51
|
Saft C, Jessen J, Hoffmann R, Lukas C, Skodda S. Speech Biomarkers in Huntington's Disease: A Longitudinal Follow-Up Study in Premanifest Mutation Carriers. J Huntingtons Dis 2024; 13:369-373. [PMID: 38995795 DOI: 10.3233/jhd-240021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Speech alterations have been reported in manifest Huntington's disease (HD) and premanifest mutation carriers (preHD). The aim of our study was to explore these alterations in preHD and whether they can be used as biomarkers. 13 preHD mutation carriers performed reading task, sustained phonation task and syllable repetition tasks at baseline and after 21 months, as well as clinical examination and MRI. Syllable repetition capacity and self-chosen velocity of single syllable repetition differed significantly between time points. There were no changes in clinical ratings or MRI volumetry. Measurements of speech might be sensitive tools for monitoring subclinical changes in preHD.
Collapse
Affiliation(s)
- Carsten Saft
- Department of Neurology, Huntington-Centre NRW, St. Josef Hospital, Ruhr-University of Bochum, Bochum, Germany
| | - Julia Jessen
- Department of Neurology, Huntington-Centre NRW, St. Josef Hospital, Ruhr-University of Bochum, Bochum, Germany
| | - Rainer Hoffmann
- Department of Neurology, Huntington-Centre NRW, St. Josef Hospital, Ruhr-University of Bochum, Bochum, Germany
- Department of Neuropsychiatry, Huntington Centre South, kbo-Isar-Amper-Klinikum, Taufkirchen (Vils), Germany
| | - Carsten Lukas
- Institute of Neuroradiology, St. Josef Hospital, Ruhr-University of Bochum, Bochum, Germany
| | - Sabine Skodda
- Department of Neurology, Knappschaftskrankenhaus, Ruhr-University of Bochum, Bochum, Germany
| |
Collapse
|
52
|
Davidson JM, Zhang L, Yue GH, Di Ieva A. Fractal Dimension Studies of the Brain Shape in Aging and Neurodegenerative Diseases. ADVANCES IN NEUROBIOLOGY 2024; 36:329-363. [PMID: 38468041 DOI: 10.1007/978-3-031-47606-8_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
The fractal dimension is a morphometric measure that has been used to investigate the changes of brain shape complexity in aging and neurodegenerative diseases. This chapter reviews fractal dimension studies in aging and neurodegenerative disorders in the literature. Research has shown that the fractal dimension of the left cerebral hemisphere increases until adolescence and then decreases with aging, while the fractal dimension of the right hemisphere continues to increase until adulthood. Studies in neurodegenerative diseases demonstrated a decline in the fractal dimension of the gray matter and white matter in Alzheimer's disease, amyotrophic lateral sclerosis, and spinocerebellar ataxia. In multiple sclerosis, the white matter fractal dimension decreases, but conversely, the fractal dimension of the gray matter increases at specific stages of disease. There is also a decline in the gray matter fractal dimension in frontotemporal dementia and multiple system atrophy of the cerebellar type and in the white matter fractal dimension in epilepsy and stroke. Region-specific changes in fractal dimension have also been found in Huntington's disease and Parkinson's disease. Associations were found between the fractal dimension and clinical scores, showing the potential of the fractal dimension as a marker to monitor brain shape changes in normal or pathological processes and predict cognitive or motor function.
Collapse
Affiliation(s)
- Jennilee M Davidson
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | | | - Guang H Yue
- Center for Mobility and Rehabilitation Engineering Research, Kessler Foundation, West Orange, NJ, USA
- Department of Physical Medicine and Rehabilitation, Rutgers New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Antonio Di Ieva
- Computational Neurosurgery (CNS) Lab, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Macquarie Park, NSW, Australia
| |
Collapse
|
53
|
Coleman A, Langan MT, Verma G, Knights H, Sturrock A, Leavitt BR, Tabrizi SJ, Scahill RI, Hobbs NZ. Assessment of Perivascular Space Morphometry Across the White Matter in Huntington's Disease Using MRI. J Huntingtons Dis 2024; 13:91-101. [PMID: 38517798 DOI: 10.3233/jhd-231508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2024]
Abstract
Background Perivascular spaces (PVS) are fluid-filled cavities surrounding small cerebral blood vessels. There are limited reports of enlarged PVS across the grey matter in manifest Huntington's disease (HD). Little is known about how PVS morphometry in the white matter may contribute to HD. Enlarged PVS have the potential to both contribute to HD pathology and affect the distribution and success of intraparenchymal and intrathecally administered huntingtin-lowering therapies. Objective To investigate PVS morphometry in the global white matter across the spectrum of HD. Relationships between PVS morphometry and disease burden and severity measures were examined. Methods White matter PVS were segmented on 3T T2 W MRI brain scans of 33 healthy controls, 30 premanifest HD (pre-HD), and 32 early manifest HD (early-HD) participants from the Vancouver site of the TRACK-HD study. PVS count and total PVS volume were measured. Results PVS total count slightly increased in pre-HD (p = 0.004), and early-HD groups (p = 0.005), compared to healthy controls. PVS volume, as a percentage of white matter volume, increased subtly in pre-HD compared to healthy controls (p = 0.044), but not in early-HD. No associations between PVS measures and HD disease burden or severity were found. Conclusions This study reveals relatively preserved PVS morphometry across the global white matter of pre-HD and early-HD. Subtle morphometric abnormalities are implied but require confirmation in a larger cohort. However, in conjunction with previous publications, further investigation of PVS in HD and its potential impact on future treatments, with a focus on subcortical grey matter, is warranted.
Collapse
Affiliation(s)
- Annabelle Coleman
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| | - Mackenzie T Langan
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Biomedical Engineering and Imaging Institute at Mount Sinai School of Medicine, New York, NY, USA
| | - Gaurav Verma
- Biomedical Engineering and Imaging Institute at Mount Sinai School of Medicine, New York, NY, USA
| | - Harry Knights
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| | - Aaron Sturrock
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Blair R Leavitt
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Sarah J Tabrizi
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| | - Rachael I Scahill
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| | - Nicola Z Hobbs
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| |
Collapse
|
54
|
Aracil-Bolaños I, Pérez-Pérez J, Martínez-Horta S, Horta-Barba A, Puig-Davi A, García-Cornet J, Olmedo-Saura G, Campolongo A, Pagonabarraga J, Kulisevsky J. Baseline Large-Scale Network Dynamics Associated with Disease Progression in Huntington's Disease. Mov Disord 2024; 39:197-203. [PMID: 38148511 DOI: 10.1002/mds.29655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/11/2023] [Accepted: 10/24/2023] [Indexed: 12/28/2023] Open
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.
Collapse
Affiliation(s)
- Ignacio Aracil-Bolaños
- Movement Disorders Unit, Neurology Department, Sant Pau Hospital, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona (U.A.B.), Barcelona, Spain
- Institut d'Investigacions Biomèdiques-Sant Pau (IIB-Sant Pau), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jesús Pérez-Pérez
- Movement Disorders Unit, Neurology Department, Sant Pau Hospital, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona (U.A.B.), Barcelona, Spain
- Institut d'Investigacions Biomèdiques-Sant Pau (IIB-Sant Pau), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Saül Martínez-Horta
- Movement Disorders Unit, Neurology Department, Sant Pau Hospital, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona (U.A.B.), Barcelona, Spain
- Institut d'Investigacions Biomèdiques-Sant Pau (IIB-Sant Pau), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Andrea Horta-Barba
- Movement Disorders Unit, Neurology Department, Sant Pau Hospital, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona (U.A.B.), Barcelona, Spain
- Institut d'Investigacions Biomèdiques-Sant Pau (IIB-Sant Pau), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Arnau Puig-Davi
- Movement Disorders Unit, Neurology Department, Sant Pau Hospital, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona (U.A.B.), Barcelona, Spain
- Institut d'Investigacions Biomèdiques-Sant Pau (IIB-Sant Pau), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Júlia García-Cornet
- Movement Disorders Unit, Neurology Department, Sant Pau Hospital, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona (U.A.B.), Barcelona, Spain
- Institut d'Investigacions Biomèdiques-Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Gonzalo Olmedo-Saura
- Movement Disorders Unit, Neurology Department, Sant Pau Hospital, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona (U.A.B.), Barcelona, Spain
- Institut d'Investigacions Biomèdiques-Sant Pau (IIB-Sant Pau), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Antonia Campolongo
- Movement Disorders Unit, Neurology Department, Sant Pau Hospital, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona (U.A.B.), Barcelona, Spain
- Institut d'Investigacions Biomèdiques-Sant Pau (IIB-Sant Pau), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Javier Pagonabarraga
- Movement Disorders Unit, Neurology Department, Sant Pau Hospital, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona (U.A.B.), Barcelona, Spain
- Institut d'Investigacions Biomèdiques-Sant Pau (IIB-Sant Pau), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jaime Kulisevsky
- Movement Disorders Unit, Neurology Department, Sant Pau Hospital, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona (U.A.B.), Barcelona, Spain
- Institut d'Investigacions Biomèdiques-Sant Pau (IIB-Sant Pau), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
55
|
Knights H, Coleman A, Hobbs NZ, Tabrizi SJ, Scahill RI. Freesurfer Software Update Significantly Impacts Striatal Volumes in the Huntington's Disease Young Adult Study and Will Influence HD-ISS Staging. J Huntingtons Dis 2024; 13:77-90. [PMID: 38489194 DOI: 10.3233/jhd-231512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
Background The Huntington's Disease Integrated Staging System (HD-ISS) defined disease onset using volumetric cut-offs for caudate and putamen derived from FreeSurfer 6 (FS6). The impact of the latest software update (FS7) on volumes remains unknown. The Huntington's Disease Young Adult Study (HD-YAS) is appropriately positioned to explore differences in FS bias when detecting early atrophy. Objective Explore the relationships and differences between raw caudate and putamen volumes, calculated total intracranial volumes (cTICV), and adjusted caudate and putamen volumes, derived from FS6 and FS7, in HD-YAS. Methods Images from 123 participants were segmented and quality controlled. Relationships and differences between volumes were explored using intraclass correlation (ICC) and Bland-Altman analysis. Results Across the whole cohort, ICC for raw caudate and putamen was 0.99, cTICV 0.93, adjusted caudate 0.87, and adjusted putamen 0.86 (all p < 0.0005). Compared to FS6, FS7 calculated: i) larger raw caudate (+0.8%, p < 0.00005) and putamen (+1.9%, p < 0.00005), with greater difference for larger volumes; and ii) smaller cTICV (-5.1%, p < 0.00005), with greater difference for smaller volumes. The systematic and proportional difference in cTICV was greater than raw volumes. When raw volumes were adjusted for cTICV, these effects compounded (adjusted caudate +7.0%, p < 0.00005; adjusted putamen +8.2%, p < 0.00005), with greater difference for larger volumes. Conclusions As new software is released, it is critical that biases are explored since differences have the potential to significantly alter the findings of HD trials. Until conversion factors are defined, the HD-ISS must be applied using FS6. This should be incorporated into the HD-ISS online calculator.
Collapse
Affiliation(s)
- Harry Knights
- Department of Neurodegenerative Disease, Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Annabelle Coleman
- Department of Neurodegenerative Disease, Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Nicola Z Hobbs
- Department of Neurodegenerative Disease, Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Sarah J Tabrizi
- Department of Neurodegenerative Disease, Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Rachael I Scahill
- Department of Neurodegenerative Disease, Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
56
|
Hobbs NZ, Papoutsi M, Delva A, Kinnunen KM, Nakajima M, Van Laere K, Vandenberghe W, Herath P, Scahill RI. Neuroimaging to Facilitate Clinical Trials in Huntington's Disease: Current Opinion from the EHDN Imaging Working Group. J Huntingtons Dis 2024; 13:163-199. [PMID: 38788082 PMCID: PMC11307036 DOI: 10.3233/jhd-240016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2024] [Indexed: 05/26/2024]
Abstract
Neuroimaging is increasingly being included in clinical trials of Huntington's disease (HD) for a wide range of purposes from participant selection and safety monitoring, through to demonstration of disease modification. Selection of the appropriate modality and associated analysis tools requires careful consideration. On behalf of the EHDN Imaging Working Group, we present current opinion on the utility and future prospects for inclusion of neuroimaging in HD trials. Covering the key imaging modalities of structural-, functional- and diffusion- MRI, perfusion imaging, positron emission tomography, magnetic resonance spectroscopy, and magnetoencephalography, we address how neuroimaging can be used in HD trials to: 1) Aid patient selection, enrichment, stratification, and safety monitoring; 2) Demonstrate biodistribution, target engagement, and pharmacodynamics; 3) Provide evidence for disease modification; and 4) Understand brain re-organization following therapy. We also present the challenges of translating research methodology into clinical trial settings, including equipment requirements and cost, standardization of acquisition and analysis, patient burden and invasiveness, and interpretation of results. We conclude, that with appropriate consideration of modality, study design and analysis, imaging has huge potential to facilitate effective clinical trials in HD.
Collapse
Affiliation(s)
- Nicola Z. Hobbs
- HD Research Centre, UCL Institute of Neurology, UCL, London, UK
| | - Marina Papoutsi
- HD Research Centre, UCL Institute of Neurology, UCL, London, UK
- IXICO plc, London, UK
| | - Aline Delva
- Department of Neurosciences, KU Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Belgium
| | | | | | - Koen Van Laere
- Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, KU Leuven, Belgium
- Division of Nuclear Medicine, University Hospitals Leuven, Belgium
| | - Wim Vandenberghe
- Department of Neurosciences, KU Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Belgium
| | | | | |
Collapse
|
57
|
Yin JH, Liu YO, Li HL, Burgunder JM, Huang Y. White Matter Microstructure Changes Revealed by Diffusion Kurtosis and Diffusion Tensor Imaging in Mutant Huntingtin Gene Carriers. J Huntingtons Dis 2024; 13:301-313. [PMID: 38905054 PMCID: PMC11494636 DOI: 10.3233/jhd-240018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2024] [Indexed: 06/23/2024]
Abstract
Background Diffusion magnetic resonance imaging (dMRI) has revealed microstructural changes in white matter (WM) in Huntington's disease (HD). Objective To compare the validities of different dMRI, i.e., diffusion kurtosis imaging (DKI) and diffusion tensor imaging (DTI) in HD. Methods 22 mutant huntingtin (mHTT) carriers and 14 controls were enrolled. Clinical assessments and dMRI were conducted. Based on CAG-Age Product (CAP) score, mHTT carriers were categorized into high CAP (hCAP) and medium and low CAP (m& lCAP) groups. Spearman analyses were used to explore correlations between imaging parameters in brain regions and clinical assessments. Receiver operating characteristic (ROC) was used to distinguish mHTT carriers from control, and define the HD patients at advanced stage. Results Compared to controls, mHTT carriers exhibited WM changes in DKI and DTI. There were 22 more regions showing significant differences in HD detected by MK than FA. Only MK in five brain regions showed significantly difference between any two group, and negatively correlated with the disease burden (r = -0.80 to -0.71). ROC analysis revealed that MK was more sensitive and FA was more specific, while Youden index showed that the integration of FA and MK gave rise to higher authenticities, in distinguishing m& lCAP from controls (Youden Index = 0.786), and discerning different phase of HD (Youden Index = 0.804). Conclusions Microstructural changes in WM occur at early stage of HD and deteriorate over the disease progression. Integrating DKI and DTI would provide the best accuracies for differentiating early HD from control and identifying advanced HD.
