism is more preserved in PD patients with ICD than patients without ICD. This metabolic preservation could be related to ICD development.


Subject(s)
Disruptive, Impulse Control, and Conduct Disorders , Parkinson Disease , Brain/diagnostic imaging , Case-Control Studies , Disruptive, Impulse Control, and Conduct Disorders/diagnostic imaging , Disruptive, Impulse Control, and Conduct Disorders/etiology , Fluorodeoxyglucose F18 , Humans , Parkinson Disease/diagnostic imaging
9.
Hum Brain Mapp ; 40(11): 3233-3242, 2019 08 01.
Article in English | MEDLINE | ID: mdl-30938027

ABSTRACT

Cognitive decline is a major disabling feature in Parkinson's disease (PD). Multimodal imaging studies have shown functional disruption in neurocognitive networks related to cognitive impairment. However, it remains unknown whether these changes are related to gray matter loss, or whether they outline network vulnerability in the early stages of cognitive impairment. In this work, we intended to assess functional connectivity and graph theoretical measures and their relation to gray matter loss in Parkinson's disease with mild cognitive impairment (PD-MCI). We recruited 53 Parkinson's disease patients and classified them for cognitive impairment using Level-1 Movement Disorders Society-Task Force Criteria. Voxel-based morphometry, functional connectivity and graph theoretical measures were obtained on a 3-Tesla MRI scanner. Loss of gray matter was observed in the default mode network (bilateral precuneus), without a corresponding disruption of functional or graph theoretical properties. However, functional and graph theoretical changes appeared in salience network nodes, without evidence of gray matter loss. Global cognition and executive scores showed a correlation with node degree in the right anterior insula. We also found a correlation between visuospatial scores and right supramarginal gyrus node degree. Our findings highlight the loss of functional connectivity and topological features without structural damage in salience network regions in PD-MCI. They also underline the importance of multimodal hubs in the transition to mild cognitive impairment. This functional disruption in the absence of gray matter atrophy suggests that the salience network is a key vulnerable system at the onset of mild cognitive impairment in PD.


Subject(s)
Brain/diagnostic imaging , Cognitive Dysfunction/diagnostic imaging , Nerve Net/diagnostic imaging , Parkinson Disease/diagnostic imaging , Aged , Cognition/physiology , Cognitive Dysfunction/psychology , Female , Gray Matter/diagnostic imaging , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neuropsychological Tests , Parkinson Disease/psychology
10.
Eur J Nucl Med Mol Imaging ; 46(5): 1111-1116, 2019 May.
Article in English | MEDLINE | ID: mdl-30627816

ABSTRACT

PURPOSE: Huntington's disease (HD) is a fatal neurodegenerative disorder with no effective treatment currently available. Although the pathological hallmark of HD is massive striatal atrophy, it has been suggested that cortical deterioration may concomitantly occur and play a major role in the patient's functional independence. Our objective was to characterize cortical structural and metabolic neurodegeneration in the transition from premanifest to early-stage Huntington's disease (HD). METHODS: Using a surface-based neuroimaging approach, we compared cortical thickness and intracortical FDG-PET uptake in 19 early-symptomatic HD patients with respect to 21 premanifest HD individuals. RESULTS: Early-HD patients showed significant cortical atrophy and intracortical hypometabolism when compared to premanifest subjects (p < 0.05, corrected for multiple comparisons). However, whereas the atrophy pattern was restricted to precentral and parieto-occipital regions, a pronounced frontotemporal hypometabolism was observed. Importantly, structural changes correlated with motor and cognitive performance, and metabolic changes were associated with the presence and severity of apathy in this population, a core neuropsychiatric feature of this disorder. CONCLUSION: Our findings reveal an asynchronous neuronal loss and metabolic compromise across the cerebral cortex in early HD. Hence, the use of structural and metabolic imaging indicators to characterize disease progression in this population should take into consideration the dissociation which occurs between cortical atrophy and hypometabolism.


