251
|
Vaca-Palomares I, Brien DC, Coe BC, Ochoa-Morales A, Martínez-Ruano L, Munoz DP, Fernandez-Ruiz J. Implicit learning impairment identified via predictive saccades in Huntington's disease correlates with extended cortico-striatal atrophy. Cortex 2019; 121:89-103. [PMID: 31550618 DOI: 10.1016/j.cortex.2019.06.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 05/02/2019] [Accepted: 06/29/2019] [Indexed: 01/10/2023]
Abstract
The ability to anticipate events and execute motor commands prior to a sensory event is an essential capability for human's everyday life. This implicitly learned anticipatory behavior depends on the past performance of repeated sensorimotor interactions timed with external cues. This kind of predictive behavior has been shown to be compromised in neurological disorders such as Huntington disease (HD), in which neural atrophy includes key cortical and basal ganglia regions. To investigate the neural basis of the anticipatory behavioral deficits in HD we used a predictive-saccade paradigm that requires predictive control to generate saccades in a metronomic temporal pattern. This is ideal because the integrity of the oculomotor network that includes the striatum and prefrontal, parietal, occipital and temporal cortices can be analyzed using structural MRI. Our results showed that the HD patients had severe predictive saccade deficits (i.e., an inability to reduce saccade reaction time in predictive condition), which are accentuated in patients with more severe motor deterioration. Structural imaging analyses revealed that these anticipatory deficits correlated with grey-matter atrophy in frontal, parietal-occipital and striatal regions. These findings indicate that the predictive saccade control deficits in HD are related to an extended cortico-striatal atrophy. This suggests that eye movement measurement could be a reliable marker of the progression of cognitive deficits in HD.
Collapse
Affiliation(s)
- Israel Vaca-Palomares
- Ciencias Cognitivas y del Comportamiento, Facultad de Psicología, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Donald C Brien
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Brian C Coe
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Adriana Ochoa-Morales
- Departamento de Genética, Instituto Nacional de Neurología y Neurocirugía, "Manuel Velasco Suarez", CDMX, Mexico
| | - Leticia Martínez-Ruano
- Departamento de Genética, Instituto Nacional de Neurología y Neurocirugía, "Manuel Velasco Suarez", CDMX, Mexico
| | - Douglas P Munoz
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Juan Fernandez-Ruiz
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico.
| |
Collapse
|
252
|
Pini L, Jacquemot C, Cagnin A, Meneghello F, Semenza C, Mantini D, Vallesi A. Aberrant brain network connectivity in presymptomatic and manifest Huntington's disease: A systematic review. Hum Brain Mapp 2019; 41:256-269. [PMID: 31532053 PMCID: PMC7268025 DOI: 10.1002/hbm.24790] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/29/2019] [Accepted: 08/26/2019] [Indexed: 12/12/2022] Open
Abstract
Resting‐state functional magnetic resonance imaging (rs‐fMRI) has the potential to shed light on the pathophysiological mechanisms of Huntington's disease (HD), paving the way to new therapeutic interventions. A systematic literature review was conducted in three online databases according to PRISMA guidelines, using keywords for HD, functional connectivity, and rs‐fMRI. We included studies investigating connectivity in presymptomatic (pre‐HD) and manifest HD gene carriers compared to healthy controls, implementing seed‐based connectivity, independent component analysis, regional property, and graph analysis approaches. Visual network showed reduced connectivity in manifest HD, while network/areas underpinning motor functions were consistently altered in both manifest HD and pre‐HD, showing disease stage‐dependent changes. Cognitive networks underlying executive and attentional functions showed divergent anterior–posterior alterations, possibly reflecting compensatory mechanisms. The involvement of these networks in pre‐HD is still unclear. In conclusion, aberrant connectivity of the sensory‐motor network is observed in the early stage of HD while, as pathology spreads, other networks might be affected, such as the visual and executive/attentional networks. Moreover, sensory‐motor and executive networks exhibit hyper‐ and hypo‐connectivity patterns following different spatiotemporal trajectories. These findings could potentially help to implement future huntingtin‐lowering interventions.
Collapse
Affiliation(s)
- Lorenzo Pini
- Department of Neuroscience & Padova Neuroscience Center, University of Padova, Padova, Italy
| | - Charlotte Jacquemot
- Département d'Etudes Cognitives, Ecole Normale Supérieure-PSL University, Paris, France.,Laboratoire de NeuroPsychologie Interventionnelle, Institut Mondor de Recherche Biomédicale, Institut National de la Santé et Recherche Médical (INSERM) U955, Equipe 01, Créteil, France.,Faculté de Médecine, Université Paris Est Créteil, Créteil, France
| | - Annachiara Cagnin
- Department of Neuroscience & Padova Neuroscience Center, University of Padova, Padova, Italy
| | - Francesca Meneghello
- Cognitive Neuroscience Research Group, IRCCS San Camillo Hospital, Venice, Italy
| | - Carlo Semenza
- Department of Neuroscience & Padova Neuroscience Center, University of Padova, Padova, Italy.,Cognitive Neuroscience Research Group, IRCCS San Camillo Hospital, Venice, Italy
| | - Dante Mantini
- Research Center for Motor Control and Neuroplasticity, KU Leuven, Leuven, Belgium.,Brain Imaging and Neural Dynamics Research Group, IRCCS San Camillo Hospital, Venice, Italy
| | - Antonino Vallesi
- Department of Neuroscience & Padova Neuroscience Center, University of Padova, Padova, Italy.,Brain Imaging and Neural Dynamics Research Group, IRCCS San Camillo Hospital, Venice, Italy
| |
Collapse
|
253
|
Sampedro F, Martínez-Horta S, Perez-Perez J, Horta-Barba A, Martin-Lahoz J, Alonso-Solís A, Corripio I, Gomez-Anson B, Kulisevsky J. Widespread Increased Diffusivity Reveals Early Cortical Degeneration in Huntington Disease. AJNR Am J Neuroradiol 2019; 40:1464-1468. [PMID: 31467235 DOI: 10.3174/ajnr.a6168] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 07/07/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND PURPOSE Huntington disease is a devastating genetic neurodegenerative disorder for which no effective treatment is yet available. Although progressive striatal atrophy is its pathologic hallmark, concomitant cortical deterioration is assumed to occur, but it is poorly characterized. Our objective was to study the loss of cortical integrity and its association with clinical indicators throughout the course of the disease. MATERIALS AND METHODS Using a cohort of 39 patients with Huntington disease and 25 controls with available MR imaging (T1WI and DTI), we compared cortical atrophy and intracortical diffusivity across disease stages. Intracortical diffusivity is a DTI-derived metric that has recently been suggested to detect incipient neuronal death because water can diffuse more freely in cortical regions with reduced neural density. RESULTS We observed progressive thinning and increasing diffusivity within the cerebral cortex of patients with Huntington disease (P < .05, corrected for multiple comparisons). Most important, in the absence of pronounced atrophy, widespread increased diffusivity was already present in individuals with premanifest Huntington disease, correlating, in turn, with clinical and disease-specific progression markers. CONCLUSIONS Intracortical diffusivity may be more sensitive than cortical thinning for tracking early neurodegeneration in Huntington disease. Moreover, our findings provide further evidence of an early cortical compromise in Huntington disease, which contributes to our understanding of its clinical phenotype and could have important therapeutic implications.
Collapse
Affiliation(s)
- F Sampedro
- From the Movement Disorders Unit (F.S., S.M.-H., J.P.-P., A.H.-B., J.M.-L., J.K.), Neurology Department.,Biomedical Research Institute (F.S., S.M.-H., J.P.-P., A.H.-B., J.M.-L., A.A.-S., I.C., J.K.), Barcelona, Spain.,Centro de Investigación en Red-Enfermedades Neurodegenerativas (F.S., S.M.-H., J.P.-P., A.H.-B., J.M.-L., J.K.), Madrid, Spain
| | - S Martínez-Horta
- From the Movement Disorders Unit (F.S., S.M.-H., J.P.-P., A.H.-B., J.M.-L., J.K.), Neurology Department.,Biomedical Research Institute (F.S., S.M.-H., J.P.-P., A.H.-B., J.M.-L., A.A.-S., I.C., J.K.), Barcelona, Spain.,Centro de Investigación en Red-Enfermedades Neurodegenerativas (F.S., S.M.-H., J.P.-P., A.H.-B., J.M.-L., J.K.), Madrid, Spain.,Universitat Autónoma de Barcelona (S.M.-H., J.P.-P., J.M.-L., B.G.-A., J.K.), Barcelona, Spain
| | - J Perez-Perez
- From the Movement Disorders Unit (F.S., S.M.-H., J.P.-P., A.H.-B., J.M.-L., J.K.), Neurology Department.,Biomedical Research Institute (F.S., S.M.-H., J.P.-P., A.H.-B., J.M.-L., A.A.-S., I.C., J.K.), Barcelona, Spain.,Centro de Investigación en Red-Enfermedades Neurodegenerativas (F.S., S.M.-H., J.P.-P., A.H.-B., J.M.-L., J.K.), Madrid, Spain.,Universitat Autónoma de Barcelona (S.M.-H., J.P.-P., J.M.-L., B.G.-A., J.K.), Barcelona, Spain
| | - A Horta-Barba
- From the Movement Disorders Unit (F.S., S.M.-H., J.P.-P., A.H.-B., J.M.-L., J.K.), Neurology Department.,Biomedical Research Institute (F.S., S.M.-H., J.P.-P., A.H.-B., J.M.-L., A.A.-S., I.C., J.K.), Barcelona, Spain.,Centro de Investigación en Red-Enfermedades Neurodegenerativas (F.S., S.M.-H., J.P.-P., A.H.-B., J.M.-L., J.K.), Madrid, Spain
| | - J Martin-Lahoz
- From the Movement Disorders Unit (F.S., S.M.-H., J.P.-P., A.H.-B., J.M.-L., J.K.), Neurology Department.,Biomedical Research Institute (F.S., S.M.-H., J.P.-P., A.H.-B., J.M.-L., A.A.-S., I.C., J.K.), Barcelona, Spain.,Centro de Investigación en Red-Enfermedades Neurodegenerativas (F.S., S.M.-H., J.P.-P., A.H.-B., J.M.-L., J.K.), Madrid, Spain.,Universitat Autónoma de Barcelona (S.M.-H., J.P.-P., J.M.-L., B.G.-A., J.K.), Barcelona, Spain
| | - A Alonso-Solís
- Biomedical Research Institute (F.S., S.M.-H., J.P.-P., A.H.-B., J.M.-L., A.A.-S., I.C., J.K.), Barcelona, Spain
| | - I Corripio
- Psychiatry Department (I.C.), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Biomedical Research Institute (F.S., S.M.-H., J.P.-P., A.H.-B., J.M.-L., A.A.-S., I.C., J.K.), Barcelona, Spain.,Centro de Investigación Biomédica en Red-Salud Mental (I.C.), Madrid, Spain
| | - B Gomez-Anson
- Neuroradiology, Radiology Department (B.G.-A.).,European Huntington's Disease Network (S.M.-H., J.P.-P., A.H.-B., J.K.), Ulm, Germany
| | - J Kulisevsky
- From the Movement Disorders Unit (F.S., S.M.-H., J.P.-P., A.H.-B., J.M.-L., J.K.), Neurology Department .,Biomedical Research Institute (F.S., S.M.-H., J.P.-P., A.H.-B., J.M.-L., A.A.-S., I.C., J.K.), Barcelona, Spain.,Centro de Investigación en Red-Enfermedades Neurodegenerativas (F.S., S.M.-H., J.P.-P., A.H.-B., J.M.-L., J.K.), Madrid, Spain.,European Huntington's Disease Network (S.M.-H., J.P.-P., A.H.-B., J.K.), Ulm, Germany
| |
Collapse
|
254
|
Yeh FC, Zaydan IM, Suski VR, Lacomis D, Richardson RM, Maroon JC, Barrios-Martinez J. Differential tractography as a track-based biomarker for neuronal injury. Neuroimage 2019; 202:116131. [PMID: 31472253 DOI: 10.1016/j.neuroimage.2019.116131] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 08/24/2019] [Accepted: 08/24/2019] [Indexed: 12/12/2022] Open
Abstract
Diffusion MRI tractography has been used to map the axonal structure of the human brain, but its ability to detect neuronal injury is yet to be explored. Here we report differential tractography, a new type of tractography that utilizes repeat MRI scans and a novel tracking strategy to map the exact segment of fiber pathways with a neuronal injury. We examined differential tractography on multiple sclerosis, Huntington's disease, amyotrophic lateral sclerosis, and epileptic patients. The results showed that the affected pathways shown by differential tractography matched well with the unique clinical symptoms of the patients, and the false discovery rate of the findings could be estimated using a sham setting to provide a reliability measurement. This novel approach enables a quantitative and objective method to monitor neuronal injury in individuals, allowing for diagnostic and prognostic evaluation of brain diseases.
