1
|
Neema M, Schultz JL, Langbehn DR, Conrad AL, Epping EA, Magnotta VA, Nopoulos PC. Mutant Huntingtin Drives Development of an Advantageous Brain Early in Life: Evidence in Support of Antagonistic Pleiotropy. Ann Neurol 2024; 96:1006-1019. [PMID: 39115048 PMCID: PMC11496017 DOI: 10.1002/ana.27046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 10/23/2024]
Abstract
OBJECTIVE Huntington's disease (HD) is a neurodegenerative disease caused by a triplet repeat expansion within the gene huntingtin (HTT). Antagonistic pleiotropy is a theory of aging that posits that some genes, facilitating individual fitness early in life through adaptive evolutionary changes, also augment detrimental aging-related processes. Antagonistic pleiotropy theory may explain a positive evolutionary pressure toward functionally advantageous brain development that is vulnerable to rapid degeneration. The current study investigated antagonistic pleiotropy in HD using a years-to-onset paradigm in a unique sample of children and young adults at risk for HD. METHODS Cognitive, behavioral, motor, and brain structural measures from premanifest gene-expanded (n = 79) and gene nonexpanded (n = 112) participants (6-21 years) in the Kids-HD study were examined. All measures in the gene-expanded group were modeled using a mixed-effects regression approach to assess years-to-onset-based changes while controlling for normal growth. Simultaneously, structure-function associations were also examined. RESULTS Decades from motor onset, gene-expanded participants showed significantly better cognitive, behavioral, and motor scores versus gene nonexpanded controls, along with larger cerebral volumes and cortical features. After this initial peak, a prolonged deterioration was observed in both functional and structural measures. Far from onset, brain measures were positively correlated with functional measures, supporting the view that functional advantages were mediated by structural differences. INTERPRETATION Mutant HTT may drive the development of a larger than normal brain that subserves superior early-life function. These findings support the antagonistic pleiotropy theory of HTT in HD, where this gene drives early advantage followed by accelerated aging processes. ANN NEUROL 2024;96:1006-1019.
Collapse
Affiliation(s)
- Mohit Neema
- Department of Psychiatry, Carver College of Medicine at the University of Iowa, 200 Hawkins Drive, Iowa City, IA
| | - Jordan L. Schultz
- Department of Psychiatry, Carver College of Medicine at the University of Iowa, 200 Hawkins Drive, Iowa City, IA
- Department of Neurology, Carver College of Medicine at the University of Iowa, 200 Hawkins Drive, Iowa City, IA
| | - Douglas R. Langbehn
- Department of Psychiatry, Carver College of Medicine at the University of Iowa, 200 Hawkins Drive, Iowa City, IA
| | - Amy L. Conrad
- Stead Family Department of Pediatrics, Carver College of Medicine at the University of Iowa, 200 Hawkins Drive, Iowa City, IA
| | - Eric A. Epping
- Department of Psychiatry, Carver College of Medicine at the University of Iowa, 200 Hawkins Drive, Iowa City, IA
| | - Vincent A. Magnotta
- Department of Radiology, Carver College of Medicine at the University of Iowa, 200 Hawkins Drive, Iowa City, IA
| | - Peggy C. Nopoulos
- Department of Psychiatry, Carver College of Medicine at the University of Iowa, 200 Hawkins Drive, Iowa City, IA
- Department of Neurology, Carver College of Medicine at the University of Iowa, 200 Hawkins Drive, Iowa City, IA
- Stead Family Department of Pediatrics, Carver College of Medicine at the University of Iowa, 200 Hawkins Drive, Iowa City, IA
| |
Collapse
|
2
|
Feleus S, Skotnicki LEM, Roos RAC, de Bot ST. Medication Use and Treatment Indications in Huntington's Disease; Analyses from a Large Cohort. Mov Disord Clin Pract 2024. [PMID: 39431460 DOI: 10.1002/mdc3.14230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 07/19/2024] [Accepted: 09/14/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND Huntington's Disease is a rare neurodegenerative disorder in which appropriate medication management is essential. While many medications are prescribed based on expert knowledge, overviews of actual medication use in HD are sparse. OBJECTIVES We provide a detailed overview of medication use and associated indications across HD disease stages, considering sex and regional differences. METHODS Data from the largest observational HD study, ENROLL-HD, were used. We created HD-related medication and indication classes to identify medication trends in manifest, premanifest and control subjects. We studied medication use in adult, childhood- and adolescent-onset HD, incorporating disease stage (including phenoconverters), sex and regional differences. RESULTS In 8546 manifest HD patients, 84.6% used medication (any type), with the average number of medications per user rising from 2.5 in premanifest HD to 5.2 in end stage disease. Antipsychotics (29.2%), SSRIs (27.5%) and painkillers (21.8%) were most often used. Medication use varied with disease progression. Several differences were observed between the sexes, and notably between Europe and Northern America as well. Medication use increased after phenoconversion (from 64.8% to 70.6%, P < 0.05), with the largest difference in antipsychotic use (4.4%-7.8%, P < 0.05). Medication patterns were different in childhood-onset HD, with no use of painkillers, less use of anti-chorea and antidepressant drugs, and more for aggression and irritability. CONCLUSIONS Medication use in HD increases with disease progression, with varying types of medications prescribed based on disease stage, sex, and region of living. Recognizing these medication trends is vital for further personalized HD management.
Collapse
Affiliation(s)
- Stephanie Feleus
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lara E M Skotnicki
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Raymund A C Roos
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Susanne T de Bot
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
3
|
Ibañez K, Jadhav B, Zanovello M, Gagliardi D, Clarkson C, Facchini S, Garg P, Martin-Trujillo A, Gies SJ, Galassi Deforie V, Dalmia A, Hensman Moss DJ, Vandrovcova J, Rocca C, Moutsianas L, Marini-Bettolo C, Walker H, Turner C, Shoai M, Long JD, Fratta P, Langbehn DR, Tabrizi SJ, Caulfield MJ, Cortese A, Escott-Price V, Hardy J, Houlden H, Sharp AJ, Tucci A. Increased frequency of repeat expansion mutations across different populations. Nat Med 2024:10.1038/s41591-024-03190-5. [PMID: 39354197 DOI: 10.1038/s41591-024-03190-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 07/11/2024] [Indexed: 10/03/2024]
Abstract
Repeat expansion disorders (REDs) are a devastating group of predominantly neurological diseases. Together they are common, affecting 1 in 3,000 people worldwide with population-specific differences. However, prevalence estimates of REDs are hampered by heterogeneous clinical presentation, variable geographic distributions and technological limitations leading to underascertainment. Here, leveraging whole-genome sequencing data from 82,176 individuals from different populations, we found an overall disease allele frequency of REDs of 1 in 283 individuals. Modeling disease prevalence using genetic data, age at onset and survival, we show that the expected number of people with REDs would be two to three times higher than currently reported figures, indicating underdiagnosis and/or incomplete penetrance. While some REDs are population specific, for example, Huntington disease-like 2 in Africans, most REDs are represented in all broad genetic ancestries (that is, Europeans, Africans, Americans, East Asians and South Asians), challenging the notion that some REDs are found only in specific populations. These results have worldwide implications for local and global health communities in the diagnosis and counseling of REDs.