Collapse
Affiliation(s)
- Jin-Hui Yin
- Human Brain & Tissue Bank, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ya-Ou Liu
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hong-Liang Li
- Department of Neurology, Aviation General Hospital, Beijing, China
| | - Jean Marc Burgunder
- Department of Neurology, Swiss Huntington’s Disease Centre, Siloah, and Department of Neurology, University Hospital, Gümligen (Muri bei Bern), Switzerland
| | - Yue Huang
- Human Brain & Tissue Bank, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Pharmacology Department, School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
| |
Collapse
|
58
|
Mirzai N, Polet K, Morisot A, Hesse S, Pesce A, Louchart de la Chapelle S, Iakimova G. Can the Ability to Recognize Facial Emotions in Individuals With Neurodegenerative Disease be Improved? A Systematic Review and Meta-analysis. Cogn Behav Neurol 2023; 36:202-218. [PMID: 37410880 PMCID: PMC10683976 DOI: 10.1097/wnn.0000000000000348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 03/30/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND Facial emotion recognition (FER) is commonly impaired in individuals with neurodegenerative disease (NDD). This impairment has been linked to an increase in behavioral disorders and caregiver burden. OBJECTIVE To identify interventions targeting the improvement of FER ability in individuals with NDD and investigate the magnitude of the efficacy of the interventions. We also wanted to explore the duration of the effects of the intervention and their possible impacts on behavioral and psychological symptoms of dementia and caregiver burden. METHOD We included 15 studies with 604 individuals who had been diagnosed with NDD. The identified interventions were categorized into three types of approach (cognitive, neurostimulation, and pharmacological) as well as a combined approach (neurostimulation with pharmacological). RESULTS The three types of approaches pooled together had a significant large effect size for FER ability improvement (standard mean difference: 1.21, 95% CI = 0.11, 2.31, z = 2.15, P = 0.03). The improvement lasted post intervention, in tandem with a decrease in behavioral disorders and caregiver burden. CONCLUSION A combination of different approaches for FER ability improvement may be beneficial for individuals with NDD and their caregivers.
Collapse
Affiliation(s)
- Naz Mirzai
- Clinical Research Unit–Memory Clinic, Princess Grace Hospital, Monaco
- Cote d’Azur University, Laboratory of Clinical, Cognitive and Social Anthropology and Psychology, Nice, France
| | - Kévin Polet
- Clinical Research Unit–Memory Clinic, Princess Grace Hospital, Monaco
| | - Adeline Morisot
- Clinical Research Unit–Memory Clinic, Princess Grace Hospital, Monaco
- Public Health Department, Cote d’Azur University, University Hospital Center of Nice, Nice, France
| | - Solange Hesse
- Clinical Research Unit–Memory Clinic, Princess Grace Hospital, Monaco
| | - Alain Pesce
- Bibliographic Research Association for Neurosciences, Nice, France
| | | | - Galina Iakimova
- Cote d’Azur University, Laboratory of Clinical, Cognitive and Social Anthropology and Psychology, Nice, France
| |
Collapse
|
59
|
Toader C, Dobrin N, Brehar FM, Popa C, Covache-Busuioc RA, Glavan LA, Costin HP, Bratu BG, Corlatescu AD, Popa AA, Ciurea AV. From Recognition to Remedy: The Significance of Biomarkers in Neurodegenerative Disease Pathology. Int J Mol Sci 2023; 24:16119. [PMID: 38003309 PMCID: PMC10671641 DOI: 10.3390/ijms242216119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 10/28/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
With the inexorable aging of the global populace, neurodegenerative diseases (NDs) like Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS) pose escalating challenges, which are underscored by their socioeconomic repercussions. A pivotal aspect in addressing these challenges lies in the elucidation and application of biomarkers for timely diagnosis, vigilant monitoring, and effective treatment modalities. This review delineates the quintessence of biomarkers in the realm of NDs, elucidating various classifications and their indispensable roles. Particularly, the quest for novel biomarkers in AD, transcending traditional markers in PD, and the frontier of biomarker research in ALS are scrutinized. Emergent susceptibility and trait markers herald a new era of personalized medicine, promising enhanced treatment initiation especially in cases of SOD1-ALS. The discourse extends to diagnostic and state markers, revolutionizing early detection and monitoring, alongside progression markers that unveil the trajectory of NDs, propelling forward the potential for tailored interventions. The synergy between burgeoning technologies and innovative techniques like -omics, histologic assessments, and imaging is spotlighted, underscoring their pivotal roles in biomarker discovery. Reflecting on the progress hitherto, the review underscores the exigent need for multidisciplinary collaborations to surmount the challenges ahead, accelerate biomarker discovery, and herald a new epoch of understanding and managing NDs. Through a panoramic lens, this article endeavors to provide a comprehensive insight into the burgeoning field of biomarkers in NDs, spotlighting the promise they hold in transforming the diagnostic landscape, enhancing disease management, and illuminating the pathway toward efficacious therapeutic interventions.
Collapse
Affiliation(s)
- Corneliu Toader
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.D.C.); (A.V.C.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Nicolaie Dobrin
- Department of Neurosurgery, Clinical Emergency Hospital “Prof. Dr. Nicolae Oblu”, 700309 Iasi, Romania
| | - Felix-Mircea Brehar
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.D.C.); (A.V.C.)
- Department of Neurosurgery, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
| | - Constantin Popa
- Department of Neurology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Neurology, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
- Medical Science Section, Romanian Academy, 060021 Bucharest, Romania
| | - Razvan-Adrian Covache-Busuioc
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.D.C.); (A.V.C.)
| | - Luca Andrei Glavan
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.D.C.); (A.V.C.)
| | - Horia Petre Costin
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.D.C.); (A.V.C.)
| | - Bogdan-Gabriel Bratu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.D.C.); (A.V.C.)
| | - Antonio Daniel Corlatescu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.D.C.); (A.V.C.)
| | - Andrei Adrian Popa
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.D.C.); (A.V.C.)
| | - Alexandru Vlad Ciurea
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (L.A.G.); (H.P.C.); (B.-G.B.); (A.D.C.); (A.V.C.)
- Medical Science Section, Romanian Academy, 060021 Bucharest, Romania
- Neurosurgery Department, Sanador Clinical Hospital, 010991 Bucharest, Romania
| |
Collapse
|
60
|
Tramutola A, Bakels HS, Perrone F, Di Nottia M, Mazza T, Abruzzese MP, Zoccola M, Pagnotta S, Carrozzo R, de Bot ST, Perluigi M, van Roon-Mom WMC, Squitieri F. GLUT-1 changes in paediatric Huntington disease brain cortex and fibroblasts: an observational case-control study. EBioMedicine 2023; 97:104849. [PMID: 37898095 PMCID: PMC10630613 DOI: 10.1016/j.ebiom.2023.104849] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 10/30/2023] Open
Abstract
BACKGROUND Paediatric Huntington disease with highly expanded mutations (HE-PHD; >80 CAG repeats) presents atypically, compared to adult-onset Huntington disease (AOHD), with neurodevelopmental delay, epilepsy, abnormal brain glucose metabolism, early striatal damage, and reduced lifespan. Since genetic GLUT-1 deficiency syndrome shows a symptom spectrum similar to HE-PHD, we investigated the potential role of the two main glucose transporters, GLUT-1 and GLUT-3, in HE-PHD. METHODS We compared GLUT-1 and GLUT-3 protein expression in HE-PHD, juvenile-onset (JOHD), and AOHD brains (n = 2; n = 3; n = 6) and periphery (n = 3; n = 2; n = 2) versus healthy adult controls (n = 6; n = 6). We also investigated mitochondrial complexes and hexokinase-II protein expression. FINDINGS GLUT-1 and GLUT-3 expression were significantly lower in HE-PHD frontal cortex (p = 0.009, 95% [CI 13.4, 14.7]; p = 0.017, 95% [CI 14.2, 14.5]) versus controls. In fibroblasts, GLUT-1 and GLUT-3 expression were lower compared to controls (p < 0.0001, 95% [CI 0.91, 1.09]; p = 0.046, 95% [CI 0.93, 1.07]). In the frontal cortex, this occurred without evidence of extensive neuronal degeneration. Patients with HE-PHD had deregulated mitochondrial complex expression, particularly complexes II-III, levels of which were lower in frontal cortex versus controls (p = 0.027, 95% [CI 17.1, 17.6]; p = 0.002, 95% CI [16.6, 16.9]) and patients with AOHD (p = 0.052, 95% [CI 17.0, 17.6]; p = 0.002, 95% [CI 16.6, 16.7]). Hexokinase-II expression was also lower in HE-PHD frontal cortex and striatum versus controls (p = 0.010, 95% [CI 17.8, 18.2]; p = 0.045, 95% [CI 18.6, 18.7]) and in frontal cortex versus patients with AOHD (p = 0.013, 95% [CI 17.7, 18.1]). Expression JOHD levels were consistently different to those of HE-PHD but similar to those of AOHD. INTERPRETATION Our data suggest a dysfunctional hypometabolic state occurring specifically in paediatric Huntington disease brains. FUNDING '5 × 1000' Personal Income Tax donation to LIRH Foundation; Italian Ministry of HealthRC2301MH04 and RF-2016-02364123 to CSS.
Collapse
Affiliation(s)
- Antonella Tramutola
- Department of Biochemical Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Hannah S Bakels
- Department of Neurology, Leiden University Medical Centre, ZA Leiden 2311, the Netherlands
| | - Federica Perrone
- Huntington and Rare Diseases Unit, IRCCS Casa Sollievo della Sofferenza (CSS) Research Hospital, San Giovanni Rotondo 71013, Italy
| | - Michela Di Nottia
- Unit of Cellular Biology and Mitochondrial Diseases, IRCCS Bambino Gesú Children's Hospital, Rome 00146, Italy
| | - Tommaso Mazza
- Bioinformatics Unit, IRCCS Casa Sollievo della Sofferenza (CSS) Research Hospital, San Giovanni Rotondo 71013, Italy
| | - Maria Pia Abruzzese
- Huntington and Rare Diseases Unit, IRCCS Casa Sollievo della Sofferenza (CSS) Research Hospital, San Giovanni Rotondo 71013, Italy
| | - Martina Zoccola
- Unit of Cellular Biology and Mitochondrial Diseases, IRCCS Bambino Gesú Children's Hospital, Rome 00146, Italy
| | - Sara Pagnotta
- Department of Biochemical Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Rosalba Carrozzo
- Unit of Cellular Biology and Mitochondrial Diseases, IRCCS Bambino Gesú Children's Hospital, Rome 00146, Italy
| | - Susanne T de Bot
- Department of Neurology, Leiden University Medical Centre, ZA Leiden 2311, the Netherlands
| | - Marzia Perluigi
- Department of Biochemical Sciences, Sapienza University of Rome, Rome 00185, Italy
| | | | - Ferdinando Squitieri
- Huntington and Rare Diseases Unit, IRCCS Casa Sollievo della Sofferenza (CSS) Research Hospital, San Giovanni Rotondo 71013, Italy; Centre for Rare Neurological Diseases (CMRN), Italian League for Research on Huntington (LIRH) Foundation, Viale di Villa Massimo 4, Rome 00161, Italy.
| |
Collapse
|
61
|
Wilton DK, Mastro K, Heller MD, Gergits FW, Willing CR, Fahey JB, Frouin A, Daggett A, Gu X, Kim YA, Faull RLM, Jayadev S, Yednock T, Yang XW, Stevens B. Microglia and complement mediate early corticostriatal synapse loss and cognitive dysfunction in Huntington's disease. Nat Med 2023; 29:2866-2884. [PMID: 37814059 PMCID: PMC10667107 DOI: 10.1038/s41591-023-02566-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 08/24/2023] [Indexed: 10/11/2023]
Abstract
Huntington's disease (HD) is a devastating monogenic neurodegenerative disease characterized by early, selective pathology in the basal ganglia despite the ubiquitous expression of mutant huntingtin. The molecular mechanisms underlying this region-specific neuronal degeneration and how these relate to the development of early cognitive phenotypes are poorly understood. Here we show that there is selective loss of synaptic connections between the cortex and striatum in postmortem tissue from patients with HD that is associated with the increased activation and localization of complement proteins, innate immune molecules, to these synaptic elements. We also found that levels of these secreted innate immune molecules are elevated in the cerebrospinal fluid of premanifest HD patients and correlate with established measures of disease burden.In preclinical genetic models of HD, we show that complement proteins mediate the selective elimination of corticostriatal synapses at an early stage in disease pathogenesis, marking them for removal by microglia, the brain's resident macrophage population. This process requires mutant huntingtin to be expressed in both cortical and striatal neurons. Inhibition of this complement-dependent elimination mechanism through administration of a therapeutically relevant C1q function-blocking antibody or genetic ablation of a complement receptor on microglia prevented synapse loss, increased excitatory input to the striatum and rescued the early development of visual discrimination learning and cognitive flexibility deficits in these models. Together, our findings implicate microglia and the complement cascade in the selective, early degeneration of corticostriatal synapses and the development of cognitive deficits in presymptomatic HD; they also provide new preclinical data to support complement as a therapeutic target for early intervention.
Collapse
Affiliation(s)
- Daniel K Wilton
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US.
| | - Kevin Mastro
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US
| | - Molly D Heller
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US
| | - Frederick W Gergits
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US
| | - Carly Rose Willing
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US
| | - Jaclyn B Fahey
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US
| | - Arnaud Frouin
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US
| | - Anthony Daggett
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Xiaofeng Gu
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Yejin A Kim
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US
| | - Richard L M Faull
- Department of Anatomy with Radiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Suman Jayadev
- Department of Neurology, University of Washington, Seattle, WA, USA
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Ted Yednock
- Annexon Biosciences, South San Francisco, CA, USA
| | - X William Yang
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Beth Stevens
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US.
- Stanley Center, Broad Institute, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
62
|
Horta-Barba A, Martinez-Horta S, Pérez-Pérez J, Puig-Davi A, de Lucia N, de Michele G, Salvatore E, Kehrer S, Priller J, Migliore S, Squitieri F, Castaldo A, Mariotti C, Mañanes V, Lopez-Sendon JL, Rodriguez N, Martinez-Descals A, Júlio F, Januário C, Delussi M, de Tommaso M, Noguera S, Ruiz-Idiago J, Sitek EJ, Wallner R, Nuzzi A, Pagonabarraga J, Kulisevsky J. Measuring cognitive impairment and monitoring cognitive decline in Huntington's disease: a comparison of assessment instruments. J Neurol 2023; 270:5408-5417. [PMID: 37462754 PMCID: PMC10576674 DOI: 10.1007/s00415-023-11804-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 10/15/2023]
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.
