1
|
Marzec P, Richer M, Lahue RS. Therapeutic targeting of mismatch repair proteins in triplet repeat expansion diseases. DNA Repair (Amst) 2025; 147:103817. [PMID: 40010080 DOI: 10.1016/j.dnarep.2025.103817] [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: 11/01/2024] [Revised: 02/07/2025] [Accepted: 02/07/2025] [Indexed: 02/28/2025]
Abstract
Triplet repeat expansion diseases are a class of ∼20 inherited neurological disorders. Many of these diseases are debilitating, sometimes fatally so, and they have unfortunately proved difficult to treat. New compelling evidence shows that somatic repeat expansions in some diseases are essential to the pathogenic process, accelerating the age of onset and the rate of disease progression. Inhibiting somatic repeat expansions, therefore, provides a therapeutic opportunity to delay or block disease onset and/or slow progression. Several key aspects enhance the appeal of this therapeutic approach. First, the proteins responsible for promoting expansions are known from human genetics and model systems, obviating the need for lengthy target searches. They include the mismatch repair proteins MSH3, PMS1 and MLH3. Second, inhibiting or downregulating any of these three proteins is attractive due to their good safety profiles. Third, having three potential targets helps mitigate risk. Fourth, another protein, the nuclease FAN1, protects against expansions; in principle, increasing FAN1 activity could be therapeutic. Fifth, therapies aimed at inhibiting somatic repeat expansions could be used against several diseases that display this shared mechanistic feature, offering the opportunity for one treatment against multiple diseases. This review will address the underlying findings and the recent therapeutic advances in targeting MSH3, PMS1, MLH3 and FAN1 in triplet repeat expansion diseases.
Collapse
Affiliation(s)
- Paulina Marzec
- LoQus23 Therapeutics Ltd, Cambridge CB22 3AT, United Kingdom
| | | | - Robert S Lahue
- Centre for Chromosome Biology, University of Galway, H91W2TY, Ireland; Galway Neuroscience Centre, University of Galway, H91W2TY, Ireland.
| |
Collapse
|
2
|
Wilkes FA, Jakabek D, Walterfang M, Velakoulis D, Poudel GR, Stout JC, Chua P, Egan GF, Looi JCL, Georgiou-Karistianis N. Hippocampal morphology in Huntington's disease, implications for plasticity and pathogenesis: The IMAGE-HD study. Psychiatry Res Neuroimaging 2023; 335:111694. [PMID: 37598529 DOI: 10.1016/j.pscychresns.2023.111694] [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: 03/10/2023] [Revised: 06/10/2023] [Accepted: 07/26/2023] [Indexed: 08/22/2023]
Abstract
While striatal changes in Huntington's Disease (HD) are well established, few studies have investigated changes in the hippocampus, a key neuronal hub. Using MRI scans obtained from the IMAGE-HD study, hippocampi were manually traced and then analysed with the Spherical Harmonic Point Distribution Method (SPHARM-PDM) in 36 individuals with presymptomatic-HD, 37 with early symptomatic-HD, and 36 healthy matched controls. There were no significant differences in overall hippocampal volume between groups. Interestingly we found decreased bilateral hippocampal volume in people with symptomatic-HD who took selective serotonin reuptake inhibitors compared to those who did not, despite no significant differences in anxiety, depressive symptoms, or motor incapacity between the two groups. In symptomatic-HD, there was also significant shape deflation in the right hippocampal head, showing the utility of using manual tracing and SPHARM-PDM to characterise subtle shape changes which may be missed by other methods. This study confirms previous findings of the lack of hippocampal volumetric differentiation in presymptomatic-HD and symptomatic-HD compared to controls. We also find novel shape and volume findings in those with symptomatic-HD, especially in relation to decreased hippocampal volume in those treated with SSRIs.
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; Neuroscience Research Australia, Sydney, Australia
| | - Nellie Georgiou-Karistianis
- School of Psychological Sciences and the Turner Institute of Brain and Mental Health, Monash University, Melbourne, Australia
| |
Collapse
|
3
|
van Eimeren T, Giehl K, Reetz K, Sampaio C, Mestre TA. Neuroimaging biomarkers in Huntington's disease: Preparing for a new era of therapeutic development. Parkinsonism Relat Disord 2023; 114:105488. [PMID: 37407343 DOI: 10.1016/j.parkreldis.2023.105488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/05/2023] [Accepted: 06/10/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND A critical challenge for Huntington's disease (HD) clinical trials in disease modification is the definition of endpoints that can capture change when clinical signs are subtle/non-existent. Reliable biomarkers are therefore urgently needed to facilitate drug development by allowing the enrichment of clinical trial populations and providing measures of benefit that can support the establishment of efficacy. METHODS By systematically examining the published literature on HD neuroimaging biomarker studies, we sought to advance knowledge to guide the validation of neuroimaging biomarkers. We started by reviewing both cross-sectional and longitudinal studies and then conducted an in-depth review to make quantitative comparisons between biomarkers using data only from longitudinal studies with samples sizes larger than ten participants in PET studies or 30 participants in MRI studies. RESULTS From a total of 2202 publications initially identified, we included 32 studies, 19 of which underwent in-depth comparative review. The majority of included studies used various MRI-based methods (manual to automatic) to longitudinally assess either the volume of the putamen or the caudate, which have been shown to undergo significant structural change during HD natural history. CONCLUSION Despite the impressively large number of neuroimaging biomarker studies, only a small number of adequately designed studies met our criteria. Among these various biomarkers, MRI-based volumetric analyses of the caudate and putamen are currently the best validated for use in the disease phase before clinical motor diagnosis. A biomarker that can be used to demonstrate a disease-modifying effect is still missing.
Collapse
Affiliation(s)
- Thilo van Eimeren
- University of Cologne, Faculty of Medicine, Department of Nuclear Medicine, Cologne, Germany; University of Cologne, Faculty of Medicine, Department of Neurology, Cologne, Germany.
| | - Kathrin Giehl
- University of Cologne, Faculty of Medicine, Department of Nuclear Medicine, Cologne, Germany; Research Center Jülich, Institute for Neuroscience and Medicine (INM-2), Jülich, Germany
| | - Kathrin Reetz
- University of Aachen, Department of Neurology, Aachen, Germany
| | | | - Tiago A Mestre
- University of Ottawa, Department of Medicine, Division of Neurology, The Ottawa Hospital Research Institute, Parkinson's Disease and Movement Disorders Center, Canada
| |
Collapse
|
4
|
Liu CF, Younes L, Tong XJ, Hinkle JT, Wang M, Phatak S, Xu X, Bu X, Looi V, Bang J, Tabrizi SJ, Scahill RI, Paulsen JS, Georgiou-Karistianis N, Faria AV, Miller MI, Ratnanather JT, Ross CA. Longitudinal imaging highlights preferential basal ganglia circuit atrophy in Huntington's disease. Brain Commun 2023; 5:fcad214. [PMID: 37744022 PMCID: PMC10516592 DOI: 10.1093/braincomms/fcad214] [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: 04/07/2022] [Revised: 05/09/2023] [Accepted: 08/17/2023] [Indexed: 09/26/2023] Open
Abstract
Huntington's disease is caused by a CAG repeat expansion in the Huntingtin gene (HTT), coding for polyglutamine in the Huntingtin protein, with longer CAG repeats causing earlier age of onset. The variable 'Age' × ('CAG'-L), where 'Age' is the current age of the individual, 'CAG' is the repeat length and L is a constant (reflecting an approximation of the threshold), termed the 'CAG Age Product' (CAP) enables the consideration of many individuals with different CAG repeat expansions at the same time for analysis of any variable and graphing using the CAG Age Product score as the X axis. Structural MRI studies have showed that progressive striatal atrophy begins many years prior to the onset of diagnosable motor Huntington's disease, confirmed by longitudinal multicentre studies on three continents, including PREDICT-HD, TRACK-HD and IMAGE-HD. However, previous studies have not clarified the relationship between striatal atrophy, atrophy of other basal ganglia structures, and atrophy of other brain regions. The present study has analysed all three longitudinal datasets together using a single image segmentation algorithm and combining data from a large number of subjects across a range of CAG Age Product score. In addition, we have used a strategy of normalizing regional atrophy to atrophy of the whole brain, in order to determine which regions may undergo preferential degeneration. This made possible the detailed characterization of regional brain atrophy in relation to CAG Age Product score. There is dramatic selective atrophy of regions involved in the basal ganglia circuit-caudate, putamen, nucleus accumbens, globus pallidus and substantia nigra. Most other regions of the brain appear to have slower but steady degeneration. These results support (but certainly do not prove) the hypothesis of circuit-based spread of pathology in Huntington's disease, possibly due to spread of mutant Htt protein, though other connection-based mechanisms are possible. Therapeutic targets related to prion-like spread of pathology or other mechanisms may be suggested. In addition, they have implications for current neurosurgical therapeutic approaches, since delivery of therapeutic agents solely to the caudate and putamen may miss other structures affected early, such as nucleus accumbens and output nuclei of the striatum, the substantia nigra and the globus pallidus.
Collapse
Affiliation(s)
- Chin-Fu Liu
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Laurent Younes
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Xiao J Tong
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore MD 21287, USA
| | - Jared T Hinkle
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Medical Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Maggie Wang
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sanika Phatak
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Xin Xu
- Division of Magnetic Resonance, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Xuan Bu
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Vivian Looi
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jee Bang
- Division of Neurobiology, Department of Psychiatry, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sarah J Tabrizi
- HD Research Centre, University College London Queen Square Institute of Neurology, UCL, London, UK
| | - Rachael I Scahill
- HD Research Centre, University College London Queen Square Institute of Neurology, UCL, London, UK
| | - Jane S Paulsen
- Department of Neurology, University of Wisconsin, Madison, WI 53705, USA
| | - Nellie Georgiou-Karistianis
- School of Psychological Sciences and The Turner Institute for Brain and Mental Health, Monash University, Melbourne, Victoria 3800, Australia
| | - Andreia V Faria
- Division of Magnetic Resonance, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Michael I Miller
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - J Tilak Ratnanather
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Christopher A Ross
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore MD 21287, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Division of Neurobiology, Department of Psychiatry, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
5
|
Hu B, Younes L, Bu X, Liu CF, Ratnanather JT, Paulsen J, Georgiou-Karistianis N, Miller MI, Ross C, Faria AV. Mixed longitudinal and cross-sectional analyses of deep gray matter and white matter using diffusion weighted images in premanifest and manifest Huntington's disease. Neuroimage Clin 2023; 39:103493. [PMID: 37582307 PMCID: PMC10448214 DOI: 10.1016/j.nicl.2023.103493] [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: 12/01/2022] [Revised: 04/29/2023] [Accepted: 08/07/2023] [Indexed: 08/17/2023]
Abstract
Changes in the brain of patients with Huntington's disease (HD) begin years before clinical onset, so it remains critical to identify biomarkers to track these early changes. Metrics derived from tensor modeling of diffusion-weighted MRIs (DTI), that indicate the microscopic brain structure, can add important information to regional volumetric measurements. This study uses two large-scale longitudinal, multicenter datasets, PREDICT-HD and IMAGE-HD, to trace changes in DTI of HD participants with a broad range of CAP scores (a product of CAG repeat expansion and age), including those with pre-manifest disease (i.e., prior to clinical onset). Utilizing a fully automated data-driven approach to study the whole brain divided in regions of interest, we traced changes in DTI metrics (diffusivity and fractional anisotropy) versus CAP scores, using sigmoidal and linear regression models. We identified points of inflection in the sigmoidal regression using change-point analysis. The deep gray matter showed more evident and earlier changes in DTI metrics over CAP scores, compared to the deep white matter. In the deep white matter, these changes were more evident and occurred earlier in superior and posterior areas, compared to anterior and inferior areas. The curves of mean diffusivity vs. age of HD participants within a fixed CAP score were different from those of controls, indicating that the disease has an additional effect to age on the microscopic brain structure. These results show the regional and temporal vulnerability of the white matter and deep gray matter in HD, with potential implications for experimental therapeutics.
Collapse
Affiliation(s)
- Beini Hu
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Laurent Younes
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA
| | - Xuan Bu
- Department of Radiology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Chin-Fu Liu
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - J Tilak Ratnanather
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jane Paulsen
- Department of Psychiatry, Neurology, Psychological Brain Sciences, University of Iowa, USA; Department Neurology, University of Wisconsin-Madison, USA
| | - Nellie Georgiou-Karistianis
- School of Psychological Sciences and Turner Institute of Brain and Mental Health, Monash University, Australia
| | - Michael I Miller
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Christopher Ross
- Department of Psychiatry, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Andreia V Faria
- Department of Radiology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
6
|
Ramirez-Garcia G, Galvez V, Diaz R, Campos-Romo A, Fernandez-Ruiz J. Montreal Cognitive Assessment (MoCA) performance in Huntington's disease patients correlates with cortical and caudate atrophy. PeerJ 2022; 10:e12917. [PMID: 35402100 PMCID: PMC8988933 DOI: 10.7717/peerj.12917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 01/20/2022] [Indexed: 01/11/2023] Open
Abstract
Huntington's Disease (HD) is an autosomal neurodegenerative disease characterized by motor, cognitive, and psychiatric symptoms. Cognitive impairment develops gradually in HD patients, progressing later into a severe cognitive dysfunction. The Montreal Cognitive Assessment (MoCA) is a brief screening test commonly employed to detect mild cognitive impairment, which has also been useful to assess cognitive decline in HD patients. However, the relationship between MoCA performance and brain structural integrity in HD patients remains unclear. Therefore, to explore this relationship we analyzed if cortical thinning and subcortical nuclei volume differences correlated with HD patients' MoCA performance. Twenty-two HD patients and twenty-two healthy subjects participated in this study. T1-weighted images were acquired to analyze cortical thickness and subcortical nuclei volumes. Group comparison analysis showed a significantly lower score in the MoCA global performance of HD patients. Also, the MoCA total score correlated with cortical thinning of fronto-parietal and temporo-occipital cortices, as well as with bilateral caudate volume differences in HD patients. These results provide new insights into the effectiveness of using the MoCA test to detect cognitive impairment and the brain atrophy pattern associated with the cognitive status of prodromal/early HD patients.