Collapse
Affiliation(s)
- Kristina Ibañez
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Bharati Jadhav
- Department of Genetics and Genomic Sciences and Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matteo Zanovello
- Department of Neuromuscular Diseases, Institute of Neurology, UCL, London, UK
| | - Delia Gagliardi
- William Harvey Research Institute, Queen Mary University of London, London, UK
- Department of Neuromuscular Diseases, Institute of Neurology, UCL, London, UK
| | | | - Stefano Facchini
- Department of Neuromuscular Diseases, Institute of Neurology, UCL, London, UK
- IRCCS Mondino Foundation, Pavia, Italy
| | - Paras Garg
- Department of Genetics and Genomic Sciences and Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alejandro Martin-Trujillo
- Department of Genetics and Genomic Sciences and Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott J Gies
- Department of Genetics and Genomic Sciences and Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | - Davina J Hensman Moss
- St George's, University of London, London, UK
- Department of Neurodegenerative Disorders, Queen Square Institute of Neurology, UCL, London, UK
| | - Jana Vandrovcova
- Department of Neuromuscular Diseases, Institute of Neurology, UCL, London, UK
| | - Clarissa Rocca
- Department of Neuromuscular Diseases, Institute of Neurology, UCL, London, UK
| | | | - Chiara Marini-Bettolo
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Helen Walker
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Chris Turner
- Centre for Neuromuscular Disease, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, London, UK
| | - Maryam Shoai
- Department of Neurodegenerative Disorders, Queen Square Institute of Neurology, UCL, London, UK
| | - Jeffrey D Long
- Departments of Psychiatry and Biostatistics, The University of Iowa, Iowa City, IA, USA
| | - Pietro Fratta
- Department of Neuromuscular Diseases, Institute of Neurology, UCL, London, UK
| | - Douglas R Langbehn
- Departments of Psychiatry and Biostatistics, The University of Iowa, Iowa City, IA, USA
| | - Sarah J Tabrizi
- UK Dementia Research Institute, UCL, London, UK
- Department of Neurodegenerative Disorders, Queen Square Institute of Neurology, UCL, London, UK
- Huntington's Disease Centre, UCL, London, UK
| | - Mark J Caulfield
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Andrea Cortese
- Department of Neuromuscular Diseases, Institute of Neurology, UCL, London, UK
- IRCCS Mondino Foundation, Pavia, Italy
| | - Valentina Escott-Price
- Department of Psychological Medicine and Clinical Neuroscience, School of Medicine, Cardiff University, Cardiff, UK
- Dementia Research Institute, Cardiff University, Cardiff, UK
| | - John Hardy
- Department of Neurodegenerative Disorders, Queen Square Institute of Neurology, UCL, London, UK
| | - Henry Houlden
- Department of Neurodegenerative Disorders, Queen Square Institute of Neurology, UCL, London, UK
- Neurogenetics Unit, National Hospital for Neurology and Neurosurgery, London, UK
| | - Andrew J Sharp
- Department of Genetics and Genomic Sciences and Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Arianna Tucci
- William Harvey Research Institute, Queen Mary University of London, London, UK.
- Department of Neuromuscular Diseases, Institute of Neurology, UCL, London, UK.
| |
Collapse
|
4
|
McDonnell EI, Xie S, Marder K, Cui F, Wang Y. Dynamic undirected graphical models for time-varying clinical symptom and neuroimaging networks. Stat Med 2024; 43:4131-4147. [PMID: 39007408 PMCID: PMC11502120 DOI: 10.1002/sim.10143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 07/16/2024]
Abstract
In this work, we propose methods to examine how the complex interrelationships between clinical symptoms and, separately, brain imaging biomarkers change over time leading up to the diagnosis of a disease in subjects with a known genetic near-certainty of disease. We propose a time-dependent undirected graphical model that ensures temporal and structural smoothness across time-specific networks to examine the trajectories of interactions between markers aligned at the time of disease onset. Specifically, we anchor subjects relative to the time of disease diagnosis (anchoring time) as in a revival process, and we estimate networks at each time point of interest relative to the anchoring time. To use all available data, we apply kernel weights to borrow information across observations that are close to the time of interest. Adaptive lasso weights are introduced to encourage temporal smoothness in edge strength, while a novel elastic fused-l 0 $$ {l}_0 $$ penalty removes spurious edges and encourages temporal smoothness in network structure. Our approach can handle practical complications such as unbalanced visit times. We conduct simulation studies to compare our approach with existing methods. We then apply our method to data from PREDICT-HD, a large prospective observational study of pre-manifest Huntington's disease (HD) patients, to identify symptom and imaging network changes that precede clinical diagnosis of HD.
Collapse
Affiliation(s)
- Erin I. McDonnell
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, New York
| | - Shanghong Xie
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, New York
- Department of Statistics, University of South Carolina, Columbia, South Carolina, USA
| | - Karen Marder
- Department of Neurology, Columbia University Medical Center, New York, New York
- Department of Psychiatry, Columbia University Medical Center, New York, New York
- The Taub Institute for Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, New York, New York
- Gertrude H. Sergievsky Center, Columbia University Medical Center, New York, New York
| | - Fanyu Cui
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, New York
| | - Yuanjia Wang
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, New York
- Department of Psychiatry, Columbia University Medical Center, New York, New York
| |
Collapse
|
5
|
Kim LJY, Kundu B, Moretti P, Lozano AM, Rahimpour S. Advancements in surgical treatments for Huntington disease: From pallidotomy to experimental therapies. Neurotherapeutics 2024:e00452. [PMID: 39304438 DOI: 10.1016/j.neurot.2024.e00452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/12/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024] Open
Abstract
Huntington disease (HD) is an autosomal dominant neurodegenerative disorder characterized by choreic movements, behavioral changes, and cognitive impairment. The pathogenesis of this process is a consequence of mutant protein toxicity in striatal and cortical neurons. Thus far, neurosurgical management of HD has largely been limited to symptomatic relief of motor symptoms using ablative and stimulation techniques. These interventions, however, do not modify the progressive course of the disease. More recently, disease-modifying experimental therapeutic strategies have emerged targeting intrastriatal infusion of neurotrophic factors, cell transplantation, HTT gene silencing, and delivery of intrabodies. Herein we review therapies requiring neurosurgical intervention, including those targeting symptom management and more recent disease-modifying agents, with a focus on safety, efficacy, and surgical considerations.
Collapse
Affiliation(s)
- Leo J Y Kim
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah, Salt Lake City, UT, USA
| | - Bornali Kundu
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah, Salt Lake City, UT, USA
| | - Paolo Moretti
- Department of Neurology, University of Utah, Salt Lake City, UT, USA; Department of Neurology, George E. Wahlen VA Medical Center, Salt Lake City, UT, USA
| | - Andres M Lozano
- Division of Neurosurgery and Toronto Western Hospital Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Shervin Rahimpour
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah, Salt Lake City, UT, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
6
|
Roos AK, Stenvall E, Kockum ES, Grönlund KÅ, Alstermark H, Wuolikainen A, Andersen PM, Nordin A, Forsberg KME. Small striatal huntingtin inclusions in patients with motor neuron disease with reduced penetrance and intermediate HTT gene expansions. Hum Mol Genet 2024:ddae137. [PMID: 39270726 DOI: 10.1093/hmg/ddae137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/22/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Short tandem repeat expansions in the human genome are overrepresented in a variety of neurological disorders. It was recently shown that huntingtin (HTT) repeat expansions with full penetrance, i.e. 40 or more CAG repeats, which normally cause Huntington's disease (HD), are overrepresented in patients with amyotrophic lateral sclerosis (ALS). Whether patients carrying HTT repeat expansions with reduced penetrance, (36-39 CAG repeats), or alleles with intermediate penetrance, (27-35 CAG repeats), have an increased risk of ALS has not yet been investigated. Here, we examined the role of HTT repeat expansions in a motor neuron disease (MND) cohort, searched for expanded HTT alleles, and investigated correlations with phenotype and neuropathology. MND patients harboring C9ORF72 hexanucleotide repeat expansions (HREs) were included, to investigate whether HTT repeat expansions were more common in this group. We found a high prevalence of intermediate (range 5.63%-6.61%) and reduced penetrance (range 0.57%-0.66%) HTT gene expansions in this cohort compared to other populations of European ancestry, but no differences between the MND cohort and the control cohort were observed, regardless of C9ORF72HRE status. Upon autopsy of three patients with intermediate or reduced penetrance HTT alleles, huntingtin inclusions were observed in the caudate nucleus and frontal lobe, but no significant somatic mosaicism was detected in different parts of the nervous system. Thus, we demonstrate, for the first time, huntingtin inclusions in individuals with MND and intermediate and reduced penetrance HTT repeat expansions but more clinicopathological investigations are needed to further understand the impact of HTT gene expansion-related pleiotropy.