Collapse
Affiliation(s)
- Andrea Horta-Barba
- Department of Medicine, Autonomous University of Barcelona (UAB), Bellaterra, Spain
- Movement Disorders Unit, Neurology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain
- Sant Pau Biomedical Research Institute (IIB Sant Pau), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- European Huntington's Disease Network (EHDN), Ulm, Germany
| | - Saul Martinez-Horta
- Department of Medicine, Autonomous University of Barcelona (UAB), Bellaterra, Spain
- Movement Disorders Unit, Neurology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain
- Sant Pau Biomedical Research Institute (IIB Sant Pau), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- European Huntington's Disease Network (EHDN), Ulm, Germany
| | - Jesús Pérez-Pérez
- Department of Medicine, Autonomous University of Barcelona (UAB), Bellaterra, Spain
- Movement Disorders Unit, Neurology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain
- Sant Pau Biomedical Research Institute (IIB Sant Pau), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- European Huntington's Disease Network (EHDN), Ulm, Germany
| | - Arnau Puig-Davi
- Movement Disorders Unit, Neurology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain
- Sant Pau Biomedical Research Institute (IIB Sant Pau), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- European Huntington's Disease Network (EHDN), Ulm, Germany
| | - Natascia de Lucia
- European Huntington's Disease Network (EHDN), Ulm, Germany
- University of Naples "Federico II", Naples, Italy
| | - Giuseppe de Michele
- European Huntington's Disease Network (EHDN), Ulm, Germany
- University of Naples "Federico II", Naples, Italy
| | - Elena Salvatore
- European Huntington's Disease Network (EHDN), Ulm, Germany
- University of Naples "Federico II", Naples, Italy
| | - Stefanie Kehrer
- European Huntington's Disease Network (EHDN), Ulm, Germany
- Department of Neuropsychiatry, Charité-Universitätsmedizin, Berlin, Germany
| | - Josef Priller
- European Huntington's Disease Network (EHDN), Ulm, Germany
- Department of Neuropsychiatry, Charité-Universitätsmedizin, Berlin, Germany
| | - Simone Migliore
- Huntington and Rare Diseases Unit, Fondazione IRCCS Casa Sollievo Della Sofferenza Research Hospital, San Giovanni Rotondo, Italy
| | - Ferdinando Squitieri
- Huntington and Rare Diseases Unit, Fondazione IRCCS Casa Sollievo Della Sofferenza Research Hospital, San Giovanni Rotondo, Italy
| | - Anna Castaldo
- European Huntington's Disease Network (EHDN), Ulm, Germany
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Caterina Mariotti
- European Huntington's Disease Network (EHDN), Ulm, Germany
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Veronica Mañanes
- European Huntington's Disease Network (EHDN), Ulm, Germany
- Department of Neurology, Hospital Universitario Ramon Y Cajal, Madrid, Spain
| | - Jose Luis Lopez-Sendon
- European Huntington's Disease Network (EHDN), Ulm, Germany
- Department of Neurology, Hospital Universitario Ramon Y Cajal, Madrid, Spain
| | - Noelia Rodriguez
- European Huntington's Disease Network (EHDN), Ulm, Germany
- Department of Neurology, Fundación Jimenez Diaz, Madrid, Spain
| | - Asunción Martinez-Descals
- European Huntington's Disease Network (EHDN), Ulm, Germany
- Department of Neurology, Fundación Jimenez Diaz, Madrid, Spain
| | - Filipa Júlio
- European Huntington's Disease Network (EHDN), Ulm, Germany
- Coimbra Institute for Biomedical Imaging and Translational Research-CIBIT, University of Coimbra, Coimbra, Portugal
- Neurology Department, Coimbra University Hospital, Coimbra, Portugal
| | - Cristina Januário
- European Huntington's Disease Network (EHDN), Ulm, Germany
- Coimbra Institute for Biomedical Imaging and Translational Research-CIBIT, University of Coimbra, Coimbra, Portugal
- Neurology Department, Coimbra University Hospital, Coimbra, Portugal
| | - Marianna Delussi
- European Huntington's Disease Network (EHDN), Ulm, Germany
- Applied Neurophysiology and Pain Unit, Apulian Center for Huntington's Disease SMBNOS Department, "Aldo Moro" University, Bari, Italy
| | - Marina de Tommaso
- European Huntington's Disease Network (EHDN), Ulm, Germany
- Applied Neurophysiology and Pain Unit, Apulian Center for Huntington's Disease SMBNOS Department, "Aldo Moro" University, Bari, Italy
| | - Sandra Noguera
- European Huntington's Disease Network (EHDN), Ulm, Germany
- Hospital Mare de Deu de La Mercè, Barcelona, Spain
| | - Jesús Ruiz-Idiago
- European Huntington's Disease Network (EHDN), Ulm, Germany
- Hospital Mare de Deu de La Mercè, Barcelona, Spain
| | - Emilia J Sitek
- European Huntington's Disease Network (EHDN), Ulm, Germany
- Department of Neurological and Psychiatric Nursing, Faculty of Health Science Medical, University of Gdansk, Gdańsk, Poland
- Department of Neurology, St. Adalbert Hospital, Copernicus, Gdańsk, Poland
| | - Renata Wallner
- Department of Psychiatry, Medical University of Wroclaw, Wroclaw, Poland
| | - Angela Nuzzi
- European Huntington's Disease Network (EHDN), Ulm, Germany
| | - Javier Pagonabarraga
- Department of Medicine, Autonomous University of Barcelona (UAB), Bellaterra, Spain
- Movement Disorders Unit, Neurology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain
- Sant Pau Biomedical Research Institute (IIB Sant Pau), Barcelona, Spain
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- European Huntington's Disease Network (EHDN), Ulm, Germany
| | - Jaime Kulisevsky
- Department of Medicine, Autonomous University of Barcelona (UAB), Bellaterra, Spain.
- Movement Disorders Unit, Neurology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain.
- Sant Pau Biomedical Research Institute (IIB Sant Pau), Barcelona, Spain.
- Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
- European Huntington's Disease Network (EHDN), Ulm, Germany.
| |
Collapse
|
63
|
Ferrari Bardile C, Radulescu CI, Pouladi MA. Oligodendrocyte pathology in Huntington's disease: from mechanisms to therapeutics. Trends Mol Med 2023; 29:802-816. [PMID: 37591764 DOI: 10.1016/j.molmed.2023.07.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 08/19/2023]
Abstract
Oligodendrocytes (OLGs), highly specialized glial cells that wrap axons with myelin sheaths, are critical for brain development and function. There is new recognition of the role of OLGs in the pathogenesis of neurodegenerative diseases (NDDs), including Huntington's disease (HD), a prototypic NDD caused by a polyglutamine tract expansion in huntingtin (HTT), which results in gain- and loss-of-function effects. Clinically, HD is characterized by a constellation of motor, cognitive, and psychiatric disturbances. White matter (WM) structures, representing myelin-rich regions of the brain, are profoundly affected in HD, and recent findings reveal oligodendroglia dysfunction as an early pathological event. Here, we focus on mechanisms that underlie oligodendroglial deficits and dysmyelination in the progression of the disease, highlighting the pathogenic contributions of mutant HTT (mHTT). We also discuss potential therapeutic implications involving these molecular pathways.
Collapse
Affiliation(s)
- Costanza Ferrari Bardile
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Djavad Mowafaghian Centre for Brain Health, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Carola I Radulescu
- UK Dementia Research Institute, Imperial College London, London, W12 0NN, UK
| | - Mahmoud A Pouladi
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Djavad Mowafaghian Centre for Brain Health, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada.
| |
Collapse
|
64
|
Wilkes FA, Jakabek D, Walterfang M, Velakoulis D, Poudel GR, Stout JC, Chua P, Egan GF, Looi JCL, Georgiou-Karistianis N. The shape of things to come. Mapping spatiotemporal progression of striatal morphology in Huntington disease: The IMAGE-HD study. Psychiatry Res Neuroimaging 2023; 335:111717. [PMID: 37751638 DOI: 10.1016/j.pscychresns.2023.111717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 09/28/2023]
Abstract
Mapping the spatiotemporal progression of neuroanatomical change in Huntington's Disease (HD) is fundamental to the development of bio-measures for prognostication. Statistical shape analysis to measure the striatum has been performed in HD, however there have been a limited number of longitudinal studies. To address these limitations, we utilised the Spherical Harmonic Point Distribution Method (SPHARM-PDM) to generate point distribution models of the striatum in individuals, and used linear mixed models to test for localised shape change over time in pre-manifest HD (pre-HD), symp-HD (symp-HD) and control individuals. Longitudinal MRI scans from the IMAGE-HD study were used (baseline, 18 and 30 months). We found significant differences in the shape of the striatum between groups. Significant group-by-time interaction was observed for the putamen bilaterally, but not for caudate. A differential rate of shape change between groups over time was observed, with more significant deflation in the symp-HD group in comparison with the pre-HD and control groups. CAG repeats were correlated with bilateral striatal shape in pre-HD and symp-HD. Robust statistical analysis of the correlates of striatal shape change in HD has confirmed the suitability of striatal morphology as a potential biomarker correlated with CAG-repeat length, and potentially, an endophenotype.
Collapse
Affiliation(s)
- Fiona A Wilkes
- Research Centre for the Neurosciences of Ageing, Academic Unit of Psychiatry and Addiction Medicine, Australian National University Medical School, Canberra Hospital, Canberra, Australia.
| | | | - Mark Walterfang
- Neuropsychiatry Unit, Royal Melbourne Hospital, Melbourne Neuropsychiatry Centre, University of Melbourne and Northwestern Mental Health, Melbourne, Australia; Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Dennis Velakoulis
- Neuropsychiatry Unit, Royal Melbourne Hospital, Melbourne Neuropsychiatry Centre, University of Melbourne and Northwestern Mental Health, Melbourne, Australia; Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Govinda R Poudel
- Mary Mackillop Institute for Health Research, Australian Catholic University, Melbourne, Australia
| | - Julie C Stout
- School of Psychological Sciences and the Turner Institute of Brain and Mental Health, Monash University, Melbourne, Australia
| | - Phyllis Chua
- Department of Psychiatry, School of Clinical Sciences, Monash University, Monash Medical Centre, Melbourne, Australia
| | - Gary F Egan
- School of Psychological Sciences and the Turner Institute of Brain and Mental Health, Monash University, Melbourne, Australia
| | - Jeffrey C L Looi
- Research Centre for the Neurosciences of Ageing, Academic Unit of Psychiatry and Addiction Medicine, Australian National University Medical School, Canberra Hospital, Canberra, Australia; Neuropsychiatry Unit, Royal Melbourne Hospital, Melbourne Neuropsychiatry Centre, University of Melbourne and Northwestern Mental Health, Melbourne, Australia
| | - Nellie Georgiou-Karistianis
- School of Psychological Sciences and the Turner Institute of Brain and Mental Health, Monash University, Melbourne, Australia
| |
Collapse
|
65
|
Lutfi Ismaeel G, Makki AlHassani OJ, S Alazragi R, Hussein Ahmed A, H Mohamed A, Yasir Jasim N, Hassan Shari F, Almashhadani HA. Genetically engineered neural stem cells (NSCs) therapy for neurological diseases; state-of-the-art. Biotechnol Prog 2023; 39:e3363. [PMID: 37221947 DOI: 10.1002/btpr.3363] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/28/2023] [Accepted: 05/01/2023] [Indexed: 05/25/2023]
Abstract
Neural stem cells (NSCs) are multipotent stem cells with remarkable self-renewal potential and also unique competencies to differentiate into neurons, astrocytes, and oligodendrocytes (ODCs) and improve the cellular microenvironment. In addition, NSCs secret diversity of mediators, including neurotrophic factors (e.g., BDNF, NGF, GDNF, CNTF, and NT-3), pro-angiogenic mediators (e.g., FGF-2 and VEGF), and anti-inflammatory biomolecules. Thereby, NSCs transplantation has become a reasonable and effective treatment for various neurodegenerative disorders by their capacity to induce neurogenesis and vasculogenesis and dampen neuroinflammation and oxidative stress. Nonetheless, various drawbacks such as lower migration and survival and less differential capacity to a particular cell lineage concerning the disease pathogenesis hinder their application. Thus, genetic engineering of NSCs before transplantation is recently regarded as an innovative strategy to bypass these hurdles. Indeed, genetically modified NSCs could bring about more favored therapeutic influences post-transplantation in vivo, making them an excellent option for neurological disease therapy. This review for the first time offers a comprehensive review of the therapeutic capability of genetically modified NSCs rather than naïve NSCs in neurological disease beyond brain tumors and sheds light on the recent progress and prospect in this context.
Collapse
Affiliation(s)
- Ghufran Lutfi Ismaeel
- Department of Pharmacology, College of Pharmacy, University of Al-Ameed, Karbala, Iraq
| | | | - Reem S Alazragi
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Ammar Hussein Ahmed
- Department of Radiology and Sonar, College of Medical Techniques, Al-Farahidi University, Baghdad, Iraq
| | - Asma'a H Mohamed
- Intelligent Medical Systems Department, Al-Mustaqbal University College, Babylon, Iraq
| | - Nisreen Yasir Jasim
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Falah Hassan Shari
- Department of Clinical Laboratory Sciences, College of Pharmacy, University of Basrah, Basrah, Iraq
| | | |
Collapse
|
66
|
Wibawa P, Walterfang M, Malpas CB, Glikmann‐Johnston Y, Poudel G, Razi A, Hannan AJ, Velakoulis D, Georgiou‐Karistianis N. Selective perforant-pathway atrophy in Huntington disease: MRI analysis of hippocampal subfields. Eur J Neurol 2023; 30:2650-2660. [PMID: 37306313 PMCID: PMC10946817 DOI: 10.1111/ene.15918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/17/2023] [Accepted: 06/08/2023] [Indexed: 06/13/2023]
Abstract
INTRODUCTION While individuals with Huntington disease (HD) show memory impairment that indicates hippocampal dysfunction, the available literature does not consistently identify structural evidence for involvement of the whole hippocampus but rather suggests that hippocampal atrophy may be confined to certain hippocampal subregions. METHODS We processed T1-weighted MRI from IMAGE-HD study using FreeSurfer 7.0 and compared the volumes of the hippocampal subfields among 36 early motor symptomatic (symp-HD), 40 pre-symptomatic (pre-HD), and 36 healthy control individuals across three timepoints over 36 months. RESULTS Mixed-model analyses revealed significantly lower subfield volumes in symp-HD, compared with pre-HD and control groups, in the subicular regions of the perforant-pathway: presubiculum, subiculum, dentate gyrus, tail, and right molecular layer. These adjoining subfields aggregated into a single principal component, which demonstrated an accelerated rate of atrophy in the symp-HD. Volumes between pre-HD and controls did not show any significant difference. In the combined HD groups, CAG repeat length and disease burden score were associated with presubiculum, molecular layer, tail, and perforant-pathway subfield volumes. Hippocampal left tail and perforant-pathway subfields were associated with motor onset in the pre-HD group. CONCLUSIONS Hippocampal subfields atrophy in early symptomatic HD affects key regions of the perforant-pathway, which may implicate the distinctive memory impairment at this stage of illness. Their volumetric associations with genetic and clinical markers suggest the selective susceptibility of these subfields to mutant Huntingtin and disease progression.
Collapse
Affiliation(s)
- Pierre Wibawa
- NeuropsychiatryRoyal Melbourne HospitalParkvilleVictoriaAustralia
- Melbourne Neuropsychiatry CenterUniversity of MelbourneParkvilleVictoriaAustralia
- School of Psychological Sciences and Turner Institute for Brain and Mental HealthMonash UniversityClaytonVictoriaAustralia
| | - Mark Walterfang
- NeuropsychiatryRoyal Melbourne HospitalParkvilleVictoriaAustralia
- Melbourne Neuropsychiatry CenterUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Charles B. Malpas
- NeuropsychiatryRoyal Melbourne HospitalParkvilleVictoriaAustralia
- Melbourne Neuropsychiatry CenterUniversity of MelbourneParkvilleVictoriaAustralia
| | - Yifat Glikmann‐Johnston
- School of Psychological Sciences and Turner Institute for Brain and Mental HealthMonash UniversityClaytonVictoriaAustralia
| | - Govinda Poudel
- Mary Mackillop Institute for Health ResearchAustralian Catholic UniversityFitzroyVictoriaAustralia
| | - Adeel Razi
- School of Psychological Sciences and Turner Institute for Brain and Mental HealthMonash UniversityClaytonVictoriaAustralia
| | - Anthony J. Hannan
- Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Dennis Velakoulis
- NeuropsychiatryRoyal Melbourne HospitalParkvilleVictoriaAustralia
- Melbourne Neuropsychiatry CenterUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Nellie Georgiou‐Karistianis
- School of Psychological Sciences and Turner Institute for Brain and Mental HealthMonash UniversityClaytonVictoriaAustralia
| |
Collapse
|
67
|
Davis MC, Hill AT, Fitzgerald PB, Bailey NW, Stout JC, Hoy KE. Neurophysiological correlates of non-motor symptoms in late premanifest and early-stage manifest huntington's disease. Clin Neurophysiol 2023; 153:166-176. [PMID: 37506604 DOI: 10.1016/j.clinph.2023.06.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 05/22/2023] [Accepted: 06/18/2023] [Indexed: 07/30/2023]
Abstract
OBJECTIVE To find sensitive neurophysiological correlates of non-motor symptoms in Huntington's disease (HD), which are essential for the development and assessment of novel treatments. METHODS We used resting state EEG to examine differences in oscillatory activity (analysing the isolated periodic as well as the complete EEG signal) and functional connectivity in 22 late premanifest and early stage people with HD and 20 neurotypical controls. We then assessed the correlations between these neurophysiological markers and clinical measures of apathy and processing speed. RESULTS Significantly lower theta and greater delta resting state power was seen in the HD group, as well as significantly greater delta connectivity. There was a significant positive correlation between theta power and processing speed, however there were no associations between the neurophysiological and apathy measures. CONCLUSIONS We speculate that these changes in oscillatory power and connectivity reflect ongoing, frontally concentrated degenerative and compensatory processes associated with HD. SIGNIFICANCE Our findings support the potential utility of quantitative EEG as a proximate marker of processing speed, but not apathy in HD.