Subject(s)
Asymptomatic Diseases , Brain/metabolism , Brain/pathology , Disease Progression , Huntington Disease/metabolism , Huntington Disease/pathology , Adult , Atrophy , Brain/diagnostic imaging , Female , Humans , Huntington Disease/diagnostic imaging , Huntington Disease/genetics , Magnetic Resonance Imaging , Male , Mutation , Positron Emission Tomography Computed Tomography
11.
Mov Disord ; 34(1): 78-86, 2019 01.
Article in English | MEDLINE | ID: mdl-30536829

ABSTRACT

BACKGROUND: Minor hallucinations and well-structured hallucinations are considered in the severity continuum of the psychotic spectrum associated with Parkinson's disease. Although their chronological relationship is largely unknown, the spatial patterns of brain atrophy in these 2 forms of hallucinations partially overlap, suggesting they share similar pathophysiological processes. Functional connectivity studies show that disruption of functional networks involved in perception and attention could be relevant in the emergence of well-structured hallucinations. However, functional neuroimaging studies in patients with isolated minor hallucinations are lacking. The objectives of this study were to explore the structural and functional changes underlying minor hallucinations. METHODS: We compared patients with (n = 18) and without (n = 14) minor hallucinations using a multimodal structural (gray-matter volume voxel-based morphometry) and functional (seed-to-whole-brain resting-state functional MRI) neuroimaging study. RESULTS: Coincident with previously described structural changes in well-structured hallucinations in Parkinson's disease, patients with minor hallucinations exhibited gray-matter atrophy with significant voxel-wise differences in visuoperceptual processing areas and core regions of the default mode network. Functional connectivity changes consisted of altered connectivity within the default mode network, reduced negative correlation with task-positive network, and aberrant connectivity between posterior regions of the default mode network and visual-processing areas. These changes are in accordance with the attentional networks hypothesis proposed for well-structured hallucinations. CONCLUSIONS: Although longitudinal studies are needed to assess the potential role of minor hallucinations as an early clinical biomarker of progression to well-structured hallucinations, the present findings show that the 2 phenomena share similar structural and functional brain correlates. © 2018 International Parkinson and Movement Disorder Society.


Subject(s)
Attention/physiology , Brain/physiopathology , Nerve Net/physiopathology , Parkinson Disease/physiopathology , Aged , Brain/pathology , Brain Mapping/methods , Cognition Disorders/physiopathology , Female , Humans , Image Processing, Computer-Assisted/methods , Male , Middle Aged , Neuropsychological Tests
12.
Hum Brain Mapp ; 39(1): 54-71, 2018 01.
Article in English | MEDLINE | ID: mdl-28990240

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder which is primarily associated with striatal degeneration. However, the alterations in connectivity of this structure in HD have been underinvestigated. In this study, we analyzed the functional and structural connectivity of the left putamen, while participants performed a finger-tapping task. Using fMRI and DW-MRI, 30 HD gene expansion carriers (HDGEC) and 29 healthy participants were scanned. Psychophysiological interaction analysis and DTI-based tractography were employed to examine functional and structural connectivity, respectively. Manifest HDGEC exhibited a reduced functional connectivity of the left putamen with the left and the right primary sensorimotor areas (SM1). Based on this result, the inhibitory functional connectivity between the left SM1 and the right SM1 was explored, appearing to be also decreased. In addition, the tract connecting these areas (motor corpus callosum), and the tract connecting the left putamen with the left SM1 appeared disrupted in HDGEC compared to controls. Significant correlations were found between measures of functional and structural connectivity of the motor corpus callosum, showing a coupling of both types of alterations in this tract. The observed reduction of functional and structural connectivity was associated with worse motor scores, which highlights the clinical relevance of these results. Hum Brain Mapp 39:54-71, 2018. © 2017 Wiley Periodicals, Inc.