Collapse
Affiliation(s)
- Anna-Karin Roos
- Department of Clinical Sciences, Neurosciences, Umeå University, Norrlands University Hospital, Building 6 M, Floor 4, Umeå SE-90184, Sweden
| | - Erica Stenvall
- Department of Medical Biosciences, Umeå University, Norrlands University Hospital, Building 6 M, Floor 2, Umeå SE-90184, Sweden
| | - Emmy Skelton Kockum
- Department of Medical Biosciences, Umeå University, Norrlands University Hospital, Building 6 M, Floor 2, Umeå SE-90184, Sweden
| | - Kornelia Åman Grönlund
- Department of Clinical Sciences, Neurosciences, Umeå University, Norrlands University Hospital, Building 6 M, Floor 4, Umeå SE-90184, Sweden
| | - Helena Alstermark
- Department of Clinical Sciences, Neurosciences, Umeå University, Norrlands University Hospital, Building 6 M, Floor 4, Umeå SE-90184, Sweden
| | - Anna Wuolikainen
- Department of Medical Sciences, Neurology, Uppsala University, Uppsala University Hospital, Entrance 85, Floor 2, Uppsala SE-75185, Sweden
| | - Peter M Andersen
- Department of Clinical Sciences, Neurosciences, Umeå University, Norrlands University Hospital, Building 6 M, Floor 4, Umeå SE-90184, Sweden
| | - Angelica Nordin
- Department of Medical Biosciences, Umeå University, Norrlands University Hospital, Building 6 M, Floor 2, Umeå SE-90184, Sweden
| | - Karin M E Forsberg
- Department of Clinical Sciences, Neurosciences, Umeå University, Norrlands University Hospital, Building 6 M, Floor 4, Umeå SE-90184, Sweden
| |
Collapse
|
7
|
Fitzgerald ES, Manousakis JE, Glikmann-Johnston Y, Rankin M, Anderson C, Stout JC, Jackson ML. Sleep fragmentation despite intact rest-activity patterns in premanifest Huntington's disease: An actigraphy study. Sleep Med 2024; 124:16-29. [PMID: 39250876 DOI: 10.1016/j.sleep.2024.08.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 09/11/2024]
Abstract
OBJECTIVE Sleep research in Huntington's disease (HD) has primarily focused on manifest HD, with significantly less attention given to premanifest HD (Pre-HD). Therefore, we investigated sleep and rest-activity patterns in people with Pre-HD versus healthy controls (HC). METHODS We conducted a cross-sectional study including 36 Pre-HD and 48 HC participants. Pre-HD participants were stratified into three groups according to their proximity to estimated diagnosis, using a cytosine-adenine-guanine (CAG) and current age-based predictive model: NEAR (<9 years to diagnosis), MID (9-15 years to diagnosis) and FAR (>15 years to diagnosis). Sleep and rest-activity patterns were assessed using wrist-worn actigraphy, a sleep diary, and sleep questionnaires. RESULTS NEAR and MID groups experienced higher fragmentation index than HC and FAR groups. NEAR and MID groups also exhibited greater WASO than the FAR group. NEAR and MID groups showed lower intra-daily variability (IV) than HC and FAR groups, with the NEAR group also being more active in the most active 10 h (M10). Groups did not differ on subjective sleep measures, inter-daily stability (IS), sleep regularity index, relative amplitude, or amount of activity in the least active 5 h (L5). Considering all Pre-HD participants, fewer years to diagnosis, higher CAG-age-product (CAP) scores (a measure of cumulative exposure to the HD-causing gene mutation) and larger CAG repeat lengths correlated with higher WASO, fragmentation index, L5, IS, and lower sleep efficiency and IV. Higher CAP score correlated with higher M10. CONCLUSIONS Despite intact rest-activity patterns and similar subjective sleep quality to HC, greater sleep fragmentation is a prominent and early feature in Pre-HD. Therefore, reducing sleep fragmentation may be a potential target for sleep intervention in HD. Longitudinal studies using larger samples are needed to assess sleep across the disease spectrum and its impact on clinical outcomes, like cognition.
Collapse
Affiliation(s)
- Emily S Fitzgerald
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Jessica E Manousakis
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Yifat Glikmann-Johnston
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Meg Rankin
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Clare Anderson
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia; Centre for Human Brain Health, School of Psychology, University of Birmingham, Edgbaston, UK
| | - Julie C Stout
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia.
| | - Melinda L Jackson
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| |
Collapse
|
8
|
Dawson J, Kay C, Black HF, Bortnick S, Javier K, Xia Q, Sandhu A, Buchanan C, Hogg V, Chang FCF, Goto J, Arning L, Saft C, Bijlsma EK, Nguyen HP, Roxburgh R, Hayden MR. The frequency and clinical impact of synonymous HTT loss-of-interruption and duplication-of-interruption variants in a diverse HD cohort. Genet Med 2024; 26:101239. [PMID: 39140258 DOI: 10.1016/j.gim.2024.101239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/04/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024] Open
Abstract
PURPOSE To determine the frequency and clinical impact of loss-of-interruption (LOI) and duplication-of-interruption modifier variants of the HTT CAG and CCG repeat in a cohort of individuals with Huntington disease (HD). METHODS We screened symptomatic HD participants from the UBC HD Biobank and 5 research sites for sequence variants. After variant identification, we examined the clinical impact and frequency in the reduced penetrance range. RESULTS Participants with CAG-CCG LOI and CCG LOI variants have a similar magnitude of earlier onset of HD, by 12.5 years. The sequence variants exhibit ancestry-specific differences. Participants with the CAG-CCG LOI variant also have a faster progression of Total Motor Score by 1.9 units per year. Symptomatic participants with the CAG-CCG LOI variant show enrichment in the reduced penetrance range. The CAG-CCG LOI variant explains the onset of 2 symptomatic HD participants with diagnostic repeats below the pathogenetic range. CONCLUSION Our findings have significant clinical implications for participants with the CAG-CCG LOI variant who receive inaccurate diagnoses near diagnostic cutoff ranges. Improved diagnostic testing approaches and clinical management are needed for these individuals. We present the largest and most diverse HTT CAG and CCG sequence variant cohort and emphasize their importance in clinical presentation in HD.
Collapse
Affiliation(s)
- Jessica Dawson
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Chris Kay
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Hailey Findlay Black
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Stephanie Bortnick
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Kyla Javier
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Qingwen Xia
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Akshdeep Sandhu
- Research Informatics, BC Children's Hospital Research Institute, Vancouver, Canada
| | | | - Virginia Hogg
- Auckland City Hospital, Health New Zealand, Auckland, New Zealand
| | - Florence C F Chang
- Huntington Disease Unit, Department of Neurology, Westmead Hospital, Westmead, New South Wales, Australia; Sydney Medical School, Westmead Campus, University of Sydney, Sydney, Australia
| | - Jun Goto
- Department of Neurology, International University of Health and Welfare, Ichikawa Hospital, Chiba, Japan
| | - Larissa Arning
- Department of Human Genetics, Medical Faculty, Ruhr University of Bochum, Bochum, Germany
| | - Carsten Saft
- Department of Neurology, Huntington Center North Rhine-Westphalia, St. Josef-Hospital Bochum, Ruhr University Bochum, Bochum, Germany
| | - Emilia K Bijlsma
- Department of Clinical Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Huu P Nguyen
- Department of Human Genetics, Medical Faculty, Ruhr University of Bochum, Bochum, Germany
| | - Richard Roxburgh
- Auckland City Hospital, Health New Zealand, Auckland, New Zealand; Department of Medicine and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Michael R Hayden
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
9
|
Farag M, Coleman A, Knights H, Murphy MJ, Rajagopal S, Touzé A, Shoai M, Hearst C, Salanio DM, Wild EJ, Tabrizi SJ. Outcomes of Percutaneous Endoscopic Gastrostomy in Huntington's Disease at a Tertiary Center. Mov Disord Clin Pract 2024; 11:998-1007. [PMID: 38853375 PMCID: PMC11329559 DOI: 10.1002/mdc3.14130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 05/19/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND Clinically assisted nutrition and hydration via percutaneous endoscopic gastrostomy (PEG) is a therapeutic option to ameliorate the difficulties associated with enhanced catabolism, weight loss, and dysphagia in Huntington's disease (HD). OBJECTIVES The objective is to provide insights into demographics, staging (Shoulson-Fahn), complications, weight trajectories, and survival rates in people with HD (pwHD) who underwent PEG. METHODS This retrospective study included 705 consecutive pwHD who attended our HD clinic between July 2006 and March 2024, of whom 52 underwent PEG. A control group (n = 52), comprising pwHD without PEG, were closely matched for sex, stage, age, CAG length, and disease burden score at PEG. The study was registered as a service evaluation at the National Hospital for Neurology and Neurosurgery. RESULTS PEG prevalence was 15.0% (n = 52/347) among manifest pwHD: 4.8% (n = 3/62) for Stage 3; 33.3% (n = 16/48) for stage 4; and 44.1% (n = 30/68) for stage 5. Commonest indications were dysphagia, weight loss, and inadequate oral intake. Complications included chest infection, tube dislodgement, and peristomal and skin infections. Modeling of weight trajectories after PEG found no difference between PEG and non-PEG groups. Mortality rate was 34.6% (n = 18/52) in the PEG and 36.5% (n = 19/52) in the non-PEG groups (P = 0.84). Treatment duration (until study endpoint or death) was 3.48 years (interquartile range = 1.71-6.02; range = 0.23-18.8), with 65.4% (n = 34/52) alive at the study endpoint. CONCLUSION PEG in pwHD at-risk for weight loss may help slow weight loss. Prospective studies are required to strengthen PEG decision-making in pwHD. PEG survival was much longer than other dementias, highlighting the need to consider PEG independently in pwHD.