Collapse
Affiliation(s)
- Marie-Claire Davis
- Central Clinical School, Department of Psychiatry, Monash University, Victoria, Australia; Statewide Progressive Neurological Disease Service, Calvary Health Care Bethlehem, Victoria, Australia.
| | - Aron T Hill
- Central Clinical School, Department of Psychiatry, Monash University, Victoria, Australia; Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Victoria, Australia.
| | - Paul B Fitzgerald
- Central Clinical School, Department of Psychiatry, Monash University, Victoria, Australia; School of Medicine and Psychology, Australian National University, Canberra, ACT, Australia.
| | - Neil W Bailey
- Central Clinical School, Department of Psychiatry, Monash University, Victoria, Australia; School of Medicine and Psychology, Australian National University, Canberra, ACT, Australia; Monarch Research Institute Monarch Mental Health Group, 225 Clarence Street, Sydney, NSW 2000, Australia.
| | - Julie C Stout
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, 18 Innovation Walk, Clayton Campus, Wellington Road, Clayton, VIC 3800, Australia.
| | - Kate E Hoy
- Central Clinical School, Department of Psychiatry, Monash University, Victoria, Australia; The Bionics Institute of Australia, 384-388 Albert St, East Melbourne, VIC 3002, Australia.
| |
Collapse
|
68
|
Le Stanc L, Youssov K, Giavazzi M, Sliwinski A, Bachoud-Lévi AC, Jacquemot C. Language disorders in patients with striatal lesions: Deciphering the role of the striatum in language performance. Cortex 2023; 166:91-106. [PMID: 37354871 DOI: 10.1016/j.cortex.2023.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/22/2023] [Accepted: 04/13/2023] [Indexed: 06/26/2023]
Abstract
The classical neural model of language refers to a cortical network involving frontal, parietal and temporal regions. However, patients with subcortical lesions of the striatum have language difficulties. We investigated whether the striatum is directly involved in language or whether its role in decision-making has an indirect effect on language performance, by testing carriers of Huntington's disease (HD) mutations and controls. HD is a genetic neurodegenerative disease primarily affecting the striatum and causing language disorders. We asked carriers of the HD mutation in the premanifest (before clinical diagnosis) and early disease stages, and controls to perform two discrimination tasks, one involving linguistic and the other non-linguistic stimuli. We used the hierarchical drift diffusion model (HDDM) to analyze the participants' responses and to assess the decision and non-decision parameters separately. We hypothesized that any language deficits related to decision-making impairments would be reflected in the decision parameters of linguistic and non-linguistic tasks. We also assessed the relative contributions of both HDDM decision and non-decision parameters to the participants' behavioral data (response time and discriminability). Finally, we investigated whether the decision and non-decision parameters of the HDDM were correlated with brain atrophy. The HDDM analysis showed that patients with early HD have impaired decision parameters relative to controls, regardless of the task. In both tasks, decision parameters better explained the variance of response time and discriminability performance than non-decision parameters. In the linguistic task, decision parameters were positively correlated with gray matter volume in the ventral striatum and putamen, whereas non-decision parameters were not. Language impairment in patients with striatal atrophy is better explained by a deficit of decision-making than by a deficit of core linguistic processing. These results suggest that the striatum is involved in language through the modulation of decision-making, presumably by regulating the process of choice between linguistic alternatives.
Collapse
Affiliation(s)
- Lorna Le Stanc
- Département d'Études Cognitives, École Normale Supérieure-PSL, Paris, France; Institut Mondor de Recherche Biomédicale, Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France; Université Paris-Est Créteil, Faculté de Médecine, Créteil, France; Université Paris Cité, LaPsyDÉ, CNRS, Paris, France
| | - Katia Youssov
- Département d'Études Cognitives, École Normale Supérieure-PSL, Paris, France; Institut Mondor de Recherche Biomédicale, Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France; Université Paris-Est Créteil, Faculté de Médecine, Créteil, France; AP-HP, Centre de Référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, Créteil, France
| | - Maria Giavazzi
- Département d'Études Cognitives, École Normale Supérieure-PSL, Paris, France; Institut Mondor de Recherche Biomédicale, Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France; Université Paris-Est Créteil, Faculté de Médecine, Créteil, France
| | - Agnès Sliwinski
- Département d'Études Cognitives, École Normale Supérieure-PSL, Paris, France; Institut Mondor de Recherche Biomédicale, Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France; Université Paris-Est Créteil, Faculté de Médecine, Créteil, France; AP-HP, Centre de Référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, Créteil, France
| | - Anne-Catherine Bachoud-Lévi
- Département d'Études Cognitives, École Normale Supérieure-PSL, Paris, France; Institut Mondor de Recherche Biomédicale, Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France; Université Paris-Est Créteil, Faculté de Médecine, Créteil, France; AP-HP, Centre de Référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, Créteil, France
| | - Charlotte Jacquemot
- Département d'Études Cognitives, École Normale Supérieure-PSL, Paris, France; Institut Mondor de Recherche Biomédicale, Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France; Université Paris-Est Créteil, Faculté de Médecine, Créteil, France.
| |
Collapse
|
69
|
Jiang A, Handley RR, Lehnert K, Snell RG. From Pathogenesis to Therapeutics: A Review of 150 Years of Huntington's Disease Research. Int J Mol Sci 2023; 24:13021. [PMID: 37629202 PMCID: PMC10455900 DOI: 10.3390/ijms241613021] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
Huntington's disease (HD) is a debilitating neurodegenerative genetic disorder caused by an expanded polyglutamine-coding (CAG) trinucleotide repeat in the huntingtin (HTT) gene. HD behaves as a highly penetrant dominant disorder likely acting through a toxic gain of function by the mutant huntingtin protein. Widespread cellular degeneration of the medium spiny neurons of the caudate nucleus and putamen are responsible for the onset of symptomology that encompasses motor, cognitive, and behavioural abnormalities. Over the past 150 years of HD research since George Huntington published his description, a plethora of pathogenic mechanisms have been proposed with key themes including excitotoxicity, dopaminergic imbalance, mitochondrial dysfunction, metabolic defects, disruption of proteostasis, transcriptional dysregulation, and neuroinflammation. Despite the identification and characterisation of the causative gene and mutation and significant advances in our understanding of the cellular pathology in recent years, a disease-modifying intervention has not yet been clinically approved. This review includes an overview of Huntington's disease, from its genetic aetiology to clinical presentation and its pathogenic manifestation. An updated view of molecular mechanisms and the latest therapeutic developments will also be discussed.
Collapse
Affiliation(s)
- Andrew Jiang
- Applied Translational Genetics Group, Centre for Brain Research, School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand; (R.R.H.); (K.L.); (R.G.S.)
| | | | | | | |
Collapse
|
70
|
Eide S, Misztal M, Feng ZP. Interleukin-6 as a marker of Huntington's disease progression: Systematic review and meta-analysis. Brain Behav Immun Health 2023; 30:100635. [PMID: 37215308 PMCID: PMC10196779 DOI: 10.1016/j.bbih.2023.100635] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/20/2023] [Accepted: 04/30/2023] [Indexed: 05/24/2023] Open
Abstract
Huntington's disease (HD) is a rare, inherited disorder with a broad spectrum of manifestations that vary with disease severity and progression. Although genetic testing can readily confirm the initial diagnosis of HD, markers sensitive to HD progression are needed to aid the development of individual treatment plans. The current analysis aims to identify plasma Interleukin-6 (IL-6) as a marker of disease progression in HD patients. A systematic search of PubMed and Medline from conception through October 2021 was conducted. Studies reporting plasma IL-6 levels of mutation-positive HD patients and healthy controls that met inclusion criteria were selected. The search strategy collected 303 studies, 9 of which met analysis inclusion criteria. From included studies, plasma IL-6 levels of 469 individuals with the HD mutation and 206 healthy controls were collected. Plasma IL-6 levels were meta-analytically compared between healthy controls and individuals with the confirmed HD mutation at all stages of disease and correlated to performance on standardized measures of total cognitive and motor function. Plasma IL-6 was significantly increased in HD groups compared to controls (g = 0.73, 95% CI = 0.31,1.16, P < 0.01) and increased significantly throughout most stages of disease progression, notably between pre-manifest and manifest (g = 0.31, 95% CI = 0.04,0.59, P < 0.05) and early and moderate HD stages (g = 0.52, 95% CI = 0.18,0.86, P < 0.01). Significant correlations between plasma IL-6 levels and HD symptomatic progression were identified, with increased cytokine levels associated with more severe motor impairments (r = 0.179, 95% CI = 0.0479,0.304, P = 0.008) and more extreme disabilities in activities of daily living and/or work tasks (r = -0.229, 95% CI = -0.334, -0.119, P < 0.001). Conclusively, plasma IL-6 levels correlate with disease and motor symptom progression and may act as a viable marker for clinical use. Analysis is limited by small study numbers and highlights the need for future work to identify definitive ranges or rates of change of plasma IL-6 levels that correlate to progressive HD disease states.
Collapse
Affiliation(s)
| | | | - Zhong-Ping Feng
- Department of Physiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| |
Collapse
|
71
|
Shiino S, van Wouwe NC, Wylie SA, Claassen DO, McDonell KE. Huntington disease exacerbates action impulses. Front Psychol 2023; 14:1186465. [PMID: 37397312 PMCID: PMC10312388 DOI: 10.3389/fpsyg.2023.1186465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/16/2023] [Indexed: 07/04/2023] Open
Abstract
Background Impulsivity is a common clinical feature of Huntington disease (HD), but the underlying cognitive dynamics of impulse control in this population have not been well-studied. Objective To investigate the temporal dynamics of action impulse control in HD patients using an inhibitory action control task. Methods Sixteen motor manifest HD patients and seventeen age-matched healthy controls (HC) completed the action control task. We applied the activation-suppression theoretical model and distributional analytic techniques to differentiate the strength of fast impulses from their top-down suppression. Results Overall, HD patients produced slower and less accurate reactions than HCs. HD patients also exhibited an exacerbated interference effect, as evidenced by a greater slowing of RT on non-corresponding compared to corresponding trials. HD patients made more fast, impulsive errors than HC, evidenced by significantly lower accuracy on their fastest reaction time trials. The slope reduction of interference effects as reactions slowed was similar between HD and controls, indicating preserved impulse suppression. Conclusion Our results indicate that patients with HD show a greater susceptibility to act rapidly on incorrect motor impulses but preserved proficiency of top-down suppression. Further research is needed to determine how these findings relate to clinical behavioral symptoms.
Collapse
Affiliation(s)
- Shuhei Shiino
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, United States
| | | | - Scott A. Wylie
- Department of Neurological Surgery, University of Louisville, Louisville, KY, United States
| | - Daniel O. Claassen
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Katherine E. McDonell
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
72
|
Abstract
Neurodegenerative diseases are characterized by the progressive loss of structure or function of neurons. In this Spotlight, we explore the idea that genetic forms of neurodegenerative disorders might be rooted in neural development. Focusing on Alzheimer's, Parkinson's and Huntington's disease, we first provide a brief overview of the pathology for these diseases. Although neurodegenerative diseases are generally thought of as late-onset diseases, we discuss recent evidence promoting the notion that they might be considered neurodevelopmental disorders. With this view in mind, we consider the suitability of animal models for studying these diseases, highlighting human-specific features of human brain development. We conclude by proposing that one such feature, human-specific regulation of neurogenic time, might be key to understanding the etiology and pathophysiology of human neurodegenerative disease.
Collapse
Affiliation(s)
- Khadijeh Shabani
- Institut du Cerveau – Paris Brain Institute – ICM, Sorbonne Université, Inserm, CNRS, Hôpital Pitié-Salpêtrière, 75013 Paris, France
| | - Bassem A. Hassan
- Institut du Cerveau – Paris Brain Institute – ICM, Sorbonne Université, Inserm, CNRS, Hôpital Pitié-Salpêtrière, 75013 Paris, France
| |
Collapse
|
73
|
Li XY, Bao YF, Xie JJ, Gao B, Qian SX, Dong Y, Wu ZY. Application Value of Serum Neurofilament Light Protein for Disease Staging in Huntington's Disease. Mov Disord 2023. [PMID: 37148558 DOI: 10.1002/mds.29430] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/06/2023] [Accepted: 04/18/2023] [Indexed: 05/08/2023] Open
Abstract
BACKGROUND Neurofilament light protein (NfL) has been proven to be a sensitive biomarker for Huntington's disease (HD). However, these studies did not include HD patients at advanced stages or with larger CAG repeats (>50), leading to a knowledge gap of the characteristics of NfL. METHODS Serum NfL (sNfL) levels were quantified using an ultrasensitive immunoassay. Participants were assessed by clinical scales and 7.0 T magnetic resonance imaging. Longitudinal samples and clinical data were obtained. RESULTS Baseline samples were available from 110 controls, 90 premanifest HD (pre-HD) and 137 HD individuals. We found levels of sNfL significantly increased in HD compared to pre-HD and controls (both P < 0.0001). The increase rates of sNfL were differed by CAG repeat lengths. However, there was no difference in sNfL levels in manifest HD from early to late stages. In addition, sNfL levels were associated with cognitive measures in pre-HD and manifest HD group, respectively. The increased levels of sNfL were also closely related to microstructural changes in white matter. In the longitudinal analysis, baseline sNfL did not correlate with subsequent clinical function decline. Random forest analysis revealed that sNfL had good power for predicting disease onset. CONCLUSIONS Although sNfL levels are independent of disease stages in manifest HD, it is still an optimal indicator for predicting disease onset and has potential use as a surrogate biomarker of treatment effect in clinical trials. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Xiao-Yan Li
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu-Feng Bao
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Juan-Juan Xie
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Bin Gao
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Shu-Xia Qian
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Dong
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhi-Ying Wu
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai, China
| |
Collapse
|
74
|
Bräuer S, Falkenburger B. [Gene Therapy for Huntington Disease]. FORTSCHRITTE DER NEUROLOGIE-PSYCHIATRIE 2023; 91:141-146. [PMID: 37040787 PMCID: PMC10089766 DOI: 10.1055/a-2042-2338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Englisch: Being one of the most common genetic neurodegenerative disease, Huntington's disease has been a model disease - also for gene therapy. Among the various options, the development of antisense oligonucleotides is the most advanced. Further options at the RNA level include micro-RNAs and modulators of RNA processing (splicing), at the DNA level zinc finger proteins. Several products are in clinical trials. These differ in their mode of application and in the extent of systemic availability. Another important difference between therapeutic strategies could be whether all forms of the huntingtin protein are targeted in the same extent, or whether a therapy preferentially targets particular toxic forms such as the exon1 protein. The results of the recently terminated GENERATION HD1 trial were somewhat sobering, most likely due to the side effect-related hydrocephalus. Therefore they represent just one step towards the development of an effective gene therapy against Huntington's disease.