Subject(s)
Cerebral Cortex/diagnostic imaging , Cerebral Cortex/physiopathology , Corpus Striatum/diagnostic imaging , Corpus Striatum/physiopathology , Huntington Disease/diagnostic imaging , Huntington Disease/physiopathology , Adult , Diffusion Tensor Imaging , Female , Fingers/physiopathology , Heterozygote , Humans , Huntington Disease/genetics , Magnetic Resonance Imaging , Male , Middle Aged , Motor Activity/physiology , Neural Pathways/diagnostic imaging , Neural Pathways/physiopathology
13.
Mov Disord ; 33(7): 1151-1159, 2018 07.
Article in English | MEDLINE | ID: mdl-29676484

ABSTRACT

BACKGROUND: Apathy is the most prevalent and characteristic neuropsychiatric feature of Huntington's disease. Congruent with the main early pathological changes, apathy is primarily associated with subcortical damage in frontal-striatal circuits. However, little is known about its precise subserving mechanisms and the contribution of regions other than the basal ganglia. OBJECTIVES: We aimed to define the neural correlates of apathy in Huntington's disease based on gray matter volume and PET/CT of 18 F-fluorodeoxyglucose metabolism. METHODS: We rated the severity of apathy in 40 mild Huntington's disease participants using the Problem Behaviors Assessment for Huntington's disease. Voxelwise regression analysis was performed, controlling for effects of potential confounders, and PET/CT results were corrected for the effects of gray matter atrophy. RESULTS: Apathy was strongly associated with decreased gray matter within a spatially distributed cortico-subcortical network, with major compromise of the bilateral amygdala and temporal cortex. PET metabolism was significantly decreased in frontotemporal and parietal regions. Metabolic uptake and gray matter values in the identified clusters showed significant correlations with multiple clinical measures. CONCLUSIONS: Our findings indicate that apathy in Huntington's disease is not exclusively a consequence of basal ganglia and related frontal-executive alterations. It is subserved by a complex cortico-subcortical network where critical reward and emotional-related prefrontal, temporal, and limbic nodes contribute strongly to its severity. This highlights the contribution of damage in regions other than the basal ganglia to the clinical expression of Huntington's disease. © 2018 International Parkinson and Movement Disorder Society.


Subject(s)
Apathy/physiology , Brain/diagnostic imaging , Brain/metabolism , Huntington Disease/pathology , Huntington Disease/physiopathology , Aged , Atrophy/etiology , Cognition Disorders/diagnostic imaging , Cognition Disorders/etiology , Female , Fluorodeoxyglucose F18/metabolism , Gray Matter/diagnostic imaging , Gray Matter/pathology , Humans , Imaging, Three-Dimensional , Male , Mental Disorders/etiology , Middle Aged , Neuropsychological Tests , Positron-Emission Tomography , Tomography Scanners, X-Ray Computed
14.
Eur J Nucl Med Mol Imaging ; 43(12): 2183-2189, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27349245

ABSTRACT

PURPOSE: To assess metabolic changes in cerebral 18F-FDG PET/CT in premanifest and manifest Huntington's disease (HD) subjects compared to a control group and to correlate 18F-FDG uptake patterns with different disease stages. MATERIALS AND METHODS: Thirty-three gene-expanded carriers (Eight males; mean age: 43 y/o; CAG > 39) were prospectively included. Based on the Unified Huntington's Disease Rating Scale Total Motor Score and the Total Functional Capacity, subjects were classified as premanifest (preHD = 15) and manifest (mHD = 18). Estimated time disease-onset was calculated using the Langbehn formula, which allowed classifying preHD as far-to (preHD-A) and close-to (PreHD-B) disease-onset. Eighteen properly matched participants were included as a control group (CG). All subjects underwent brain 18F-FDG PET/CT and MRI. 18F-FDG PET/CT were initially assessed by two nuclear medicine physicians identifying qualitative metabolic changes in the striatum. Quantitative analysis was performed using SPM8 with gray matter atrophy correction using the BPM toolbox. RESULTS: Visual analysis showed a marked striatal hypometabolism in mHD. A normal striatal distribution of 18F-FDG uptake was observed for most of the preHD subjects. Quantitative analysis showed a significant striatal hypometabolism in mHD subjects compared to CG (p < 0.001 uncorrected, k = 50 voxels). In both preHD groups we observed a significant striatal hypometabolism with respect to CG (p < 0.001 uncorrected, k = 50 voxels). In mHD subjects we observed a significant striatal hypometabolism with respect to both preHD groups (p < 0.001 uncorrected, k = 50 voxels). CONCLUSION: 18F-FDG PET/CT might be a helpful tool to identify patterns of glucose metabolism in the striatum across the stages of HD and might be relevant in assessing the clinical status of gene-expanded HD carriers due to the fact that dysfunctional glucose metabolism begins at early preHD stages of the disease. 18F-FDG PET/CT appears as a promising method to monitor the response to disease-modifying therapies even if applied in premanifest subjects.