Collapse
Affiliation(s)
- Mena Farag
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- The National Hospital for Neurology and Neurosurgery (NHNN), Queen Square, London, United Kingdom
| | - Annabelle Coleman
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- The National Hospital for Neurology and Neurosurgery (NHNN), Queen Square, London, United Kingdom
| | - Harry Knights
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- The National Hospital for Neurology and Neurosurgery (NHNN), Queen Square, London, United Kingdom
| | - Michael J Murphy
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- The National Hospital for Neurology and Neurosurgery (NHNN), Queen Square, London, United Kingdom
| | - Sangeerthana Rajagopal
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- The National Hospital for Neurology and Neurosurgery (NHNN), Queen Square, London, United Kingdom
| | - Alexiane Touzé
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- The National Hospital for Neurology and Neurosurgery (NHNN), Queen Square, London, United Kingdom
| | - Maryam Shoai
- Department of Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Cara Hearst
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- The National Hospital for Neurology and Neurosurgery (NHNN), Queen Square, London, United Kingdom
| | - Desiree M Salanio
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- The National Hospital for Neurology and Neurosurgery (NHNN), Queen Square, London, United Kingdom
| | - Edward J Wild
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- The National Hospital for Neurology and Neurosurgery (NHNN), Queen Square, London, United Kingdom
| | - Sarah J Tabrizi
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- The National Hospital for Neurology and Neurosurgery (NHNN), Queen Square, London, United Kingdom
| |
Collapse
|
10
|
Stanisławska-Sachadyn A, Krzemiński M, Zielonka D, Krygier M, Ziętkiewicz E, Sławek J, Limon J. Sex contribution to average age at onset of Huntington's disease depends on the number of (CAG) n repeats. Sci Rep 2024; 14:15729. [PMID: 38977715 PMCID: PMC11231309 DOI: 10.1038/s41598-024-64105-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 06/05/2024] [Indexed: 07/10/2024] Open
Abstract
Huntington's disease (HD) is a hereditary neurodegenerative disorder caused by the extension of the CAG repeats in exon 1 of the HTT gene and is transmitted in a dominant manner. The present study aimed to assess whether patients' sex, in the context of mutated and normal allele length, contributes to age on onset (AO) of HD. The study population comprised a large cohort of 3723 HD patients from the European Huntington's Disease Network's REGISTRY database collected at 160 sites across 17 European countries and in one location outside Europe. The data were analyzed using regression models and factorial analysis of variance (ANOVA) considering both mutated allele length and sex as predictors of patients' AO. AO, as described by the rater's estimate, was found to be later in affected women than in men across the whole population. This difference was most pronounced in a subgroup of 1273 patients with relatively short variants of the mutated allele (40-45 CAG repeats) and normal alleles in a higher half of length distribution-namely, more than 17 CAG repeats; however, it was also observed in each group. Our results presented in this observational study point to sex-related differences in AO, most pronounced in the presence of the short mutated and long normal allele, which may add to understanding the dynamics of AO in Huntington's Disease.Trial registration: ClinicalTrials.gov identifier NCT01590589.
Collapse
Affiliation(s)
- Anna Stanisławska-Sachadyn
- Department of Biotechnology and Microbiology, Gdańsk University of Technology, 80-233, Gdańsk, Poland.
- Department of Biology and Medical Genetics, Medical University of Gdańsk, 80-211, Gdańsk, Poland.
- BioTechMed Center, Gdańsk University of Technology, Narutowicza 11/12, 80-233, Gdańsk, Poland.
| | - Michał Krzemiński
- Institute of Applied Mathematics , Gdańsk University of Technology, 80-233, Gdańsk, Poland
| | - Daniel Zielonka
- Department of Public Health, Poznań University of Medical Sciences, 60-812, Poznan, Poland
| | - Magdalena Krygier
- Department of Developmental Neurology, Medical University of Gdansk, 80-952, Gdańsk, Poland
| | - Ewa Ziętkiewicz
- Institute of Human Genetics, Polish Academy of Sciences, 60-479, Poznan, Poland
| | - Jarosław Sławek
- Department of Neurology, St. Adalbert Hospital, Copernicus PL, 80-462,, Gdańsk, Poland
- Department of Neurological and Psychiatric Nursing, Faculty of Health Sciences, Medical University of Gdańsk, 80-211, Gdańsk, Poland
| | - Janusz Limon
- Department of Medical Ethics, Medical University of Gdańsk, 80-211, Gdańsk, Poland
| |
Collapse
|
11
|
Chenain L, Riad R, Fraisse N, Jubin C, Morgado G, Youssov K, Lunven M, Bachoud-Levi AC. Graph methods to infer spatial disturbances: Application to Huntington's Disease's speech. Cortex 2024; 176:144-160. [PMID: 38795650 DOI: 10.1016/j.cortex.2024.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 03/07/2024] [Accepted: 04/25/2024] [Indexed: 05/28/2024]
Abstract
OBJECTIVE Huntington's Disease (HD) is an inherited neurodegenerative disease caused by the mutation of the Htt gene, impacting all aspects of living and functioning. Among cognitive disabilities, spatial capacities are impaired, but their monitoring remains scarce as limited by lengthy experts' assessments. Language offers an alternative medium to evaluate patients' performance in HD. Yet, its capacities to assess HD's spatial abilities are unknown. Here, we aimed to bring proof-of-concept that HD's spatial deficits can be assessed through speech. METHODS We developed the Spatial Description Model to graphically represent spatial relations described during the Cookie Theft Picture (CTP) task. We increased the sensitivity of our model by using only sentences with spatial terms, unlike previous studies in Alzheimer's disease. 78 carriers of the mutant Htt, including 56 manifest and 22 premanifest individuals, as well as 25 healthy controls were included from the BIOHD & (NCT01412125) & Repair-HD (NCT03119246) cohorts. The convergence and divergence of the model were validated using the SelfCog battery. RESULTS Our Spatial Description Model was the only one among the four assessed approaches, revealing that individuals with manifest HD expressed fewer spatial relations and engaged in less spatial exploration compared to healthy controls. Their graphs correlated with both visuospatial and language SelfCog performances, but not with motor, executive nor memory functions. CONCLUSIONS We provide the proof-of-concept using our Spatial Description Model that language can grasp HD patient's spatial disturbances. By adding spatial capabilities to the panel of functions tested by the language, it paves the way for eventual remote clinical application.