Collapse
Affiliation(s)
- Fang-Cheng Yeh
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States.
| | - Islam M Zaydan
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Valerie R Suski
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - David Lacomis
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Live Like Lou Center for ALS Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - R Mark Richardson
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Joseph C Maroon
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jessica Barrios-Martinez
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
255
|
Heath CJ, O'Callaghan C, Mason SL, Phillips BU, Saksida LM, Robbins TW, Barker RA, Bussey TJ, Sahakian BJ. A Touchscreen Motivation Assessment Evaluated in Huntington's Disease Patients and R6/1 Model Mice. Front Neurol 2019; 10:858. [PMID: 31447770 PMCID: PMC6696591 DOI: 10.3389/fneur.2019.00858] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/24/2019] [Indexed: 12/18/2022] Open
Abstract
Apathy is pervasive across many neuropsychiatric disorders but is poorly characterized mechanistically, so targeted therapeutic interventions remain elusive. A key impediment has been the lack of validated assessment tools to facilitate translation of promising findings between preclinical disease models and patients. Apathy is a common symptom in Huntington's disease. Due to its established genetic basis and the availability of defined animal models, this disease offers a robust translational framework for linking motivated behavior with underlying neurobiology and an ideal context in which to evaluate a quantitative, translational apathy assessment method. In this study we therefore aimed to demonstrate the validity of using touchscreen-delivered progressive ratio tasks to mirror apathy assessment in Huntington's disease patients and a representative mouse model. To do this we evaluated Huntington's disease patients (n = 23) and age-matched healthy controls (n = 20), and male R6/1 mice (n = 23) and wildtype controls (n = 29) for apathy-like behavior using touchscreen-delivered progressive ratio tasks. The primary outcome measure of the assessment was breakpoint, defined as the highest number of touchscreen responses emitted before task engagement ceased. Patients and R6/1 mice were both found to exhibit significantly reduced breakpoints relative to their respective control groups, consistent with apathy-like behavior. This performance was also not associated with motoric differences in either species. These data demonstrate the utility of touchscreen-delivered progressive ratio tasks in detecting clinically relevant motivational deficits in Huntington's disease. This approach may offer a platform from which clinically relevant mechanistic insights concerning motivation symptoms can be derived and provide an effective route for translation of promising preclinical findings into viable therapeutic interventions.
Collapse
Affiliation(s)
- Christopher J. Heath
- Department of Psychology, Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Claire O'Callaghan
- Department of Psychology, Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
| | - Sarah L. Mason
- John van Geest Centre for Brain Repair, Addenbrooke's Hospital, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Benjamin U. Phillips
- Department of Psychology, Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom
| | - Lisa M. Saksida
- Department of Psychology, Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Trevor W. Robbins
- Department of Psychology, Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom
| | - Roger A. Barker
- John van Geest Centre for Brain Repair, Addenbrooke's Hospital, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Timothy J. Bussey
- Department of Psychology, Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Barbara J. Sahakian
- Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| |
Collapse
|
256
|
van der Plas E, Langbehn DR, Conrad AL, Koscik TR, Tereshchenko A, Epping EA, Magnotta VA, Nopoulos PC. Abnormal brain development in child and adolescent carriers of mutant huntingtin. Neurology 2019; 93:e1021-e1030. [PMID: 31371571 DOI: 10.1212/wnl.0000000000008066] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 05/20/2019] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE The huntingtin gene is critical for the formation and differentiation of the CNS, which raises questions about the neurodevelopmental effect of CAG expansion mutations within this gene (mHTT) that cause Huntington disease (HD). We sought to test the hypothesis that child and adolescent carriers of mHTT exhibit different brain growth compared to peers without the mutation by conducting structural MRI in youth who are at risk for HD. We also explored whether the length of CAG expansion affects brain development. METHODS Children and adolescents (age 6-18) with a parent or grandparent diagnosed with HD underwent MRI and blinded genetic testing to confirm the presence or absence of mHTT. Seventy-five individuals were gene-expanded (GE) and 97 individuals were gene-nonexpanded (GNE). The GE group was estimated to be on average 35 years from clinical onset. Following an accelerated longitudinal design, age-related changes in brain regions were estimated. RESULTS Age-related striatal volume changes differed significantly between the GE and GNE groups, with initial hypertrophy and more rapid volume decline in GE. This pattern was exaggerated with CAG expansion length for CAG > 50. A similar age-dependent group difference was observed for the globus pallidus, but not in other major regions. CONCLUSION Our results suggest that pathogenesis of HD begins with abnormal brain development. An understanding of potential neurodevelopmental features associated with mHTT may be needed for optimized implementation of preventative gene silencing therapies, such that normal aspects of neurodevelopment are preserved as neurodegeneration is forestalled.
Collapse
Affiliation(s)
- Ellen van der Plas
- From the Department of Psychiatry (E.v.d.P., T.R.K.), University of Iowa Hospitals & Clinics; and the Departments of Psychiatry (D.R.L., A.T., E.A.E., P.C.N.), Biostatistics (D.R.L., A.T.), and Radiology (V.A.M.) and Stead Family Department of Pediatrics (A.L.C.), University of Iowa, Iowa City.
| | - Douglas R Langbehn
- From the Department of Psychiatry (E.v.d.P., T.R.K.), University of Iowa Hospitals & Clinics; and the Departments of Psychiatry (D.R.L., A.T., E.A.E., P.C.N.), Biostatistics (D.R.L., A.T.), and Radiology (V.A.M.) and Stead Family Department of Pediatrics (A.L.C.), University of Iowa, Iowa City
| | - Amy L Conrad
- From the Department of Psychiatry (E.v.d.P., T.R.K.), University of Iowa Hospitals & Clinics; and the Departments of Psychiatry (D.R.L., A.T., E.A.E., P.C.N.), Biostatistics (D.R.L., A.T.), and Radiology (V.A.M.) and Stead Family Department of Pediatrics (A.L.C.), University of Iowa, Iowa City
| | - Timothy R Koscik
- From the Department of Psychiatry (E.v.d.P., T.R.K.), University of Iowa Hospitals & Clinics; and the Departments of Psychiatry (D.R.L., A.T., E.A.E., P.C.N.), Biostatistics (D.R.L., A.T.), and Radiology (V.A.M.) and Stead Family Department of Pediatrics (A.L.C.), University of Iowa, Iowa City
| | - Alexander Tereshchenko
- From the Department of Psychiatry (E.v.d.P., T.R.K.), University of Iowa Hospitals & Clinics; and the Departments of Psychiatry (D.R.L., A.T., E.A.E., P.C.N.), Biostatistics (D.R.L., A.T.), and Radiology (V.A.M.) and Stead Family Department of Pediatrics (A.L.C.), University of Iowa, Iowa City
| | - Eric A Epping
- From the Department of Psychiatry (E.v.d.P., T.R.K.), University of Iowa Hospitals & Clinics; and the Departments of Psychiatry (D.R.L., A.T., E.A.E., P.C.N.), Biostatistics (D.R.L., A.T.), and Radiology (V.A.M.) and Stead Family Department of Pediatrics (A.L.C.), University of Iowa, Iowa City
| | - Vincent A Magnotta
- From the Department of Psychiatry (E.v.d.P., T.R.K.), University of Iowa Hospitals & Clinics; and the Departments of Psychiatry (D.R.L., A.T., E.A.E., P.C.N.), Biostatistics (D.R.L., A.T.), and Radiology (V.A.M.) and Stead Family Department of Pediatrics (A.L.C.), University of Iowa, Iowa City
| | - Peggy C Nopoulos
- From the Department of Psychiatry (E.v.d.P., T.R.K.), University of Iowa Hospitals & Clinics; and the Departments of Psychiatry (D.R.L., A.T., E.A.E., P.C.N.), Biostatistics (D.R.L., A.T.), and Radiology (V.A.M.) and Stead Family Department of Pediatrics (A.L.C.), University of Iowa, Iowa City
| |
Collapse
|
257
|
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
|
258
|
McLauchlan DJ, Lancaster T, Craufurd D, Linden DEJ, Rosser AE. Insensitivity to loss predicts apathy in huntington's disease. Mov Disord 2019; 34:1381-1391. [PMID: 31361357 DOI: 10.1002/mds.27787] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/03/2019] [Accepted: 06/10/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Apathy is a deficit in goal-directed behavior that significantly affects quality of life and function. It is common in Huntington's disease and other disorders affecting corticostriatal pathways. Deficits in processing of reward, altered effort, and executive dysfunction are associated with apathy in other disorders, but the cognitive processes leading to apathy in Huntington's disease remain largely unknown. A previously reported deficit in learning from losses in Huntington's disease raises the possibility of a hitherto unrecognized mechanism leading to apathy. This study's objective was to delineate the cognitive processes associated with apathy in HD. METHODS We tested 51 Huntington's disease participants and 26 controls on a battery of novel and established measures to assess the contribution to apathy in Huntington's disease of executive function, reward value, reward-effort calculations, instrumental learning, and response to reward and loss. RESULTS Huntington's disase participants had deficits in instrumental learning with impaired response to loss, but no evidence to suggest altered reward-related behavior or effort. We also saw an executive dysfunction contribution to apathy in Huntington's disease. DISCUSSION We report the novel finding that apathy in Huntington's disease is associated with blunted responses to losses and impaired instrumental learning. This association is consistent with the known early degeneration of the indirect pathway and amygdala involvement in apathy in Huntington's disease, but is previously unreported in any disorder. In keeping with the comparative preservation of the ventral striatum and orbitofrontal cortex in Huntington's disease, reward valuation and reward-effort calculations did not contribute to apathy. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Duncan J McLauchlan
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK.,MRC Center for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK.,Cardiff University Brain Research Imaging Center, Cardiff University, Cardiff, UK
| | - Thomas Lancaster
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK.,MRC Center for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK.,Cardiff University Brain Research Imaging Center, Cardiff University, Cardiff, UK
| | - David Craufurd
- Manchester Center for Genomic Medicine, Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Center, Manchester, UK.,St. Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Center, Manchester, UK
| | - David E J Linden
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK.,MRC Center for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK.,Cardiff University Brain Research Imaging Center, Cardiff University, Cardiff, UK.,School for Mental Health and Neuroscience, Fac. Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Anne E Rosser
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK.,MRC Center for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK.,Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
259
|
Radulescu CI, Garcia-Miralles M, Sidik H, Bardile CF, Yusof NABM, Lee HU, Ho EXP, Chu CW, Layton E, Low D, De Sessions PF, Pettersson S, Ginhoux F, Pouladi MA. Manipulation of microbiota reveals altered callosal myelination and white matter plasticity in a model of Huntington disease. Neurobiol Dis 2019; 127:65-75. [DOI: 10.1016/j.nbd.2019.02.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 02/02/2019] [Accepted: 02/20/2019] [Indexed: 01/08/2023] Open
|
260
|
Rudenko O, Springer C, Skov LJ, Madsen AN, Hasholt L, Nørremølle A, Holst B. Ghrelin-mediated improvements in the metabolic phenotype in the R6/2 mouse model of Huntington's disease. J Neuroendocrinol 2019; 31:e12699. [PMID: 30776164 DOI: 10.1111/jne.12699] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/17/2019] [Accepted: 02/14/2019] [Indexed: 12/18/2022]
Abstract
Huntington's disease (HD) is a heritable neurodegenerative disorder, characterised by metabolic disturbances, along with cognitive and psychiatric impairments. Targeting metabolic HD dysfunction via the maintenance of body weight and fat mass and restoration of peripheral energy metabolism can improve the progression of neurological symptoms. In this respect, we focused on the therapeutic potential of the orexigenic peptide hormone ghrelin, which plays an important role in promoting a positive energy balance. In the present study, we found a significant disruption of circadian metabolic regulation in a R6/2 mouse HD model in the late stage of disease. Daily circadian rhythms of activity, energy expenditure, respiratory exchange ratio and feeding were strongly attenuated in R6/2 mice. During the rest phase, R6/2 mice had a higher total activity, elevated energy expenditure and excessive water consumption compared to control mice. We also found that, in the late stage of disease, R6/2 mice had ghrelin axis deficiency as a result of low circulating ghrelin levels, in addition to down-regulation of the ghrelin receptor and several key signalling molecules in the hypothalamus, as well as a reduced responsiveness to exogenous peripheral ghrelin. We demonstrated that, in pre-symptomatic mice, responsiveness to ghrelin is preserved. Chronic ghrelin treatment efficiently increased lean body mass and decreased the energy expenditure and fat utilisation of R6/2 mice in the early stage of disease. In addition, ghrelin treatment was also effective in the normalisation of drinking behaviour and the rest activity of these mice. Ghrelin treatment could provide a novel therapeutic possibility for delaying disease progression; however, deficiency in ghrelin receptor expression could limit its therapeutic potential in the late stage of disease.