Collapse
Affiliation(s)
- Gabriel Ramirez-Garcia
- Departamento de Fisiología, Universidad Nacional Autónoma de Mexico, Ciudad de Mexico, Mexico
| | - Victor Galvez
- Escuela de Psicología, Universidad Panamericana, Ciudad de Mexico, Mexico
| | - Rosalinda Diaz
- Departamento de Fisiología, Universidad Nacional Autónoma de Mexico, Ciudad de Mexico, Mexico
| | - Aurelio Campos-Romo
- Facultad de Medicina, Unidad Periférica de Neurociencias, Universidad Nacional Autónoma de México/Instituto Nacional de Neurologia y Neurocirugia, Ciudad de Mexico, Mexico
| | - Juan Fernandez-Ruiz
- Departamento de Fisiología, Universidad Nacional Autónoma de Mexico, Ciudad de Mexico, Mexico
| |
Collapse
|
7
|
Nagtegaal S, David S, van Grinsven E, van Zandvoort M, Seravalli E, Snijders T, Philippens M, Verhoeff J. Morphological changes after cranial fractionated photon radiotherapy: Localized loss of white matter and grey matter volume with increasing dose. Clin Transl Radiat Oncol 2021; 31:14-20. [PMID: 34504960 PMCID: PMC8416633 DOI: 10.1016/j.ctro.2021.08.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 08/04/2021] [Accepted: 08/25/2021] [Indexed: 12/21/2022] Open
Abstract
PURPOSE Numerous brain MR imaging studies have been performed to understand radiation-induced cognitive decline. However, many of them focus on a single region of interest, e.g. cerebral cortex or hippocampus. In this study, we use deformation-based morphometry (DBM) and voxel-based morphometry (VBM) to measure the morphological changes in patients receiving fractionated photon RT, and relate these to the dose. Additionally, we study tissue specific volume changes in white matter (WM), grey matter (GM), cerebrospinal fluid and total intracranial volume (TIV). METHODS AND MATERIALS From our database, we selected 28 patients with MRI of high quality available at baseline and 1 year after RT. Scans were rigidly registered to each other, and to the planning CT and dose file. We used DBM to study non-tissue-specific volumetric changes, and VBM to study volume loss in grey matter. Observed changes were then related to the applied radiation dose (in EQD2). Additionally, brain tissue was segmented into WM, GM and cerebrospinal fluid, and changes in these volumes and TIV were tested. RESULTS Performing DBM resulted in clusters of dose-dependent volume loss 1 year after RT seen throughout the brain. Both WM and GM were affected; within the latter both cerebral cortex and subcortical nuclei show volume loss. Volume loss rates ranging from 5.3 to 15.3%/30 Gy were seen in the cerebral cortical regions in which more than 40% of voxels were affected. In VBM, similar loss rates were seen in the cortex and nuclei. The total volume of WM and GM significantly decreased with rates of 5.8% and 2.1%, while TIV remained unchanged as expected. CONCLUSIONS Radiotherapy is associated with dose-dependent intracranial morphological changes throughout the entire brain. Therefore, we will consider to revise sparing of organs at risk based on future cognitive and neurofunctional data.
Collapse
Key Words
- Brain neoplasms
- CAT12, Computational Anatomy Toolbox 12
- CSF, cerebrospinal fluid
- CT, computed tomography
- DBM, deformation based morphometry
- FWER, family-wise error rate
- GM, grey matter
- Gray matter
- IMPT, intensity modulated proton therapy
- MNI, Montreal Neurological Institute
- MRI, magnetic resonance imaging
- PALM, permutation analysis of linear models
- PTV, planning target volume
- RT, radiotherapy
- Radiotherapy
- SNR, signal to noise ratio
- TFCE, Threshold-Free Cluster Enhancement
- TFE, turbo fast echo
- TIV, total intracranial volume
- VBM, voxel-based morphometry
- VMAT, volumetric modulated arc therapy
- White matter
Collapse
Affiliation(s)
- S.H.J. Nagtegaal
- Department of Radiation Oncology, University Medical Center, HP Q 00.3.11, PO Box 85500, 3508 GA Utrecht, the Netherlands
| | - S David
- Department of Radiation Oncology, University Medical Center, HP Q 00.3.11, PO Box 85500, 3508 GA Utrecht, the Netherlands
| | - E.E. van Grinsven
- Department of Radiation Oncology, University Medical Center, HP Q 00.3.11, PO Box 85500, 3508 GA Utrecht, the Netherlands
| | - M.J.E. van Zandvoort
- UMC Utrecht Brain Center, Department of Neurology & Neurosurgery, University Medical Center, HP L 01.310, PO Box 85500, 3508 GA Utrecht, the Netherlands
| | - E. Seravalli
- Department of Radiation Oncology, University Medical Center, HP Q 00.3.11, PO Box 85500, 3508 GA Utrecht, the Netherlands
| | - T.J Snijders
- UMC Utrecht Brain Center, Department of Neurology & Neurosurgery, University Medical Center, HP L 01.310, PO Box 85500, 3508 GA Utrecht, the Netherlands
| | - M.E.P. Philippens
- Department of Radiation Oncology, University Medical Center, HP Q 00.3.11, PO Box 85500, 3508 GA Utrecht, the Netherlands
| | - J.J.C. Verhoeff
- Department of Radiation Oncology, University Medical Center, HP Q 00.3.11, PO Box 85500, 3508 GA Utrecht, the Netherlands
| |
Collapse
|
8
|
Kinnunen KM, Schwarz AJ, Turner EC, Pustina D, Gantman EC, Gordon MF, Joules R, Mullin AP, Scahill RI, Georgiou-Karistianis N. Volumetric MRI-Based Biomarkers in Huntington's Disease: An Evidentiary Review. Front Neurol 2021; 12:712555. [PMID: 34621236 PMCID: PMC8490802 DOI: 10.3389/fneur.2021.712555] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/10/2021] [Indexed: 01/02/2023] Open
Abstract
Huntington's disease (HD) is an autosomal-dominant inherited neurodegenerative disorder that is caused by expansion of a CAG-repeat tract in the huntingtin gene and characterized by motor impairment, cognitive decline, and neuropsychiatric disturbances. Neuropathological studies show that disease progression follows a characteristic pattern of brain atrophy, beginning in the basal ganglia structures. The HD Regulatory Science Consortium (HD-RSC) brings together diverse stakeholders in the HD community—biopharmaceutical industry, academia, nonprofit, and patient advocacy organizations—to define and address regulatory needs to accelerate HD therapeutic development. Here, the Biomarker Working Group of the HD-RSC summarizes the cross-sectional evidence indicating that regional brain volumes, as measured by volumetric magnetic resonance imaging, are reduced in HD and are correlated with disease characteristics. We also evaluate the relationship between imaging measures and clinical change, their longitudinal change characteristics, and within-individual longitudinal associations of imaging with disease progression. This analysis will be valuable in assessing pharmacodynamics in clinical trials and supporting clinical outcome assessments to evaluate treatment effects on neurodegeneration.
Collapse
Affiliation(s)
| | - Adam J Schwarz
- Takeda Pharmaceuticals, Ltd., Cambridge, MA, United States
| | | | - Dorian Pustina
- CHDI Management/CHDI Foundation, Princeton, NJ, United States
| | - Emily C Gantman
- CHDI Management/CHDI Foundation, Princeton, NJ, United States
| | - Mark F Gordon
- Teva Pharmaceuticals, West Chester, PA, United States
| | | | - Ariana P Mullin
- Critical Path Institute, Tucson, AZ, United States.,Wave Life Sciences, Ltd., Cambridge, MA, United States
| | - Rachael I Scahill
- Huntington's Disease Research Centre, UCL Institute of Neurology, London, United Kingdom
| | - Nellie Georgiou-Karistianis
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia
| | | |
Collapse
|
9
|
Kim R, Müller S, Garcia TP. svReg: Structural varying-coefficient regression to differentiate how regional brain atrophy affects motor impairment for Huntington disease severity groups. Biom J 2021; 63:1254-1271. [PMID: 33871905 PMCID: PMC9012319 DOI: 10.1002/bimj.202000312] [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: 10/19/2020] [Revised: 02/10/2021] [Accepted: 03/06/2021] [Indexed: 11/06/2022]
Abstract
For Huntington disease, identification of brain regions related to motor impairment can be useful for developing interventions to alleviate the motor symptom, the major symptom of the disease. However, the effects from the brain regions to motor impairment may vary for different groups of patients. Hence, our interest is not only to identify the brain regions but also to understand how their effects on motor impairment differ by patient groups. This can be cast as a model selection problem for a varying-coefficient regression. However, this is challenging when there is a pre-specified group structure among variables. We propose a novel variable selection method for a varying-coefficient regression with such structured variables and provide a publicly available R package svreg for implementation of our method. Our method is empirically shown to select relevant variables consistently. Also, our method screens irrelevant variables better than existing methods. Hence, our method leads to a model with higher sensitivity, lower false discovery rate and higher prediction accuracy than the existing methods. Finally, we found that the effects from the brain regions to motor impairment differ by disease severity of the patients. To the best of our knowledge, our study is the first to identify such interaction effects between the disease severity and brain regions, which indicates the need for customized intervention by disease severity.
Collapse
Affiliation(s)
- Rakheon Kim
- Department of Statistics, Texas A&M University, TX, USA
| | - Samuel Müller
- Department of Mathematics and Statistics, Sydney, Australia
- School of Mathematics and Statistics, The University of Sydney, Sydney, Australia
| | - Tanya P. Garcia
- Department of Biostatistics, Gillings School of Public Health, UNC Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
10
|
Fotiadis P, Pasi M, Charidimou A, Warren AD, Schwab KM, Rosand J, van der Grond J, van Buchem MA, Viswanathan A, Gurol ME, Greenberg SM. Decreased Basal Ganglia Volume in Cerebral Amyloid Angiopathy. J Stroke 2021; 23:223-233. [PMID: 34102757 PMCID: PMC8189850 DOI: 10.5853/jos.2020.04280] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 01/06/2021] [Indexed: 11/25/2022] Open
Abstract
Background and Purpose Cerebral amyloid angiopathy (CAA) is a common pathology of the leptomeningeal and cortical small vessels associated with hemorrhagic and non-hemorrhagic brain injury. Given previous evidence for CAA-related loss of cortical thickness and white matter volume, we hypothesized that CAA might also cause tissue loss in the basal ganglia.
Methods We compared basal ganglia volumes expressed as a percentage of total intracranial volume (pBGV) of non-demented patients with sporadic and hereditary CAA to age-matched healthy control (HC) and Alzheimer’s disease (AD) cohorts.
Results Patients with sporadic CAA had lower pBGV (n=80, 1.16%±0.14%) compared to HC (n=80, 1.30%±0.13%, P<0.0001) and AD patients (n=80, 1.23%±0.11%, P=0.001). Similarly, patients with hereditary CAA demonstrated lower pBGV (n=25, 1.26%±0.17%) compared to their matched HC (n=25, 1.36%±0.15%, P=0.036). Using a measurement of normalized basal ganglia width developed for analysis of clinical-grade magnetic resonance images, we found smaller basal ganglia width in patients with CAA-related lobar intracerebral hemorrhage (ICH; n=93, 12.35±1.47) compared to age-matched patients with hypertension-related deep ICH (n=93, 13.46±1.51, P<0.0001) or HC (n=93, 15.45±1.22, P<0.0001). Within the sporadic CAA research cohort, decreased basal ganglia volume was independently correlated with greater cortical gray matter atrophy (r=0.45, P<0.0001), increased basal ganglia fractional anisotropy (r=–0.36, P=0.001), and worse performance on language processing (r=0.35, P=0.003), but not with cognitive tests of executive function or processing speed.
Conclusions These findings suggest an independent effect of CAA on basal ganglia tissue loss, indicating a novel mechanism for CAA-related brain injury and neurologic dysfunction.