Collapse
Affiliation(s)
- Lucie Chenain
- Département d'Etudes Cognitives, École normale supérieure, PSL University, NeuroPsychologie Interventionnelle, 75005 Paris, France; Univ Paris Est Créteil, INSERM U955, Institut Mondor de Recherche Biomédicale, Equipe NeuroPsychologie Interventionnelle, F-94010 Créteil, France; NeurATRIS Créteil, France; ALMAnaCH, INRIA, 75012 Paris, France; Learning Planet Institute, Université de Paris, 75004 Paris, France
| | - Rachid Riad
- Département d'Etudes Cognitives, École normale supérieure, PSL University, NeuroPsychologie Interventionnelle, 75005 Paris, France; Univ Paris Est Créteil, INSERM U955, Institut Mondor de Recherche Biomédicale, Equipe NeuroPsychologie Interventionnelle, F-94010 Créteil, France; NeurATRIS Créteil, France
| | - Nicolas Fraisse
- Département d'Etudes Cognitives, École normale supérieure, PSL University, NeuroPsychologie Interventionnelle, 75005 Paris, France; Univ Paris Est Créteil, INSERM U955, Institut Mondor de Recherche Biomédicale, Equipe NeuroPsychologie Interventionnelle, F-94010 Créteil, France; NeurATRIS Créteil, France; AP-HP, Hôpital Henri Mondor-Albert Chenevier, Centre de référence Maladie de Huntington, Service de Neurologie, F-94010 Créteil, France
| | - Cécilia Jubin
- Département d'Etudes Cognitives, École normale supérieure, PSL University, NeuroPsychologie Interventionnelle, 75005 Paris, France; Univ Paris Est Créteil, INSERM U955, Institut Mondor de Recherche Biomédicale, Equipe NeuroPsychologie Interventionnelle, F-94010 Créteil, France; NeurATRIS Créteil, France; AP-HP, Hôpital Henri Mondor-Albert Chenevier, Centre de référence Maladie de Huntington, Service de Neurologie, F-94010 Créteil, France
| | - Graça Morgado
- Inserm, Centre d'Investigation Clinique 1430, AP-HP, Hôpital Henri Mondor, Créteil, France
| | - Katia Youssov
- Département d'Etudes Cognitives, École normale supérieure, PSL University, NeuroPsychologie Interventionnelle, 75005 Paris, France; Univ Paris Est Créteil, INSERM U955, Institut Mondor de Recherche Biomédicale, Equipe NeuroPsychologie Interventionnelle, F-94010 Créteil, France; NeurATRIS Créteil, France; AP-HP, Hôpital Henri Mondor-Albert Chenevier, Centre de référence Maladie de Huntington, Service de Neurologie, F-94010 Créteil, France; Inserm, Centre d'Investigation Clinique 1430, AP-HP, Hôpital Henri Mondor, Créteil, France
| | - Marine Lunven
- Département d'Etudes Cognitives, École normale supérieure, PSL University, NeuroPsychologie Interventionnelle, 75005 Paris, France; Univ Paris Est Créteil, INSERM U955, Institut Mondor de Recherche Biomédicale, Equipe NeuroPsychologie Interventionnelle, F-94010 Créteil, France; NeurATRIS Créteil, France.
| | - Anne-Catherine Bachoud-Levi
- Département d'Etudes Cognitives, École normale supérieure, PSL University, NeuroPsychologie Interventionnelle, 75005 Paris, France; Univ Paris Est Créteil, INSERM U955, Institut Mondor de Recherche Biomédicale, Equipe NeuroPsychologie Interventionnelle, F-94010 Créteil, France; NeurATRIS Créteil, France; AP-HP, Hôpital Henri Mondor-Albert Chenevier, Centre de référence Maladie de Huntington, Service de Neurologie, F-94010 Créteil, France; Inserm, Centre d'Investigation Clinique 1430, AP-HP, Hôpital Henri Mondor, Créteil, France
| |
Collapse
|
12
|
Ruiz de Sabando A, Ciosi M, Galbete A, Cumming SA, Monckton DG, Ramos-Arroyo MA. Somatic CAG repeat instability in intermediate alleles of the HTT gene and its potential association with a clinical phenotype. Eur J Hum Genet 2024; 32:770-778. [PMID: 38433266 PMCID: PMC11220145 DOI: 10.1038/s41431-024-01546-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 01/08/2024] [Accepted: 01/17/2024] [Indexed: 03/05/2024] Open
Abstract
Huntington disease (HD) is a neurodegenerative disorder caused by ≥36 CAGs in the HTT gene. Intermediate alleles (IAs) (27-35 CAGs) are not considered HD-causing, but their potential association with neurocognitive symptoms remains controversial. As HTT somatic CAG expansion influences HD onset, we hypothesised that IAs are somatically unstable, and that somatic CAG expansion may drive phenotypic presentation in some IA carriers. We quantified HTT somatic CAG expansions by MiSeq sequencing in the blood DNA of 164 HD subjects and 191 IA (symptomatic and control) carriers, and in the brain DNA of a symptomatic 33 CAG carrier. We also performed genotype-phenotype analysis. The phenotype of symptomatic IA carriers was characterised by motor (85%), cognitive (27%) and/or behavioural (29%) signs, with a late (58.7 ± 18.6 years), but not CAG-dependent, age at onset. IAs displayed somatic expansion that were CAG and age-dependent in blood DNA, with 0.4% and 0.01% of DNA molecules expanding by CAG and year, respectively. Somatic expansions of +1 and +2 CAGs were detected in the brain of the individual with 33 CAGs, with the highest expansion frequency in the putamen (10.3%) and the lowest in the cerebellum (4.8%). Somatic expansion in blood DNA was not different in symptomatic vs. control IA carriers. In conclusion, we show that HTT IAs are somatically unstable, but we found no association with HD-like phenotypes. It is plausible, however, that some IAs, close to the HD pathological threshold and with a predisposing genetic background, could manifest with neurocognitive symptoms.
Collapse
Affiliation(s)
- Ainara Ruiz de Sabando
- Department of Medical Genetics, Hospital Universitario de Navarra, IdiSNA, 31008, Pamplona, Spain
- Department of Health Sciences, Universidad Pública de Navarra, IdiSNA, 31008, Pamplona, Spain
- Fundación Miguel Servet-Navarrabiomed, IdiSNA, 31008, Pamplona, Spain
| | - Marc Ciosi
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Arkaitz Galbete
- Department of Statistics, Informatics and Mathematics, Universidad Pública de Navarra, IdiSNA, 31006, Pamplona, Spain
| | - Sarah A Cumming
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Darren G Monckton
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Maria A Ramos-Arroyo
- Department of Medical Genetics, Hospital Universitario de Navarra, IdiSNA, 31008, Pamplona, Spain.
- Fundación Miguel Servet-Navarrabiomed, IdiSNA, 31008, Pamplona, Spain.
| |
Collapse
|
13
|
Le Stanc L, Lunven M, Giavazzi M, Sliwinski A, Youssov K, Bachoud-Lévi AC, Jacquemot C. Cognitive reserve involves decision making and is associated with left parietal and hippocampal hypertrophy in neurodegeneration. Commun Biol 2024; 7:741. [PMID: 38890487 PMCID: PMC11189446 DOI: 10.1038/s42003-024-06416-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/05/2024] [Indexed: 06/20/2024] Open
Abstract
Cognitive reserve is the ability to actively cope with brain deterioration and delay cognitive decline in neurodegenerative diseases. It operates by optimizing performance through differential recruitment of brain networks or alternative cognitive strategies. We investigated cognitive reserve using Huntington's disease (HD) as a genetic model of neurodegeneration to compare premanifest HD, manifest HD, and controls. Contrary to manifest HD, premanifest HD behave as controls despite neurodegeneration. By decomposing the cognitive processes underlying decision making, drift diffusion models revealed a response profile that differs progressively from controls to premanifest and manifest HD. Here, we show that cognitive reserve in premanifest HD is supported by an increased rate of evidence accumulation compensating for the abnormal increase in the amount of evidence needed to make a decision. This higher rate is associated with left superior parietal and hippocampal hypertrophy, and exhibits a bell shape over the course of disease progression, characteristic of compensation.
Collapse
Affiliation(s)
- Lorna Le Stanc
- Département d'Études Cognitives, École Normale Supérieure-PSL, Paris, France
- Institut Mondor de Recherche Biomédicale, Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France
- Université Paris-Est Créteil, Faculté de Santé, Créteil, France
- Université Paris Cité, LaPsyDÉ, CNRS, F-75005 Paris, France
| | - Marine Lunven
- Département d'Études Cognitives, École Normale Supérieure-PSL, Paris, France
- Institut Mondor de Recherche Biomédicale, Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France
- Université Paris-Est Créteil, Faculté de Santé, Créteil, France
| | - Maria Giavazzi
- Département d'Études Cognitives, École Normale Supérieure-PSL, Paris, France
- Institut Mondor de Recherche Biomédicale, Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France
- Université Paris-Est Créteil, Faculté de Santé, Créteil, France
| | - Agnès Sliwinski
- Département d'Études Cognitives, École Normale Supérieure-PSL, Paris, France
- Institut Mondor de Recherche Biomédicale, Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France
- Université Paris-Est Créteil, Faculté de Santé, Créteil, France
- AP-HP, Centre de Référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, Créteil, France
| | - Katia Youssov
- Département d'Études Cognitives, École Normale Supérieure-PSL, Paris, France
- Institut Mondor de Recherche Biomédicale, Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France
- Université Paris-Est Créteil, Faculté de Santé, Créteil, France
- AP-HP, Centre de Référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, Créteil, France
| | - Anne-Catherine Bachoud-Lévi
- Département d'Études Cognitives, École Normale Supérieure-PSL, Paris, France
- Institut Mondor de Recherche Biomédicale, Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France
- Université Paris-Est Créteil, Faculté de Santé, Créteil, France
- AP-HP, Centre de Référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, Créteil, France
| | - Charlotte Jacquemot
- Département d'Études Cognitives, École Normale Supérieure-PSL, Paris, France.