Collapse
Affiliation(s)
- Olga Rudenko
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Cecilie Springer
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Louisa J Skov
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Andreas N Madsen
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Lis Hasholt
- Medical Genetics Program, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Nørremølle
- Medical Genetics Program, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte Holst
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
261
|
Shishegar R, Rajapakse S, Georgiou-Karistianis N. Altered Cortical Morphometry in Pre-manifest Huntington's Disease: Cross-sectional Data from the IMAGE-HD Study. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2019; 2019:2844-2847. [PMID: 31946485 DOI: 10.1109/embc.2019.8857240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Huntington's disease (HD) is an inherited progressive neurodegenerative disease mainly associated with subcortical striatal atrophy. There is also strong evidence showing cerebral atrophy and cortical thinning; however, limited research has investigated altered patterns of cortical folding in this disease. Here, we investigated cortical morphometry via both gyrification index (GI, a measure of cortical folding) and cortical thinning. The localized GI was examined using a novel GI, namely LB-GI. As part of a cross-sectional study, pre-manifest (pre-HD) individuals (n = 29) and matched controls (n = 29) underwent T1-MRI using data from the IMAGE-HD study. Compared to controls, pre-HD individuals demonstrated significantly lower GI in the left superior parietal and the right superior temporal regions and greater cortical thinning in the bilateral pre-central and the superior frontal gyri and left caudal middle frontal gyrus, as well as the superior parietal region. For the first time, we report evidence of abnormal localized cortical folding in pre-HD. We also provide evidence that cortical folding impacts different regions of the cortical surface more so than cortical thickness. As a result, we propose a potential new biological marker that may increase our understanding of the neuropathology of HD. Greater understanding of brain changes could inform new therapeutic approaches and target points for clinical trials.
Collapse
|
262
|
Cortical neurodevelopment in pre-manifest Huntington's disease. NEUROIMAGE-CLINICAL 2019; 23:101913. [PMID: 31491822 PMCID: PMC6627026 DOI: 10.1016/j.nicl.2019.101913] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 11/20/2022]
Abstract
Background The expression of the HTT CAG repeat expansion mutation causes neurodegeneration in Huntington's disease (HD). Objectives: In light of the – mainly in-vitro – evidence suggesting an additional role of huntingtin in neurodevelopment we used 3T MRI to test the hypothesis that in CAG-expanded individuals without clinical signs of HD (preHD) there is evidence for neurodevelopmental abnormalities. Methods We specifically investigated the complexity of cortical folding, a measure of cortical neurodevelopment, employing a novel method to quantify local fractal dimension (FD) measures that uses spherical harmonic reconstructions. Results The complexity of cortical folding differed at a group level between preHD (n = 57) and healthy volunteers (n = 57) in areas of the motor and visual system as well as temporal cortical areas. However, there was no association between the complexity of cortical folding and the loss in putamen volume that was clearly evident in preHD. Conclusions Our results suggest that HTT CAG repeat length may have an influence on cortical folding without evidence that this leads to developmental pathology or was clinically meaningful. This suggests that the HTT CAG-repeat expansion mutation may influence the processes governing cortical neurodevelopment; however, that influence seems independent of the events that lead to neurodegeneration. Measures of cortical neurodevelopment in preclinical Huntington's disease (HD) gene carriers differ from healthy volunteers The influence on cortical folding of the HD gene was not associated with developmental pathology or clinically meaningful The influence of the HD gene on cortical neurodevelopment may differ from that on neurodegeneration
Collapse
|
263
|
Flower M, Lomeikaite V, Ciosi M, Cumming S, Morales F, Lo K, Hensman Moss D, Jones L, Holmans P, Monckton DG, Tabrizi SJ. MSH3 modifies somatic instability and disease severity in Huntington's and myotonic dystrophy type 1. Brain 2019; 142:awz115. [PMID: 31216018 PMCID: PMC6598626 DOI: 10.1093/brain/awz115] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 01/31/2019] [Accepted: 02/27/2019] [Indexed: 12/22/2022] Open
Abstract
The mismatch repair gene MSH3 has been implicated as a genetic modifier of the CAG·CTG repeat expansion disorders Huntington's disease and myotonic dystrophy type 1. A recent Huntington's disease genome-wide association study found rs557874766, an imputed single nucleotide polymorphism located within a polymorphic 9 bp tandem repeat in MSH3/DHFR, as the variant most significantly associated with progression in Huntington's disease. Using Illumina sequencing in Huntington's disease and myotonic dystrophy type 1 subjects, we show that rs557874766 is an alignment artefact, the minor allele for which corresponds to a three-repeat allele in MSH3 exon 1 that is associated with a reduced rate of somatic CAG·CTG expansion (P = 0.004) and delayed disease onset (P = 0.003) in both Huntington's disease and myotonic dystrophy type 1, and slower progression (P = 3.86 × 10-7) in Huntington's disease. RNA-Seq of whole blood in the Huntington's disease subjects found that repeat variants are associated with MSH3 and DHFR expression. A transcriptome-wide association study in the Huntington's disease cohort found increased MSH3 and DHFR expression are associated with disease progression. These results suggest that variation in the MSH3 exon 1 repeat region influences somatic expansion and disease phenotype in Huntington's disease and myotonic dystrophy type 1, and suggests a common DNA repair mechanism operates in both repeat expansion diseases.
Collapse
Affiliation(s)
- Michael Flower
- Department of Neurodegenerative Disease and Dementia Research Institute, UCL, UK
| | - Vilija Lomeikaite
- Institute of Molecular, Cell and Systems Biology, University of Glasgow, UK
| | - Marc Ciosi
- Institute of Molecular, Cell and Systems Biology, University of Glasgow, UK
| | - Sarah Cumming
- Institute of Molecular, Cell and Systems Biology, University of Glasgow, UK
| | - Fernando Morales
- Institute of Molecular, Cell and Systems Biology, University of Glasgow, UK
- Instituto de Investigaciones en Salud (INISA), Universidad de Costa Rica, San José, Costa Rica
| | - Kitty Lo
- School of Mathematics and Statistics, University of Sydney, Australia
| | - Davina Hensman Moss
- Department of Neurodegenerative Disease and Dementia Research Institute, UCL, UK
| | - Lesley Jones
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, UK
| | - Peter Holmans
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, UK
| | - Darren G Monckton
- Institute of Molecular, Cell and Systems Biology, University of Glasgow, UK
| | - Sarah J Tabrizi
- Department of Neurodegenerative Disease and Dementia Research Institute, UCL, UK
| |
Collapse
|
264
|
Tabrizi SJ, Leavitt BR, Landwehrmeyer GB, Wild EJ, Saft C, Barker RA, Blair NF, Craufurd D, Priller J, Rickards H, Rosser A, Kordasiewicz HB, Czech C, Swayze EE, Norris DA, Baumann T, Gerlach I, Schobel SA, Paz E, Smith AV, Bennett CF, Lane RM. Targeting Huntingtin Expression in Patients with Huntington's Disease. N Engl J Med 2019; 380:2307-2316. [PMID: 31059641 DOI: 10.1056/nejmoa1900907] [Citation(s) in RCA: 466] [Impact Index Per Article: 77.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Huntington's disease is an autosomal-dominant neurodegenerative disease caused by CAG trinucleotide repeat expansion in HTT, resulting in a mutant huntingtin protein. IONIS-HTTRx (hereafter, HTTRx) is an antisense oligonucleotide designed to inhibit HTT messenger RNA and thereby reduce concentrations of mutant huntingtin. METHODS We conducted a randomized, double-blind, multiple-ascending-dose, phase 1-2a trial involving adults with early Huntington's disease. Patients were randomly assigned in a 3:1 ratio to receive HTTRx or placebo as a bolus intrathecal administration every 4 weeks for four doses. Dose selection was guided by a preclinical model in mice and nonhuman primates that related dose level to reduction in the concentration of huntingtin. The primary end point was safety. The secondary end point was HTTRx pharmacokinetics in cerebrospinal fluid (CSF). Prespecified exploratory end points included the concentration of mutant huntingtin in CSF. RESULTS Of the 46 patients who were enrolled in the trial, 34 were randomly assigned to receive HTTRx (at ascending dose levels of 10 to 120 mg) and 12 were randomly assigned to receive placebo. Each patient received all four doses and completed the trial. Adverse events, all of grade 1 or 2, were reported in 98% of the patients. No serious adverse events were seen in HTTRx-treated patients. There were no clinically relevant adverse changes in laboratory variables. Predose (trough) concentrations of HTTRx in CSF showed dose dependence up to doses of 60 mg. HTTRx treatment resulted in a dose-dependent reduction in the concentration of mutant huntingtin in CSF (mean percentage change from baseline, 10% in the placebo group and -20%, -25%, -28%, -42%, and -38% in the HTTRx 10-mg, 30-mg, 60-mg, 90-mg, and 120-mg dose groups, respectively). CONCLUSIONS Intrathecal administration of HTTRx to patients with early Huntington's disease was not accompanied by serious adverse events. We observed dose-dependent reductions in concentrations of mutant huntingtin. (Funded by Ionis Pharmaceuticals and F. Hoffmann-La Roche; ClinicalTrials.gov number, NCT02519036.).