Collapse
Affiliation(s)
- Panagiotis Fotiadis
- Department of Neurology, J.P. Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marco Pasi
- Department of Neurology, J.P. Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Stroke Unit, Department of Neurology, University of Lille, INSERM U1171, CHU Lille, Lille, France
| | - Andreas Charidimou
- Department of Neurology, J.P. Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew D Warren
- Department of Neurology, J.P. Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kristin M Schwab
- Department of Neurology, J.P. Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Jonathan Rosand
- Department of Neurology, J.P. Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jeroen van der Grond
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Mark A van Buchem
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Anand Viswanathan
- Department of Neurology, J.P. Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - M Edip Gurol
- Department of Neurology, J.P. Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Steven M Greenberg
- Department of Neurology, J.P. Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
Ravalia AS, Lau J, Barron JC, Purchase SLM, Southwell AL, Hayden MR, Nafar F, Parsons MP. Super-resolution imaging reveals extrastriatal synaptic dysfunction in presymptomatic Huntington disease mice. Neurobiol Dis 2021; 152:105293. [PMID: 33556538 DOI: 10.1016/j.nbd.2021.105293] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 01/14/2021] [Accepted: 02/03/2021] [Indexed: 12/17/2022] Open
Abstract
Synaptic structure and function are compromised prior to cell death and symptom onset in a variety of neurodegenerative diseases. In Huntington disease (HD), a CAG repeat expansion in the gene encoding the huntingtin protein results in a presymptomatic stage that typically spans multiple decades and is followed by striking degeneration of striatal tissue and the progression of debilitating motor symptoms. Many lines of evidence demonstrate that the HD presymptomatic window is associated with injurious effects to striatal synapses, many of which appear to be prerequisites to subsequent cell death. While the striatum is the most vulnerable region in the HD brain, it is widely recognized that HD is a brain-wide disease, affecting numerous extrastriatal regions that contribute to debilitating non-motor symptoms including cognitive dysfunction. Currently, we have a poor understanding of the synaptic integrity, or lack thereof, in extrastriatal regions in the presymptomatic HD brain. If early therapeutic intervention seeks to maintain healthy synaptic function, it is important to understand early HD-associated synaptopathy at a brain-wide, rather than striatal-exclusive, level. Here, we focused on the hippocampus as this structure is generally thought to be affected only in manifest HD despite the subtle cognitive deficits known to emerge in prodromal HD. We used super-resolution microscopy and multi-electrode array electrophysiology as sensitive measures of excitatory synapse structure and function, respectively, in the hippocampus of presymptomatic heterozygous HD mice (Q175FDN model). We found clear evidence for enhanced AMPA receptor subunit clustering and hyperexcitability well before the onset of a detectable HD-like behavioral phenotype. In addition, activity-dependent re-organization of synaptic protein nanostructure, and functional measures of synaptic plasticity were impaired in presymptomatic HD mice. These data demonstrate that synaptic abnormalities in the presymptomatic HD brain are not exclusive to the striatum, and highlight the need to better understand the region-dependent complexities of early synaptopathy in the HD brain.
Collapse
Affiliation(s)
- Adam S Ravalia
- Division of Biomedical Science, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| | - James Lau
- Division of Biomedical Science, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| | - Jessica C Barron
- Division of Biomedical Science, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| | - Stephanie L M Purchase
- Division of Biomedical Science, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| | - Amber L Southwell
- University of Central Florida, College of Medicine, Burnett School of Biomedical Sciences, Orlando, FL, USA
| | - Michael R Hayden
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Firoozeh Nafar
- Division of Biomedical Science, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| | - Matthew P Parsons
- Division of Biomedical Science, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada.
| |
Collapse
|
12
|
Estevez-Fraga C, Scahill RI, Durr A, Leavitt BR, Roos RAC, Langbehn DR, Rees G, Gregory S, Tabrizi SJ. Composite UHDRS Correlates With Progression of Imaging Biomarkers in Huntington's Disease. Mov Disord 2021; 36:1259-1264. [PMID: 33471951 DOI: 10.1002/mds.28489] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 11/10/2020] [Accepted: 12/07/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The composite Unified Huntington's Disease Rating Scale (cUHDRS) is a multidimensional measure of progression in Huntington's disease (HD) being used as a primary outcome in clinical trials investigating potentially disease-modifying huntingtin-lowering therapies. OBJECTIVE Evaluating volumetric and structural connectivity correlates of the cUHDRS. METHODS One hundred and nineteen premanifest and 119 early-HD participants were included. Gray and white matter (WM) volumes were correlated with cUHDRS cross-sectionally and longitudinally using voxel-based morphometry. Correlations between baseline fractional anisotropy (FA); mean, radial, and axial diffusivity; and baseline cUHDRS were examined using tract-based spatial statistics. RESULTS Worse performance in the cUHDRS over time correlated with longitudinal volume decreases in the occipito-parietal cortex and centrum semiovale, whereas lower baseline scores correlated with decreased volume in the basal ganglia and surrounding WM. Lower cUHDRS scores were also associated with reduced FA and increased diffusivity at baseline. CONCLUSION The cUHDRS correlates with imaging biomarkers and tracks atrophy progression in HD supporting its biological relevance. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Carlos Estevez-Fraga
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Rachael I Scahill
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Alexandra Durr
- Sorbonne Université, Paris Brain Institute (ICM), AP-HP, Inserm, CNRS, Pitié-Salpêtrière University Hospital, Paris, France
| | - Blair R Leavitt
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Raymund A C Roos
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Geraint Rees
- Wellcome Centre for Neuroimaging, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Sarah Gregory
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Sarah J Tabrizi
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
13
|
Tan B, Shishegar R, Poudel GR, Fornito A, Georgiou-Karistianis N. Cortical morphometry and neural dysfunction in Huntington's disease: a review. Eur J Neurol 2020; 28:1406-1419. [PMID: 33210786 DOI: 10.1111/ene.14648] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/22/2020] [Accepted: 11/12/2020] [Indexed: 01/09/2023]
Abstract
Numerous neuroimaging techniques have been used to identify biomarkers of disease progression in Huntington's disease (HD). To date, the earliest and most sensitive of these is caudate volume; however, it is becoming increasingly evident that numerous changes to cortical structures, and their interconnected networks, occur throughout the course of the disease. The mechanisms by which atrophy spreads from the caudate to these cortical regions remains unknown. In this review, the neuroimaging literature specific to T1-weighted and diffusion-weighted magnetic resonance imaging is summarized and new strategies for the investigation of cortical morphometry and the network spread of degeneration in HD are proposed. This new avenue of research may enable further characterization of disease pathology and could add to a suite of biomarker/s of disease progression for patient stratification that will help guide future clinical trials.
Collapse
Affiliation(s)
- Brendan Tan
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia
| | - Rosita Shishegar
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia.,Australian e-Health Research Centre, CSIRO, Melbourne, VIC, Australia.,Monash Biomedical Imaging, Melbourne, VIC, Australia
| | - Govinda R Poudel
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia.,Sydney Imaging, Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia.,Australian Catholic University, Melbourne, VIC, Australia
| | - Alex Fornito
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia.,Monash Biomedical Imaging, Melbourne, VIC, Australia
| | - Nellie Georgiou-Karistianis
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
14
|
Dose-dependent volume loss in subcortical deep grey matter structures after cranial radiotherapy. Clin Transl Radiat Oncol 2020; 26:35-41. [PMID: 33294645 PMCID: PMC7691672 DOI: 10.1016/j.ctro.2020.11.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022] Open
Abstract
Subcortical grey matter is susceptible to dose-dependent volume loss after RT. Hippocampal age increases 1 year after radiotherapy, by a median of 11 years. We may need to reconsider current sparing strategies in RT for brain tumours. Future studies should examine the impact of deep GM volume loss on cognition.
Background and purpose The relation between radiotherapy (RT) dose to the brain and morphological changes in healthy tissue has seen recent increased interest. There already is evidence for changes in the cerebral cortex and white matter, as well as selected subcortical grey matter (GM) structures. We studied this relation in all deep GM structures, to help understand the aetiology of post-RT neurocognitive symptoms. Materials and methods We selected 31 patients treated with RT for grade II-IV glioma. Pre-RT and 1 year post-RT 3D T1-weighted MRIs were automatically segmented, and the changes in volume of the following structures were assessed: amygdala, nucleus accumbens, caudate nucleus, hippocampus, globus pallidus, putamen, and thalamus. The volumetric changes were related to the mean RT dose received by each structure. Hippocampal volumes were entered into a population-based nomogram to estimate hippocampal age. Results A significant relation between RT dose and volume loss was seen in all examined structures, except the caudate nucleus. The volume loss rates ranged from 0.16 to 1.37%/Gy, corresponding to 4.9–41.2% per 30 Gy. Hippocampal age, as derived from the nomogram, was seen to increase by a median of 11 years. Conclusion Almost all subcortical GM structures are susceptible to radiation-induced volume loss, with higher volume loss being observed with increasing dose. Volume loss of these structures is associated with neurological deterioration, including cognitive decline, in neurodegenerative diseases. To support a causal relationship between radiation-induced deep GM loss and neurocognitive functioning in glioma patients, future studies are needed that directly correlate volumetrics to clinical outcomes.
Collapse
Key Words
- Amygdala
- Brain neoplasms
- CAT12, computational anatomy toolbox 12
- CT, computed tomography
- Caudate nucleus
- FWER, family-wise error rate
- GM, grey matter
- Globus pallidus
- Gray matter
- Hippocampus
- MRI, magnetic resonance imaging
- Nucleus accumbens
- PALM, permutation analysis of linear models
- PTV, planning target volume
- Putamen
- RT, radiotherapy
- Radiotherapy
- SPM, statistical parametric mapping
- TFE, turbo fast echo
- Thalamus
- WBRT, whole-brain radiotherapy
Collapse
|
15
|
Palomar-Garcia A, Camara E. SeSBAT: Single Subject Brain Analysis Toolbox. Application to Huntington's Disease as a Preliminary Study. Front Syst Neurosci 2020; 14:488652. [PMID: 33117135 PMCID: PMC7550747 DOI: 10.3389/fnsys.2020.488652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 08/21/2020] [Indexed: 12/02/2022] Open
Abstract
Magnetic resonance imaging (MRI) biomarkers require complex processing routines that are time-consuming and labor-intensive for clinical users. The Single Subject Brain Analysis Toolbox (SeSBAT) is a fully automated MATLAB toolbox with a graphical user interface (GUI) that offers standardized and optimized protocols for the pre-processing and analysis of anatomical MRI data at the single-subject level. In this study, the two-fold strategy provided by SeSBAT is illustrated through its application on a cohort of 42 patients with Huntington’s disease (HD), in pre-manifest and early manifest stages, as a suitable model of neurodegenerative processes. On the one hand, hypothesis-driven analysis can be used to extract biomarkers of neurodegeneration in specific brain regions of interest (ROI-based analysis). On the other hand, an exploratory voxel-based morphometry (VBM) approach can detect volume changes due to neurodegeneration throughout the whole brain (whole-brain analysis). That illustration reveals the potential of SeSBAT in providing potential prognostic biomarkers in neurodegenerative processes in clinics, which could be critical to overcoming the limitations of current qualitative evaluation strategies, and thus improve the diagnosis and monitoring of neurodegenerative disorders. Furthermore, the importance of the availability of tools for characterization at the single-subject level has been emphasized, as there is high interindividual variability in the pattern of neurodegeneration. Thus, tools like SeSBAT could pave the way towards more effective and personalized medicine.
Collapse
Affiliation(s)
- Alicia Palomar-Garcia
- Cognition and Brain Plasticity Unit, IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), Barcelona, Spain
| | - Estela Camara
- Department of Cognition, Development and Educational Psychology, University of Barcelona, Barcelona, Spain
| |
Collapse
|
16
|
Caobi A, Dutta RK, Garbinski LD, Esteban-Lopez M, Ceyhan Y, Andre M, Manevski M, Ojha CR, Lapierre J, Tiwari S, Parira T, El-Hage N. The Impact of CRISPR-Cas9 on Age-related Disorders: From Pathology to Therapy. Aging Dis 2020; 11:895-915. [PMID: 32765953 PMCID: PMC7390517 DOI: 10.14336/ad.2019.0927] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 09/27/2019] [Indexed: 12/11/2022] Open
Abstract
With advances in medical technology, the number of people over the age of 60 is on the rise, and thus, increasing the prevalence of age-related pathologies within the aging population. Neurodegenerative disorders, cancers, metabolic and inflammatory diseases are some of the most prevalent age-related pathologies affecting the growing population. It is imperative that a new treatment to combat these pathologies be developed. Although, still in its infancy, the CRISPR-Cas9 system has become a potent gene-editing tool capable of correcting gene-mediated age-related pathology, and therefore ameliorating or eliminating disease symptoms. Deleting target genes using the CRISPR-Cas9 system or correcting for gene mutations may ameliorate many different neurodegenerative disorders detected in the aging population. Cancer cells targeted by the CRISPR-Cas9 system may result in an increased sensitivity to chemotherapeutics, lower proliferation, and higher cancer cell death. Finally, reducing gene targeting inflammatory molecules production through microRNA knockout holds promise as a therapeutic strategy for both arthritis and inflammation. Here we present a review based on how the expanding world of genome editing can be applied to disorders and diseases affecting the aging population.