Collapse
Affiliation(s)
- Stefan Bräuer
- Klinik und Poliklinik für Neurologie, Universitätsklinikum an der TU Dresden, Dresden, Germany
| | - Björn Falkenburger
- Klinik und Poliklinik für Neurologie, Universitätsklinikum an der TU Dresden, Dresden, Germany
| |
Collapse
|
75
|
Abstract
Huntington's disease (HD) is a fatal, monogenic, autosomal dominant neurodegenerative disease caused by a polyglutamine-encoding CAG expansion in the huntingtin (HTT) gene that results in mutant huntingtin proteins (mHTT) in cells throughout the body. Although large parts of the central nervous system (CNS) are affected, the striatum is especially vulnerable and undergoes marked atrophy. Astrocytes are abundant within the striatum and contain mHTT in HD, as well as in mouse models of the disease. We focus on striatal astrocytes and summarize how they participate in, and contribute to, molecular pathophysiology and disease-related phenotypes in HD model mice. Where possible, reference is made to pertinent astrocyte alterations in human HD. Astrocytic dysfunctions related to cellular morphology, extracellular ion and neurotransmitter homeostasis, and metabolic support all accompany the development and progression of HD, in both transgenic mouse and human cellular and chimeric models of HD. These findings reveal the potential for the therapeutic targeting of astrocytes so as to restore synaptic as well as tissue homeostasis in HD. Elucidation of the mechanisms by which astrocytes contribute to HD pathogenesis may inform a broader understanding of the role of glial pathology in neurodegenerative disorders and, by so doing, enable new strategies of glial-directed therapeutics.
Collapse
Affiliation(s)
- Baljit S. Khakh
- Department of Physiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Steven A. Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
| |
Collapse
|
76
|
El Haj M, Caillaud M, Moustafa A, Prundean A, Scherer C, Verny C, Allain P. "Ten euros now" temporal discounting in Huntington disease. Neurol Sci 2023:10.1007/s10072-023-06775-z. [PMID: 36964316 DOI: 10.1007/s10072-023-06775-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/21/2023] [Indexed: 03/26/2023]
Abstract
BACKGROUND When making decisions, one often faces a trade-off between immediate and long-term rewards. In these situations, people may prefer immediate over later rewards, even if immediate rewards are smaller than later ones; a phenomenon known as temporal discounting. In this study, we, for the first time, assessed temporal discounting in three populations: participants with manifest Huntington disease (HD), participants with premanifest HD, and control participants. METHODS Using the temporal discounting task, we invited participants to choose between small immediate amount of money vs. delayed, but larger amount of money (e.g., "Which do you prefer: you get 10 euros right now or 50 euros in a month?"). We also measured inhibition in order to test if it impacts discounting performance. RESULTS Analysis demonstrated higher temporal discounting (i.e., a preference for the immediate rewards) in participants with manifest HD compared to those with premanifest HD or control participants, but no significant differences were observed in participants with premanifest HD and control participants. Analysis also demonstrated significant correlations between temporal discounting and scores on an inhibition test in participants with manifest HD, but not in those with premanifest HD or in control participants. DISCUSSION We suggest that, when making decisions, patients with manifest HD may have difficulties with suppressing the temptation of smaller, but immediate, rewards.
Collapse
Affiliation(s)
- Mohamad El Haj
- Nantes Université, Univ Angers, Laboratoire de Psychologie des Pays de la Loire (LPPL - EA 4638), F-44000, Nantes, France.
- CHU Nantes, Clinical Gerontology Department, 41 rue Pierre et Marie Curie, 44093, Nantes, France.
- Institut Universitaire de France, Paris, France.
- Faculté de Psychologie, LPPL - Laboratoire de Psychologie des Pays de la Loire, Université Nantes, Chemin de la Censive du Tertre, BP 81227, 44312, Nantes Cedex 3, France.
| | - Marie Caillaud
- University of Texas, Clinical Neuroscience Lab, 108 East Dean Keeton St., Austin, TX, 78712, USA
| | - Ahmed Moustafa
- School of Psychology, Faculty of Society and Design, Bond University, Gold Coast, Queensland, Australia
- Department of Human Anatomy and Physiology, the Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | | | | | | | - Philippe Allain
- Département de Neurologie, CHU Angers, Angers, France
- Laboratoire de Psychologie des Pays de la Loire, LPPL EA 4638 SFR Confluences, UNIV Angers, Nantes Université, Maison de la recherche Germaine Tillion, 5 bis Boulevard Lavoisier, 49045, Angers Cedex 01, France
| |
Collapse
|
77
|
Cincotta MC, Walker RH. Diagnostic Uncertainties: Chorea. Semin Neurol 2023; 43:65-80. [PMID: 36882120 DOI: 10.1055/s-0043-1763506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Chorea is a hyperkinetic movement disorder with a multitude of potential etiologies, both acquired and inherited. Although the differential diagnosis for new-onset chorea is extensive, there are often clues in the history, exam, and basic testing that can help to narrow the options. Evaluation for treatable or reversible causes should take priority, as rapid diagnosis can lead to more favorable outcomes. While Huntington's disease is most common genetic cause of chorea, multiple phenocopies also exist and should be considered if Huntington gene testing is negative. The decision of what additional genetic testing to pursue should be based on both clinical and epidemiological factors. The following review provides an overview of the many possible etiologies as well as a practical approach for a patient presenting with new-onset chorea.
Collapse
Affiliation(s)
- Molly C Cincotta
- Department of Neurology, Temple University, Philadelphia, Pennsylvania
| | - Ruth H Walker
- Department of Neurology, James J. Peters Veterans Affairs Medical Center and Mount Sinai School of Medicine, Bronx, New York
| |
Collapse
|
78
|
Horta-Barba A, Martinez-Horta S, Sampedro F, Pérez-Pérez J, Pagonabarraga J, Kulisevsky J. Structural and metabolic brain correlates of arithmetic word-problem solving in Huntington's disease. J Neurosci Res 2023; 101:990-999. [PMID: 36807154 DOI: 10.1002/jnr.25174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 01/03/2023] [Accepted: 01/20/2023] [Indexed: 02/22/2023]
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.
Collapse
Affiliation(s)
- Andrea Horta-Barba
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Medicine, Autonomous University of Barcelona (UAB), Bellaterra, Spain.,European Huntington's Disease Network (EHDN), Bellaterra, Spain
| | - Saul Martinez-Horta
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Medicine, Autonomous University of Barcelona (UAB), Bellaterra, Spain.,European Huntington's Disease Network (EHDN), Bellaterra, Spain
| | - Frederic Sampedro
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jesús Pérez-Pérez
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Medicine, Autonomous University of Barcelona (UAB), Bellaterra, Spain.,European Huntington's Disease Network (EHDN), Bellaterra, Spain
| | - Javier Pagonabarraga
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Medicine, Autonomous University of Barcelona (UAB), Bellaterra, Spain.,European Huntington's Disease Network (EHDN), Bellaterra, Spain
| | - Jaime Kulisevsky
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Medicine, Autonomous University of Barcelona (UAB), Bellaterra, Spain.,European Huntington's Disease Network (EHDN), Bellaterra, Spain
| |
Collapse
|
79
|
Kouba T, Frank W, Tykalova T, Mühlbäck A, Klempíř J, Lindenberg KS, Landwehrmeyer GB, Rusz J. Speech biomarkers in Huntington's disease: A cross-sectional study in pre-symptomatic, prodromal and early manifest stages. Eur J Neurol 2023; 30:1262-1271. [PMID: 36732902 DOI: 10.1111/ene.15726] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/25/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND PURPOSE Motor speech alterations are a prominent feature of clinically manifest Huntington's disease (HD). Objective acoustic analysis of speech can quantify speech alterations. It is currently unknown, however, at what stage of HD speech alterations can be reliably detected. We aimed to explore the patterns and extent of speech alterations using objective acoustic analysis in HD and to assess correlations with both rater-assessed phenotypical features and biological determinants of HD. METHODS Speech samples were acquired from 44 premanifest (29 pre-symptomatic and 15 prodromal) and 25 manifest HD gene expansion carriers, and 25 matched healthy controls. A quantitative automated acoustic analysis of 10 speech dimensions was performed. RESULTS Automated speech analysis allowed us to differentiate between participants with HD and controls, with areas under the curve of 0.74 for pre-symptomatic, 0.92 for prodromal, and 0.97 for manifest stages. In addition to irregular alternating motion rates and prolonged pauses seen only in manifest HD, both prodromal and manifest HD displayed slowed articulation rate, slowed alternating motion rates, increased loudness variability, and unstable steady-state position of articulators. In participants with premanifest HD, speech alteration severity was associated with cognitive slowing (r = -0.52, p < 0.001) and the extent of bradykinesia (r = 0.43, p = 0.004). Speech alterations correlated with a measure of exposure to mutant gene products (CAG-age-product score; r = 0.60, p < 0.001). CONCLUSION Speech abnormalities in HD are associated with other motor and cognitive deficits and are measurable already in premanifest stages of HD. Therefore, automated speech analysis might represent a quantitative HD biomarker with potential for assessing disease progression.
Collapse
Affiliation(s)
- Tomas Kouba
- Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Wiebke Frank
- Department of Neurology, University Ulm, Ulm, Germany
| | - Tereza Tykalova
- Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Alzbeta Mühlbäck
- Department of Neurology, University Ulm, Ulm, Germany.,Department of Neuropsychiatry, Huntington Center South, kbo-Isar-Amper-Klinikum Taufkirchen (Vils), Taufkirchen, Germany.,Department of Neurology and Center of Clinical Neuroscience, 1st Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Jiří Klempíř
- Department of Neurology and Center of Clinical Neuroscience, 1st Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | | | | | - Jan Rusz
- Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic.,Department of Neurology and Center of Clinical Neuroscience, 1st Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic.,Department of Neurology & ARTORG Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
80
|
Gangwani MR, Soto JS, Jami-Alahmadi Y, Tiwari S, Kawaguchi R, Wohlschlegel JA, Khakh BS. Neuronal and astrocytic contributions to Huntington's disease dissected with zinc finger protein transcriptional repressors. Cell Rep 2023; 42:111953. [PMID: 36640336 PMCID: PMC9898160 DOI: 10.1016/j.celrep.2022.111953] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 12/15/2022] [Accepted: 12/15/2022] [Indexed: 01/09/2023] Open
Abstract
Huntington's disease (HD) is caused by expanded CAG repeats in the huntingtin gene (HTT) resulting in expression of mutant HTT proteins (mHTT) with extended polyglutamine tracts, including in striatal neurons and astrocytes. It is unknown whether pathophysiology in vivo can be attenuated by lowering mHTT in either cell type throughout the brain, and the relative contributions of neurons and astrocytes to HD remain undefined. We use zinc finger protein (ZFP) transcriptional repressors to cell-selectively lower mHTT in vivo. Astrocytes display loss of essential functions such as cholesterol metabolism that are partly driven by greater neuronal dysfunctions, which encompass neuromodulation, synaptic, and intracellular signaling pathways. Using transcriptomics, proteomics, electrophysiology, and behavior, we dissect neuronal and astrocytic contributions to HD pathophysiology. Remarkably, brain-wide delivery of neuronal ZFPs results in strong mHTT lowering, rescue of HD-associated behavioral and molecular phenotypes, and significant extension of lifespan, findings that support translational development.
Collapse
Affiliation(s)
- Mohitkumar R. Gangwani
- Department of Physiology, University of California Los Angeles. Los Angeles, CA 90095-1751, USA
| | - Joselyn S. Soto
- Department of Physiology, University of California Los Angeles. Los Angeles, CA 90095-1751, USA
| | - Yasaman Jami-Alahmadi
- Department of Biological Chemistry, University of California Los Angeles. Los Angeles, CA 90095-1751, USA
| | - Srushti Tiwari
- Department of Physiology, University of California Los Angeles. Los Angeles, CA 90095-1751, USA
| | - Riki Kawaguchi
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles. Los Angeles, CA 90095-1751, USA
| | - James A. Wohlschlegel
- Department of Biological Chemistry, University of California Los Angeles. Los Angeles, CA 90095-1751, USA
| | - Baljit S. Khakh
- Department of Physiology, University of California Los Angeles. Los Angeles, CA 90095-1751, USA,Department of Neurobiology, University of California Los Angeles. Los Angeles, CA 90095-1751, USA
| |
Collapse
|
81
|
Cunningham CN, Jenkins LC, Chang WJ, McAuley JH, Schabrun SM. Relative and absolute reliability of somatosensory evoked potentials in response to non-noxious electrical stimulation of the paraspinal muscles in healthy participants at an interval of 3-months. Int J Neurosci 2023; 133:103-109. [PMID: 33663320 DOI: 10.1080/00207454.2021.1893722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Somatosensory evoked potentials (SEPs) are used extensively to quantify cortical activity in response to noxious and/or non-noxious sensory stimuli. However, data demonstrating the reliability of SEP measures in response to non-noxious stimulation over time are scarce. AIM We investigated the relative and absolute reliability, and the smallest detectable change at 95% confidence (SDC95) for SEPs evoked by non-noxious electrical stimulation of the paraspinal muscles in thirty-nine healthy participants at a 3-month interval. METHODS SEPs were evoked at an intensity three-times that of each participant's perceptual threshold and recorded from a single electrode placed over the primary somatosensory cortex (S1). RESULTS Our analyses reveal that i) latency, as a measure of activity onset, has poor relative reliability but good absolute reliability; ii) area, as a measure of cortical activity, has good relative and absolute reliability (except for the N150 component) and iii) perceptual threshold and stimulation intensity was not reliable over time. CONCLUSION These findings suggest that the area of the N80 and P260 SEP components, and the area of the N80-N150-P260 SEP complex, can be utilised in future studies as reliable markers of cortical activity.
Collapse
Affiliation(s)
- Chelsea N Cunningham
- Centre for Pain IMPACT, Neuroscience Research Australia, Randwick, NSW, Australia
| | - Luke C Jenkins
- Centre for Pain IMPACT, Neuroscience Research Australia, Randwick, NSW, Australia.,School of Science and Health, Western Sydney University, Penrith, NSW, Australia
| | - Wei-Ju Chang
- Centre for Pain IMPACT, Neuroscience Research Australia, Randwick, NSW, Australia
| | - James H McAuley
- Centre for Pain IMPACT, Neuroscience Research Australia, Randwick, NSW, Australia
| | - Siobhan M Schabrun
- Centre for Pain IMPACT, Neuroscience Research Australia, Randwick, NSW, Australia
| |
Collapse
|
82
|
Fitzgerald ES, Stout JC, Glikmann-Johnston Y, Anderson C, Jackson ML. Sleep, Circadian Rhythms, and Cognitive Dysfunction in Huntington's Disease. J Huntingtons Dis 2023; 12:293-304. [PMID: 37599535 DOI: 10.3233/jhd-230578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
BACKGROUND In healthy people, sleep and circadian disruption are linked to cognitive deficits. People with Huntington's disease (HD), who have compromised brain function and sleep and circadian disturbances, may be even more susceptible to these cognitive effects. OBJECTIVE To conduct a comprehensive review and synthesis of the literature in HD on the associations of cognitive dysfunction with disturbed sleep and circadian rhythms. METHODS We searched MEDLINE via OVID, CINAHL Plus, EMBASE via OVID, and PubMed in May 2023. The first author then screened by title and abstract and conducted a full review of remaining articles. RESULTS Eight studies investigating the influence of sleep and/or circadian rhythms on cognitive function in HD were found. In manifest HD, poorer sleep was associated with worse cognitive function. For behavioral 24-hour (circadian) rhythms, two studies indicated that later wake times correlated with poorer cognitive function. No reported studies in HD examined altered physiological 24-hour (circadian) rhythms and cognitive impairment. CONCLUSION Some associations exist between poor sleep and cognitive dysfunction in manifest HD, yet whether these associations are present before clinical diagnosis is unknown. Whether circadian disturbances relate to cognitive impairment in HD also remains undetermined. To inform sleep and circadian interventions aimed at improving cognitive symptoms in HD, future research should include a range of disease stages, control for external factors, and utilize robust cognitive batteries targeted to the aspects of cognitive function known to be adversely affected in HD.