Collapse
Affiliation(s)
- Sarah J Tabrizi
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Blair R Leavitt
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - G Bernhard Landwehrmeyer
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Edward J Wild
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Carsten Saft
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Roger A Barker
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Nick F Blair
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - David Craufurd
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Josef Priller
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Hugh Rickards
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Anne Rosser
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Holly B Kordasiewicz
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Christian Czech
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Eric E Swayze
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Daniel A Norris
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Tiffany Baumann
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Irene Gerlach
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Scott A Schobel
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Erika Paz
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Anne V Smith
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - C Frank Bennett
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Roger M Lane
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| |
Collapse
|
265
|
Poudel GR, Harding IH, Egan GF, Georgiou-Karistianis N. Network spread determines severity of degeneration and disconnection in Huntington's disease. Hum Brain Mapp 2019; 40:4192-4201. [PMID: 31187915 DOI: 10.1002/hbm.24695] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 03/13/2019] [Accepted: 05/27/2019] [Indexed: 02/01/2023] Open
Abstract
Trans-neuronal propagation of mutant huntingtin protein contributes to the organised spread of cortico-striatal degeneration and disconnection in Huntington's disease (HD). We investigated whether the network diffusion model, which models transneuronal spread as diffusion of pathological proteins via the brain connectome, can determine the severity of neural degeneration and disconnection in HD. We used structural magnetic resonance imaging (MRI) and high-angular resolution diffusion weighted imaging (DWI) data from symptomatic Huntington's disease (HD) (N = 26) and age-matched healthy controls (N = 26) to measure neural degeneration and disconnection in HD. The network diffusion model was used to test whether disease spread, via the human brain connectome, is a viable mechanism to explain the distribution of pathology across the brain. We found that an eigenmode identified in the healthy human brain connectome Laplacian matrix, accurately predicts the cortico-striatal spatial pattern of degeneration in HD. Furthermore, the spread of neural degeneration from sub-cortical brain regions, including the accumbens and thalamus, generates a spatial pattern which represents the typical neurodegenerative characteristics in HD. The white matter connections connecting the nodes with the highest amount of disease factors, when diffusion based disease spread is initiated from the striatum, were found to be most vulnerable to disconnection in HD. These findings suggest that trans-neuronal diffusion of mutant huntingtin protein across the human brain connectome may explain the pattern of gray matter degeneration and white matter disconnection that are hallmarks of HD.
Collapse
Affiliation(s)
- Govinda R Poudel
- Mary Mackillop Institute for Health Research, Australian Catholic University, Melbourne, Australia
| | - Ian H Harding
- School of Psychological Sciences & Monash Institute of Cognitive and Clinical Neurosciences, Monash University, Clayton, Victoria, Australia
| | - Gary F Egan
- School of Psychological Sciences & Monash Institute of Cognitive and Clinical Neurosciences, Monash University, Clayton, Victoria, Australia.,Monash Biomedical Imaging (MBI), Monash University, Melbourne, VIC, Australia.,ARC Centre of Excellence for Integrative Brain Function, Monash University, Clayton, Victoria, Australia
| | - Nellie Georgiou-Karistianis
- School of Psychological Sciences & Monash Institute of Cognitive and Clinical Neurosciences, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
266
|
Patassini S, Begley P, Xu J, Church SJ, Kureishy N, Reid SJ, Waldvogel HJ, Faull RLM, Snell RG, Unwin RD, Cooper GJS. Cerebral Vitamin B5 (D-Pantothenic Acid) Deficiency as a Potential Cause of Metabolic Perturbation and Neurodegeneration in Huntington's Disease. Metabolites 2019; 9:E113. [PMID: 31212603 PMCID: PMC6630497 DOI: 10.3390/metabo9060113] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 05/31/2019] [Accepted: 05/31/2019] [Indexed: 12/15/2022] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by an expanded CAG repeat in exon 1 of the HTT gene. HD usually manifests in mid-life with loss of GABAergic projection neurons from the striatum accompanied by progressive atrophy of the putamen followed by other brain regions, but linkages between the genetics and neurodegeneration are not understood. We measured metabolic perturbations in HD-human brain in a case-control study, identifying pervasive lowering of vitamin B5, the obligatory precursor of coenzyme A (CoA) that is essential for normal intermediary metabolism. Cerebral pantothenate deficiency is a newly-identified metabolic defect in human HD that could potentially: (i) impair neuronal CoA biosynthesis; (ii) stimulate polyol-pathway activity; (iii) impair glycolysis and tricarboxylic acid cycle activity; and (iv) modify brain-urea metabolism. Pantothenate deficiency could lead to neurodegeneration/dementia in HD that might be preventable by treatment with vitamin B5.
Collapse
Affiliation(s)
- Stefano Patassini
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M19 9NT, UK.
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland 1142, New Zealand.
- Owlstone Medical, Cambridge Science Park, Cambridge CB4 0GJ, UK.
| | - Paul Begley
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M19 9NT, UK.
| | - Jingshu Xu
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M19 9NT, UK.
- Manchester Cancer Research Centre Building, The University of Manchester, Manchester M20 4GJ, UK.
| | - Stephanie J Church
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M19 9NT, UK.
| | - Nina Kureishy
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M19 9NT, UK.
| | - Suzanne J Reid
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland 1142, New Zealand.
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand.
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand.
| | - Richard L M Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand.
| | - Russell G Snell
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland 1142, New Zealand.
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand.
| | - Richard D Unwin
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M19 9NT, UK.
| | - Garth J S Cooper
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M19 9NT, UK.
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland 1142, New Zealand.
- Manchester Cancer Research Centre Building, The University of Manchester, Manchester M20 4GJ, UK.
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland 1142, New Zealand.
| |
Collapse
|
267
|
Quantitative grip force assessment of muscular weakness in chronic inflammatory demyelinating polyneuropathy. BMC Neurol 2019; 19:118. [PMID: 31176377 PMCID: PMC6556046 DOI: 10.1186/s12883-019-1339-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 05/22/2019] [Indexed: 11/10/2022] Open
Abstract
Background In patients suffering from Chronic Inflammatory Demyelinating Polyneuropathy (CIDP) disease severity is assessed by Medical Research Counsil (MRC) Scale or Inflammatory Neuropathy Cause and Treatment (INCAT) disability score. However, none of these methods is appropriate to objectively assess muscle weakness or to detect very small subclinical changes. More objective and quantitative measures are needed in order to evaluate treatment efficiency or to detect subclinical affection of upper limps for early diagnosis. The goal of our study was to objectively quantify muscular weakness in CIDP patients with the non-invasive Quantitative Motor (Q-Motor) test of Grip Force Assessment (QGFA) as well as the Involuntary Movement Assessment (QIMA) and to search for differences between typical and atypical CIDP variants. In addition, we hypothesized that Q-Motor findings correlate with disease severity scales such as MRC or INCAT score. Methods In this cross-sectional exploratory proof-of-concept study subjects with confirmed diagnosis of typical or atypical CIDP were examined and compared to healthy controls (HC). For Q-Motor tests all subjects had to lift a device (250 g and 500 g) equipped with an electromagnetic sensor that measured grip force (GF) and three-dimensional changes in position and orientation. The measures “grip force variability” (GFV), “position index” (PI) and “orientation index” (OI) were provided to assess involuntary movements due to muscular weakness. Results 33 patients with CIDP and 28 HC were included. All measures were significantly elevated in CIDP patients for both devices in the right and left hand compared to healthy controls. Subgroup analysis revealed no differences between typical and atypical CIDP variants. INCAT score only weakly correlated with OI and PI. However, there was a stronger correlation between MRC and QIMA parameters in both hands. Conclusion Q-Motor assessments were capable to objectively assess muscular weakness in CIDP. In particular, QIMA measures detected subclinical generalized muscle weakness even in patients with milder disability. Sensitivity and rater-independence of Q-Motor assessments support a further exploration of QIMA measures as potential endpoints for future clinical trials in CIDP.
Collapse
|
268
|
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
|
269
|
Johnson EB, Gregory S. Huntington's disease: Brain imaging in Huntington's disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 165:321-369. [PMID: 31481169 DOI: 10.1016/bs.pmbts.2019.04.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Huntington's disease (HD) gene-carriers show prominent neuronal loss by end-stage disease, and the use of magnetic resonance imaging (MRI) has been increasingly used to quantify brain changes during earlier stages of the disease. MRI offers an in vivo method of measuring structural and functional brain change. The images collected via MRI are processed to measure different anatomical features, such as brain volume, macro- and microstructural changes within white matter and functional brain activity. Structural imaging has demonstrated significant volume loss across multiple white and gray matter regions in HD, particularly within subcortical structures. There also appears to be increasing disorganization of white matter tracts and between-region connectivity with increasing disease progression. Finally, functional changes are thought to represent changes in brain activity underlying compensatory mechanisms in HD. This chapter will provide an overview of the principles of MRI and practicalities associated with using MRI in HD studies, and summarize findings from MRI studies investigating brain structure and function in HD.
Collapse
Affiliation(s)
- Eileanoir B Johnson
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Sarah Gregory
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.
| |
Collapse
|
270
|
Modulation of Beta Oscillations for Implicit Motor Timing in Primate Sensorimotor Cortex during Movement Preparation. Neurosci Bull 2019; 35:826-840. [PMID: 31062334 DOI: 10.1007/s12264-019-00387-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 12/09/2018] [Indexed: 01/03/2023] Open
Abstract
Motor timing is an important part of sensorimotor control. Previous studies have shown that beta oscillations embody the process of temporal perception in explicit timing tasks. In contrast, studies focusing on beta oscillations in implicit timing tasks are lacking. In this study, we set up an implicit motor timing task and found a modulation pattern of beta oscillations with temporal perception during movement preparation. We trained two macaques in a repetitive visually-guided reach-to-grasp task with different holding intervals. Spikes and local field potentials were recorded from microelectrode arrays in the primary motor cortex, primary somatosensory cortex, and posterior parietal cortex. We analyzed the association between beta oscillations and temporal interval in fixed-duration experiments (500 ms as the Short Group and 1500 ms as the Long Group) and random-duration experiments (500 ms to 1500 ms). The results showed that the peak beta frequencies in both experiments ranged from 15 Hz to 25 Hz. The beta power was higher during the hold period than the movement (reach and grasp) period. Further, in the fixed-duration experiments, the mean power as well as the maximum rate of change of beta power in the first 300 ms were higher in the Short Group than in the Long Group when aligned with the Center Hit event. In contrast, in the random-duration experiments, the corresponding values showed no statistical differences among groups. The peak latency of beta power was shorter in the Short Group than in the Long Group in the fixed-duration experiments, while no consistent modulation pattern was found in the random-duration experiments. These results indicate that beta oscillations can modulate with temporal interval in their power mode. The synchronization period of beta power could reflect the cognitive set maintaining working memory of the temporal structure and attention.
Collapse
|
271
|
Lai HY, Saavedra-Pena G, Sodini CG, Sze V, Heldt T. Measuring Saccade Latency Using Smartphone Cameras. IEEE J Biomed Health Inform 2019; 24:885-897. [PMID: 31056528 DOI: 10.1109/jbhi.2019.2913846] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Accurate quantification of neurodegenerative disease progression is an ongoing challenge that complicates efforts to understand and treat these conditions. Clinical studies have shown that eye movement features may serve as objective biomarkers to support diagnosis and tracking of disease progression. Here, we demonstrate that saccade latency-an eye movement measure of reaction time-can be measured robustly outside of the clinical environment with a smartphone camera. METHODS To enable tracking of saccade latency in large cohorts of patients and control subjects, we combined a deep convolutional neural network for gaze estimation with a model-based approach for saccade onset determination that provides automated signal-quality quantification and artifact rejection. RESULTS Simultaneous recordings with a smartphone and a high-speed camera resulted in negligible differences in saccade latency distributions. Furthermore, we demonstrated that the constraint of chinrest support can be removed when recording healthy subjects. Repeat smartphone-based measurements of saccade latency in 11 self-reported healthy subjects resulted in an intraclass correlation coefficient of 0.76, showing our approach has good to excellent test-retest reliability. Additionally, we conducted more than 19 000 saccade latency measurements in 29 self-reported healthy subjects and observed significant intra- and inter-subject variability, which highlights the importance of individualized tracking. Lastly, we showed that with around 65 measurements we can estimate mean saccade latency to within less-than-10-ms precision, which takes within 4 min with our setup. CONCLUSION AND SIGNIFICANCE By enabling repeat measurements of saccade latency and its distribution in individual subjects, our framework opens the possibility of quantifying patient state on a finer timescale in a broader population than previously possible.