Collapse
Affiliation(s)
- Allen Caobi
- 1Departments of Immunology and Nano-medicine
| | | | - Luis D Garbinski
- 3Cell Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Valdés Hernández MDC, Abu-Hussain J, Qiu X, Priller J, Parra Rodríguez M, Pino M, Báez S, Ibáñez A. Structural neuroimaging differentiates vulnerability from disease manifestation in colombian families with Huntington's disease. Brain Behav 2019; 9:e01343. [PMID: 31276317 PMCID: PMC6710228 DOI: 10.1002/brb3.1343] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 04/29/2019] [Accepted: 05/28/2019] [Indexed: 01/18/2023] Open
Abstract
INTRODUCTION The volume of the striatal structures has been associated with disease progression in individuals with Huntington's disease (HD) from North America, Europe, and Australia. However, it is not known whether the gray matter (GM) volume in the striatum is also sensitive in differentiating vulnerability from disease manifestation in HD families from a South-American region known to have high incidence of the disease. In addition, the association of enlarged brain perivascular spaces (PVS) with cognitive, behavioral, and motor symptoms of HD is unknown. MATERIALS AND METHODS We have analyzed neuroimaging indicators of global atrophy, PVS burden, and GM tissue volume in the basal ganglia and thalami, in relation to behavioral, motor, and cognitive scores, in 15 HD patients with overt disease manifestation and 14 first-degree relatives not genetically tested, which represent a vulnerable group, from the region of Magdalena, Colombia. RESULTS Poor fluid intelligence as per the Raven's Standard Progressive Matrices was associated with global brain atrophy (p = 0.002) and PVS burden (p ≤ 0.02) in HD patients, where the GM volume in all subcortical structures, with the exception of the right globus pallidus, was associated with motor or cognitive scores. Only the GM volume in the right putamen was associated with envy and MOCA scores (p = 0.008 and 0.015 respectively) in first-degree relatives. CONCLUSION Striatal GM volume, global brain atrophy and PVS burden may serve as differential indicators of disease manifestation in HD. The Raven's Standard Progressive Matrices could be a cognitive test worth to consider in the differentiation of vulnerability versus overt disease in HD.
Collapse
Affiliation(s)
- Maria Del C Valdés Hernández
- Department of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Janna Abu-Hussain
- College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Xinyi Qiu
- Glan Clwyd Hospital, North Wales, UK
| | - Josef Priller
- Dementia Research Institute, University of Edinburgh, Edinburgh, UK.,Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Mario Parra Rodríguez
- School of Psychological Sciences and Health, Strathclyde University, Glasgow, UK.,Department of Psychology, Universidad Autónoma del Caribe, Barranquilla, Colombia
| | - Mariana Pino
- Department of Psychology, Universidad Autónoma del Caribe, Barranquilla, Colombia
| | - Sandra Báez
- Department of Psychology, Universidad de Los Andes, Bogotá, Colombia
| | - Agustín Ibáñez
- Department of Psychology, Universidad Autónoma del Caribe, Barranquilla, Colombia.,Institute of Cognitive and Translational Neuroscience (INCYT), INECO Foundation, Favaloro University, Buenos Aires, Argentina.,National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina.,Centre of Excellence in Cognition and its Disorders, Australian Research Council (ARC), Sydney, NSW, Australia.,Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibáñez, Santiago, Chile
| |
Collapse
|
18
|
Banez-Coronel M, Ranum LPW. Repeat-associated non-AUG (RAN) translation: insights from pathology. J Transl Med 2019; 99:929-942. [PMID: 30918326 PMCID: PMC7219275 DOI: 10.1038/s41374-019-0241-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 01/29/2019] [Indexed: 12/14/2022] Open
Abstract
More than 40 different neurological diseases are caused by microsatellite repeat expansions. Since the discovery of repeat-associated non-AUG (RAN) translation by Zu et al. in 2011, nine expansion disorders have been identified as RAN-positive diseases. RAN proteins are translated from different types of nucleotide repeat expansions and can be produced from both sense and antisense transcripts. In some diseases, RAN proteins have been shown to accumulate in affected brain regions. Here we review the pathological and molecular aspects associated with RAN protein accumulation for each particular disorder, the correlation between disease pathology and the available in vivo models and the common aspects shared by some of the newly discovered RAN proteins.
Collapse
Affiliation(s)
- Monica Banez-Coronel
- Center for NeuroGenetics, University of Florida, Gainesville, FL, 32610, USA
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, 32610, USA
| | - Laura P W Ranum
- Center for NeuroGenetics, University of Florida, Gainesville, FL, 32610, USA.
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, 32610, USA.
- Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA.
- Genetics Institute, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
19
|
Garcia-Gorro C, Llera A, Martinez-Horta S, Perez-Perez J, Kulisevsky J, Rodriguez-Dechicha N, Vaquer I, Subira S, Calopa M, Muñoz E, Santacruz P, Ruiz-Idiago J, Mareca C, Beckmann CF, de Diego-Balaguer R, Camara E. Specific patterns of brain alterations underlie distinct clinical profiles in Huntington's disease. Neuroimage Clin 2019; 23:101900. [PMID: 31255947 PMCID: PMC6606833 DOI: 10.1016/j.nicl.2019.101900] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/11/2019] [Accepted: 06/13/2019] [Indexed: 12/16/2022]
Abstract
Huntington's disease (HD) is a genetic neurodegenerative disease which involves a triad of motor, cognitive and psychiatric disturbances. However, there is great variability in the prominence of each type of symptom across individuals. The neurobiological basis of such variability remains poorly understood but would be crucial for better tailored treatments. Multivariate multimodal neuroimaging approaches have been successful in disentangling these profiles in other disorders. Thus we applied for the first time such approach to HD. We studied the relationship between HD symptom domains and multimodal measures sensitive to grey and white matter structural alterations. Forty-three HD gene carriers (23 manifest and 20 premanifest individuals) were scanned and underwent behavioural assessments evaluating motor, cognitive and psychiatric domains. We conducted a multimodal analysis integrating different structural neuroimaging modalities measuring grey matter volume, cortical thickness and white matter diffusion indices - fractional anisotropy and radial diffusivity. All neuroimaging measures were entered into a linked independent component analysis in order to obtain multimodal components reflecting common inter-subject variation across imaging modalities. The relationship between multimodal neuroimaging independent components and behavioural measures was analysed using multiple linear regression. We found that cognitive and motor symptoms shared a common neurobiological basis, whereas the psychiatric domain presented a differentiated neural signature. Behavioural measures of different symptom domains correlated with different neuroimaging components, both the brain regions involved and the neuroimaging modalities most prominently associated with each type of symptom showing differences. More severe cognitive and motor signs together were associated with a multimodal component consisting in a pattern of reduced grey matter, cortical thickness and white matter integrity in cognitive and motor related networks. In contrast, depressive symptoms were associated with a component mainly characterised by reduced cortical thickness pattern in limbic and paralimbic regions. In conclusion, using a multivariate multimodal approach we were able to disentangle the neurobiological substrates of two distinct symptom profiles in HD: one characterised by cognitive and motor features dissociated from a psychiatric profile. These results open a new view on a disease classically considered as a uniform entity and initiates a new avenue for further research considering these qualitative individual differences.
Collapse
Affiliation(s)
- Clara Garcia-Gorro
- Cognition and Brain Plasticity Unit, L'Hospitalet de Llobregat (Barcelona), IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), Spain
- Department of Cognition, Development and Educational Psychology, University of Barcelona, Barcelona, Spain
| | - Alberto Llera
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Radboud University, Nijmegen, the Netherlands
| | - Saul Martinez-Horta
- Movement Disorders Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain
- CIBERNED (Center for Networked Biomedical Research on Neurodegenerative Diseases), Carlos III Institute, Madrid, Spain
| | - Jesus Perez-Perez
- Movement Disorders Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain
- CIBERNED (Center for Networked Biomedical Research on Neurodegenerative Diseases), Carlos III Institute, Madrid, Spain
| | - Jaime Kulisevsky
- Movement Disorders Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain
- CIBERNED (Center for Networked Biomedical Research on Neurodegenerative Diseases), Carlos III Institute, Madrid, Spain
- Universidad Autónoma de Barcelona, Barcelona, Spain
| | | | - Irene Vaquer
- Hestia Duran i Reynals, Hospital Duran i Reynals, Hospitalet de Llobregat (Barcelona), Spain
| | - Susana Subira
- Hestia Duran i Reynals, Hospital Duran i Reynals, Hospitalet de Llobregat (Barcelona), Spain
- Department of Clinical and Health Psychology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Matilde Calopa
- Movement Disorders Unit, Neurology Service, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Esteban Muñoz
- Movement Disorders Unit, Neurology Service, Hospital Clínic, Barcelona, Spain
- IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Barcelona, Spain
- Facultat de Medicina, University of Barcelona, Barcelona, Spain
| | - Pilar Santacruz
- Movement Disorders Unit, Neurology Service, Hospital Clínic, Barcelona, Spain
| | - Jesus Ruiz-Idiago
- Hospital Mare de Deu de la Mercè, Barcelona, Spain
- Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Celia Mareca
- Hospital Mare de Deu de la Mercè, Barcelona, Spain
| | - Christian F. Beckmann
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Radboud University, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ruth de Diego-Balaguer
- Cognition and Brain Plasticity Unit, L'Hospitalet de Llobregat (Barcelona), IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), Spain
- Department of Cognition, Development and Educational Psychology, University of Barcelona, Barcelona, Spain
- The Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- ICREA (Catalan Institute for Research and Advanced Studies), Barcelona, Spain
| | - Estela Camara
- Cognition and Brain Plasticity Unit, L'Hospitalet de Llobregat (Barcelona), IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), Spain
| |
Collapse
|
20
|
Zeun P, Scahill RI, Tabrizi SJ, Wild EJ. Fluid and imaging biomarkers for Huntington's disease. Mol Cell Neurosci 2019; 97:67-80. [PMID: 30807825 DOI: 10.1016/j.mcn.2019.02.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/25/2019] [Accepted: 02/12/2019] [Indexed: 01/18/2023] Open
Abstract
Huntington's disease is a chronic progressive neurodegenerative condition for which there is no disease-modifying treatment. The known genetic cause of Huntington's disease makes it possible to identify individuals destined to develop the disease and instigate treatments before the onset of symptoms. Multiple trials are already underway that target the cause of HD, yet clinical measures are often insensitive to change over typical clinical trial duration. Robust biomarkers of drug target engagement, disease severity and progression are required to evaluate the efficacy of treatments and concerted efforts are underway to achieve this. Biofluid biomarkers have potential advantages of direct quantification of biological processes at the molecular level, whilst imaging biomarkers can quantify related changes at a structural level in the brain. The most robust biofluid and imaging biomarkers can offer complementary information, providing a more comprehensive evaluation of disease stage and progression to inform clinical trial design and endpoints.
Collapse
Affiliation(s)
- Paul Zeun
- Huntington's Disease Centre, University College London (UCL) Institute of Neurology, London WC1N 3BG, United Kingdom.
| | - Rachael I Scahill
- Huntington's Disease Centre, University College London (UCL) Institute of Neurology, London WC1N 3BG, United Kingdom.
| | - Sarah J Tabrizi
- Huntington's Disease Centre, University College London (UCL) Institute of Neurology, London WC1N 3BG, United Kingdom.
| | - Edward J Wild
- Huntington's Disease Centre, University College London (UCL) Institute of Neurology, London WC1N 3BG, United Kingdom.
| |
Collapse
|
21
|
Ciarochi JA, Johnson HJ, Calhoun VD, Liu J, Espinoza FA, Bockholt HJ, Misiura M, Caprihan A, Plis S, Paulsen JS, Turner JA. Concurrent Cross-Sectional and Longitudinal Analyses of Multivariate White Matter Profiles and Clinical Functioning in Pre-Diagnosis Huntington Disease. J Huntingtons Dis 2019; 8:199-219. [PMID: 30932891 DOI: 10.3233/jhd-180332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Gray matter (GM) atrophy in the striatum and across the brain is a consistently reported feature of the Huntington Disease (HD) prodrome. More recently, widespread prodromal white matter (WM) degradation has also been detected. However, longitudinal WM studies are limited and conflicting, and most analyses comparing WM and clinical functioning have also been cross-sectional. OBJECTIVE We simultaneously assessed changes in WM and cognitive and motor functioning at various prodromal HD stages. METHODS Data from 1,336 (1,047 prodromal, 289 control) PREDICT-HD participants were analyzed (3,700 sessions). MRI images were used to create GM, WM, and cerebrospinal fluid probability maps. Using source-based morphometry, independent component analysis was applied to WM probability maps to extract covarying spatial patterns and their subject profiles. WM profiles were analyzed in two sets of linear mixed model (LMM) analyses: one to compare WM profiles across groups cross-sectionally and longitudinally, and one to concurrently compare WM profiles and clinical variables cross-sectionally and longitudinally within each group. RESULTS Findings illustrate widespread prodromal changes in GM-adjacent-WM, with premotor, supplementary motor, middle frontal and striatal changes early in the prodrome that subsequently extend sub-gyrally with progression. Motor functioning agreed most with WM until the near-onset prodromal stage, when Stroop interference was the best WM indicator. Across groups, Trail-Making Test part A outperformed other cognitive variables in its similarity to WM, particularly cross-sectionally. CONCLUSIONS Results suggest that distinct regions coincide with cognitive compared to motor functioning. Furthermore, at different prodromal stages, distinct regions appear to align best with clinical functioning. Thus, the informativeness of clinical measures may vary according to the type of data available (cross-sectional or longitudinal) as well as age and CAG-number.