- Institut Mondor de Recherche Biomédicale, Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France.
- Université Paris-Est Créteil, Faculté de Santé, Créteil, France.
| |
Collapse
|
14
|
Sprenger GP, van Zwet EW, Bakels HS, Achterberg WP, Roos RA, de Bot ST. Prevalence and burden of pain across the entire spectrum of Huntington's disease. J Neurol Neurosurg Psychiatry 2024; 95:647-655. [PMID: 38290837 DOI: 10.1136/jnnp-2023-332992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 01/09/2024] [Indexed: 02/01/2024]
Abstract
BACKGROUND Pain is an important symptom in Huntington's disease (HD), however, not systematically studied and understood. The objective of the current study is to assess the prevalence of pain, pain interference in daily activities, painful conditions, analgesic use and the severity of the pain burden across different disease stages and 'Age at symptom Onset' groups. Additionally, the association between pain and disease burden was investigated. METHODS A cross-sectional analysis was conducted within two large data sets, which included different types of pain scales. Multivariable logistic regression analyses and analyses of variance were performed to compare the pain levels with those in the general population. The analyses were adjusted for sex and age. Locally Estimated Scatterplot Smoothing was used to test the association between pain and the HD pathology score: a measure of disease burden. RESULTS The mean prevalence of pain in the HD population was 40% and for pain interference around 35% in both data sets. Patients in the early, middle and late stage of HD experience more pain burden compared with what is reported in patients with chronic pain (p<0.01). A positive and significant association was demonstrated between pain and disease burden. Patients in late stage HD with pain use significantly less analgesics compared with the general population (5% vs 13%, respectively (p<0.01)). CONCLUSIONS Pain is a prevalent and important symptom in HD. Severe pain burden in the HD population is present and positively associated with disease burden. Risk for undertreatment with analgesics is nevertheless present. Awareness of pain in HD needs to be increased, both clinically and scientifically.
Collapse
Affiliation(s)
- Gregory P Sprenger
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
- Huntington Center, Amstelring, Amsterdam, The Netherlands
| | - Erik W van Zwet
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Hannah S Bakels
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Wilco P Achterberg
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, The Netherlands
- Huntington Center overduin, Topaz, Leiden, The Netherlands
| | - Raymund A Roos
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Susanne T de Bot
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
15
|
Raschka T, Li Z, Gaßner H, Kohl Z, Jukic J, Marxreiter F, Fröhlich H. Unraveling progression subtypes in people with Huntington's disease. EPMA J 2024; 15:275-287. [PMID: 38841617 PMCID: PMC11148000 DOI: 10.1007/s13167-024-00368-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/09/2024] [Indexed: 06/07/2024]
Abstract
Background Huntington's disease (HD) is a progressive neurodegenerative disease caused by a CAG trinucleotide expansion in the huntingtin gene. The length of the CAG repeat is inversely correlated with disease onset. HD is characterized by hyperkinetic movement disorder, psychiatric symptoms, and cognitive deficits, which greatly impact patient's quality of life. Despite this clear genetic course, high variability of HD patients' symptoms can be observed. Current clinical diagnosis of HD solely relies on the presence of motor signs, disregarding the other important aspects of the disease. By incorporating a broader approach that encompasses motor as well as non-motor aspects of HD, predictive, preventive, and personalized (3P) medicine can enhance diagnostic accuracy and improve patient care. Methods Multisymptom disease trajectories of HD patients collected from the Enroll-HD study were first aligned on a common disease timescale to account for heterogeneity in disease symptom onset and diagnosis. Following this, the aligned disease trajectories were clustered using the previously published Variational Deep Embedding with Recurrence (VaDER) algorithm and resulting progression subtypes were clinically characterized. Lastly, an AI/ML model was learned to predict the progression subtype from only first visit data or with data from additional follow-up visits. Results Results demonstrate two distinct subtypes, one large cluster (n = 7122) showing a relative stable disease progression and a second, smaller cluster (n = 411) showing a dramatically more progressive disease trajectory. Clinical characterization of the two subtypes correlates with CAG repeat length, as well as several neurobehavioral, psychiatric, and cognitive scores. In fact, cognitive impairment was found to be the major difference between the two subtypes. Additionally, a prognostic model shows the ability to predict HD subtypes from patients' first visit only. Conclusion In summary, this study aims towards the paradigm shift from reactive to preventive and personalized medicine by showing that non-motor symptoms are of vital importance for predicting and categorizing each patients' disease progression pattern, as cognitive decline is oftentimes more reflective of HD progression than its motor aspects. Considering these aspects while counseling and therapy definition will personalize each individuals' treatment. The ability to provide patients with an objective assessment of their disease progression and thus a perspective for their life with HD is the key to improving their quality of life. By conducting additional analysis on biological data from both subtypes, it is possible to gain a deeper understanding of these subtypes and uncover the underlying biological factors of the disease. This greatly aligns with the goal of shifting towards 3P medicine. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-024-00368-2.
Collapse
Affiliation(s)
- Tamara Raschka
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53757 Sankt Augustin, Germany
| | - Zexin Li
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53757 Sankt Augustin, Germany
- Bonn-Aachen International Center for IT, University of Bonn, Friedrich-Hirzebruch-Allee 6, 53115 Bonn, Germany
| | - Heiko Gaßner
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
- Fraunhofer IIS, Fraunhofer Institute for Integrated Circuits IIS, Am Wolfsmantel 33, 91058 Erlangen, Germany
| | - Zacharias Kohl
- Department of Neurology, University of Regensburg, Regensburg, Germany
| | - Jelena Jukic
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
- Center for Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Franz Marxreiter
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
- Center for Movement Disorders, Passauer Wolf, 93333 Bad Gögging, Germany
- Center for Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Holger Fröhlich
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53757 Sankt Augustin, Germany
- Bonn-Aachen International Center for IT, University of Bonn, Friedrich-Hirzebruch-Allee 6, 53115 Bonn, Germany
| |
Collapse
|
16
|
Korpela S, Sundblom J, Zetterberg H, Constantinescu R, Svenningsson P, Paucar M, Niemelä V. Cerebrospinal fluid glial fibrillary acidic protein, in contrast to amyloid beta protein, is associated with disease symptoms in Huntington's disease. J Neurol Sci 2024; 459:122979. [PMID: 38569376 DOI: 10.1016/j.jns.2024.122979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/07/2024] [Accepted: 03/25/2024] [Indexed: 04/05/2024]
Abstract
INTRODUCTION Huntington's disease (HD) is a hereditary neurodegenerative disease, currently lacking disease-modifying treatments. Biomarkers are needed for objective assessment of disease progression. Evidence supports both complex protein aggregation and astrocyte activation in HD. This study assesses the 42 amino acid long amyloid beta (Aβ42) and glial fibrillary acidic protein (GFAP) as potential biomarkers in the cerebrospinal fluid (CSF) of HD mutation carriers. METHODS CSF from participants was obtained from three sites in Sweden. Clinical symptoms were graded with the composite Unified Huntington's disease rating scale (cUHDRS). Protein concentrations were measured using ELISA. Pearson correlations were calculated to assess disease progression association. Results were adjusted for age and collection site. RESULTS The study enrolled 28 manifest HD patients (ManHD), 13 premanifest HD gene-expansion carriers (PreHD) and 20 controls. Aβ42 levels did not differ between groups and there was no correlation with measures of disease progression. GFAP concentration was higher in ManHD (424 ng/l, SD 253) compared with both PreHD (266 ng/l, SD 92.4) and controls (208 ng/l, SD 83.7). GFAP correlated with both cUHDRS (r = -0.77, p < 0.001), and 5-year risk of disease onset (r = 0.70, p = 0.008). CONCLUSION We provide evidence that indicates CSF Aβ42 has limited potential as a biomarker for HD. GFAP is a potential biomarker of progression in HD. Validation in larger cohorts measuring GFAP in blood and CSF would be of interest.