Collapse
|
272
|
Olfactory bulb atrophy and caspase activation observed in the BACHD rat models of Huntington disease. Neurobiol Dis 2019; 125:219-231. [DOI: 10.1016/j.nbd.2019.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 12/14/2018] [Accepted: 02/04/2019] [Indexed: 01/08/2023] Open
|
273
|
Osborne-Crowley K, Andrews SC, Labuschagne I, Nair A, Scahill R, Craufurd D, Tabrizi SJ, Stout JC. Apathy Associated With Impaired Recognition of Happy Facial Expressions in Huntington's Disease. J Int Neuropsychol Soc 2019; 25:453-461. [PMID: 30767839 PMCID: PMC6542690 DOI: 10.1017/s1355617718001224] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Previous research has demonstrated an association between emotion recognition and apathy in several neurological conditions involving fronto-striatal pathology, including Parkinson's disease and brain injury. In line with these findings, we aimed to determine whether apathetic participants with early Huntington's disease (HD) were more impaired on an emotion recognition task compared to non-apathetic participants and healthy controls. METHODS We included 43 participants from the TRACK-HD study who reported apathy on the Problem Behaviours Assessment - short version (PBA-S), 67 participants who reported no apathy, and 107 controls matched for age, sex, and level of education. During their baseline TRACK-HD visit, participants completed a battery of cognitive and psychological tests including an emotion recognition task, the Hospital Depression and Anxiety Scale (HADS) and were assessed on the PBA-S. RESULTS Compared to the non-apathetic group and the control group, the apathetic group were impaired on the recognition of happy facial expressions, after controlling for depression symptomology on the HADS and general disease progression (Unified Huntington's Disease Rating Scale total motor score). This was despite no difference between the apathetic and non-apathetic group on overall cognitive functioning assessed by a cognitive composite score. CONCLUSIONS Impairment of the recognition of happy expressions may be part of the clinical picture of apathy in HD. While shared reliance on frontostriatal pathways may broadly explain associations between emotion recognition and apathy found across several patient groups, further work is needed to determine what relationships exist between recognition of specific emotions, distinct subtypes of apathy and underlying neuropathology. (JINS, 2019, 25, 453-461).
Collapse
Affiliation(s)
- Katherine Osborne-Crowley
- 1Huntington's Disease Centre,University College London,Institute of Neurology, and National Hospital for Neurology and Neurosurgery,London,United Kingdom
| | - Sophie C Andrews
- 2Monash Institute of Cognitive and Clinical Neurosciences,School of Psychological Sciences,Monash University,Melbourne,Australia
| | - Izelle Labuschagne
- 3Cognition and Emotion Research Centre,School of Psychology,Australian Catholic University,Melbourne,Australia
| | - Akshay Nair
- 1Huntington's Disease Centre,University College London,Institute of Neurology, and National Hospital for Neurology and Neurosurgery,London,United Kingdom
| | - Rachael Scahill
- 1Huntington's Disease Centre,University College London,Institute of Neurology, and National Hospital for Neurology and Neurosurgery,London,United Kingdom
| | - David Craufurd
- 4Manchester Centre for Genomic Medicine,Division of Evolution and Genomic Sciences,School of Biological Sciences, Faculty of Biology,Medicine and Health,University of Manchester,Manchester Academic Health Science Centre,Manchester,United Kingdom
| | - Sarah J Tabrizi
- 1Huntington's Disease Centre,University College London,Institute of Neurology, and National Hospital for Neurology and Neurosurgery,London,United Kingdom
| | - Julie C Stout
- 2Monash Institute of Cognitive and Clinical Neurosciences,School of Psychological Sciences,Monash University,Melbourne,Australia
| |
Collapse
|
274
|
Intrinsic mutant HTT-mediated defects in oligodendroglia cause myelination deficits and behavioral abnormalities in Huntington disease. Proc Natl Acad Sci U S A 2019; 116:9622-9627. [PMID: 31015293 DOI: 10.1073/pnas.1818042116] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
White matter abnormalities are a nearly universal pathological feature of neurodegenerative disorders including Huntington disease (HD). A long-held assumption is that this white matter pathology is simply a secondary outcome of the progressive neuronal loss that manifests with advancing disease. Using a mouse model of HD, here we show that white matter and myelination abnormalities are an early disease feature appearing before the manifestation of any behavioral abnormalities or neuronal loss. We further show that selective inactivation of mutant huntingtin (mHTT) in the NG2+ oligodendrocyte progenitor cell population prevented myelin abnormalities and certain behavioral deficits in HD mice. Strikingly, the improvements in behavioral outcomes were seen despite the continued expression of mHTT in nonoligodendroglial cells including neurons, astrocytes, and microglia. Using RNA-seq and ChIP-seq analyses, we implicate a pathogenic mechanism that involves enhancement of polycomb repressive complex 2 (PRC2) activity by mHTT in the intrinsic oligodendroglial dysfunction and myelination deficits observed in HD. Our findings challenge the long-held dogma regarding the etiology of white matter pathology in HD and highlight the contribution of epigenetic mechanisms to the observed intrinsic oligodendroglial dysfunction. Our results further suggest that ameliorating white matter pathology and oligodendroglial dysfunction may be beneficial for HD.
Collapse
|
275
|
D'Aurizio G, Migliore S, Curcio G, Squitieri F. Safer Attitude to Risky Decision-Making in Premanifest Huntington's Disease Subjects. Front Psychol 2019; 10:846. [PMID: 31057466 PMCID: PMC6477566 DOI: 10.3389/fpsyg.2019.00846] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 03/29/2019] [Indexed: 01/18/2023] Open
Abstract
Huntington’s disease (HD) is an inherited neurodegenerative disorder characterized by involuntary, jerky movements, incoordination, behavioral changes and subtle executive and cognitive impairment starting before motor symptoms. Our study aimed to assess the risky decision-making process in premanifest (pre) HD subjects, by means Game of Dice Task (GDT). As dependent variables, several GDT outcomes have been taken into consideration. We recruited 30 subjects (15 females) with preHD (i.e., Diagnosis Confidence Level < 4; Total Motor Score < 10), and 21 age, gender and education matched neurologically normal subjects (11 females). GDT is a computer-guided task where subjects are invited to watch the digits on which to bet and to evaluate the related potential risk to win or loss. Our results showed that decision and feedback times were longer in preHD than in neurologically normal group in both disadvantageous and advantageous choices. PreHD subjects provided a greater number of “safe” strategies, taken with longer decision-making time than neurologically normal subjects, showing a reduced propensity to risk. Such behavior, characterized by increased slowness in acting and providing answers, might contribute to delineate a behavioral and cognitive profile in preHD.
Collapse
Affiliation(s)
- Giulia D'Aurizio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Simone Migliore
- Huntington and Rare Diseases Unit, Fondazione IRCCS Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Italy
| | - Giuseppe Curcio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Ferdinando Squitieri
- Huntington and Rare Diseases Unit, Fondazione IRCCS Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Italy
| |
Collapse
|
276
|
Espinoza FA, Liu J, Ciarochi J, Turner JA, Vergara VM, Caprihan A, Misiura M, Johnson HJ, Long JD, Bockholt JH, Paulsen JS, Calhoun VD. Dynamic functional network connectivity in Huntington's disease and its associations with motor and cognitive measures. Hum Brain Mapp 2019; 40:1955-1968. [PMID: 30618191 PMCID: PMC6865767 DOI: 10.1002/hbm.24504] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/12/2018] [Accepted: 12/14/2018] [Indexed: 02/03/2023] Open
Abstract
Dynamic functional network connectivity (dFNC) is an expansion of traditional, static FNC that measures connectivity variation among brain networks throughout scan duration. We used a large resting-state fMRI (rs-fMRI) sample from the PREDICT-HD study (N = 183 Huntington disease gene mutation carriers [HDgmc] and N = 78 healthy control [HC] participants) to examine whole-brain dFNC and its associations with CAG repeat length as well as the product of scaled CAG length and age, a variable representing disease burden. We also tested for relationships between functional connectivity and motor and cognitive measurements. Group independent component analysis was applied to rs-fMRI data to obtain whole-brain resting state networks. FNC was defined as the correlation between RSN time-courses. Dynamic FNC behavior was captured using a sliding time window approach, and FNC results from each window were assigned to four clusters representing FNC states, using a k-means clustering algorithm. HDgmc individuals spent significantly more time in State-1 (the state with the weakest FNC pattern) compared to HC. However, overall HC individuals showed more FNC dynamism than HDgmc. Significant associations between FNC states and genetic and clinical variables were also identified. In FNC State-4 (the one that most resembled static FNC), HDgmc exhibited significantly decreased connectivity between the putamen and medial prefrontal cortex compared to HC, and this was significantly associated with cognitive performance. In FNC State-1, disease burden in HDgmc participants was significantly associated with connectivity between the postcentral gyrus and posterior cingulate cortex, as well as between the inferior occipital gyrus and posterior parietal cortex.
Collapse
Affiliation(s)
- Flor A. Espinoza
- Department of Translational Neuroscience, The Mind Research NetworkAlbuquerqueNew Mexico
| | - Jingyu Liu
- Department of Translational Neuroscience, The Mind Research NetworkAlbuquerqueNew Mexico
| | - Jennifer Ciarochi
- Department of Psychology and NeuroscienceGeorgia State UniversityAtlantaGeorgia
| | - Jessica A. Turner
- Department of Psychology and NeuroscienceGeorgia State UniversityAtlantaGeorgia
| | - Victor M. Vergara
- Department of Translational Neuroscience, The Mind Research NetworkAlbuquerqueNew Mexico
| | - Arvind Caprihan
- Department of Translational Neuroscience, The Mind Research NetworkAlbuquerqueNew Mexico
| | - Maria Misiura
- Department of Psychology and NeuroscienceGeorgia State UniversityAtlantaGeorgia
| | - Hans J. Johnson
- Department of Electrical and Computer EngineeringUniversity of IowaIowa CityIowa
- Department of PsychiatryUniversity of IowaIowa CityIowa
| | - Jeffrey D. Long
- Department of PsychiatryUniversity of IowaIowa CityIowa
- Department of BiostatisticsUniversity of IowaIowa CityIowa
| | - Jeremy H. Bockholt
- Department of Translational Neuroscience, The Mind Research NetworkAlbuquerqueNew Mexico
| | | | - Vince D. Calhoun
- Department of Translational Neuroscience, The Mind Research NetworkAlbuquerqueNew Mexico
- Department of Psychology and NeuroscienceGeorgia State UniversityAtlantaGeorgia
- Department of Electrical and Computer EngineeringUniversity of New MexicoAlbuquerqueNew Mexico
| |
Collapse
|
277
|
Abstract
In this study we longitudinally investigated the rate of microstructural alterations in the occipital cortex in different stages of Huntington's disease (HD) by applying an automated atlas-based approach to diffusion MRI data. Twenty-two premanifest (preHD), 10 early manifest HD (early HD) and 24 healthy control subjects completed baseline and two year follow-up scans. The preHD group was stratified based on the predicted years to disease onset into a far (preHD-A) and near (preHD-B) to disease onset group. Clinical and behavioral measures were collected per assessment time point. An automated atlas-based DTI analysis approach was used to obtain the mean, axial and radial diffusivities of the occipital cortex. We found that the longitudinal rate of diffusivity change in the superior occipital gyrus (SOG), middle occipital gyrus (MOG), and inferior occipital gyrus (IOG) was significantly higher in early HD compared to both preHD and controls (all p's ≤ 0.005), which can be interpreted as an increased rate of microstructural degeneration. Furthermore, the change rate in the diffusivity of the MOG could significantly discriminate between preHD-B compared to preHD-A and the other groups (all p's ≤ 0.04). Finally, we found an inverse correlation between the Stroop Word Reading task and diffusivities in the SOG and MOG (all p's ≤ 0.01). These findings suggest that measures obtained from the occipital cortex can serve as sensitive longitudinal biomarkers for disease progression in preHD-B and early HD. These could in turn be used to assess potential effects of proposed disease modifying therapies.