Collapse
Affiliation(s)
| | - Hans J Johnson
- Department of Electrical and Computer Engineering, 1402 Seamans Center for the Engineering Arts and Science, The University of Iowa, Iowa City, IA, USA
| | - Vince D Calhoun
- The Mind Research Network, Albuquerque, NM, USA
- Department of Electrical and Computer Engineering, University of New Mexico, Albuquerque, NM, USA
| | - Jingyu Liu
- The Mind Research Network, Albuquerque, NM, USA
| | | | | | - Maria Misiura
- Department of Psychology, Georgia State University, Atlanta, GA, USA
| | | | - Sergey Plis
- The Mind Research Network, Albuquerque, NM, USA
| | - Jane S Paulsen
- Department of Psychiatry, Iowa Mental Health Clinical Research Center, University of Iowa, IA, USA
- Departments of Neurology and Psychology, University of Iowa, IA, USA
| | - Jessica A Turner
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
- Department of Psychology, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
22
|
Structural Magnetic Resonance Imaging in Huntington's Disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2018; 142:335-380. [PMID: 30409258 DOI: 10.1016/bs.irn.2018.09.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder, caused by expansion of the CAG repeat in the huntingtin gene. HD is characterized clinically by progressive motor, cognitive and neuropsychiatric symptoms. There are currently no disease modifying treatments available for HD, and there is a great need for biomarkers to monitor disease progression and identify new targets for therapeutic intervention. Neuroimaging techniques provide a powerful tool for assessing disease pathology and progression in premanifest stages, before the onset of overt motor symptoms. Structural magnetic resonance imaging (MRI) is non-invasive imaging techniques which have been employed to study structural and microstructural changes in premanifest and manifest HD gene carriers. This chapter described structural imaging techniques and analysis methods employed across HD MRI studies. Current evidence for structural MRI abnormalities in HD, and associations between atrophy, structural white matter changes, iron deposition and clinical performance are discussed; together with the use of structural MRI measures as a diagnostic tool, to assess longitudinal changes, and as potential biomarkers and endpoints for clinical trials.
Collapse
|
23
|
Kloos AD, Kegelmeyer DA, Fritz NE, Daley AM, Young GS, Kostyk SK. Cognitive Dysfunction Contributes to Mobility Impairments in Huntington's Disease. J Huntingtons Dis 2018; 6:363-370. [PMID: 29254103 PMCID: PMC5757646 DOI: 10.3233/jhd-170279] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background: Huntington’s disease (HD) is a progressive neurodegenerative disorder that results in a gradual decline in mobility and balance. Increasing evidence has documented an important role of executive function in the safe ambulation of the elderly and people with a variety of neurological disorders. Little is known about the contribution of cognitive deficits to decline in mobility over time in HD. Objective: This study examined the relationships of mobility, motor and cognitive function measures at baseline, and of mobility and cognitive measures over four years. Methods: A retrospective chart review was performed on 70 patients with genetically confirmed HD (age 20–75 years old) across 121 HD clinic visits. Correlations between Unified Huntington’s Disease Rating Scale – Total Motor, Tinetti Mobility Test (TMT), and cognitive measures (Letter Verbal Fluency, Symbol Digit Modalities Test (SDMT), and Stroop Test) were analyzed. Longitudinal relationships between TMT and cognitive measures were examined using mixed effect regression models. Results: Gait and balance measures representing domains of mobility (TMT scores) were significantly correlated with each of the cognitive measures with the exception of the Verbal Fluency score. Mixed effects regression modeling showed that the Stroop Interference sub-test and SDMT were significant predictors (p-values <0.01) of TMT total scores. Conclusions: Impairments in executive function measures correlate highly with measures of gait, balance and mobility in individuals with HD. Interventions designed to improve mobility and decrease fall risk should also address issues of cognitive impairments with particular consideration given to interventions that may focus on motor-cognitive dual task training.
Collapse
Affiliation(s)
- Anne D Kloos
- Division of Physical Therapy, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Deb A Kegelmeyer
- Division of Physical Therapy, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Nora E Fritz
- Division of Physical Therapy, College of Medicine, The Ohio State University, Columbus, OH, USA.,Department of Neurology, Program in Physical Therapy, College of Pharmacy and Health Sciences, College of Medicine, Wayne State University, Detroit, MI, USA
| | - Allison M Daley
- Department of Neurology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Gregory S Young
- Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | - Sandra K Kostyk
- Department of Neurology, College of Medicine, The Ohio State University, Columbus, OH, USA.,Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
24
|
Podvin S, Reardon HT, Yin K, Mosier C, Hook V. Multiple clinical features of Huntington's disease correlate with mutant HTT gene CAG repeat lengths and neurodegeneration. J Neurol 2018; 266:551-564. [PMID: 29956026 DOI: 10.1007/s00415-018-8940-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/06/2018] [Accepted: 06/07/2018] [Indexed: 12/28/2022]
Abstract
Huntington's disease (HD) is a fatal neurodegenerative disease caused by mutant HTT gene expansions of CAG triplet repeat numbers that are inherited in an autosomal dominant manner. HD patients display multiple clinical features that are correlated with HTT CAG repeat numbers that include age of disease onset, motor dysfunction, cognitive deficits, compromised daily living capacity, and brain neurodegeneration. It is important to understand the significant relationships of the multiple HD clinical deficits correlated with the number of mutant HTT CAG expansions that are the genetic basis for HD disabilities. Therefore, this review highlights the significant correlations of the HD clinical features of age of onset, motor and cognitive disabilities, decline in living capabilities, weight loss, risk of death, and brain neurodegeneration with respect to their associations with CAG repeat lengths of the HTT gene. Quantitative HTT gene expression patterns analyzed in normal adult human brain regions demonstrated its distribution in areas known to undergo neurodegeneration in HD, as well as in other brain regions. Future investigation of the relationships of the spectrum of clinical HD features with mutant HTT molecular mechanisms will be important to gain understanding of how mutant CAG expansions of the HTT gene result in the devastating disabilities of HD patients.
Collapse
Affiliation(s)
- Sonia Podvin
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, 9500 Gilman Drive, MC0719, La Jolla, San Diego, CA, 92093-0719, USA
| | - Holly T Reardon
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, 9500 Gilman Drive, MC0719, La Jolla, San Diego, CA, 92093-0719, USA
| | - Katrina Yin
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, 9500 Gilman Drive, MC0719, La Jolla, San Diego, CA, 92093-0719, USA
| | - Charles Mosier
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, 9500 Gilman Drive, MC0719, La Jolla, San Diego, CA, 92093-0719, USA
| | - Vivian Hook
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, 9500 Gilman Drive, MC0719, La Jolla, San Diego, CA, 92093-0719, USA.
- Department of Neurosciences, University of California, 9500 Gilman Drive, MC0719, La Jolla, San Diego, CA, 92093-0719, USA.
- Department of Pharmacology, University of California, 9500 Gilman Drive, MC0719, La Jolla, San Diego, CA, 92093-0719, USA.
| |
Collapse
|
25
|
|
26
|
Liu J, Ciarochi J, Calhoun VD, Paulsen JS, Bockholt HJ, Johnson HJ, Long JD, Lin D, Espinoza FA, Misiura MB, Caprihan A, Turner JA. Genetics Modulate Gray Matter Variation Beyond Disease Burden in Prodromal Huntington's Disease. Front Neurol 2018; 9:190. [PMID: 29651271 PMCID: PMC5884935 DOI: 10.3389/fneur.2018.00190] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 03/12/2018] [Indexed: 12/13/2022] Open
Abstract
Huntington’s disease (HD) is a neurodegenerative disorder caused by an expansion mutation of the cytosine–adenine–guanine (CAG) trinucleotide in the HTT gene. Decline in cognitive and motor functioning during the prodromal phase has been reported, and understanding genetic influences on prodromal disease progression beyond CAG will benefit intervention therapies. From a prodromal HD cohort (N = 715), we extracted gray matter (GM) components through independent component analysis and tested them for associations with cognitive and motor functioning that cannot be accounted for by CAG-induced disease burden (cumulative effects of CAG expansion and age). Furthermore, we examined genetic associations (at the genomic, HD pathway, and candidate region levels) with the GM components that were related to functional decline. After accounting for disease burden, GM in a component containing cuneus, lingual, and middle occipital regions was positively associated with attention and working memory performance, and the effect size was about a tenth of that of disease burden. Prodromal participants with at least one dystonia sign also had significantly lower GM volume in a bilateral inferior parietal component than participants without dystonia, after controlling for the disease burden. Two single-nucleotide polymorphisms (SNPs: rs71358386 in NCOR1 and rs71358386 in ADORA2B) in the HD pathway were significantly associated with GM volume in the cuneus component, with minor alleles being linked to reduced GM volume. Additionally, homozygous minor allele carriers of SNPs in a candidate region of ch15q13.3 had significantly higher GM volume in the inferior parietal component, and one minor allele copy was associated with a total motor score decrease of 0.14 U. Our findings depict an early genetical GM reduction in prodromal HD that occurs irrespective of disease burden and affects regions important for cognitive and motor functioning.
Collapse
Affiliation(s)
- Jingyu Liu
- The Mind Research Network & Lovelace Biomedical and Environmental Research Institute (LBERI), Albuquerque, NM, United States.,Department of Electrical and Computer Engineering, University of New Mexico, Albuquerque, NM, United States
| | - Jennifer Ciarochi
- Department of Psychology, Georgia State University, Atlanta, GA, United States.,Department of Neuroscience, Georgia State University, Atlanta, GA, United States
| | - Vince D Calhoun
- The Mind Research Network & Lovelace Biomedical and Environmental Research Institute (LBERI), Albuquerque, NM, United States.,Department of Electrical and Computer Engineering, University of New Mexico, Albuquerque, NM, United States
| | - Jane S Paulsen
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States.,Department of Neurology, University of Iowa, Iowa City, IA, United States.,Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, United States
| | - H Jeremy Bockholt
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States.,Department of Neurology, University of Iowa, Iowa City, IA, United States.,Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, United States
| | - Hans J Johnson
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States.,Department of Electrical and Computer Engineering, University of Iowa, Iowa City, IA, United States
| | - Jeffrey D Long
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States.,Department of Biostatistics, University of Iowa, Iowa City, IA, United States
| | - Dongdong Lin
- The Mind Research Network & Lovelace Biomedical and Environmental Research Institute (LBERI), Albuquerque, NM, United States
| | - Flor A Espinoza
- The Mind Research Network & Lovelace Biomedical and Environmental Research Institute (LBERI), Albuquerque, NM, United States
| | - Maria B Misiura
- Department of Psychology, Georgia State University, Atlanta, GA, United States.,Department of Neuroscience, Georgia State University, Atlanta, GA, United States
| | - Arvind Caprihan
- The Mind Research Network & Lovelace Biomedical and Environmental Research Institute (LBERI), Albuquerque, NM, United States
| | - Jessica A Turner
- The Mind Research Network & Lovelace Biomedical and Environmental Research Institute (LBERI), Albuquerque, NM, United States.,Department of Psychology, Georgia State University, Atlanta, GA, United States.,Department of Neuroscience, Georgia State University, Atlanta, GA, United States
| | | |
Collapse
|
27
|
Langfelder P, Gao F, Wang N, Howland D, Kwak S, Vogt TF, Aaronson JS, Rosinski J, Coppola G, Horvath S, Yang XW. MicroRNA signatures of endogenous Huntingtin CAG repeat expansion in mice. PLoS One 2018; 13:e0190550. [PMID: 29324753 PMCID: PMC5764268 DOI: 10.1371/journal.pone.0190550] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 12/15/2017] [Indexed: 12/16/2022] Open
Abstract
In Huntington's disease (HD) patients and in model organisms, messenger RNA transcriptome has been extensively studied; in contrast, comparatively little is known about expression and potential role of microRNAs. Using RNA-sequencing, we have quantified microRNA expression in four brain regions and liver, at three different ages, from an allelic series of HD model mice with increasing CAG length in the endogenous Huntingtin gene. Our analyses reveal CAG length-dependent microRNA expression changes in brain, with 159 microRNAs selectively altered in striatum, 102 in cerebellum, 51 in hippocampus, and 45 in cortex. In contrast, a progressive CAG length-dependent microRNA dysregulation was not observed in liver. We further identify microRNAs whose transcriptomic response to CAG length expansion differs significantly among the brain regions and validate our findings in data from a second, independent cohort of mice. Using existing mRNA expression data from the same animals, we assess the possible relationships between microRNA and mRNA expression and highlight candidate microRNAs that are negatively correlated with, and whose predicted targets are enriched in, CAG-length dependent mRNA modules. Several of our top microRNAs (Mir212/Mir132, Mir218, Mir128 and others) have been previously associated with aspects of neuronal development and survival. This study provides an extensive resource for CAG length-dependent changes in microRNA expression in disease-vulnerable and -resistant brain regions in HD mice, and provides new insights for further investigation of microRNAs in HD pathogenesis and therapeutics.