Subject(s)
Brain Diseases, Metabolic/metabolism , Corpus Striatum/metabolism , Fluorodeoxyglucose F18/pharmacokinetics , Huntington Disease/diagnostic imaging , Huntington Disease/metabolism , Positron Emission Tomography Computed Tomography/methods , Adult , Aged , Brain Diseases, Metabolic/complications , Brain Diseases, Metabolic/diagnostic imaging , Corpus Striatum/diagnostic imaging , Early Diagnosis , Female , Humans , Huntington Disease/complications , Male , Middle Aged , Radiopharmaceuticals/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity , Young Adult
15.
J Extracell Vesicles ; 13(10): e12522, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39377487

ABSTRACT

Despite the advances in the understanding of Huntington's disease (HD), there is a need for molecular biomarkers to categorize mutation carriers during the preclinical stage of the disease preceding functional decline. Small RNAs (sRNAs) are a promising source of biomarkers since their expression levels are highly sensitive to pathobiological processes. Here, using an optimized method for plasma extracellular vesicles (EVs) purification and an exhaustive analysis pipeline of sRNA sequencing data, we show that EV-sRNAs are downregulated early in mutation carriers and that this deregulation is associated with premanifest cognitive performance. Seven candidate sRNAs (tRF-Glu-CTC, tRF-Gly-GCC, miR-451a, miR-21-5p, miR-26a-5p, miR-27a-3p and let7a-5p) were validated in additional subjects, showing a significant diagnostic accuracy at premanifest stages. Of these, miR-21-5p was significantly decreased over time in a longitudinal study; and miR-21-5p and miR-26a-5p levels correlated with cognitive changes in the premanifest cohort. In summary, the present results suggest that deregulated plasma EV-sRNAs define an early biosignature in mutation carriers with specific species highlighting the progression and cognitive changes occurring at the premanifest stage.


Subject(s)
Biomarkers , Extracellular Vesicles , Huntington Disease , MicroRNAs , Huntington Disease/blood , Huntington Disease/genetics , Humans , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Biomarkers/blood , MicroRNAs/blood , MicroRNAs/genetics , Male , Female , Adult , Middle Aged , Mutation , Longitudinal Studies
16.
Ann Clin Transl Neurol ; 11(5): 1160-1171, 2024 May.
Article in English | MEDLINE | ID: mdl-38544341

ABSTRACT

OBJECTIVE: The clinical phenotype of Huntington's disease (HD) can be very heterogeneous between patients, even when they share equivalent CAG repeat length, age, or disease burden. This heterogeneity is especially evident in terms of the cognitive profile and related brain changes. To shed light on the mechanisms participating in this heterogeneity, the present study delves into the association between Tau pathology and more severe cognitive phenotypes and brain damage in HD. METHODS: We used a comprehensive neuropsychological examination to characterize the cognitive phenotype of a sample of 30 participants with early-to-middle HD for which we also obtained 3 T structural magnetic resonance image (MRI) and cerebrospinal fluid (CSF). We quantified CSF levels of neurofilament light chain (NfL), total Tau (tTau), and phosphorylated Tau-231 (pTau-231). Thanks to the cognitive characterization carried out, we subsequently explored the relationship between different levels of biomarkers, the cognitive phenotype, and brain integrity. RESULTS: The results confirmed that more severe forms of cognitive deterioration in HD extend beyond executive dysfunction and affect processes with clear posterior-cortical dependence. This phenotype was in turn associated with higher CSF levels of tTau and pTau-231 and to a more pronounced pattern of posterior-cortical atrophy in specific brain regions closely linked to the cognitive processes affected by Tau. INTERPRETATION: Our findings reinforce the association between Tau pathology, cognition, and neurodegeneration in HD, emphasizing the need to explore the role of Tau in the cognitive heterogeneity of the disease.