Collapse
|
278
|
Teo RTY, Ferrari Bardile C, Tay YL, Yusof NABM, Kreidy CA, Tan LJ, Pouladi MA. Impaired Remyelination in a Mouse Model of Huntington Disease. Mol Neurobiol 2019; 56:6873-6882. [PMID: 30937636 DOI: 10.1007/s12035-019-1579-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 03/20/2019] [Indexed: 01/26/2023]
Abstract
White matter (WM) abnormalities are a well-established feature of Huntington disease (HD), although their nature is not fully understood. Here, we asked whether remyelination as a measure of WM plasticity is impaired in a model of HD. Using the cuprizone assay, we examined demyelination and remyelination responses in YAC128 HD mice. Treatment with 0.2% cuprizone (CPZ) for 6 weeks resulted in significant reduction in mature (GSTπ-positive) oligodendrocyte counts and FluoroMyelin staining in the corpus callosum, leading to similar demyelination states in YAC128 and wild-type (WT) mice. Six weeks following cessation of CPZ, we observed robust remyelination in WT mice as indicated by an increase in mature oligodendrocyte counts and FluoroMyelin staining. In contrast, YAC128 mice exhibited an impaired remyelination response. The increase in mature oligodendrocyte counts in YAC128 HD mice following CPZ cessation was lower than that of WT. Furthermore, there was no increase in FluoroMyelin staining compared to the demyelinated state in YAC128 mice. We confirmed these findings using electron microscopy where the CPZ-induced reduction in myelinated axons was reversed following CPZ cessation in WT but not YAC128 mice. Our findings demonstrate that remyelination is impaired in YAC128 mice and suggest that WM plasticity may be compromised in HD.
Collapse
Affiliation(s)
- Roy Tang Yi Teo
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore
| | - Costanza Ferrari Bardile
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore
| | - Yi Lin Tay
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore
| | - Nur Amirah Binte Mohammad Yusof
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore
| | - Charbel A Kreidy
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore
| | - Liang Juin Tan
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore
| | - Mahmoud A Pouladi
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore.
- Department of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Department of Physiology, National University of Singapore, Singapore, 117597, Singapore.
| |
Collapse
|
279
|
Júlio F, Ribeiro MJ, Patrício M, Malhão A, Pedrosa F, Gonçalves H, Simões M, van Asselen M, Simões MR, Castelo-Branco M, Januário C. A Novel Ecological Approach Reveals Early Executive Function Impairments in Huntington's Disease. Front Psychol 2019; 10:585. [PMID: 30967810 PMCID: PMC6438896 DOI: 10.3389/fpsyg.2019.00585] [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] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 03/01/2019] [Indexed: 11/13/2022] Open
Abstract
Introduction: Impairments in executive functions are common in neurogenetic disorders such as Huntington's disease (HD) and are thought to significantly influence the patient's functional status. Reliable tools with higher ecological validity that can assess and predict the impact of executive dysfunction in daily-life performance are needed. This study aimed to develop and validate a novel non-immersive virtual reality task ("EcoKitchen") created with the purpose of capturing cognitive and functional changes shown by HD carriers without clinical manifestations of the disease (Premanifest HD), in a more realistic setting. Materials and Methods: We designed a virtual reality task with three blocks of increasing executive load. The performance of three groups (Controls, CTRL; Premanifest HD individuals, HP; Early Manifest HD patients, HD) was compared in four main components of the study protocol: the EcoKitchen; a subjective (self-report) measure - "The Adults and Older Adults Functional Assessment Inventory (IAFAI)"; the "Behavioural Assessment of Dysexecutive Syndrome battery (BADS)"; and a conventional neuropsychological test battery. We also examined statistical associations between EcoKitchen and the other executive, functional and clinical measures used. Results: The HD group showed deficits in all the assessment methods used. In contrast, the HP group was only found to be impaired in the EcoKitchen task, particularly in the most cognitively demanding blocks, where they showed a higher number of errors compared to the CTRL group. Statistically significant correlations were identified between the EcoKitchen, measures of the other assessment tools, and HD clinical features. Discussion: The EcoKitchen task, developed as an ecological executive function assessment tool, was found to be sensitive to early deficits in this domain. Critically, in premanifest HD individuals, it identifies dysfunction prior to symptom onset. Further it adds a potential tool for diagnosis and management of the patients' real-life problems.
Collapse
Affiliation(s)
- Filipa Júlio
- Faculty of Psychology and Education Sciences, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Maria J. Ribeiro
- Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Miguel Patrício
- Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Alexandre Malhão
- Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Fábio Pedrosa
- Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Hélio Gonçalves
- Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Marco Simões
- Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Marieke van Asselen
- Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Mário R. Simões
- Faculty of Psychology and Education Sciences, University of Coimbra, Coimbra, Portugal
- Center for Research in Neuropsychology and Cognitive Behavioral Intervention, Faculty of Psychology and Education Sciences, University of Coimbra, Coimbra, Portugal
| | - Miguel Castelo-Branco
- Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
- Institute of Nuclear Sciences Applied to Health, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Cristina Januário
- Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Coimbra University Hospital, Coimbra, Portugal
| |
Collapse
|
280
|
Thorley EM, Iyer RG, Wicks P, Curran C, Gandhi SK, Abler V, Anderson KE, Carlozzi NE. Understanding How Chorea Affects Health-Related Quality of Life in Huntington Disease: An Online Survey of Patients and Caregivers in the United States. PATIENT-PATIENT CENTERED OUTCOMES RESEARCH 2019; 11:547-559. [PMID: 29750428 PMCID: PMC6132452 DOI: 10.1007/s40271-018-0312-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Chorea is the hallmark motor feature of Huntington disease (HD) and can negatively impact daily functioning and health-related quality of life (HRQoL). OBJECTIVE The objective of this study was to evaluate how chorea impacts HRQoL and overall functioning among HD patients participating on the PatientsLikeMe website ( www.PatientsLikeMe.com ). METHODS A survey was provided to HD participants and/or their caregivers via PatientsLikeMe (9 February 2017-22 March 2017), comprising multiple-choice and open-ended questions designed to assess how chorea impacts HRQoL and overall functioning, and the importance of treating chorea. The HDQLIFE measurement system was used to evaluate patient-reported outcomes of chorea and compare Anxiety and Stigma scores in participants with high chorea versus those with low chorea [HDQLIFE Chorea scores ≥ 60 (n = 45) vs. < 60 (n = 38)]. RESULTS A total of 115 participants (n = 35 caregivers; n = 80 individuals with HD) were included in this study. Among those experiencing chorea (n = 83, 74% of respondents), 66% indicated it was 'Very Important' to manage chorea; however, only 47% agreed that their current medication regimen helped manage their movements. In general, respondents reported that chorea negatively affected HRQoL [HDQLIFE Chorea mean score (standard deviation): 59.3 (6.1)]. Consistent with this, significantly higher Anxiety (P = 0.0423) and stigma (P < 0.0001) scores were observed among respondents with high chorea than in those with low chorea. CONCLUSIONS These results highlight the negative impact of chorea on HRQoL and overall functioning in individuals with HD. Better chorea treatment options are needed to successfully manage symptoms and to help improve HRQoL in these individuals, and patient experiences of anxiety and stigma should be considered in treatment plans.
Collapse
Affiliation(s)
| | - Ravi G Iyer
- Teva Pharmaceutical Industries, 41 Moores Rd, Frazer, PA, 19355, USA
| | - Paul Wicks
- PatientsLikeMe, 160 2nd Street, Cambridge, MA, 02142, USA
| | - Chris Curran
- PatientsLikeMe, 160 2nd Street, Cambridge, MA, 02142, USA
| | - Sanjay K Gandhi
- Teva Pharmaceutical Industries, 41 Moores Rd, Frazer, PA, 19355, USA
| | - Victor Abler
- Teva Pharmaceutical Industries, 41 Moores Rd, Frazer, PA, 19355, USA
| | - Karen E Anderson
- Department of Psychiatry, Georgetown University, Washington, DC, USA
| | - Noelle E Carlozzi
- Department of Physical Medicine and Rehabilitation, University of Michigan, 500 S. State Street, Ann Arbor, MI, 48109, USA
| |
Collapse
|
281
|
Purcell NL, Goldman JG, Ouyang B, Bernard B, O'Keefe JA. The Effects of Dual-Task Cognitive Interference and Environmental Challenges on Balance in Huntington's Disease. Mov Disord Clin Pract 2019; 6:202-212. [PMID: 30949551 PMCID: PMC6417749 DOI: 10.1002/mdc3.12720] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/05/2018] [Accepted: 12/07/2018] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Huntington's disease (HD) is characterized by chorea, balance and gait impairments, and cognitive deficits, which increase fall risk. Dual task (DT) and environmentally challenging paradigms reflect balance related to everyday life. Furthermore, the impact of cognitive deficits on balance dysfunction and falls in HD is unknown. OBJECTIVE To determine the impact of DT interference, sensory feedback, and cognitive performance on balance and falls in HD. METHODS Seventeen participants with HD (55 ± 9.7 years) and 17 age-matched controls (56.5 ± 9.3 years) underwent quantitative balance testing with APDM inertial sensors. Postural sway was assessed during conditions of manipulated stance, vision, proprioception, and cognitive demand. The DT was a concurrent verbal fluency task. Neuropsychological assessments testing multiple cognitive domains were also administered. RESULTS HD participants exhibited significantly greater total sway area, jerk, and variability under single-task (ST) and DT conditions compared to controls (P = 0.0002 - < 0.0001). They also demonstrated greater DT interference with vision removed for total sway area (P = 0.01) and variability (P = 0.02). Significantly worse postural control was observed in HD with vision removed and reduced proprioception (P = 0.001 - 0.01). Decreased visuospatial performance correlated with greater total sway and jerk (P = 0.01; 0.009). No balance parameters correlated with retrospective falls in HD. CONCLUSIONS HD participants have worse postural control under DT, limited proprioception/vision, and greater DT interference with a narrowed base and no visual input. These findings may have implications for designing motor and cognitive strategies to improve balance in HD.
Collapse
Affiliation(s)
| | - Jennifer G. Goldman
- Department of Neurological Sciences, Section of Parkinson Disease and Movement DisordersRush University Medical CenterChicagoILUSA
| | - Bichun Ouyang
- Department of Neurological Sciences, Section of Parkinson Disease and Movement DisordersRush University Medical CenterChicagoILUSA
| | - Bryan Bernard
- Department of Neurological Sciences, Section of Parkinson Disease and Movement DisordersRush University Medical CenterChicagoILUSA
| | - Joan A. O'Keefe
- Department of Cell and Molecular MedicineRush University Medical CenterChicagoILUSA
- Department of Neurological Sciences, Section of Parkinson Disease and Movement DisordersRush University Medical CenterChicagoILUSA
| |
Collapse
|
282
|
Martínez-Horta S, Moreu A, Perez-Perez J, Sampedro F, Horta-Barba A, Pagonabarraga J, Gomez-Anson B, Lozano-Martinez GA, Lopez-Mora DA, Camacho V, Fernández-León A, Carrió I, Kulisevsky J. The impact of bilingualism on brain structure and function in Huntington's disease. Parkinsonism Relat Disord 2019; 60:92-97. [DOI: 10.1016/j.parkreldis.2018.09.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 09/01/2018] [Accepted: 09/16/2018] [Indexed: 10/28/2022]
|
283
|
Spatial memory in Huntington’s disease: A comparative review of human and animal data. Neurosci Biobehav Rev 2019; 98:194-207. [DOI: 10.1016/j.neubiorev.2019.01.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 11/26/2018] [Accepted: 01/14/2019] [Indexed: 12/24/2022]
|
284
|
Lane RM, Smith A, Baumann T, Gleichmann M, Norris D, Bennett CF, Kordasiewicz H. Translating Antisense Technology into a Treatment for Huntington's Disease. Methods Mol Biol 2019; 1780:497-523. [PMID: 29856033 DOI: 10.1007/978-1-4939-7825-0_23] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Advances in molecular biology and genetics have been used to elucidate the fundamental genetic mechanisms underlying central nervous system (CNS) diseases, yet disease-modifying therapies are currently unavailable for most CNS conditions. Antisense oligonucleotides (ASOs) are synthetic single stranded chains of nucleic acids that bind to a specific sequence on ribonucleic acid (RNA) and regulate posttranscriptional gene expression. Decreased gene expression with ASOs might be able to reduce production of the disease-causing protein underlying dominantly inherited neurodegenerative disorders. Huntington's disease (HD), which is caused by a CAG repeat expansion in exon 1 of the huntingtin (HTT) gene and leads to the pathogenic expansion of a polyglutamine (PolyQ ) tract in the N terminus of the huntingtin protein (Htt), is a prime candidate for ASO therapy.State-of-the art translational science techniques can be applied to the development of an ASO targeting HTT RNA, allowing for a data-driven, stepwise progression through the drug development process. A deep and wide-ranging understanding of the basic, preclinical, clinical, and epidemiologic components of drug development will improve the likelihood of success. This includes characterizing the natural history of the disease, including evolution of biomarkers indexing the underlying pathology; using predictive preclinical models to assess the putative gain-of-function of mutant Htt protein and any loss-of-function of the wild-type protein; characterizing toxicokinetic and pharmacodynamic effects of ASOs in predictive animal models; developing sensitive and reliable biomarkers to monitor target engagement and effects on pathology that translate from animal models to patients with HD; establishing a drug delivery method that ensures reliable distribution to relevant CNS tissue; and designing clinical trials that move expeditiously from proof of concept to proof of efficacy. This review focuses on the translational science techniques that allow for efficient and informed development of an ASO for the treatment of HD.