Collapse
Affiliation(s)
- Sara Korpela
- Department of Medicine, Neurology, Västerås Central Hospital, Västerås, Sweden
| | - Jimmy Sundblom
- Department of Medical Sciences, Neurosurgery, Uppsala University, Uppsala, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; UK Dementia Research Institute at UCL, London, UK; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Radu Constantinescu
- Institute of Neuroscience and Physiology, Clinical Neuroscience, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Martin Paucar
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Valter Niemelä
- Department of Medical Sciences, Neurology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
17
|
Gijs M, Jorna N, Datson N, Beekman C, Dansokho C, Weiss A, Linden DEJ, Oosterloo M. High Levels of Mutant Huntingtin Protein in Tear Fluid From Huntington's Disease Gene Expansion Carriers. J Mov Disord 2024; 17:181-188. [PMID: 38379425 PMCID: PMC11082600 DOI: 10.14802/jmd.24014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/17/2024] [Accepted: 02/21/2024] [Indexed: 02/22/2024] Open
Abstract
OBJECTIVE Huntington's disease (HD) is an autosomal dominant, fully penetrant, neurodegenerative disease that most commonly affects middle-aged adults. HD is caused by a CAG repeat expansion in the HTT gene, resulting in the expression of mutant huntingtin (mHTT). Our aim was to detect and quantify mHTT in tear fluid, which, to our knowledge, has never been measured before. METHODS We recruited 20 manifest and 13 premanifest HD gene expansion carriers, and 20 age-matched controls. All patients underwent detailed assessments, including the Unified Huntington's Disease Rating Scale (UHDRS) total motor score (TMS) and total functional capacity (TFC) score. Tear fluid was collected using paper Schirmer's strips. The level of tear mHTT was determined using single-molecule counting SMCxPRO technology. RESULTS The average tear mHTT levels in manifest (67,223 ± 80,360 fM) and premanifest patients (55,561 ± 45,931 fM) were significantly higher than those in controls (1,622 ± 2,179 fM). We noted significant correlations between tear mHTT levels and CAG repeat length, "estimated years to diagnosis," disease burden score and UHDRS TMS and TFC. The receiver operating curve demonstrated an almost perfect score (area under the curve [AUC] = 0.9975) when comparing controls to manifest patients. Similarly, the AUC between controls and premanifest patients was 0.9846. The optimal cutoff value for distinguishing between controls and manifest patients was 4,544 fM, whereas it was 6,596 fM for distinguishing between controls and premanifest patients. CONCLUSION Tear mHTT has potential for early and noninvasive detection of alterations in HD patients and could be integrated into both clinical trials and clinical diagnostics.
Collapse
Affiliation(s)
- Marlies Gijs
- University Eye Clinic Maastricht, Mental Health and Neuroscience Research Institute (MHeNs), Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Nynke Jorna
- Department of Neurology, Mental Health and Neuroscience Research Institute (MHeNs), Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | | | | | | | | | - David E. J. Linden
- Department of Neurology, Mental Health and Neuroscience Research Institute (MHeNs), Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Mayke Oosterloo
- Department of Neurology, Mental Health and Neuroscience Research Institute (MHeNs), Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| |
Collapse
|
18
|
Novy C, Busk ØL, Tysnes OB, Landa SS, Aanjesen TN, Alstadhaug KB, Bjerknes TL, Bjørnå IK, Bråthen G, Dahl E, Demic N, Fahlström M, Flemmen HØ, Hallerstig E, HogenEsch I, Kampman MT, Kleveland G, Kvernmo HB, Ljøstad U, Maniaol A, Morsund AH, Nakken O, Olsen CG, Schlüter K, Utvik MS, Yaseen R, Holla ØL, Holmøy T, Høyer H. Repeat expansions in AR, ATXN1, ATXN2 and HTT in Norwegian patients diagnosed with amyotrophic lateral sclerosis. Brain Commun 2024; 6:fcae087. [PMID: 38585669 PMCID: PMC10998343 DOI: 10.1093/braincomms/fcae087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/23/2024] [Accepted: 03/12/2024] [Indexed: 04/09/2024] Open
Abstract
Genetic repeat expansions cause neuronal degeneration in amyotrophic lateral sclerosis as well as other neurodegenerative disorders such as spinocerebellar ataxia, Huntington's disease and Kennedy's disease. Repeat expansions in the same gene can cause multiple clinical phenotypes. We aimed to characterize repeat expansions in a Norwegian amyotrophic lateral sclerosis cohort. Norwegian amyotrophic lateral sclerosis patients (n = 414) and neurologically healthy controls adjusted for age and gender (n = 713) were investigated for repeat expansions in AR, ATXN1, ATXN2 and HTT using short read exome sequencing and the ExpansionHunter software. Five amyotrophic lateral sclerosis patients (1.2%) and two controls (0.3%) carried ≥36 repeats in HTT (P = 0.032), and seven amyotrophic lateral sclerosis patients (1.7%) and three controls (0.4%) carried ≥29 repeats in ATXN2 (P = 0.038). One male diagnosed with amyotrophic lateral sclerosis carried a pathogenic repeat expansion in AR, and his diagnosis was revised to Kennedy's disease. In ATXN1, 50 amyotrophic lateral sclerosis patients (12.1%) and 96 controls (13.5%) carried ≥33 repeats (P = 0.753). None of the patients with repeat expansions in ATXN2 or HTT had signs of Huntington's disease or spinocerebellar ataxia type 2, based on a re-evaluation of medical records. The diagnosis of amyotrophic lateral sclerosis was confirmed in all patients, with the exception of one patient who had primary lateral sclerosis. Our findings indicate that repeat expansions in HTT and ATXN2 are associated with increased likelihood of developing amyotrophic lateral sclerosis. Further studies are required to investigate the potential relationship between HTT repeat expansions and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Camilla Novy
- Department of Medical Genetics, Telemark Hospital Trust, 3710 Skien, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0316 Oslo, Norway
| | - Øyvind L Busk
- Department of Medical Genetics, Telemark Hospital Trust, 3710 Skien, Norway
| | - Ole-Bjørn Tysnes
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5009 Bergen, Norway
| | - Sigve S Landa
- Department of Medical Genetics, Telemark Hospital Trust, 3710 Skien, Norway
| | - Tori N Aanjesen
- Department of Neurology, Akershus University Hospital, 1478 Lørenskog, Norway
| | | | - Tale L Bjerknes
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5009 Bergen, Norway
- Institute of Clinical Medicine, University of Bergen, 5007 Bergen, Norway
| | - Ingrid K Bjørnå
- Department of Neurology, Vestre Viken Hospital Trust, 3004 Drammen, Norway
| | - Geir Bråthen
- Department of Neurology and Clinical Neurophysiology, St. Olavs Hospital, Trondheim University Hospital, 7030 Trondheim, Norway
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, 7034 Trondheim, Norway
| | - Elin Dahl
- Department of Neurology, Telemark Hospital Trust, 3710 Skien, Norway
| | - Natasha Demic
- Department of Neurology, Vestfold Hospital Trust, 3103 Tønsberg, Norway
| | - Maria Fahlström
- Department of Medical Genetics, Telemark Hospital Trust, 3710 Skien, Norway
| | - Heidi Ø Flemmen
- Department of Neurology, Telemark Hospital Trust, 3710 Skien, Norway
| | - Erika Hallerstig
- Department of Neurology, Østfold Hospital Trust, 1714 Grålum, Norway
| | - Ineke HogenEsch
- Department of Neurology, Fonna Hospital Trust, 5528 Haugesund, Norway
| | - Margitta T Kampman
- Department of Neurology, University Hospital of North Norway, 9019 Tromsø, Norway
| | - Grethe Kleveland
- Department of Neurology, Innlandet Hospital Trust, 2609 Lillehammer, Norway
| | - Helene B Kvernmo
- Department of Neurology and Clinical Neurophysiology, St. Olavs Hospital, Trondheim University Hospital, 7030 Trondheim, Norway
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, 7034 Trondheim, Norway
| | - Unn Ljøstad
- Institute of Clinical Medicine, University of Bergen, 5007 Bergen, Norway
- Department of Neurology, Sørlandet Hospital Trust, 4615 Kristiansand, Norway
| | - Angelina Maniaol
- Department of Neurology, Oslo University Hospital, 0450 Oslo, Norway
| | | | - Ola Nakken
- Department of Neurology, Akershus University Hospital, 1478 Lørenskog, Norway
| | - Cathrine G Olsen