Collapse
Affiliation(s)
- Peter Langfelder
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience & Human Behavior, University of California Los Angeles (UCLA), Los Angeles, CA, United States of America
| | - Fuying Gao
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience & Human Behavior, University of California Los Angeles (UCLA), Los Angeles, CA, United States of America
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Nan Wang
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience & Human Behavior, University of California Los Angeles (UCLA), Los Angeles, CA, United States of America
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
- UCLA Brain Research Institute, Los Angeles, CA, United States of America
| | - David Howland
- CHDI Foundation/CHDI Management Inc., Princeton, NJ, United States of America
| | - Seung Kwak
- CHDI Foundation/CHDI Management Inc., Princeton, NJ, United States of America
| | - Thomas F. Vogt
- CHDI Foundation/CHDI Management Inc., Princeton, NJ, United States of America
| | - Jeffrey S. Aaronson
- CHDI Foundation/CHDI Management Inc., Princeton, NJ, United States of America
| | - Jim Rosinski
- CHDI Foundation/CHDI Management Inc., Princeton, NJ, United States of America
| | - Giovanni Coppola
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience & Human Behavior, University of California Los Angeles (UCLA), Los Angeles, CA, United States of America
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
- UCLA Brain Research Institute, Los Angeles, CA, United States of America
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
- Department of Biostatistics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - X. William Yang
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience & Human Behavior, University of California Los Angeles (UCLA), Los Angeles, CA, United States of America
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
- UCLA Brain Research Institute, Los Angeles, CA, United States of America
| |
Collapse
|
28
|
Faulty neuronal determination and cell polarization are reverted by modulating HD early phenotypes. Proc Natl Acad Sci U S A 2018; 115:E762-E771. [PMID: 29311338 DOI: 10.1073/pnas.1715865115] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Increasing evidence suggests that early neurodevelopmental defects in Huntington's disease (HD) patients could contribute to the later adult neurodegenerative phenotype. Here, by using HD-derived induced pluripotent stem cell lines, we report that early telencephalic induction and late neural identity are affected in cortical and striatal populations. We show that a large CAG expansion causes complete failure of the neuro-ectodermal acquisition, while cells carrying shorter CAGs repeats show gross abnormalities in neural rosette formation as well as disrupted cytoarchitecture in cortical organoids. Gene-expression analysis showed that control organoid overlapped with mature human fetal cortical areas, while HD organoids correlated with the immature ventricular zone/subventricular zone. We also report that defects in neuroectoderm and rosette formation could be rescued by molecular and pharmacological approaches leading to a recovery of striatal identity. These results show that mutant huntingtin precludes normal neuronal fate acquisition and highlights a possible connection between mutant huntingtin and abnormal neural development in HD.
Collapse
|
29
|
Grey matter volume loss is associated with specific clinical motor signs in Huntington's disease. Parkinsonism Relat Disord 2018; 46:56-61. [DOI: 10.1016/j.parkreldis.2017.11.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 09/12/2017] [Accepted: 11/01/2017] [Indexed: 11/15/2022]
|
30
|
Downing NR, Lourens S, De Soriano I, Long JD, Paulsen JS. Phenotype Characterization of HD Intermediate Alleles in PREDICT-HD. J Huntingtons Dis 2017; 5:357-368. [PMID: 27983559 DOI: 10.3233/jhd-160185] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Huntington disease (HD) is a neurodegenerative disease caused by a CAG repeat expansion on chromosome 4. Pathology is associated with CAG repeat length. Prior studies examining people in the intermediate allele (IA) range found subtle differences in motor, cognitive, and behavioral domains compared to controls. OBJECTIVE The purpose of this study was to examine baseline and longitudinal differences in motor, cognitive, behavioral, functional, and imaging outcomes between persons with CAG repeats in three ranges: normal (≤26), intermediate (27-35), and reduced penetrance (36-39). METHODS We examined longitudinal data from 389 participants in three allele groups: 280 normal controls (NC), 21 intermediate allele [IA], and 88 reduced penetrance [RP]. We used linear mixed models to identify differences in baseline and longitudinal outcomes between groups. Three models were tested: 1) no baseline or longitudinal differences; 2) baseline differences but no longitudinal differences; and 3) baseline and longitudinal differences. RESULTS Model 1 was the best fitting model for most outcome variables. Models 2 and 3 were best fitting for some of the variables. We found baseline and longitudinal trends of declining performance across increasing CAG repeat length groups, but no significant differences between the NC and IA groups. CONCLUSION We did not find evidence to support differences in the IA group compared to the NC group. These findings are limited by a small IA sample size.
Collapse
Affiliation(s)
| | - Spencer Lourens
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Isabella De Soriano
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Jeffrey D Long
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Department of Biostatistics, College of Public Health, The University of Iowa, Iowa City, IA, USA
| | - Jane S Paulsen
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Department of Neurology, Carver College of Medicine, The University of Iowa, Iowa City, IA, USA.,Department of Psychological and Brain Sciences, The University of Iowa, Iowa City, IA, USA
| | | |
Collapse
|
31
|
Abstract
OBJECTIVE Psychomotor slowing is a common cognitive complication in type 1 diabetes (T1D), but its neuroanatomical correlates and risk factors are unclear. In nondiabetic adults, smaller gray matter volume (GMV) and presence of white matter hyperintensities are associated with psychomotor slowing. We hypothesize that smaller GMV in prefronto-parietal regions explains T1D-related psychomotor slowing. We also inspect the contribution of microvascular disease and hyperglycemia. METHODS GMV, white matter hyperintensities (WMH), and glucose levels were measured concurrently with a test of psychomotor speed (Digit Symbol Substitution Test [DSST]) in 95 adults with childhood-onset T1D (mean age/duration = 49/41 years) and 135 similarly aged non-T1D adults. Linear regression models tested associations between DSST and regional GMV, controlling for T1D, sex, and education; a bootstrapping method tested whether regional GMV explained between-group differences in DSST. For the T1D cohort, voxel-based and a priori regions-of-interest methods further tested associations between GMV and DSST, adjusting for WMH, hyperglycemia, and age. RESULTS Bilateral putamen, but no other regions examined, significantly attenuated DSST differences between the cohorts (bootstrapped unstandardized indirect effects: -3.49, -3.26; 95% confidence interval = -5.49 to -1.80, -5.29 to -1.44, left and right putamen, respectively). Among T1D, DSST was positively associated with GMV of bilateral putamen and left thalamus. Neither WMH, hyperglycemia, age, nor other factors substantially modified these relationships. CONCLUSIONS For middle-aged adults with T1D and cerebral microvascular disease, GMV of basal ganglia may play a critical role in regulating psychomotor speed, as measured via DSST. Studies to quantify the impact of basal ganglia atrophy concurrent with WMH progression on psychomotor slowing are warranted.
Collapse
|
32
|
Neuroimaging as a tool to study the sources of phenotypic heterogeneity in Huntington's disease. Curr Opin Neurol 2017; 30:398-404. [PMID: 28509681 DOI: 10.1097/wco.0000000000000461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Huntington's disease is a neurodegenerative disorder characterized by a triad of motor, cognitive and psychiatric disturbances. There is great variability regarding the prominence and evolution of each type of clinical sign. One possible source of phenotypic heterogeneity could be the more prominent degeneration of specific brain circuits. The scope of this review is to highlight the most recent neuroimaging studies that have analysed the relationship between brain changes and motor, cognitive and psychiatric alterations in Huntington's disease. RECENT FINDINGS The results from recent neuroimaging studies are heterogeneous. Although there is a great overlap between the different regions associated with each symptomatic domain, there is some degree of differentiation. For example, the motor network is associated with motor impairment, whereas the ventral striatum is especially involved in emotional deficits related with psychiatric problems. SUMMARY Motor, cognitive and psychiatric impairments are associated with structural and functional brain biomarkers. However, the specificity of the regions involved remains unknown, because these studies focused on specific regions and symptoms. In order to tease apart the neural substrates that underlie the phenotypic heterogeneity in Huntington's disease, multivariate approaches combining brain and behavioural measures related to all symptomatic domains should be considered in the future.
Collapse
|
33
|
Ma Q, Yang J, Milner TA, Vonsattel JPG, Palko ME, Tessarollo L, Hempstead BL. SorCS2-mediated NR2A trafficking regulates motor deficits in Huntington's disease. JCI Insight 2017; 2:88995. [PMID: 28469074 PMCID: PMC5414556 DOI: 10.1172/jci.insight.88995] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 03/30/2017] [Indexed: 12/14/2022] Open
Abstract
Motor dysfunction is a prominent and disabling feature of Huntington's disease (HD), but the molecular mechanisms that dictate its onset and progression are unknown. The N-methyl-D-aspartate receptor 2A (NR2A) subunit regulates motor skill development and synaptic plasticity in medium spiny neurons (MSNs) of the striatum, cells that are most severely impacted by HD. Here, we document reduced NR2A receptor subunits on the dendritic membranes and at the synapses of MSNs in zQ175 mice that model HD. We identify that SorCS2, a vacuolar protein sorting 10 protein-domain (VPS10P-domain) receptor, interacts with VPS35, a core component of retromer, thereby regulating surface trafficking of NR2A in MSNs. In the zQ175 striatum, SorCS2 is markedly decreased in an age- and allele-dependent manner. Notably, SorCS2 selectively interacts with mutant huntingtin (mtHTT), but not WT huntingtin (wtHTT), and is mislocalized to perinuclear clusters in striatal neurons of human HD patients and zQ175 mice. Genetic deficiency of SorCS2 accelerates the onset and exacerbates the motor coordination deficit of zQ175 mice. Together, our results identify SorCS2 as an interacting protein of mtHTT and demonstrate that impaired SorCS2-mediated NR2A subunit trafficking to dendritic surface of MSNs is, to our knowledge, a novel mechanism contributing to motor coordination deficits of HD.
Collapse
Affiliation(s)
- Qian Ma
- Graduate Program of Neuroscience
| | - Jianmin Yang
- Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Teresa A. Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York, USA
| | - Jean-Paul G. Vonsattel
- The New York Brain Bank/Taub Institute Columbia University, Children’s Hospital, New York, New York, USA
| | - Mary Ellen Palko
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, Maryland, USA
| | - Lino Tessarollo
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, Maryland, USA
| | - Barbara L. Hempstead
- Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
34
|
Developmental alterations in Huntington's disease neural cells and pharmacological rescue in cells and mice. Nat Neurosci 2017; 20:648-660. [PMID: 28319609 PMCID: PMC5610046 DOI: 10.1038/nn.4532] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 02/23/2017] [Indexed: 12/11/2022]
Abstract
Neural cultures derived from Huntington's disease (HD) patient-derived induced pluripotent stem cells were used for 'omics' analyses to identify mechanisms underlying neurodegeneration. RNA-seq analysis identified genes in glutamate and GABA signaling, axonal guidance and calcium influx whose expression was decreased in HD cultures. One-third of gene changes were in pathways regulating neuronal development and maturation. When mapped to stages of mouse striatal development, the profiles aligned with earlier embryonic stages of neuronal differentiation. We observed a strong correlation between HD-related histone marks, gene expression and unique peak profiles associated with dysregulated genes, suggesting a coordinated epigenetic program. Treatment with isoxazole-9, which targets key dysregulated pathways, led to amelioration of expanded polyglutamine repeat-associated phenotypes in neural cells and of cognitive impairment and synaptic pathology in HD model R6/2 mice. These data suggest that mutant huntingtin impairs neurodevelopmental pathways that could disrupt synaptic homeostasis and increase vulnerability to the pathologic consequence of expanded polyglutamine repeats over time.
Collapse
|
35
|
Egocentric and allocentric visuospatial working memory in premotor Huntington's disease: A double dissociation with caudate and hippocampal volumes. Neuropsychologia 2017; 101:57-64. [PMID: 28427989 DOI: 10.1016/j.neuropsychologia.2017.04.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 03/21/2017] [Accepted: 04/15/2017] [Indexed: 12/13/2022]
Abstract
Our brains represent spatial information in egocentric (self-based) or allocentric (landmark-based) coordinates. Rodent studies have demonstrated a critical role for the caudate in egocentric navigation and the hippocampus in allocentric navigation. We administered tests of egocentric and allocentric working memory to individuals with premotor Huntington's disease (pmHD), which is associated with early caudate nucleus atrophy, and controls. Each test had 80 trials during which subjects were asked to remember 2 locations over 1-sec delays. The only difference between these otherwise identical tests was that locations could only be coded in self-based or landmark-based coordinates. We applied a multiatlas-based segmentation algorithm and computed point-wise Jacobian determinants to measure regional variations in caudate and hippocampal volumes from 3T MRI. As predicted, the pmHD patients were significantly more impaired on egocentric working memory. Only egocentric accuracy correlated with caudate volumes, specifically the dorsolateral caudate head, right more than left, a region that receives dense efferents from dorsolateral prefrontal cortex. In contrast, only allocentric accuracy correlated with hippocampal volumes, specifically intermediate and posterior regions that connect strongly with parahippocampal and posterior parietal cortices. These results indicate that the distinction between egocentric and allocentric navigation applies to working memory. The dorsolateral caudate is important for egocentric working memory, which can explain the disproportionate impairment in pmHD. Allocentric working memory, in contrast, relies on the hippocampus and is relatively spared in pmHD.