Subject(s)
Cognitive Dysfunction , Huntington Disease , Phenotype , tau Proteins , Humans , Huntington Disease/cerebrospinal fluid , tau Proteins/cerebrospinal fluid , Male , Middle Aged , Female , Adult , Cognitive Dysfunction/cerebrospinal fluid , Cognitive Dysfunction/etiology , Cognitive Dysfunction/physiopathology , Neurofilament Proteins/cerebrospinal fluid , Magnetic Resonance Imaging , Biomarkers/cerebrospinal fluid , Atrophy/pathology , Neuropsychological Tests
17.
Neurol Ther ; 2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39370480

ABSTRACT

BACKGROUND: Limited information is available on patients' experience living with Huntington's disease (HD). The primary objective of this study was to assess the health-related quality of life and well being of patients with HD. METHODS: A non-interventional, cross-sectional study was conducted in 17 hospitals-based movement disorders units in Spain. Patients aged ≥ 18 years, genetically HD diagnosed [with a diagnostic confidence level score of 4, and an Independence Scale (IS) score ≥ 70] were included. The primary variables were the Huntington's Disease Health-related Quality of Life (HDQLIFE) scores and results of the Satisfaction with Life Scale (SWLS). Secondary outcomes include the Unified HD Rating Scale (UHDRS), Beck Hopelessness Scale (BHS), Stigma Scale for Chronic Illness (SSCI-8), Beck Depression Inventory-Fast Screen (BDI-FS) and Problem Behaviours Assessment for HD short Version (PBA-S). RESULTS: A total of 102 patients were included. The mean age (SD) was 53.1 (12.1) years and 56% were male. Most of the patients (99.0%) showed motor symptoms (87.3%), behavioural and psychiatric disturbances (59.8%), or cognitive impairment (20.6%). HDQLIFE domain score means (SD) includes concern with death and dying 45.97 (9.60) end-of-life planning 37.91 (8.84), and meaning and purpose 44.74 (9.05). SWLS score mean was 24.25 (7.33). Depressive symptoms were found in 37.4% of patients and moderate-to-severe feelings of hopelessness in 32.9%. The prevalence of stigma was 55.9% (n = 57). CONCLUSION: HD impacted quality of life, with prevalent motor, psychiatric symptoms and cognitive impairment. Patient perspectives may provide complementary information to implement specific interventions.

18.
Mov Disord Clin Pract ; 10(6): 1004-1012, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37332648

ABSTRACT

Background: Magnesium is an important intracellular cation involved in essential enzymatic reactions. It is necessary for neuronal function and its depletion can produce neurological symptoms such as cramps or seizures. Clinical consequences of its deficit in the cerebellum are less known and the diagnosis can be delayed because of the lack of awareness on this condition. Cases: We present three cases of cerebellar syndrome (CS) due to hypomagnesemia: A midline CS with myoclonus and ocular flutter and two cases of hemispheric CS, one of them entailed a Schmahmann's syndrome and the other suffered a seizure. MRI findings revealed cerebellar vasogenic edema and the symptoms improved after magnesium replacement in all cases. Literature Review: We reviewed 22 cases of CS due to hypomagnesemia, all with subacute onset (days to weeks). Encephalopathy and/or epileptic seizures were common. MRI findings were vasogenic edema involving the cerebellar hemispheres, the vermis, or the nodule. Up to 50% of patients presented hypocalcemia and/or hypokalemia. All the patients showed symptomatic improvement after magnesium replacement, but 50% showed significant sequelae, and 46% relapsed. Conclusions: Hypomagnesaemia should always be considered in the differential diagnosis of CS as it has a potential treatment, and its early recognition can avoid recurrences and permanent cerebellar impairment.