Collapse
Affiliation(s)
- Emily S Fitzgerald
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Julie C Stout
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Yifat Glikmann-Johnston
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Clare Anderson
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Melinda L Jackson
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| |
Collapse
|
83
|
Cvetanovic M, Gray M. Contribution of Glial Cells to Polyglutamine Diseases: Observations from Patients and Mouse Models. Neurotherapeutics 2023; 20:48-66. [PMID: 37020152 PMCID: PMC10119372 DOI: 10.1007/s13311-023-01357-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2023] [Indexed: 04/07/2023] Open
Abstract
Neurodegenerative diseases are broadly characterized neuropathologically by the degeneration of vulnerable neuronal cell types in a specific brain region. The degeneration of specific cell types has informed on the various phenotypes/clinical presentations in someone suffering from these diseases. Prominent neurodegeneration of specific neurons is seen in polyglutamine expansion diseases including Huntington's disease (HD) and spinocerebellar ataxias (SCA). The clinical manifestations observed in these diseases could be as varied as the abnormalities in motor function observed in those who have Huntington's disease (HD) as demonstrated by a chorea with substantial degeneration of striatal medium spiny neurons (MSNs) or those with various forms of spinocerebellar ataxia (SCA) with an ataxic motor presentation primarily due to degeneration of cerebellar Purkinje cells. Due to the very significant nature of the degeneration of MSNs in HD and Purkinje cells in SCAs, much of the research has centered around understanding the cell autonomous mechanisms dysregulated in these neuronal cell types. However, an increasing number of studies have revealed that dysfunction in non-neuronal glial cell types contributes to the pathogenesis of these diseases. Here we explore these non-neuronal glial cell types with a focus on how each may contribute to the pathogenesis of HD and SCA and the tools used to evaluate glial cells in the context of these diseases. Understanding the regulation of supportive and harmful phenotypes of glia in disease could lead to development of novel glia-focused neurotherapeutics.
Collapse
Affiliation(s)
- Marija Cvetanovic
- Department of Neuroscience, Institute for Translational Neuroscience, University of Minnesota, Minneapolis, USA
| | - Michelle Gray
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
84
|
Taghian T, Gallagher J, Batcho E, Pullan C, Kuchel T, Denney T, Perumal R, Moore S, Muirhead R, Herde P, Johns D, Christou C, Taylor A, Passler T, Pulaparthi S, Hall E, Chandra S, O’Neill CA, Gray-Edwards H. Brain Alterations in Aged OVT73 Sheep Model of Huntington's Disease: An MRI Based Approach. J Huntingtons Dis 2022; 11:391-406. [PMID: 36189602 PMCID: PMC9837686 DOI: 10.3233/jhd-220526] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Huntington's disease (HD) is a fatal neurodegenerative autosomal dominant disorder with prevalence of 1 : 20000 that has no effective treatment to date. Translatability of candidate therapeutics could be enhanced by additional testing in large animal models because of similarities in brain anatomy, size, and immunophysiology. These features enable realistic pre-clinical studies of biodistribution, efficacy, and toxicity. OBJECTIVE AND METHODS Here we non-invasively characterized alterations in brain white matter microstructure, neurochemistry, neurological status, and mutant Huntingtin protein (mHTT) levels in cerebrospinal fluid (CSF) of aged OVT73 HD sheep. RESULTS Similar to HD patients, CSF mHTT differentiates HD from normal sheep. Our results are indicative of a decline in neurological status, and alterations in brain white matter diffusion and spectroscopy metric that are more severe in aged female HD sheep. Longitudinal analysis of aged female HD sheep suggests that the decline is detectable over the course of a year. In line with reports of HD human studies, white matter alterations in corpus callosum correlates with a decline in gait of HD sheep. Moreover, alterations in the occipital cortex white matter correlates with a decline in clinical rating score. In addition, the marker of energy metabolism in striatum of aged HD sheep, shows a correlation with decline of clinical rating score and eye coordination. CONCLUSION This data suggests that OVT73 HD sheep can serve as a pre-manifest large animal model of HD providing a platform for pre-clinical testing of HD therapeutics and non-invasive tracking of the efficacy of the therapy.
Collapse
Affiliation(s)
- Toloo Taghian
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA,
Department of Radiology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jillian Gallagher
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Erin Batcho
- Department of Electrical and Computer Engineering, Auburn University, Auburn, AL, USA
| | - Caitlin Pullan
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Tim Kuchel
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Thomas Denney
- Department of Electrical and Computer Engineering, Auburn University, Auburn, AL, USA
| | - Raj Perumal
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Shamika Moore
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Robb Muirhead
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Paul Herde
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Daniel Johns
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Chris Christou
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Amanda Taylor
- Department of Clinical Sciences, Auburn University, Auburn, AL, USA
| | - Thomas Passler
- Department of Electrical and Computer Engineering, Auburn University, Auburn, AL, USA
| | - Sanjana Pulaparthi
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Erin Hall
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Sundeep Chandra
- Sana Biotechnology, South San Francisco, CA, USA,Bio Marin Pharmaceutical Inc., San Rafael, CA, USA
| | | | - Heather Gray-Edwards
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA,
Department of Radiology, University of Massachusetts Medical School, Worcester, MA, USA,Correspondence to: Heather L. Gray-Edwards, DVM, PhD, University of Massachusetts Medical School, Department of Radiology and Horae Gene Therapy Center, 368 Plantation Street, ASC6-2055, Worcester, MA 01605, USA. Tel.: +1 508 856 4051; Fax: +1 508 856 1552; E-mail:
| |
Collapse
|
85
|
Hendel RK, Hellem MNN, Hjermind LE, Nielsen JE, Vogel A. An Exploratory Study Investigating Autonomy in Huntington's Disease Gene Expansion Carriers. J Huntingtons Dis 2022; 11:373-381. [PMID: 35964199 DOI: 10.3233/jhd-220540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Autonomy describes a psychological state of self-regulation of motivation and action, which is a central characteristic of healthy functioning. In neurodegenerative diseases measures of self-perception have been found to be affected by the disease. However, it has never been investigated whether measures of self-perception, like autonomy, is affected in Huntington's disease. OBJECTIVE We investigated whether autonomy is affected in Huntington's disease and if the degree of autonomy is associated with motor function, neuropsychiatric symptoms, cognitive impairments, and apathy. METHODS We included 44 premanifest and motor-manifest Huntington's disease gene expansion carriers and 19 controls. Autonomy was examined using two self-report questionnaires, the Autonomy-Connectedness Scale-30 and the Index of Autonomous Functioning. All participants were examined according to motor function, cognitive impairments, and neuropsychiatric symptoms, including apathy. RESULTS Statistically significant differences were found between motor-manifest Huntington's disease gene expansion carriers and premanifest Huntington's disease gene expansion carriers or controls on two measures of autonomy. Between 25-38% of motor-manifest Huntington's disease gene expansion carriers scored significantly below the normal level on subscales of autonomy as compared to controls. One autonomy subscale was associated with apathy (r = -0.65), but not with other symptoms of Huntington's disease. CONCLUSION This study provides evidence for impaired autonomy in individuals with Huntington's disease and an association between autonomy and apathy. The results underline the importance of maintaining patient autonomy and involvement in care throughout the disease.
Collapse
Affiliation(s)
- Rebecca K Hendel
- Department of Neurology, Danish Dementia Research Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark.,Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | - Marie N N Hellem
- Department of Neurology, Danish Dementia Research Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Lena E Hjermind
- Department of Neurology, Danish Dementia Research Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Jørgen E Nielsen
- Department of Neurology, Danish Dementia Research Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Asmus Vogel
- Department of Neurology, Danish Dementia Research Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark.,Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
86
|
Humbert S, Barnat M. Huntington's disease and brain development. C R Biol 2022; 345:77-90. [PMID: 36847466 DOI: 10.5802/crbiol.93] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
Huntington's disease is a rare inherited neurological disorder that generally manifests in mild-adulthood. The disease is characterized by the dysfunction and the degeneration of specific brain structures leading progressively to psychiatric, cognitive and motor disorders. The disease is caused by a mutation in the gene coding for huntingtin and, although it appears in adulthood, embryos carry the mutated gene from their development in utero. Studies based on mouse models and human stem cells have reported altered developmental mechanisms in disease conditions. However, does the mutation affect development in humans? Focusing on the early stages of brain development in human fetuses carrying the HD mutation, we have identified abnormalities in the development of the neocortex, the structure that ensure higher cerebral functions. Altogether, these studies suggests that developmental defects could contribute to the onset symptoms in adults, changing the perspective on disease and thus the health care of patients.
Collapse
|
87
|
Tan B, Shishegar R, Oldham S, Fornito A, Poudel G, Georgiou-Karistianis N. Investigating longitudinal changes to frontal cortico-striatal tracts in Huntington's disease: the IMAGE-HD study. Brain Imaging Behav 2022; 16:2457-2466. [PMID: 35768755 PMCID: PMC9712302 DOI: 10.1007/s11682-022-00699-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2022] [Indexed: 11/28/2022]
Abstract
The striatum is the principal site of disease pathology in Huntington's disease and contains neural connections to numerous cortical brain regions. Studies examining abnormalities to neural connections find that white matter integrity is compromised in HD; however, further regional, and longitudinal investigation is required. This paper is the first longitudinal investigation into region-based white-matter integrity changes in Huntington's Disease. The aim of this study was to better understand how disease progression impacts white matter tracts connecting the striatum to the prefrontal and motor cortical regions in HD. We used existing neuroimaging data from IMAGE-HD, comprised of 25 pre-symptomatic, 27 symptomatic, and 25 healthy controls at three separate time points (baseline, 18-months, 30-months). Fractional anisotropy, axial diffusivity and radial diffusivity were derived as measures of white matter microstructure. The anatomical regions of interest were identified using the Desikan-Killiany brain atlas. A Group by Time repeated measures ANCOVA was conducted for each tract of interest and for each measure. We found significantly lower fractional anisotropy and significantly higher radial diffusivity in the symptomatic group, compared to both the pre-symptomatic group and controls (the latter two groups did not differ from each other), in the rostral middle frontal and superior frontal tracts; as well as significantly higher axial diffusivity in the rostral middle tracts only. We did not find a Group by Time interaction for any of the white matter integrity measures. These findings demonstrate that whilst the microstructure of white matter tracts, extending from the striatum to these regions of interest, are compromised during the symptomatic stages of Huntington's disease, 36-month follow-up did not show progressive changes in these measures. Additionally, no correlations were found between clinical measures and tractography changes, indicating further investigations into the relationship between tractography changes and clinical symptoms in Huntington's disease are required.
Collapse
Affiliation(s)
- Brendan Tan
- School of Psychological Sciences and The Turner Institute for Brain and Mental Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton Campus, Melbourne, Victoria, 3800, Australia
| | - Rosita Shishegar
- School of Psychological Sciences and The Turner Institute for Brain and Mental Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton Campus, Melbourne, Victoria, 3800, Australia
- The Australian E-Health Research Centre, CSIRO, Melbourne, Australia
- Monash Biomedical Imaging, 770 Blackburn Road, Melbourne, Victoria, 3800, Australia
| | - Stuart Oldham
- Monash Biomedical Imaging, 770 Blackburn Road, Melbourne, Victoria, 3800, Australia
- Developmental Imaging, Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC, 3052, Australia
| | - Alex Fornito
- School of Psychological Sciences and The Turner Institute for Brain and Mental Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton Campus, Melbourne, Victoria, 3800, Australia
- Monash Biomedical Imaging, 770 Blackburn Road, Melbourne, Victoria, 3800, Australia
| | - Govinda Poudel
- School of Psychological Sciences and The Turner Institute for Brain and Mental Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton Campus, Melbourne, Victoria, 3800, Australia
- Sydney Imaging, Brain and Mind Centre, the University of Sydney, Sydney, New South Wales, 2050, Australia
- The Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, 3000, Australia
| | - Nellie Georgiou-Karistianis
- School of Psychological Sciences and The Turner Institute for Brain and Mental Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton Campus, Melbourne, Victoria, 3800, Australia.
| |
Collapse
|
88
|
Castro E, Polosecki P, Pustina D, Wood A, Sampaio C, Cecchi GA. Predictive Modeling of Huntington's Disease Unfolds Thalamic and Caudate Atrophy Dissociation. Mov Disord 2022; 37:2407-2416. [PMID: 36173150 DOI: 10.1002/mds.29219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/16/2022] [Accepted: 07/28/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Atrophy in the striatum is a hallmark of Huntington's disease (HD), including the period before clinical motor diagnosis (before-CMD), but it extends to other subcortical structures. The study of the covariation of these structures could improve the detection of disease-related longitudinal progression before-CMD, provide mechanistic insights of the disease, and potentially be used to obtain accurate prospective estimates of atrophy before-CMD and early after-CMD. METHODS We analyzed data from 337 before-CMD individuals, 236 healthy control subjects, and 95 early after-CMD individuals from three studies, and we used nine subcortical regions volumes in two analyses. First, we discriminated before-CMD from healthy control trajectories by integrating volume changes from these regions. Second, we estimated prospective atrophy before-CMD and early after-CMD by considering the influence of a region's present volume over the future volume of another one. RESULTS Before-CMD progression was robustly detected across studies. Indeed, detection of before-CMD progression improved when multiple structures were integrated, as opposed to analyzing the striatum alone, likely because of the reduced partial correlation between caudate and thalamic volume change before-CMD. Our multivariate atrophy prediction model found a thalamus-caudate association that is consistent with this pattern, which yields an improved caudate atrophy prediction in early after-CMD. CONCLUSIONS This study is the first attempt to validate before-CMD multivariate subcortical change detection across studies and to do multivariate prospective atrophy prediction in HD. These models achieve improved performance by detecting a dissociation between caudate and thalamic atrophy trajectories, and they provide a possible mechanistic understanding of the dynamics of HD. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Eduardo Castro
- Digital Health, IBM T.J. Watson Research Center, Yorktown Heights, New York, USA
| | - Pablo Polosecki
- Digital Health, IBM T.J. Watson Research Center, Yorktown Heights, New York, USA
| | - Dorian Pustina
- CHDI Management/CHDI Foundation, Princeton, New Jersey, USA
| | - Andrew Wood
- CHDI Management/CHDI Foundation, Princeton, New Jersey, USA
| | | | - Guillermo A Cecchi
- Digital Health, IBM T.J. Watson Research Center, Yorktown Heights, New York, USA
| |
Collapse
|
89
|
Abdollah Zadegan S, Coco HM, Reddy KS, Anderson KM, Teixeira AL, Stimming EF. Frequency and Pathophysiology of Apathy in Huntington Disease: A Systematic Review and Meta-Analysis. J Neuropsychiatry Clin Neurosci 2022; 35:121-132. [PMID: 36353818 DOI: 10.1176/appi.neuropsych.20220033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Apathy is a common behavioral symptom of Huntington disease (HD). This systematic review describes current evidence on the pathophysiology, assessment, and frequency of apathy in HD. METHODS This systematic review was conducted in accordance with PRISMA guidelines. Using a comprehensive search strategy, the investigators searched the MEDLINE, Embase, and PsycINFO databases. All studies that evaluated apathy in HD patients with a valid scale and reported apathy frequency or scores were included. Apathy scores were analyzed by mean or standardized mean differences in accordance with Cochrane guidelines. RESULTS A total of 1,085 records were screened and 80 studies were ultimately included. The Problem Behaviors Assessment-Short was the most frequently used apathy assessment tool. Apathy frequency generally ranged from 10%-33% in premanifest HD to 24%-76% in manifest HD. A meta-analysis of 5,311 records of patients with premanifest HD showed significantly higher apathy scores, with a standardized mean difference of 0.41 (CI=0.29-0.52; p<0.001). A comparison of 1,247 patients showed significantly higher apathy scores in manifest than premanifest HD, with a mean difference of 1.87 (CI=1.48-2.26; p<0.001). There was evidence of involvement of various cortical and subcortical brain regions in HD patients with apathy. CONCLUSIONS Apathy was more frequent among individuals with premanifest HD compared with those in a control group and among individuals with manifest HD compared with those with premanifest HD. Considering the complexity and unique pattern of development in neurodegenerative disease, further studies are required to explore the pathophysiology of apathy in HD.