Collapse
|
36
|
Cognitive Control, Learning, and Clinical Motor Ratings Are Most Highly Associated with Basal Ganglia Brain Volumes in the Premanifest Huntington's Disease Phenotype. J Int Neuropsychol Soc 2017; 23:159-170. [PMID: 28205498 PMCID: PMC5803794 DOI: 10.1017/s1355617716001132] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVES Huntington's disease (HD) is a debilitating genetic disorder characterized by motor, cognitive and psychiatric abnormalities associated with neuropathological decline. HD pathology is the result of an extended chain of CAG (cytosine, adenine, guanine) trinucleotide repetitions in the HTT gene. Clinical diagnosis of HD requires the presence of an otherwise unexplained extrapyramidal movement disorder in a participant at risk for HD. Over the past 15 years, evidence has shown that cognitive, psychiatric, and subtle motor dysfunction is evident decades before traditional motor diagnosis. This study examines the relationships among subcortical brain volumes and measures of emerging disease phenotype in prodromal HD, before clinical diagnosis. METHODS The dataset includes 34 cognitive, motor, psychiatric, and functional variables and five subcortical brain volumes from 984 prodromal HD individuals enrolled in the PREDICT HD study. Using cluster analyses, seven distinct clusters encompassing cognitive, motor, psychiatric, and functional domains were identified. Individual cluster scores were then regressed against the subcortical brain volumetric measurements. RESULTS Accounting for site and genetic burden (the interaction of age and CAG repeat length) smaller caudate and putamen volumes were related to clusters reflecting motor symptom severity, cognitive control, and verbal learning. CONCLUSIONS Variable reduction of the HD phenotype using cluster analysis revealed biologically related domains of HD and are suitable for future research with this population. Our cognitive control cluster scores show sensitivity to changes in basal ganglia both within and outside the striatum that may not be captured by examining only motor scores. (JINS, 2017, 23, 159-170).
Collapse
|
37
|
Scahill RI, Andre R, Tabrizi SJ, Aylward EH. Structural imaging in premanifest and manifest Huntington disease. HANDBOOK OF CLINICAL NEUROLOGY 2017; 144:247-261. [PMID: 28947121 DOI: 10.1016/b978-0-12-801893-4.00020-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Huntington disease (HD) neuropathology has a devastating effect on brain structure and consequently brain function; neuroimaging provides a means to assess these effects in gene carriers. In this chapter we first outline the unique utility of structural imaging in understanding HD and discuss some of the acquisition and analysis techniques currently available. We review the existing literature to summarize what we know so far about structural brain changes across the spectrum of disease from premanifest through to manifest disease. We then consider how these neuroimaging findings relate to patient function and nonimaging biomarkers, and can be used to predict disease onset. Finally we review the utility of imaging measures for assessment of treatment efficacy in clinical trials.
Collapse
Affiliation(s)
- Rachael I Scahill
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, United Kingdom
| | - Ralph Andre
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, United Kingdom
| | - Sarah J Tabrizi
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, United Kingdom.
| | - Elizabeth H Aylward
- Center for Integrative Brain Research, Seattle Children's Research Institute, University of Washington, Seattle, WA, United States
| |
Collapse
|
38
|
Arteaga-Bracho EE, Gulinello M, Winchester ML, Pichamoorthy N, Petronglo JR, Zambrano AD, Inocencio J, De Jesus CD, Louie JO, Gokhan S, Mehler MF, Molero AE. Postnatal and adult consequences of loss of huntingtin during development: Implications for Huntington's disease. Neurobiol Dis 2016; 96:144-155. [PMID: 27623015 DOI: 10.1016/j.nbd.2016.09.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 08/25/2016] [Accepted: 09/06/2016] [Indexed: 12/25/2022] Open
Abstract
The mutation in huntingtin (mHtt) leads to a spectrum of impairments in the developing forebrain of Huntington's disease (HD) mouse models. Whether these developmental alterations are due to loss- or gain-of-function mechanisms and contribute to HD pathogenesis is unknown. We examined the role of selective loss of huntingtin (Htt) function during development on postnatal vulnerability to cell death. We employed mice expressing very low levels of Htt throughout embryonic life to postnatal day 21 (Hdhd•hyp). We demonstrated that Hdhd•hyp mice exhibit: (1) late-life striatal and cortical neuronal degeneration; (2) neurological and skeletal muscle alterations; and (3) white matter tract impairments and axonal degeneration. Hdhd•hyp embryos also exhibited subpallial heterotopias, aberrant striatal maturation and deregulation of gliogenesis. These results indicate that developmental deficits associated with Htt functions render cells present at discrete neural foci increasingly susceptible to cell death, thus implying the potential existence of a loss-of-function developmental component to HD pathogenesis.
Collapse
Affiliation(s)
- Eduardo E Arteaga-Bracho
- Roslyn and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA; Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, NY, USA; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Maria Gulinello
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, NY, USA; Behavioral Core Facility, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Michael L Winchester
- Roslyn and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA; Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, NY, USA; The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA; Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Nandini Pichamoorthy
- Roslyn and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA; Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, NY, USA; The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA; Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jenna R Petronglo
- Roslyn and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA; Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, NY, USA; The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA; Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alicia D Zambrano
- Roslyn and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA; Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, NY, USA; The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Julio Inocencio
- The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Chirstopher D De Jesus
- Roslyn and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA; Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, NY, USA; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Joseph O Louie
- The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Solen Gokhan
- Roslyn and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA; Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, NY, USA; The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA; Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mark F Mehler
- Roslyn and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA; Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, NY, USA; The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA; Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, NY, USA; Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA; Ruth L. and David S. Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA; Center for Epigenomics, Albert Einstein College of Medicine, Bronx, NY, USA; Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aldrin E Molero
- Roslyn and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA; Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, NY, USA; The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA; Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
39
|
Horvath S, Langfelder P, Kwak S, Aaronson J, Rosinski J, Vogt TF, Eszes M, Faull RL, Curtis MA, Waldvogel HJ, Choi OW, Tung S, Vinters HV, Coppola G, Yang XW. Huntington's disease accelerates epigenetic aging of human brain and disrupts DNA methylation levels. Aging (Albany NY) 2016; 8:1485-512. [PMID: 27479945 PMCID: PMC4993344 DOI: 10.18632/aging.101005] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 07/12/2016] [Indexed: 01/10/2023]
Abstract
Age of Huntington's disease (HD) motoric onset is strongly related to the number of CAG trinucleotide repeats in the huntingtin gene, suggesting that biological tissue age plays an important role in disease etiology. Recently, a DNA methylation based biomarker of tissue age has been advanced as an epigenetic aging clock. We sought to inquire if HD is associated with an accelerated epigenetic age. DNA methylation data was generated for 475 brain samples from various brain regions of 26 HD cases and 39 controls. Overall, brain regions from HD cases exhibit a significant epigenetic age acceleration effect (p=0.0012). A multivariate model analysis suggests that HD status increases biological age by 3.2 years. Accelerated epigenetic age can be observed in specific brain regions (frontal lobe, parietal lobe, and cingulate gyrus). After excluding controls, we observe a negative correlation (r=-0.41, p=5.5×10-8) between HD gene CAG repeat length and the epigenetic age of HD brain samples. Using correlation network analysis, we identify 11 co-methylation modules with a significant association with HD status across 3 broad cortical regions. In conclusion, HD is associated with an accelerated epigenetic age of specific brain regions and more broadly with substantial changes in brain methylation levels.
Collapse
Affiliation(s)
- Steve Horvath
- Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Biostatistics, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Peter Langfelder
- Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Seung Kwak
- CHDI Management/CHDI Foundation, Princeton, NJ 08540, USA
| | - Jeff Aaronson
- CHDI Management/CHDI Foundation, Princeton, NJ 08540, USA
| | - Jim Rosinski
- CHDI Management/CHDI Foundation, Princeton, NJ 08540, USA
| | - Thomas F. Vogt
- CHDI Management/CHDI Foundation, Princeton, NJ 08540, USA
| | - Marika Eszes
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Science (FMHS), University of Auckland, Auckland, New Zealand
- Centre for Brain Research, Faculty of Medical and Health Science (FMHS), University of Auckland, Auckland, New Zealand
| | - Richard L.M. Faull
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Science (FMHS), University of Auckland, Auckland, New Zealand
- Centre for Brain Research, Faculty of Medical and Health Science (FMHS), University of Auckland, Auckland, New Zealand
| | - Maurice A. Curtis
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Science (FMHS), University of Auckland, Auckland, New Zealand
- Centre for Brain Research, Faculty of Medical and Health Science (FMHS), University of Auckland, Auckland, New Zealand
| | - Henry J. Waldvogel
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Science (FMHS), University of Auckland, Auckland, New Zealand
- Centre for Brain Research, Faculty of Medical and Health Science (FMHS), University of Auckland, Auckland, New Zealand
| | - Oi-Wa Choi
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience & Human Behavior, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Spencer Tung
- Pathology and Laboratory Medicine, and Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Harry V. Vinters
- Pathology and Laboratory Medicine, and Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Giovanni Coppola
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience & Human Behavior, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- UCLA Brain Research Institute, Los Angeles, CA 90095, USA
| | - X. William Yang
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience & Human Behavior, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- UCLA Brain Research Institute, Los Angeles, CA 90095, USA
| |
Collapse
|
40
|
Deng YP, Reiner A. Cholinergic interneurons in the Q140 knockin mouse model of Huntington's disease: Reductions in dendritic branching and thalamostriatal input. J Comp Neurol 2016; 524:3518-3529. [PMID: 27219491 DOI: 10.1002/cne.24013] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 03/29/2016] [Accepted: 04/06/2016] [Indexed: 12/19/2022]
Abstract
We have previously found that thalamostriatal axodendritic terminals are reduced as early as 1 month of age in heterozygous Q140 HD mice (Deng et al. [] Neurobiol Dis 60:89-107). Because cholinergic interneurons are a major target of thalamic axodendritic terminals, we examined the VGLUT2-immunolabeled thalamic input to striatal cholinergic interneurons in heterozygous Q140 males at 1 and 4 months of age, using choline acetyltransferase (ChAT) immunolabeling to identify cholinergic interneurons. Although blinded neuron counts showed that ChAT+ perikarya were in normal abundance in Q140 mice, size measurements indicated that they were significantly smaller. Sholl analysis further revealed the dendrites of Q140 ChAT+ interneurons were significantly fewer and shorter. Consistent with the light microscopic data, ultrastructural analysis showed that the number of ChAT+ dendritic profiles per unit area of striatum was significantly decreased in Q140 striata, as was the abundance of VGLUT2+ axodendritic terminals making synaptic contact with ChAT+ dendrites per unit area of striatum. The density of thalamic terminals along individual cholinergic dendrites was, however, largely unaltered, indicating that the reduction in the areal striatal density of axodendritic thalamic terminals on cholinergic neurons was due to their dendritic territory loss. These results show that the abundance of thalamic input to individual striatal cholinergic interneurons is reduced early in the life span of Q140 mice, raising the possibility that this may occur in human HD as well. Because cholinergic interneurons differentially affect striatal direct vs. indirect pathway spiny projection neurons, their reduced thalamic excitatory drive may contribute to early abnormalities in movement in HD. J. Comp. Neurol. 524:3518-3529, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yun-Ping Deng
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee, 38163
| | - Anton Reiner
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee, 38163.
| |
Collapse
|
41
|
Wallace M, Downing N, Lourens S, Mills J, Kim JI, Long J, Paulsen J. Is There an Association of Physical Activity with Brain Volume, Behavior, and Day-to-day Functioning? A Cross Sectional Design in Prodromal and Early Huntington Disease. PLOS CURRENTS 2016; 8. [PMID: 27818843 PMCID: PMC4866530 DOI: 10.1371/currents.hd.cba6ea74972cf8412a73ce52eb018c1e] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Huntington disease (HD) is a genetic neurodegenerative disease leading to progressive motor, cognitive, and behavioral decline. Subtle changes in these domains are detectable up to 15 years before a definitive motor diagnosis is made. This period, called prodromal HD, provides an opportunity to examine lifestyle behaviors that may impact disease progression. THEORETICAL FRAMEWORK Physical activity relates to decreased rates of brain atrophy and improved cognitive and day-to-day functioning in Alzheimer disease and healthy aging populations. Previous research has yielded mixed results regarding the impact of physical activity on disease progression in HD and paid little attention to the prodromal phase. METHODS We conducted analyses of associations among current physical activity level, current and retrospective rate of change for hippocampus and striatum volume, and cognitive, motor, and day-to-day functioning variables. Participants were 48 gene-expanded cases with prodromal and early-diagnosed HD and 27 nongene-expanded control participants. Participants wore Fitbit Ultra activity monitors for three days and completed the self-reported International Physical Activity Questionnaire (IPAQ). Hippocampal and striatal white matter volumes were measured using magnetic resonance imaging. Cognitive tests included the Stroop Color and Word Test, and the Symbol Digit Modalities Test (SDMT). Motor function was assessed using the Unified Huntington's Disease Rating Scale total motor score (TMS). Day-to-day functioning was measured using the World Health Organization Disability Assessment Schedule (WHODAS) version 2.0. RESULTS Higher Fitbit activity scores were significantly related to better scores on the SDMT and WHODAS in case participants but not in controls. Fitbit activity scores tracked better with TMS scores in the group as a whole, though the association did not reach statistical significance in the case participants. Higher Fitbit activity scores related to less day-to-day functioning decline in retrospective slope analyses. Fitbit activity scores did not differ significantly between cases and controls. CONCLUSIONS This is the first known study examining the associations between activity level and imaging, motor, cognitive, and day-to-day functioning outcomes in prodromal and early HD. Preliminary results suggest physical activity positively correlates with improved cognitive and day-to-day functioning and possibly motor function in individuals in the prodromal and early phase of the condition.