19.
J Neurol ; 270(11): 5408-5417, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37462754

ABSTRACT

BACKGROUND: Progressive cognitive decline is an inevitable feature of Huntington's disease (HD) but specific criteria and instruments are still insufficiently developed to reliably classify patients into categories of cognitive severity and to monitor the progression of cognitive impairment. METHODS: We collected data from a cohort of 180 positive gene-carriers: 33 with premanifest HD and 147 with manifest HD. Using a specifically developed gold-standard for cognitive status we classified participants into those with normal cognition, those with mild cognitive impairment, and those with dementia. We administered the Parkinson's Disease-Cognitive Rating Scale (PD-CRS), the MMSE and the UHDRS cogscore at baseline, and at 6-month and 12-month follow-up visits. Cutoff scores discriminating between the three cognitive categories were calculated for each instrument. For each cognitive group and instrument we addressed cognitive progression, sensitivity to change, and the minimally clinical important difference corresponding to conversion from one category to another. RESULTS: The PD-CRS cutoff scores for MCI and dementia showed excellent sensitivity and specificity ratios that were not achieved with the other instruments. Throughout follow-up, in all cognitive groups, PD-CRS captured the rate of conversion from one cognitive category to another and also the different patterns in terms of cognitive trajectories. CONCLUSION: The PD-CRS is a valid and reliable instrument to capture MCI and dementia syndromes in HD. It captures the different trajectories of cognitive progression as a function of cognitive status and shows sensitivity to change in MCI and dementia.


Subject(s)
Cognitive Dysfunction , Huntington Disease , Parkinson Disease , Humans , Huntington Disease/complications , Huntington Disease/diagnosis , Huntington Disease/genetics , Neuropsychological Tests , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/etiology , Cognitive Dysfunction/psychology , Cognition , Parkinson Disease/complications , Parkinson Disease/diagnosis
20.
J Neurol ; 269(7): 3541-3549, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35061089

ABSTRACT

BACKGROUND: Patients with Huntington's disease (HD) exhibit a variable predominance of cognitive, behavioral and motor symptoms. A specific instrument focusing on the impact of cognitive impairment in HD over functional capacity is lacking. OBJECTIVE: To address the need for a brief and specifically developed HD questionnaire able to capture functional aspects suspected to be sensitive to cognitive impairment. METHODS: We developed and validated the "Huntington's Disease-Cognitive Functional Rating Scale" (HD-CFRS) in 78 symptomatic carriers of the Huntington's disease mutation. We also administered the HD-CFRS to a knowledgeable informant to measure the level of agreement. To explore the association between HD-CFRS scores and participants' cognitive status, we administered objective measures of cognition. Participants were classified as cognitively preserved (HD-NC), as having mild cognitive impairment (HD-MCI), or as having dementia (HD-Dem). RESULTS: The HD-CFRS showed concurrent validity and internal consistency in the three groups. HD carriers and informants in the HD-NC group obtained similar HD-CFRS scores. However, in patients with mild cognitive impairment and dementia, informers reported greater functional impairment than HD participants. The HD-CFRS total score showed strong correlations with measures assessing cognition. CONCLUSIONS: These findings support the utility of the HD-CFRS as a brief and reliable instrument to measure functional defects associated with cognitive impairment in HD. We believe this questionnaire could be a useful tool both for clinical practice and research.


Subject(s)
Cognition Disorders , Cognitive Dysfunction , Huntington Disease , Cognition , Cognition Disorders/complications , Cognitive Dysfunction/complications , Cognitive Dysfunction/etiology , Humans , Neuropsychological Tests , Surveys and Questionnaires
SELECTION OF CITATIONS
SEARCH DETAIL