Collapse
Affiliation(s)
| | - Anne Smith
- Ionis Pharmaceuticals, Carlsbad, CA, USA
| | | | | | - Dan Norris
- Ionis Pharmaceuticals, Carlsbad, CA, USA
| | | | | |
Collapse
|
285
|
Hua J, Liu P, Kim T, Donahue M, Rane S, Chen JJ, Qin Q, Kim SG. MRI techniques to measure arterial and venous cerebral blood volume. Neuroimage 2019; 187:17-31. [PMID: 29458187 PMCID: PMC6095829 DOI: 10.1016/j.neuroimage.2018.02.027] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/12/2018] [Accepted: 02/14/2018] [Indexed: 12/14/2022] Open
Abstract
The measurement of cerebral blood volume (CBV) has been the topic of numerous neuroimaging studies. To date, however, most in vivo imaging approaches can only measure CBV summed over all types of blood vessels, including arterial, capillary and venous vessels in the microvasculature (i.e. total CBV or CBVtot). As different types of blood vessels have intrinsically different anatomy, function and physiology, the ability to quantify CBV in different segments of the microvascular tree may furnish information that is not obtainable from CBVtot, and may provide a more sensitive and specific measure for the underlying physiology. This review attempts to summarize major efforts in the development of MRI techniques to measure arterial (CBVa) and venous CBV (CBVv) separately. Advantages and disadvantages of each type of method are discussed. Applications of some of the methods in the investigation of flow-volume coupling in healthy brains, and in the detection of pathophysiological abnormalities in brain diseases such as arterial steno-occlusive disease, brain tumors, schizophrenia, Huntington's disease, Alzheimer's disease, and hypertension are demonstrated. We believe that the continual development of MRI approaches for the measurement of compartment-specific CBV will likely provide essential imaging tools for the advancement and refinement of our knowledge on the exquisite details of the microvasculature in healthy and diseased brains.
Collapse
Affiliation(s)
- Jun Hua
- Neurosection, Div. of MRI Research, Dept. of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA.
| | - Peiying Liu
- Neurosection, Div. of MRI Research, Dept. of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Tae Kim
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Manus Donahue
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Swati Rane
- Radiology, University of Washington Medical Center, Seattle, WA, USA
| | - J Jean Chen
- Rotman Research Institute, Baycrest Centre, Canada; Department of Medical Biophysics, University of Toronto, Canada
| | - Qin Qin
- Neurosection, Div. of MRI Research, Dept. of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Seong-Gi Kim
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, South Korea; Department of Biomedical Engineering, Sungkyunkwan University, Suwon, South Korea
| |
Collapse
|
286
|
Jeppesen Kragh F, Bruun M, Budtz-Jørgensen E, Hjermind LE, Schubert R, Reilmann R, Nielsen JE, Hasselbalch SG. Quantitative Measurements of Motor Function in Alzheimer's Disease, Frontotemporal Dementia, and Dementia with Lewy Bodies: A Proof-of-Concept Study. Dement Geriatr Cogn Disord 2019; 46:168-179. [PMID: 30257254 DOI: 10.1159/000492860] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 08/10/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND This study examines the efficacy of using quantitative measurements of motor dysfunction, compared to clinical ratings, in Alzheimer's disease (AD), frontotemporal dementia (FTD), and dementia with Lewy bodies (DLB). METHODS In this cross-sectional study, 49 patients with a diagnosis of AD (n = 17), FTD (n = 19), or DLB (n = 13) were included and underwent cognitive testing, clinical motor evaluation, and quantitative motor tests: pronation/supination hand tapping, grasping and lifting, and finger and foot tapping. RESULTS Our results revealed significantly higher Q-Motor values in pronation/supination and in grip lift force assessment in AD, FTD, and DLB compared to healthy controls (HC). Q-Motor values detected significant differences between AD and HC, while clinical ratings did not. CONCLUSION Our results suggest that quantitative measurements provide more objective and sensitive measurements of motor dysfunction in dementia.
Collapse
Affiliation(s)
- Frederikke Jeppesen Kragh
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Marie Bruun
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Esben Budtz-Jørgensen
- Department of Public Health, Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - Lena Elisabeth Hjermind
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | - Ralf Reilmann
- The George Huntington Institute, Münster, Germany.,Department of Clinical Radiology, University of Münster, Münster, Germany.,Department of Neurodegenerative Diseases and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Jørgen Erik Nielsen
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Steen Gregers Hasselbalch
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen,
| |
Collapse
|
287
|
Jensen MP, Barker RA. Disease-Modification in Huntington's Disease: Moving Away from a Single-Target Approach. J Huntingtons Dis 2019; 8:9-22. [PMID: 30636742 DOI: 10.3233/jhd-180320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
To date, no candidate intervention has demonstrated a disease-modifying effect in Huntington's disease, despite promising results in preclinical studies. In this commentary we discuss disease-modifying therapies that have been trialled in Huntington's disease and speculate that these failures may be attributed, in part, to the assumption that a single drug selectively targeting one aspect of disease pathology will be universally effective, regardless of disease stage or "subtype". We therefore propose an alternative approach for effective disease-modification that uses 1) a combination approach rather than monotherapy, and 2) targets the disease process early on - before it is clinically manifest. Finally, we will consider whether this change in approach that we propose will be relevant in the future given the recent shift to targeting more proximal disease processes-e.g., huntingtin gene expression; a timely question given Roche's recent decision to take on the clinical development of a promising new drug candidate in Huntington's disease, IONIS-HTTRx.
Collapse
Affiliation(s)
- Melanie P Jensen
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Roger A Barker
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Cambridge Stem Cell Institute, Cambridge, UK
| |
Collapse
|
288
|
Paulsen JS, Lourens S, Kieburtz K, Zhang Y. Sample enrichment for clinical trials to show delay of onset in huntington disease. Mov Disord 2019; 34:274-280. [PMID: 30644132 DOI: 10.1002/mds.27595] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 10/19/2018] [Accepted: 11/26/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Disease-modifying clinical trials in persons without symptoms are often limited in methods to assess the impact associated with experimental therapeutics. This study suggests sample enrichment approaches to facilitate preventive trials to delay disease onset in individuals with the dominant gene for Huntington disease. METHODS Using published onset prediction indexes, we conducted the receiver operating curve analysis for diagnosis within a 3-year clinical trial time frame. We determined optimal cut points on the indexes for participant recruitment and then conducted sample size and power calculations to detect varying effect sizes for treatment efficacy in reducing 3-year rates of disease onset (or diagnosis). RESULTS Area under the curve for 3 onset prediction indexes all demonstrated excellent value in sample enrichment methodology, with the best-performing index being the multivariate risk score (MRS). CONCLUSIONS This study showed that conducting an intervention trial in premanifest and prodromal individuals with the gene expansion for Huntington disease is highly feasible using sample enrichment recruitment methods. Ongoing natural history studies are highly likely to indicate additional markers of disease prior to diagnosis. Statistical modeling of identified markers can facilitate participant enrichment to increase the likelihood of detecting a difference between treatment arms in a cost-effective and efficient manner. Such variations may expedite translation of emerging therapies to persons in an earlier phase of the disease. TRIAL REGISTRATION PREDICT-HD is registered with www.clinicaltrials.gov, number NCT00051324. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Jane S Paulsen
- Departments of Neurology, Psychiatry, and Psychological and Brain Sciences, University of Iowa, Iowa City, Iowa, USA
| | - Spencer Lourens
- Department of Biostatistics, Indiana University Fairbanks School of Public Health and School of Medicine, Indianapolis, Indiana, USA
| | - Karl Kieburtz
- Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA
| | - Ying Zhang
- Department of Biostatistics, Indiana University Fairbanks School of Public Health and School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
289
|
Cortical atrophic-hypometabolic dissociation in the transition from premanifest to early-stage Huntington’s disease. Eur J Nucl Med Mol Imaging 2019; 46:1111-1116. [DOI: 10.1007/s00259-018-4257-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 12/27/2018] [Indexed: 02/05/2023]
|
290
|
Anderson DG, Haagensen M, Ferreira-Correia A, Pierson R, Carr J, Krause A, Margolis RL. Emerging differences between Huntington's disease-like 2 and Huntington's disease: A comparison using MRI brain volumetry. Neuroimage Clin 2019; 21:101666. [PMID: 30682531 PMCID: PMC6350216 DOI: 10.1016/j.nicl.2019.101666] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/18/2018] [Accepted: 01/04/2019] [Indexed: 01/18/2023]
Abstract
Huntington's Disease-Like 2 (HDL2), caused by a CTG/CAG expansion in JPH3 on chromosome 16q24, is the most common Huntington's Disease (HD) phenocopy in populations with African ancestry. Qualitatively, brain MRIs of HDL2 patients have been indistinguishable from HD. To determine brain regions most affected in HDL2 a cross-sectional study using MRI brain volumetry was undertaken to compare the brains of nine HDL2, 11 HD and nine age matched control participants. Participants were ascertained from the region in South Africa with the world's highest HDL2 incidence. The HDL2 and HD patient groups showed no significant differences with respect to mean age at MRI, disease duration, abnormal triplet repeat length, or age at disease onset. Overall, intracerebral volumes were smaller in both affected groups compared to the control group. Comparing the HDL2 and HD groups across multiple covariates, cortical and subcortical volumes were similar with the exception that the HDL2 thalamic volumes were smaller. Consistent with other similarities between the two diseases, these results indicate a pattern of neurodegeneration in HDL2 that is remarkably similar to HD. However smaller thalamic volumes in HDL2 raises intriguing questions into the pathogenesis of both disorders, and how these volumetric differences relate to their respective phenotypes.