Collapse
Affiliation(s)
- Shayan Abdollah Zadegan
- Department of Neurology (Zadegan, Furr Stimming), Huntington's Disease Society of America Center of Excellence (Zadegan, Anderson, Teixeira, Furr Stimming), McGovern Medical School (Coco, Reddy), Department of Psychiatry and Behavioral Sciences (Anderson, Teixeira), all at the University of Texas Health Science Center at Houston
| | - Hannah M Coco
- Department of Neurology (Zadegan, Furr Stimming), Huntington's Disease Society of America Center of Excellence (Zadegan, Anderson, Teixeira, Furr Stimming), McGovern Medical School (Coco, Reddy), Department of Psychiatry and Behavioral Sciences (Anderson, Teixeira), all at the University of Texas Health Science Center at Houston
| | - Kirthan S Reddy
- Department of Neurology (Zadegan, Furr Stimming), Huntington's Disease Society of America Center of Excellence (Zadegan, Anderson, Teixeira, Furr Stimming), McGovern Medical School (Coco, Reddy), Department of Psychiatry and Behavioral Sciences (Anderson, Teixeira), all at the University of Texas Health Science Center at Houston
| | - Kendra M Anderson
- Department of Neurology (Zadegan, Furr Stimming), Huntington's Disease Society of America Center of Excellence (Zadegan, Anderson, Teixeira, Furr Stimming), McGovern Medical School (Coco, Reddy), Department of Psychiatry and Behavioral Sciences (Anderson, Teixeira), all at the University of Texas Health Science Center at Houston
| | - Antonio L Teixeira
- Department of Neurology (Zadegan, Furr Stimming), Huntington's Disease Society of America Center of Excellence (Zadegan, Anderson, Teixeira, Furr Stimming), McGovern Medical School (Coco, Reddy), Department of Psychiatry and Behavioral Sciences (Anderson, Teixeira), all at the University of Texas Health Science Center at Houston
| | - Erin Furr Stimming
- Department of Neurology (Zadegan, Furr Stimming), Huntington's Disease Society of America Center of Excellence (Zadegan, Anderson, Teixeira, Furr Stimming), McGovern Medical School (Coco, Reddy), Department of Psychiatry and Behavioral Sciences (Anderson, Teixeira), all at the University of Texas Health Science Center at Houston
| |
Collapse
|
90
|
Koval I, Dighiero-Brecht T, Tobin AJ, Tabrizi SJ, Scahill RI, Tezenas du Montcel S, Durrleman S, Durr A. Forecasting individual progression trajectories in Huntington disease enables more powered clinical trials. Sci Rep 2022; 12:18928. [PMID: 36344508 PMCID: PMC9640581 DOI: 10.1038/s41598-022-18848-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 08/22/2022] [Indexed: 11/09/2022] Open
Abstract
Variability in neurodegenerative disease progression poses great challenges for the evaluation of potential treatments. Identifying the persons who will experience significant progression in the short term is key for the implementation of trials with smaller sample sizes. We apply here disease course mapping to forecast biomarker progression for individual carriers of the pathological CAG repeat expansions responsible for Huntington disease. We used data from two longitudinal studies (TRACK-HD and TRACK-ON) to synchronize temporal progression of 15 clinical and imaging biomarkers from 290 participants with Huntington disease. We used then the resulting HD COURSE MAP to forecast clinical endpoints from the baseline data of 11,510 participants from ENROLL-HD, an external validation cohort. We used such forecasts to select participants at risk for progression and compute the power of trials for such an enriched population. HD COURSE MAP forecasts biomarkers 5 years after the baseline measures with a maximum mean absolute error of 10 points for the total motor score and 2.15 for the total functional capacity. This allowed reducing sample sizes in trial up to 50% including participants with a higher risk for progression ensuring a more homogeneous group of participants.
Collapse
Affiliation(s)
- Igor Koval
- Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, Sorbonne Université, 75013, Paris, France
| | - Thomas Dighiero-Brecht
- Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, Sorbonne Université, 75013, Paris, France
| | - Allan J Tobin
- Biological Adaptation and Ageing, Sorbonne Université, Paris, France
- Brain Research Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Sarah J Tabrizi
- UCL Queen Square Institute of Neurology, University College London, Queen Square, London, UK
| | - Rachael I Scahill
- UCL Queen Square Institute of Neurology, University College London, Queen Square, London, UK
| | - Sophie Tezenas du Montcel
- Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, Sorbonne Université, 75013, Paris, France
| | - Stanley Durrleman
- Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, Sorbonne Université, 75013, Paris, France.
| | - Alexandra Durr
- Department of Neurology, DMU Neurosciences, Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, 75013, Paris, France.
| |
Collapse
|
91
|
Pellegrini M, Bergonzoni G, Perrone F, Squitieri F, Biagioli M. Current Diagnostic Methods and Non-Coding RNAs as Possible Biomarkers in Huntington's Disease. Genes (Basel) 2022; 13:2017. [PMID: 36360254 PMCID: PMC9689996 DOI: 10.3390/genes13112017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022] Open
Abstract
Whether as a cause or a symptom, RNA transcription is recurrently altered in pathologic conditions. This is also true for non-coding RNAs, with regulatory functions in a variety of processes such as differentiation, cell identity and metabolism. In line with their increasingly recognized roles in cellular pathways, RNAs are also currently evaluated as possible disease biomarkers. They could be informative not only to follow disease progression and assess treatment efficacy in clinics, but also to aid in the development of new therapeutic approaches. This is especially important for neurological and genetic disorders, where the administration of appropriate treatment during the disease prodromal stage could significantly delay, if not halt, disease progression. In this review we focus on the current status of biomarkers in Huntington's Disease (HD), a fatal hereditary and degenerative disease condition. First, we revise the sources and type of wet biomarkers currently in use. Then, we explore the feasibility of different RNA types (miRNA, ncRNA, circRNA) as possible biomarker candidates, discussing potential advantages, disadvantages, sources of origin and the ongoing investigations on this topic.
Collapse
Affiliation(s)
- Miguel Pellegrini
- Department of Cellular, Computational and Integrative Biology, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Guendalina Bergonzoni
- Department of Cellular, Computational and Integrative Biology, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Federica Perrone
- Huntington and Rare Diseases Unit, IRCCS Casa Sollievo Della Sofferenza Research Hospital, Viale Cappuccini, 71013 San Giovanni Rotondo, Italy
| | - Ferdinando Squitieri
- Huntington and Rare Diseases Unit, IRCCS Casa Sollievo Della Sofferenza Research Hospital, Viale Cappuccini, 71013 San Giovanni Rotondo, Italy
| | - Marta Biagioli
- Department of Cellular, Computational and Integrative Biology, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| |
Collapse
|
92
|
Chu M, Chen Z, Nie B, Liu L, Xie K, Cui Y, Chen K, Rosa-Neto P, Wu L. A longitudinal 18F-FDG PET/MRI study in asymptomatic stage of genetic Creutzfeldt-Jakob disease linked to G114V mutation. J Neurol 2022; 269:6094-6103. [PMID: 35864212 PMCID: PMC9553814 DOI: 10.1007/s00415-022-11288-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 10/31/2022]
Abstract
BACKGROUND Pathogenic prion protein may start to deposit in some brain regions and cause functional alterations in the asymptomatic stage in Creutzfeldt-Jakob disease. The study aims to determine the trajectory of the brain metabolic changes for prion protein diseases at the preclinical stage. METHODS At baseline, we enrolled five asymptomatic PRNP G114V mutation carriers, six affected genetic PRNP E200K CJD patients and 23 normal controls. All participants completed clinical, diffusion-weighted imaging (DWI) and 18F fluorodeoxyglucose-positron emission tomography (18F-FDG-PET) examinations. Longitudinal follow-up was completed in five asymptomatic mutation carriers. We set three-time points to identify the changing trajectory in the asymptomatic carriers group including baseline, 2-year and 4-year follow-up. RESULTS At baseline, DWI signals, the cerebral glucose standardized uptake value rate ratio (SUVR) and clinical status in 5 asymptomatic cases were normal. At the follow-up period, mild hypometabolism on PET images was found in asymptomatic carriers without any DWI abnormal signal. Further group quantitatively analysis showed hypometabolic brain regions in the asymptomatic genetic CJD group were in the insula, frontal, parietal, and temporal lobes in 4-year follow-up. The SUVR changing trajectories of all asymptomatic cases were within the range between the normal controls and affected patients. Notably, the SUVR of one asymptomatic individual whose baseline age was older showed a rapid decline at the last follow-up. CONCLUSIONS Our study illustrates that the neurodegenerative process associated with genetic CJD may initiate before the clinical presentation of the disease.
Collapse
Affiliation(s)
- Min Chu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Changchun Street 45, Beijing, 100053, China
| | - Zhongyun Chen
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Changchun Street 45, Beijing, 100053, China
| | - Binbin Nie
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Li Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Changchun Street 45, Beijing, 100053, China
| | - Kexin Xie
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Changchun Street 45, Beijing, 100053, China
| | - Yue Cui
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Changchun Street 45, Beijing, 100053, China
| | - Kewei Chen
- Banner Alzheimer's Institute, Phoenix, AZ, USA
- School of Mathematics and Statistics, Arizona State University, Phoenix, USA
| | - Pedro Rosa-Neto
- Alzheimer's Disease Research Unit, McGill Centre for Studies in Aging, Montreal, H4H 1R3, Canada
| | - Liyong Wu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Changchun Street 45, Beijing, 100053, China.
| |
Collapse
|
93
|
Papoutsi M, Flower M, Hensman Moss DJ, Holmans P, Estevez-Fraga C, Johnson EB, Scahill RI, Rees G, Langbehn D, Tabrizi SJ. Intellectual enrichment and genetic modifiers of cognition and brain volume in Huntington's disease. Brain Commun 2022; 4:fcac279. [PMID: 36519153 PMCID: PMC9732861 DOI: 10.1093/braincomms/fcac279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 08/22/2022] [Accepted: 10/27/2022] [Indexed: 12/14/2022] Open
Abstract
An important step towards the development of treatments for cognitive impairment in ageing and neurodegenerative diseases is to identify genetic and environmental modifiers of cognitive function and understand the mechanism by which they exert an effect. In Huntington's disease, the most common autosomal dominant dementia, a small number of studies have identified intellectual enrichment, i.e. a cognitively stimulating lifestyle and genetic polymorphisms as potential modifiers of cognitive function. The aim of our study was to further investigate the relationship and interaction between genetic factors and intellectual enrichment on cognitive function and brain atrophy in Huntington's disease. For this purpose, we analysed data from Track-HD, a multi-centre longitudinal study in Huntington's disease gene carriers and focused on the role of intellectual enrichment (estimated at baseline) and the genes FAN1, MSH3, BDNF, COMT and MAPT in predicting cognitive decline and brain atrophy. We found that carrying the 3a allele in the MSH3 gene had a positive effect on global cognitive function and brain atrophy in multiple cortical regions, such that 3a allele carriers had a slower rate of cognitive decline and atrophy compared with non-carriers, in agreement with its role in somatic instability. No other genetic predictor had a significant effect on cognitive function and the effect of MSH3 was independent of intellectual enrichment. Intellectual enrichment also had a positive effect on cognitive function; participants with higher intellectual enrichment, i.e. those who were better educated, had higher verbal intelligence and performed an occupation that was intellectually engaging, had better cognitive function overall, in agreement with previous studies in Huntington's disease and other dementias. We also found that intellectual enrichment interacted with the BDNF gene, such that the positive effect of intellectual enrichment was greater in Met66 allele carriers than non-carriers. A similar relationship was also identified for changes in whole brain and caudate volume; the positive effect of intellectual enrichment was greater for Met66 allele carriers, rather than for non-carriers. In summary, our study provides additional evidence for the beneficial role of intellectual enrichment and carrying the 3a allele in MSH3 in cognitive function in Huntington's disease and their effect on brain structure.
Collapse
Affiliation(s)
- Marina Papoutsi
- UCL Huntington’s Disease Centre, Queen Square Institute of Neurology, University College London, London, UK
- Ixico plc, London, UK
| | - Michael Flower
- UCL Huntington’s Disease Centre, Queen Square Institute of Neurology, University College London, London, UK
| | - Davina J Hensman Moss
- UCL Huntington’s Disease Centre, Queen Square Institute of Neurology, University College London, London, UK
| | - Peter Holmans
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Carlos Estevez-Fraga
- UCL Huntington’s Disease Centre, Queen Square Institute of Neurology, University College London, London, UK
| | - Eileanoir B Johnson
- UCL Huntington’s Disease Centre, Queen Square Institute of Neurology, University College London, London, UK
| | - Rachael I Scahill
- UCL Huntington’s Disease Centre, Queen Square Institute of Neurology, University College London, London, UK
| | - Geraint Rees
- Wellcome Centre for Human Neuroimaging, Queen Square Institute of Neurology, University College London, London, UK
- Institute of Cognitive Neuroscience, University College London, London, UK
| | - Douglas Langbehn
- Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Sarah J Tabrizi
- UCL Huntington’s Disease Centre, Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute at University College London, London, UK
| |
Collapse
|
94
|
Sun Y, Tong H, Yang T, Liu L, Li XJ, Li S. Insights into White Matter Defect in Huntington's Disease. Cells 2022; 11:3381. [PMID: 36359783 PMCID: PMC9656068 DOI: 10.3390/cells11213381] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/21/2022] [Accepted: 10/21/2022] [Indexed: 08/05/2023] Open
Abstract
Huntington's disease (HD) is an autosomal-dominant inherited progressive neurodegenerative disorder. It is caused by a CAG repeat expansion in the Huntingtin gene that is translated to an expanded polyglutamine (PolyQ) repeat in huntingtin protein. HD is characterized by mood swings, involuntary movement, and cognitive decline in the late disease stage. HD patients often die 15-20 years after disease onset. Currently, there is no cure for HD. Due to the striking neuronal loss in HD, most studies focused on the investigation of the predominantly neuronal degeneration in specific brain regions. However, the pathology of the white matter area in the brains of HD patients was also reported by clinical imaging studies, which showed white matter abnormalities even before the clinical onset of HD. Since oligodendrocytes form myelin sheaths around the axons in the brain, white matter lesions are likely attributed to alterations in myelin and oligodendrocyte-associated changes in HD. In this review, we summarized the evidence for white matter, myelin, and oligodendrocytes alterations that were previously observed in HD patients and animal models. We also discussed potential mechanisms for white matter changes and possible treatment to prevent glial dysfunction in HD.