Collapse
Affiliation(s)
- McKenzie Wallace
- Frances Payne Bolton School of Nursing, Case Western Reserve University, Iowa City, Iowa, USA
| | - Nancy Downing
- College of Nursing, The University of Iowa, Iowa City, Iowa, USA
| | - Spencer Lourens
- School of Medicine, Indiana University Purdue University at Indianapolis, Indianapolis, Indiana, USA
| | - James Mills
- Department on Psychiatry, University of Iowa, Iowa City, Iowa, USA
| | - Ji-In Kim
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Jeffrey Long
- Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Jane Paulsen
- Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
42
|
Faria AV, Ratnanather JT, Tward DJ, Lee DS, van den Noort F, Wu D, Brown T, Johnson H, Paulsen JS, Ross CA, Younes L, Miller MI. Linking white matter and deep gray matter alterations in premanifest Huntington disease. Neuroimage Clin 2016; 11:450-460. [PMID: 27104139 PMCID: PMC4827723 DOI: 10.1016/j.nicl.2016.02.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 02/17/2016] [Accepted: 02/22/2016] [Indexed: 01/07/2023]
Abstract
Huntington disease (HD) is a fatal progressive neurodegenerative disorder for which only symptomatic treatment is available. A better understanding of the pathology, and identification of biomarkers will facilitate the development of disease-modifying treatments. HD is potentially a good model of a neurodegenerative disease for development of biomarkers because it is an autosomal-dominant disease with complete penetrance, caused by a single gene mutation, in which the neurodegenerative process can be assessed many years before onset of signs and symptoms of manifest disease. Previous MRI studies have detected abnormalities in gray and white matter starting in premanifest stages. However, the understanding of how these abnormalities are related, both in time and space, is still incomplete. In this study, we combined deep gray matter shape diffeomorphometry and white matter DTI analysis in order to provide a better mapping of pathology in the deep gray matter and subcortical white matter in premanifest HD. We used 296 MRI scans from the PREDICT-HD database. Atrophy in the deep gray matter, thalamus, hippocampus, and nucleus accumbens was analyzed by surface based morphometry, and while white matter abnormalities were analyzed in (i) regions of interest surrounding these structures, using (ii) tractography-based analysis, and using (iii) whole brain atlas-based analysis. We detected atrophy in the deep gray matter, particularly in putamen, from early premanifest stages. The atrophy was greater both in extent and effect size in cases with longer exposure to the effects of the CAG expansion mutation (as assessed by greater CAP-scores), and preceded detectible abnormalities in the white matter. Near the predicted onset of manifest HD, the MD increase was widespread, with highest indices in the deep and posterior white matter. This type of in-vivo macroscopic mapping of HD brain abnormalities can potentially indicate when and where therapeutics could be targeted to delay the onset or slow the disease progression.
Collapse
Affiliation(s)
- Andreia V Faria
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - J Tilak Ratnanather
- Center for Imaging Science, The Johns Hopkins University, Baltimore, MD, USA; Institute for Computational Medicine, The Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Daniel J Tward
- Center for Imaging Science, The Johns Hopkins University, Baltimore, MD, USA; Institute for Computational Medicine, The Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - David Soobin Lee
- Center for Imaging Science, The Johns Hopkins University, Baltimore, MD, USA; Institute for Computational Medicine, The Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Frieda van den Noort
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Dan Wu
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Timothy Brown
- Center for Imaging Science, The Johns Hopkins University, Baltimore, MD, USA
| | - Hans Johnson
- Department of Psychiatry, The University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Jane S Paulsen
- Department of Psychiatry, The University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Christopher A Ross
- Division of Neurobiology, Department of Psychiatry, and Departments of Neurology, Neuroscience and Pharmacology, Johns Hopkins University, Baltimore, MD, USA
| | - Laurent Younes
- Center for Imaging Science, The Johns Hopkins University, Baltimore, MD, USA; Institute for Computational Medicine, The Johns Hopkins University, Baltimore, MD, USA; Department of Applied Mathematics and Statistics, The Johns Hopkins University, Baltimore, MD, USA
| | - Michael I Miller
- Center for Imaging Science, The Johns Hopkins University, Baltimore, MD, USA; Institute for Computational Medicine, The Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
43
|
Integrated genomics and proteomics define huntingtin CAG length-dependent networks in mice. Nat Neurosci 2016; 19:623-33. [PMID: 26900923 PMCID: PMC5984042 DOI: 10.1038/nn.4256] [Citation(s) in RCA: 268] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 01/28/2016] [Indexed: 12/14/2022]
Abstract
To gain insight into how mutant huntingtin (mHtt) CAG repeat length modifies Huntington’s disease (HD) pathogenesis, we profiled mRNA in over 600 brain and peripheral tissue samples from HD knock-in mice with increasing CAG repeat lengths. We find repeat length dependent transcriptional signatures are prominent in the striatum, less so in cortex, and minimal in the liver. Co-expression network analyses reveal 13 striatal and 5 cortical modules that are highly correlated with CAG length and age, and that are preserved in HD models and some in the patients. Top striatal modules implicate mHtt CAG length and age in graded impairment of striatal medium spiny neuron identity gene expression and in dysregulation of cAMP signaling, cell death, and protocadherin genes. Importantly, we used proteomics to confirm 790 genes and 5 striatal modules with CAG length-dependent dysregulation at both RNA and protein levels, and validated 22 striatal module genes as modifiers of mHtt toxicities in vivo.
Collapse
|
44
|
Pavese N, Tai YF. Genetic and degenerative disorders primarily causing other movement disorders. HANDBOOK OF CLINICAL NEUROLOGY 2016; 135:507-523. [PMID: 27432681 DOI: 10.1016/b978-0-444-53485-9.00025-8] [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: 06/06/2023]
Abstract
In this chapter, we will discuss the contributions of structural and functional imaging to the diagnosis and management of genetic and degenerative diseases that lead to the occurrence of movement disorders. We will mainly focus on Huntington's disease, Wilson's disease, dystonia, and neurodegeneration with brain iron accumulation, as they are the more commonly encountered clinical conditions within this group.
Collapse
Affiliation(s)
- Nicola Pavese
- Division of Brain Sciences, Imperial College London, UK; Aarhus University, Denmark.
| | - Yen F Tai
- Division of Brain Sciences, Imperial College London, UK
| |
Collapse
|
45
|
Crittenden J, Graybiel A. Disease-Associated Changes in the Striosome and Matrix Compartments of the Dorsal Striatum. HANDBOOK OF BEHAVIORAL NEUROSCIENCE 2016. [DOI: 10.1016/b978-0-12-802206-1.00039-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
46
|
Schramke S, Schuldenzucker V, Schubert R, Frank F, Wirsig M, Ott S, Motlik J, Fels M, Kemper N, Hölzner E, Reilmann R. Behavioral phenotyping of minipigs transgenic for the Huntington gene. J Neurosci Methods 2015; 265:34-45. [PMID: 26688470 DOI: 10.1016/j.jneumeth.2015.11.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 11/17/2015] [Accepted: 11/19/2015] [Indexed: 12/27/2022]
Abstract
BACKGROUND While several novel therapeutic approaches for HD are in development, resources to conduct clinical trials are limited. Large animal models have been proposed to improve assessment of safety, tolerability and especially to increase translational reliability of efficacy signals obtained in preclinical studies. They may thus help to select candidates for translation to human studies. We here introduce a battery of novel tests designed to assess the motor, cognitive and behavioral phenotype of a transgenic (tg) HD minipig model. NEW METHODS A group of tgHD and wildtype (wt) Libechov minipigs (n=36) was available for assessment with (1) a gait test using the GAITRite(®) automated acquisition system, (2) a hurdle-test, (3) a tongue coordination test, (4) a color discrimination test, (5) a startbox back and forth test and (6) a dominance test. Performance of all tests and definition of measures obtained is presented. RESULTS Minipigs were able to learn performance of all tests. All tests were safe, well tolerated and feasible. Exploratory between group comparisons showed no differences between groups of tgHD and wt minipigs assessed, but low variability within and between groups. COMPARISON WITH EXISTING METHOD(S) So far there are no established or validated assessments to test minipigs in the domains described. CONCLUSIONS The data shows that the tests presented are safe, well tolerated and all measures defined can be assessed. Prospective longitudinal application of these tests is warranted to determine their test-retest reliability, sensitivity and validity in assessing motor, cognitive and behavioral features of tg and wt minipigs.
Collapse
Affiliation(s)
- Sarah Schramke
- George-Huntington-Institute, Technology Park Muenster, Johann-Krane Weg 27 48149, Muenster, Germany; Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour, University of Veterinary Medicine Hannover, Bischofsholer Damm 15 30173, Hannover, Germany
| | - Verena Schuldenzucker
- George-Huntington-Institute, Technology Park Muenster, Johann-Krane Weg 27 48149, Muenster, Germany
| | - Robin Schubert
- George-Huntington-Institute, Technology Park Muenster, Johann-Krane Weg 27 48149, Muenster, Germany
| | - Frauke Frank
- George-Huntington-Institute, Technology Park Muenster, Johann-Krane Weg 27 48149, Muenster, Germany; Laboratory of Cell Regeneration and Plasticity, Institute of Animal Physiology and Genetics, v.v.i., AS CR, Libechov, Czech Republic
| | - Maike Wirsig
- George-Huntington-Institute, Technology Park Muenster, Johann-Krane Weg 27 48149, Muenster, Germany
| | - Stefanie Ott
- George-Huntington-Institute, Technology Park Muenster, Johann-Krane Weg 27 48149, Muenster, Germany
| | - Jan Motlik
- Laboratory of Cell Regeneration and Plasticity, Institute of Animal Physiology and Genetics, v.v.i., AS CR, Libechov, Czech Republic
| | - Michaela Fels
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour, University of Veterinary Medicine Hannover, Bischofsholer Damm 15 30173, Hannover, Germany
| | - Nicole Kemper
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour, University of Veterinary Medicine Hannover, Bischofsholer Damm 15 30173, Hannover, Germany
| | - Eva Hölzner
- George-Huntington-Institute, Technology Park Muenster, Johann-Krane Weg 27 48149, Muenster, Germany
| | - Ralf Reilmann
- George-Huntington-Institute, Technology Park Muenster, Johann-Krane Weg 27 48149, Muenster, Germany; Department of Radiology, Universitaetsklinikum Muenster, Albert-Schweitzer Campus 1 48149, Muenster, Germany; Dept of Neurology Muenster, Germany; Department of Neurodegenerative Diseases and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Hoppe-Seyler Str. 3 72076 Tuebingen, Germany.
| |
Collapse
|
47
|
Mo C, Hannan AJ, Renoir T. Environmental factors as modulators of neurodegeneration: Insights from gene–environment interactions in Huntington's disease. Neurosci Biobehav Rev 2015; 52:178-92. [DOI: 10.1016/j.neubiorev.2015.03.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 02/13/2015] [Accepted: 03/03/2015] [Indexed: 12/11/2022]
|
48
|
Aylward EH. Magnetic resonance imaging striatal volumes: a biomarker for clinical trials in Huntington's disease. Mov Disord 2014; 29:1429-33. [PMID: 25164586 DOI: 10.1002/mds.26013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 07/25/2014] [Accepted: 08/03/2014] [Indexed: 11/06/2022] Open
Abstract
An abundance of research shows that magnetic resonance imaging (MRI) striatal volumes decrease long before diagnosis of Huntington's disease (HD) and closely track disease progression. Additional research indicates that these volumetric measures meet important criteria for a biomarker that can be used in clinical trials: They are 1) objectively measureable; 2) able to predict known endpoints; and 3) associated with known mechanisms of pathology of the disease. Researchers should consider formal application to regulatory agencies for biomarker status of volumetric MRI striatal measures, because these measures are anticipated to contribute significantly in the assessment of treatment effectiveness in HD.
Collapse
|
49
|
Reilmann R, Leavitt BR, Ross CA. Diagnostic criteria for Huntington's disease based on natural history. Mov Disord 2014; 29:1335-41. [PMID: 25164527 DOI: 10.1002/mds.26011] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 08/06/2014] [Indexed: 01/10/2023] Open
Abstract
Huntington's disease (HD) is currently diagnosed based on the presence of motor signs indicating 99% "diagnostic confidence" for HD. Recent advances in the understanding of HD natural history and neurobiology indicate that disease-related brain changes begin at least 12 to 15 years before the formal diagnosis based on motor onset. Furthermore, subtle motor dysfunction, cognitive changes, and behavioral alterations are often seen before diagnosis made according to the current criteria. As disease-modifying treatments are developed, likely beginning therapy early will be desirable. We therefore suggest that expanded diagnostic criteria for HD should be adapted to better reflect the natural history of the disease, to enable the conduct of clinical trials in premanifest subjects targeting prevention of neurodegeneration, and to facilitate earlier symptomatic treatment. We propose a new set of criteria for HD diagnostic categories in the International Classification of Diseases that reflect our current understanding of HD natural history and pathogenesis. Based on defined criteria, for example, the Diagnostic Confidence Level and the Total Functional Capacity scales of the Unified Huntington's Disease Rating Scale, HD should be divided in the categories "genetically confirmed" with the subcategories "presymptomatic," "prodromal," and "manifest" and "not genetically confirmed" subdivided into "clinically at risk," "clinically prodromal," and "clinically manifest."
Collapse
Affiliation(s)
- Ralf Reilmann
- George-Huntington-Institute, Technology-Park, Muenster, Germany; Department of Neurodegenerative Diseases and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | | | | |
Collapse
|