Collapse
Affiliation(s)
- David G Anderson
- The University of the Witwatersrand Donald Gordon Medical Centre, Neurology, Johannesburg, South Africa; Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, The University of the Witwatersrand, Johannesburg, South Africa.
| | - Mark Haagensen
- The University of the Witwatersrand Donald Gordon Medical Centre, Radiology Department, Johannesburg, South Africa
| | - Aline Ferreira-Correia
- Department of Psychology, School of Human and Community Development, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Jonathan Carr
- Division of Neurology, Department of Medicine, University of Stellenbosch, Cape Town, South Africa
| | - Amanda Krause
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, The University of the Witwatersrand, Johannesburg, South Africa
| | - Russell L Margolis
- Departments of Psychiatry and Neurology, Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
291
|
Selection of Reference Regions to Model Neurodegeneration in Huntington Disease by 18F-FDG PET/CT Using Imaging and Clinical Parameters. Clin Nucl Med 2019; 44:e1-e5. [DOI: 10.1097/rlu.0000000000002329] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
292
|
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
|
293
|
Chatzikonstantinou S, Dagklis I, Kazis D, Karantali E, Bostantjopoulou S. Huntington's disease with comorbid myasthenia gravis: a case report. Hippokratia 2019; 23:28-29. [PMID: 32256036 PMCID: PMC7124878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
BACKGROUND In the literature, several reports are describing the coexistence of Huntington's disease (HD) or myasthenia gravis (MG) with other neurodegenerative and autoimmune disorders. Herein, we report a rare case of HD in a 66-year-old male with MG. Description of the case: The diagnosis of MG was established by acetylcholine receptor antibodies testing and compatible clinical presentation. The diagnosis of HD was based on clinical features, family history, and DNA testing. Several immunologic mechanisms have been proposed regarding the pathogenesis of HD and MG, respectively. Sharing a common autoimmune aspect could be an uncertain but potential association between the two disorders. CONCLUSION The probability of HD and MG occurring in the same patient is extremely small. While a number of neurological and autoimmune disorders have been reported with HD and MG, this is the first described coexistence of these two entities. HIPPOKRATIA 2019, 23(1): 28-29.
Collapse
Affiliation(s)
- S Chatzikonstantinou
- 3Department of Neurology, Papanikolaou General Hospital, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - I Dagklis
- 3Department of Neurology, Papanikolaou General Hospital, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - D Kazis
- 3Department of Neurology, Papanikolaou General Hospital, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - E Karantali
- 3Department of Neurology, Papanikolaou General Hospital, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - S Bostantjopoulou
- 3Department of Neurology, Papanikolaou General Hospital, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
294
|
|
295
|
Abstract
Huntington's disease (HD) is a dominantly inherited neurodegenerative disease that results in motor, cognitive and psychiatric dysfunction. It is caused by a polyglutamine repeat expansion mutation in the widely expressed HTT protein. The clinical manifestations of HD have been largely attributed to the neurodegeneration of specific neuronal cell types in the brain. However, it has become clear that other cell types, including astrocytes, play important roles in the pathogenesis of HD. The mutant HTT (mHTT) protein is present in neuronal and non-neuronal cell types throughout the nervous system. Studies designed to understand the contribution of mHTT expression in non-neuronal cell types to HD pathogenesis has lagged considerably behind those focused on neurons. However, the role of astrocytes in HD has received more attention over the last 5-10 years. In this chapter we present an overview of HD and our current understanding of astrocytic involvement in this disease. We describe the neuropathological features of HD and provide evidence of morphological and molecular changes in mHTT expressing astrocytes. We review data from animal models and HD patients that implicate mHTT expressing astrocytes to the progression of HD.
Collapse
Affiliation(s)
- Michelle Gray
- Department of Neurology and Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, 1720 2nd Ave S, CIRC 425B, Birmingham, AL 35294, USA.
| |
Collapse
|
296
|
Jędrak P, Mozolewski P, Węgrzyn G, Więckowski MR. Mitochondrial alterations accompanied by oxidative stress conditions in skin fibroblasts of Huntington's disease patients. Metab Brain Dis 2018; 33:2005-2017. [PMID: 30120672 PMCID: PMC6244791 DOI: 10.1007/s11011-018-0308-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/12/2018] [Indexed: 01/08/2023]
Abstract
Huntington disease (HD) is an autosomal dominant neurodegenerative disorder manifesting as progressive impairment of motor function and different neuropsychiatric symptoms caused by an expansion of CAG repeats in huntingtin gene (HTT). Mitochondrial dysfunction and bioenergetic defects can contribute to the course of the disease, however, the molecular mechanism underlying this process is still largely unknown. In this study, we aimed to determine several mitochondrial parameters in HD fibroblasts and assess their relevance to the disease progression as well as to value mitochondrial pathology in peripheral cells as disease potential biomarker. We showed that HD fibroblasts demonstrate significantly lower growth rate compared to control fibroblasts despite the lack of cell cycle perturbations. In order to investigate mitochondrial contribution to cell growth differences between HD and healthy cells, we provided insight into various mitochondrial parameters. Conducted experiments have revealed a significant reduction of the ATP level in HD fibroblasts accompanied by a decrease in mitochondrial metabolic activity in relation to the cells from healthy donors. Importantly, there were no differences in the mitochondrial membrane potential (mtΔΨ) and OXPHOS complexes' levels. Slightly increased level of mitochondrial superoxide (mt. O2•-), but not cytosolic reactive oxygen species (cyt. ROS), has been demonstrated. We have also observed significantly elevated levels of some antioxidant enzymes (SOD2 and GR) which may serve as an indicator of antioxidant defense system in HD patients. Thus, we suggest that mitochondrial alterations in skin fibroblasts of Huntington's disease patients might be helpful in searching for novel disease biomarkers.
Collapse
Affiliation(s)
- Paulina Jędrak
- Department of Molecular Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdańsk, Poland
| | - Paweł Mozolewski
- Department of Medical Biology and Genetics, University of Gdańsk, Gdańsk, Poland
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdańsk, Poland.
| | - Mariusz R Więckowski
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093, Warsaw, Poland.
| |
Collapse
|
297
|
Coppen EM, van der Grond J, Hafkemeijer A, Barkey Wolf JJH, Roos RAC. Structural and functional changes of the visual cortex in early Huntington's disease. Hum Brain Mapp 2018; 39:4776-4786. [PMID: 30144208 PMCID: PMC6866293 DOI: 10.1002/hbm.24322] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 06/19/2018] [Accepted: 07/11/2018] [Indexed: 01/13/2023] Open
Abstract
Huntington's disease (HD) is an autosomal-dominant inherited neurodegenerative disorder characterized by motor disturbances, psychiatric disturbances, and cognitive impairment. Visual cognitive deficits and atrophy of the posterior cerebral cortex are additionally present in early disease stages. This study aimed to assess the extent of structural and functional brain alterations of the visual cortex in HD gene carriers using different neuroimaging modalities. Structural and functional magnetic resonance imaging data were acquired from 18 healthy controls, 21 premanifest, and 20 manifest HD gene carriers. Voxel-based morphometry (VBM) analysis and cortical thickness measurements were performed to assess structural changes in the visual cortex. Brain function was measured by assessing neuronal connectivity changes in response to visual stimulation and at rest in visual resting-state networks. Multiple linear regression analyses were performed to examine the relationship between visual cognitive function and structural imaging measures. Compared to controls, pronounced atrophy and decreased neuronal function at rest were present in associative visual cortices in manifest HD. The primary visual cortex did not show group differences in cortical thickness and in vascular activity after visual stimulation. Thinning of the associative visual cortex was related to worse visual perceptual function. Premanifest HD gene carriers did not show any differences in brain structure or function compared to controls. This study improves the knowledge on posterior brain changes in HD, as our findings suggest that the primary visual cortex remains preserved, both structurally and functionally, while atrophy of associative visual cortices is present in early HD and linked to clinical visual deficits.
Collapse
Affiliation(s)
- Emma M. Coppen
- Department of NeurologyLeiden University Medical CenterLeidenthe Netherlands
| | | | - Anne Hafkemeijer
- Department of RadiologyLeiden University Medical CenterLeidenthe Netherlands
- Department of Methodology and Statistics, Institute of PsychologyLeiden UniversityLeidenthe Netherlands
- Leiden Institute for Brain and CognitionLeiden UniversityLeidenthe Netherlands
| | - Jurriaan J. H. Barkey Wolf
- Department of RadiologyLeiden University Medical CenterLeidenthe Netherlands
- Department of Molecular EpidemiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Raymund A. C. Roos
- Department of NeurologyLeiden University Medical CenterLeidenthe Netherlands
| |
Collapse
|
298
|
Giavazzi M, Daland R, Palminteri S, Peperkamp S, Brugières P, Jacquemot C, Schramm C, Cleret de Langavant L, Bachoud-Lévi AC. The role of the striatum in linguistic selection: Evidence from Huntington's disease and computational modeling. Cortex 2018; 109:189-204. [DOI: 10.1016/j.cortex.2018.08.031] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 07/04/2018] [Accepted: 08/05/2018] [Indexed: 11/29/2022]
|
299
|
Long JD, Mills JA. Joint modeling of multivariate longitudinal data and survival data in several observational studies of Huntington's disease. BMC Med Res Methodol 2018; 18:138. [PMID: 30445915 PMCID: PMC6240282 DOI: 10.1186/s12874-018-0592-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 10/29/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Joint modeling is appropriate when one wants to predict the time to an event with covariates that are measured longitudinally and are related to the event. An underlying random effects structure links the survival and longitudinal submodels and allows for individual-specific predictions. Multiple time-varying and time-invariant covariates can be included to potentially increase prediction accuracy. The goal of this study was to estimate a multivariate joint model on several longitudinal observational studies of Huntington's disease, examine external validity performance, and compute individual-specific predictions for characterizing disease progression. Emphasis was on the survival submodel for predicting the hazard of motor diagnosis. METHODS Data from four observational studies was analyzed: Enroll-HD, PREDICT-HD, REGISTRY, and Track-HD. A Bayesian approach to estimation was adopted, and external validation was performed using a time-varying AUC measure. Individual-specific cumulative hazard predictions were computed based on a simulation approach. The cumulative hazard was used for computing predicted age of motor onset and also for a deviance residual indicating the discrepancy between observed diagnosis status and model-based status. RESULTS The joint model trained in a single study had very good performance in discriminating among diagnosed and pre-diagnosed participants in the remaining test studies, with the 5-year mean AUC = .83 (range .77-.90), and the 10-year mean AUC = .86 (range .82-.92). Graphical analysis of the predicted age of motor diagnosis showed an expected strong relationship with the trinucleotide expansion that causes Huntington's disease. Graphical analysis of the deviance-type residual revealed there were individuals who converted to a diagnosis despite having relatively low model-based risk, others who had not yet converted despite having relatively high risk, and the majority falling between the two extremes. CONCLUSIONS Joint modeling is an improvement over traditional survival modeling because it considers all the longitudinal observations of covariates that are predictive of an event. Predictions from joint models can have greater accuracy because they are tailored to account for individual variability. These predictions can provide relatively accurate characterizations of individual disease progression, which might be important in the timing of interventions, determining the qualification for appropriate clinical trials, and general genotypic analysis.
Collapse
Affiliation(s)
- Jeffrey D. Long
- Department of Psychiatry, Carver College of Medicine, University of Iowa, 500 Newton Road, Iowa City, IA 52242-1000 USA
- Department of Biostatistics, Department of Public Health, University of Iowa, 145 N. Riverside Drive, Iowa City, IA 52242-1000 USA
| | - James A. Mills
- Department of Psychiatry, Carver College of Medicine, University of Iowa, 500 Newton Road, Iowa City, IA 52242-1000 USA
| |
Collapse
|
300
|
Molecular Imaging in Huntington's Disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2018; 142:289-333. [PMID: 30409256 DOI: 10.1016/bs.irn.2018.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Huntington's disease (HD) is a rare monogenic neurodegenerative disorder caused by a trinucleotide CAG repeat expansion in the huntingtin gene resulting in the formation of intranuclear inclusions of mutated huntingtin. The accumulation of mutated huntingtin leads to loss of GABAergic medium spiny neurons (MSNs); subsequently resulting in the development of chorea, cognitive dysfunction and psychiatric symptoms. Premanifest HD gene expansion carriers, provide a unique cohort to examine very early molecular changes, occurring before the development of overt symptoms, to elucidate disease pathophysiology and identify reliable biomarkers of HD progression. Positron emission tomography (PET) is a non-invasive molecular imaging technique allowing the evaluation of specific molecular targets in vivo. Selective PET radioligands provide invaluable tools to investigate the role of the dopaminergic system, brain metabolism, microglial activation, phosphodiesterase 10A, and cannabinoid, GABA, adenosine and opioid receptors in HD. PET has been employed to monitor disease progression aiming to identify a reliable biomarker to predict phenoconversion from premanifest to manifest HD.
Collapse
|