Collapse
|
95
|
Ly S, Didiot MC, Ferguson CM, Coles AH, Miller R, Chase K, Echeverria D, Wang F, Sadri-Vakili G, Aronin N, Khvorova A. Mutant huntingtin messenger RNA forms neuronal nuclear clusters in rodent and human brains. Brain Commun 2022; 4:fcac248. [PMID: 36458209 PMCID: PMC9707646 DOI: 10.1093/braincomms/fcac248] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 07/14/2022] [Accepted: 10/12/2022] [Indexed: 01/25/2023] Open
Abstract
Mutant messenger RNA (mRNA) and protein contribute to the clinical manifestation of many repeat-associated neurological disorders, with the presence of nuclear RNA clusters being a common pathological feature. Yet, investigations into Huntington's disease-caused by a CAG repeat expansion in exon 1 of the huntingtin (HTT) gene-have primarily focused on toxic protein gain-of-function as the primary disease-causing feature. To date, mutant HTT mRNA has not been identified as an in vivo hallmark of Huntington's disease. Here, we report that, in two Huntington's disease mouse models (YAC128 and BACHD-97Q-ΔN17), mutant HTT mRNA is retained in the nucleus. Widespread formation of large mRNA clusters (∼0.6-5 µm3) occurred in 50-75% of striatal and cortical neurons. Cluster formation was independent of age and driven by expanded repeats. Clusters associate with chromosomal transcriptional sites and quantitatively co-localize with the aberrantly processed N-terminal exon 1-intron 1 mRNA isoform, HTT1a. HTT1a mRNA clusters are observed in a subset of neurons from human Huntington's disease post-mortem brain and are likely caused by somatic expansion of repeats. In YAC128 mice, clusters, but not individual HTT mRNA, are resistant to antisense oligonucleotide treatment. Our findings identify mutant HTT/HTT1a mRNA clustering as an early, robust molecular signature of Huntington's disease, providing in vivo evidence that Huntington's disease is a repeat expansion disease with mRNA involvement.
Collapse
Affiliation(s)
| | | | | | - Andrew H Coles
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Rachael Miller
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Kathryn Chase
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Dimas Echeverria
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Feng Wang
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Ghazaleh Sadri-Vakili
- Mass General Institute for Neurodegenerative Disease, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Neil Aronin
- Correspondence may also be addressed to: Neil Aronin 368 Plantation Street, Albert Sherman Center Worcester, MA 01655, USA. E-mail:
| | - Anastasia Khvorova
- Correspondence to: Anastasia Khvorova 368 Plantation Street, Albert Sherman Center Worcester, MA 01655, USA E-mail:
| |
Collapse
|
96
|
Weiss AR, Liguore WA, Brandon K, Wang X, Liu Z, Domire JS, Button D, Srinivasan S, Kroenke CD, McBride JL. A novel rhesus macaque model of Huntington's disease recapitulates key neuropathological changes along with motor and cognitive decline. eLife 2022; 11:e77568. [PMID: 36205397 PMCID: PMC9545527 DOI: 10.7554/elife.77568] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 09/06/2022] [Indexed: 11/25/2022] Open
Abstract
We created a new nonhuman primate model of the genetic neurodegenerative disorder Huntington's disease (HD) by injecting a mixture of recombinant adeno-associated viral vectors, serotypes AAV2 and AAV2.retro, each expressing a fragment of human mutant HTT (mHTT) into the caudate and putamen of adult rhesus macaques. This modeling strategy results in expression of mutant huntingtin protein (mHTT) and aggregate formation in the injected brain regions, as well as dozens of other cortical and subcortical brain regions affected in human HD patients. We queried the disruption of cortico-basal ganglia circuitry for 30 months post-surgery using a variety of behavioral and imaging readouts. Compared to controls, mHTT-treated macaques developed working memory decline and progressive motor impairment. Multimodal imaging revealed circuit-wide white and gray matter degenerative processes in several key brain regions affected in HD. Taken together, we have developed a novel macaque model of HD that may be used to develop disease biomarkers and screen promising therapeutics.
Collapse
Affiliation(s)
- Alison R Weiss
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
| | - William A Liguore
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
| | - Kristin Brandon
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
| | - Xiaojie Wang
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
- Advanced Imaging Research Center, Oregon Health and Science UniversityPortlandUnited States
| | - Zheng Liu
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
- Advanced Imaging Research Center, Oregon Health and Science UniversityPortlandUnited States
| | - Jacqueline S Domire
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
| | - Dana Button
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
| | - Sathya Srinivasan
- Imaging and Morphology Support Core, Oregon National Primate Research CenterBeavertonUnited States
| | - Christopher D Kroenke
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
- Advanced Imaging Research Center, Oregon Health and Science UniversityPortlandUnited States
- Department of Behavioral Neuroscience, Oregon Health and Science UniversityPortlandUnited States
| | - Jodi L McBride
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
- Department of Behavioral Neuroscience, Oregon Health and Science UniversityPortlandUnited States
| |
Collapse
|
97
|
Feigin A, Evans EE, Fisher TL, Leonard JE, Smith ES, Reader A, Mishra V, Manber R, Walters KA, Kowarski L, Oakes D, Siemers E, Kieburtz KD, Zauderer M. Pepinemab antibody blockade of SEMA4D in early Huntington's disease: a randomized, placebo-controlled, phase 2 trial. Nat Med 2022; 28:2183-2193. [PMID: 35941373 PMCID: PMC9361919 DOI: 10.1038/s41591-022-01919-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 06/27/2022] [Indexed: 12/18/2022]
Abstract
SIGNAL is a multicenter, randomized, double-blind, placebo-controlled phase 2 study (no. NCT02481674) established to evaluate pepinemab, a semaphorin 4D (SEMA4D)-blocking antibody, for treatment of Huntington's disease (HD). The trial enrolled a total of 265 HD gene expansion carriers with either early manifest (EM, n = 179) or late prodromal (LP, n = 86) HD, randomized (1:1) to receive 18 monthly infusions of pepinemab (n = 91 EM, 41 LP) or placebo (n = 88 EM, 45 LP). Pepinemab was generally well tolerated, with a relatively low frequency of serious treatment-emergent adverse events of 5% with pepinemab compared to 9% with placebo, including both EM and LP participants. Coprimary efficacy outcome measures consisted of assessments within the EM cohort of (1) a two-item HD cognitive assessment family comprising one-touch stockings of Cambridge (OTS) and paced tapping (PTAP) and (2) clinical global impression of change (CGIC). The differences between pepinemab and placebo in mean change (95% confidence interval) from baseline at month 17 for OTS were -1.98 (-4.00, 0.05) (one-sided P = 0.028), and for PTAP 1.43 (-0.37, 3.23) (one-sided P = 0.06). Similarly, because a significant treatment effect was not observed for CGIC, the coprimary endpoint, the study did not meet its prespecified primary outcomes. Nevertheless, a number of other positive outcomes and post hoc subgroup analyses-including additional cognitive measures and volumetric magnetic resonance imaging and fluorodeoxyglucose-positron-emission tomography imaging assessments-provide rationale and direction for the design of a phase 3 study and encourage the continued development of pepinemab in patients diagnosed with EM HD.
Collapse
Affiliation(s)
- Andrew Feigin
- New York University Langone Health and The Marlene and Paolo Fresco Institute for Parkinson's and Movement Disorders, New York, NY, USA
| | | | | | | | | | | | | | | | | | - Lisa Kowarski
- WCG Statistics Collaborative, Inc., Washington, DC, USA
| | - David Oakes
- University of Rochester Medical Center, Rochester, NY, USA
| | | | | | | |
Collapse
|
98
|
Teismann H, Schubert R, Reilmann R, Berger K. Effects of age and sex on outcomes of the Q-Motor speeded finger tapping and grasping and lifting tests-findings from the population-based BiDirect Study. Front Neurol 2022; 13:965031. [PMID: 36247774 PMCID: PMC9561931 DOI: 10.3389/fneur.2022.965031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/30/2022] [Indexed: 12/03/2022] Open
Abstract
Background Q-Motor is a suite of motor tests originally designed to assess motor symptoms in Huntington's disease. Among others, Q-Motor encompasses a finger tapping task and a grasping and lifting task. To date, there are no systematic investigations regarding effects of variables which may affect the performance in specific Q-Motor tests per se, and normative Q-Motor data based on a large population-based sample are not yet available. Objective We investigated effects of age and sex on five selected Q-Motor outcomes representing the two core Q-Motor tasks speeded finger tapping and grasping and lifting in a community sample of middle-aged to elderly adults. Furthermore, we explored effects of the potentially mediating variables educational attainment, alcohol consumption, smoking status, and depressive symptoms. Moreover, we explored inter-examiner variability. Finally, we compared the findings to findings for the Purdue Pegboard test. Methods Based on a sample of 726 community-dwelling adults and using multiple (Gaussian) regression analysis, we modeled the motor outcomes using age, sex, years in full-time education, depressive symptoms in the past seven days, alcohol consumption in the past seven days, and smoking status as explanatory variables. Results With regard to the Q-Motor tests, we found that more advanced age was associated with reduced tapping speed, male sex was associated with increased tapping speed and less irregularity, female sex was associated with less involuntary movement, more years of education were associated with increased tapping speed and less involuntary movement, never smoking was associated with less involuntary movement compared to current smoking, and more alcohol consumed was associated with more involuntary movement. Conclusion The present results show specific effects of age and sex on Q-Motor finger tapping and grasping and lifting performance. In addition, besides effects of education, there also were specific effects of smoking status and alcohol consumption. Importantly, the present study provides normative Q-Motor data based on a large population-based sample. Overall, the results are in favor of the feasibility and validity of Q-Motor finger tapping and grasping and lifting for large observational studies. Due to their low task-complexity and lack of placebo effects, Q-Motor tests may generate additional value in particular with regard to clinical conditions such as Huntington's or Parkinson's disease.
Collapse
Affiliation(s)
- Henning Teismann
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
- *Correspondence: Henning Teismann
| | | | - Ralf Reilmann
- George-Huntington-Institute, Münster, Germany
- Institute of Clinical Radiology, University of Münster, Münster, Germany
- Department of Neurodegenerative Diseases and Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Klaus Berger
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| |
Collapse
|
99
|
Ghafouri-Fard S, Khoshbakht T, Hussen BM, Taheri M, Ebrahimzadeh K, Noroozi R. The emerging role of long non-coding RNAs, microRNAs, and an accelerated epigenetic age in Huntington’s disease. Front Aging Neurosci 2022; 14:987174. [PMID: 36185471 PMCID: PMC9520620 DOI: 10.3389/fnagi.2022.987174] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
Abstract
Huntington’s disease (HD) is a dominantly inherited neurodegenerative disease with variable clinical manifestations. Recent studies highlighted the contribution of epigenetic alterations to HD progress and onset. The potential crosstalk between different epigenetic layers and players such as aberrant expression of non-coding RNAs and methylation alterations has been found to affect the pathogenesis of HD or mediate the effects of trinucleotide expansion in its pathophysiology. Also, microRNAs have been assessed for their roles in the modulation of HD manifestations, among them are miR-124, miR-128a, hsa-miR-323b-3p, miR-432, miR-146a, miR-19a, miR-27a, miR-101, miR-9*, miR-22, miR-132, and miR-214. Moreover, long non-coding RNAs such as DNM3OS, NEAT1, Meg3, and Abhd11os are suggested to be involved in the pathogenesis of HD. An accelerated DNA methylation age is another epigenetic signature reported recently for HD. The current literature search collected recent findings of dysregulation of miRNAs or lncRNAs as well as methylation changes and epigenetic age in HD.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tayyebeh Khoshbakht
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Iraq
- Center of Research and Strategic Studies, Lebanese French University, Erbil, Iraq
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kaveh Ebrahimzadeh
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- *Correspondence: Kaveh Ebrahimzadeh,
| | - Rezvan Noroozi
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
- Rezvan Noroozi,
| |
Collapse
|
100
|
Neueder A, Kojer K, Hering T, Lavery DJ, Chen J, Birth N, Hallitsch J, Trautmann S, Parker J, Flower M, Sethi H, Haider S, Lee JM, Tabrizi SJ, Orth M. Abnormal molecular signatures of inflammation, energy metabolism, and vesicle biology in human Huntington disease peripheral tissues. Genome Biol 2022; 23:189. [PMID: 36071529 PMCID: PMC9450392 DOI: 10.1186/s13059-022-02752-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 08/18/2022] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND A major challenge in neurodegenerative diseases concerns identifying biological disease signatures that track with disease progression or respond to an intervention. Several clinical trials in Huntington disease (HD), an inherited, progressive neurodegenerative disease, are currently ongoing. Therefore, we examine whether peripheral tissues can serve as a source of readily accessible biological signatures at the RNA and protein level in HD patients. RESULTS We generate large, high-quality human datasets from skeletal muscle, skin and adipose tissue to probe molecular changes in human premanifest and early manifest HD patients-those most likely involved in clinical trials. The analysis of the transcriptomics and proteomics data shows robust, stage-dependent dysregulation. Gene ontology analysis confirms the involvement of inflammation and energy metabolism in peripheral HD pathogenesis. Furthermore, we observe changes in the homeostasis of extracellular vesicles, where we find consistent changes of genes and proteins involved in this process. In-depth single nucleotide polymorphism data across the HTT gene are derived from the generated primary cell lines. CONCLUSIONS Our 'omics data document the involvement of inflammation, energy metabolism, and extracellular vesicle homeostasis. This demonstrates the potential to identify biological signatures from peripheral tissues in HD suitable as biomarkers in clinical trials. The generated data, complemented by the primary cell lines established from peripheral tissues, and a large panel of iPSC lines that can serve as human models of HD are a valuable and unique resource to advance the current understanding of molecular mechanisms driving HD pathogenesis.
Collapse
Affiliation(s)
- Andreas Neueder
- Department of Neurology, Ulm University, 89081, Ulm, Germany
| | - Kerstin Kojer
- Department of Neurology, Ulm University, 89081, Ulm, Germany
| | - Tanja Hering
- Department of Neurology, Ulm University, 89081, Ulm, Germany
| | - Daniel J Lavery
- CHDI Foundation, Princeton, NJ, 08540, USA
- Loulou Foundation, Orphan Disease Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jian Chen
- CHDI Foundation, Princeton, NJ, 08540, USA
| | - Nathalie Birth
- Department of Neurology, Ulm University, 89081, Ulm, Germany
| | | | - Sonja Trautmann
- Department of Neurology, Ulm University, 89081, Ulm, Germany
| | - Jennifer Parker
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK
| | - Michael Flower
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK
| | - Huma Sethi
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK
| | - Salman Haider
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK
| | - Jong-Min Lee
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Neurology, Harvard Medical School, Boston, MA, 02115, USA
| | - Sarah J Tabrizi
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK
| | - Michael Orth
- Department of Neurology, Ulm University, 89081, Ulm, Germany.
- Swiss Huntington Centre, Neurozentrum, Siloah AG, Worbstr. 312, 3073, Gümligen, Switzerland.
- University Hospital of Old Age Psychiatry and Psychotherapy, Bern University, Bern, Switzerland.
| |
Collapse
|