- Department of Medical Genetics, Telemark Hospital Trust, 3710 Skien, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0316 Oslo, Norway
| | - Katrin Schlüter
- Department of Neurology, Stavanger University Hospital, 4019 Stavanger, Norway
| | - May-Sissel Utvik
- Department of Neurology, Namsos Hospital Trust, 7803 Namsos, Norway
| | - Ryaz Yaseen
- Department of Neurology, Oslo University Hospital, 0450 Oslo, Norway
| | - Øystein L Holla
- Department of Medical Genetics, Telemark Hospital Trust, 3710 Skien, Norway
| | - Trygve Holmøy
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0316 Oslo, Norway
- Department of Neurology, Akershus University Hospital, 1478 Lørenskog, Norway
| | - Helle Høyer
- Department of Medical Genetics, Telemark Hospital Trust, 3710 Skien, Norway
| |
Collapse
|
19
|
Paz-Rodríguez F, Chávez-Oliveros M, Bernal-Pérez A, Ochoa-Morales A, Martínez-Ruano L, Camacho-Molina A, Rodríguez-Agudelo Y. Neuropsychological performance and disease burden in individuals at risk of developing Huntington disease. Neurologia 2024; 39:127-134. [PMID: 38272259 DOI: 10.1016/j.nrleng.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/31/2021] [Accepted: 04/06/2021] [Indexed: 01/27/2024] Open
Abstract
INTRODUCTION Huntington disease (HD) is a hereditary neurodegenerative disorder. Thanks to predictive diagnosis, incipient clinical characteristics have been described in the prodromal phase. OBJECTIVE To compare performance in cognitive tasks of carriers (HDC) and non-carriers (non-HDC) of the huntingtin gene and to analyse the variability in performance as a function of disease burden and proximity to the manifest stage (age of symptom onset). METHOD A sample of 146 participants in a predictive diagnosis of HD programme were divided into the HDC (41.1%) and non-HDC groups (58.9%). Mathematical formulae were used to calculate disease burden and proximity to the manifest stage in the HDC group; these parameters were correlated with neuropsychological performance. RESULTS Significant differences were observed between groups in performance on the Mini-Mental State Examination (MMSE), Stroop-B, Symbol-Digit Modalities Test (SDMT), and phonological fluency. In the HDC group, correlations were observed between disease burden and performance on the MMSE, Stroop-B, and SDMT. The group of patients close to the manifest stage scored lowest on the MMSE, Stroop-B, Stroop-C, SDMT, and semantic verbal fluency. According to the multivariate analysis of covariance, the MMSE effect shows statistically significant differences in disease burden and proximity to onset of symptoms. CONCLUSIONS Members of the HDC group close to the manifest phase performed more poorly on tests assessing information processing speed and attention. Prefrontal cognitive dysfunction appears early, several years before the motor diagnosis of HD.
Collapse
Affiliation(s)
- F Paz-Rodríguez
- Laboratorio de Neuropsicología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| | - M Chávez-Oliveros
- Laboratorio de Neuropsicología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| | - A Bernal-Pérez
- Laboratorio de Neuropsicología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| | - A Ochoa-Morales
- Departamento de Genética, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| | - L Martínez-Ruano
- Departamento de Genética, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| | - A Camacho-Molina
- Departamento de Genética, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| | - Y Rodríguez-Agudelo
- Laboratorio de Neuropsicología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico.
| |
Collapse
|
20
|
Dalene Skarping K, Arning L, Petersén Å, Nguyen HP, Gebre-Medhin S. Attenuated huntingtin gene CAG nucleotide repeat size in individuals with Lynch syndrome. Sci Rep 2024; 14:4300. [PMID: 38383663 PMCID: PMC10881568 DOI: 10.1038/s41598-024-54277-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 02/10/2024] [Indexed: 02/23/2024] Open
Abstract
DNA mismatch repair (MMR) is thought to contribute to the onset and progression of Huntington disease (HD) by promoting somatic expansion of the pathogenic CAG nucleotide repeat in the huntingtin gene (HTT). Here we have studied constitutional HTT CAG repeat size in two cohorts of individuals with Lynch syndrome (LS) carrying heterozygous loss-of-function variants in the MMR genes MLH1 (n = 12/60; Lund cohort/Bochum cohort, respectively), MSH2 (n = 15/88), MSH6 (n = 21/23), and controls (n = 19/559). The sum of CAG repeats for both HTT alleles in each individual was calculated due to unknown segregation with the LS allele. In the larger Bochum cohort, the sum of CAG repeats was lower in the MLH1 subgroup compared to controls (MLH1 35.40 CAG repeats ± 3.6 vs. controls 36.89 CAG repeats ± 4.5; p = 0.014). All LS genetic subgroups in the Bochum cohort displayed lower frequencies of unstable HTT intermediate alleles and lower HTT somatic CAG repeat expansion index values compared to controls. Collectively, our results indicate that MMR gene haploinsufficiency could have a restraining impact on constitutional HTT CAG repeat size and support the notion that the MMR pathway is a driver of nucleotide repeat expansion diseases.
Collapse
Affiliation(s)
- Karin Dalene Skarping
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Department of Clinical Genetics and Pathology, Office for Medical Service, 221 85, Lund, Sweden
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Larissa Arning
- Department of Human Genetics, Faculty of Medicine, Ruhr University Bochum, Universitätsstr. 150, 44801, Bochum, Germany
| | - Åsa Petersén
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Huu Phuc Nguyen
- Department of Human Genetics, Faculty of Medicine, Ruhr University Bochum, Universitätsstr. 150, 44801, Bochum, Germany.
| | - Samuel Gebre-Medhin
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden.
- Department of Clinical Genetics and Pathology, Office for Medical Service, 221 85, Lund, Sweden.
| |
Collapse
|
21
|
Yao J, Morrison MA, Jakary A, Avadiappan S, Rowley P, Glueck J, Driscoll T, Geschwind MD, Nelson AB, Possin KL, Xu D, Hess CP, Lupo JM. Altered Iron and Microstructure in Huntington's Disease Subcortical Nuclei: Insight From 7T MRI. J Magn Reson Imaging 2024:10.1002/jmri.29195. [PMID: 38206986 PMCID: PMC11521114 DOI: 10.1002/jmri.29195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND Pathophysiological changes of Huntington's disease (HD) can precede symptom onset by decades. Robust imaging biomarkers are needed to monitor HD progression, especially before the clinical onset. PURPOSE To investigate iron dysregulation and microstructure alterations in subcortical regions as HD imaging biomarkers, and to associate such alterations with motor and cognitive impairments. STUDY TYPE Prospective. POPULATION Fourteen individuals with premanifest HD (38.0 ± 11.0 years, 9 females; far-from-onset N = 6, near-onset N = 8), 21 manifest HD patients (49.1 ± 12.1 years, 11 females), and 33 age-matched healthy controls (43.9 ± 12.2 years, 17 females). FIELD STRENGTH/SEQUENCE 7 T, T1 -weighted imaging, quantitative susceptibility mapping, and diffusion tensor imaging. ASSESSMENT Volume, susceptibility, fractional anisotropy (FA), and mean diffusivity (MD) within subcortical brain structures were compared across groups, used to establish HD classification models, and correlated to clinical measures and cognitive assessments. STATISTICAL TESTS Generalized linear model, multivariate logistic regression, receiver operating characteristics with the area under the curve (AUC), and likelihood ratio test comparing a volumetric model to one that also includes susceptibility and diffusion metrics, Wilcoxon paired signed-rank test, and Pearson's correlation. A P-value <0.05 after Benjamini-Hochberg correction was considered statistically significant. RESULTS Significantly higher striatal susceptibility and FA were found in premanifest and manifest HD preceding atrophy, even in far-from-onset premanifest HD compared to controls (putamen susceptibility: 0.027 ± 0.022 vs. 0.018 ± 0.013 ppm; FA: 0.358 ± 0.048 vs. 0.313 ± 0.039). The model with additional susceptibility, FA, and MD features showed higher AUC compared to volume features alone when differentiating premanifest HD from HC (0.83 vs. 0.66), and manifest from premanifest HD (0.94 vs. 0.83). Higher striatal susceptibility significantly correlated with cognitive deterioration in HD (executive function: r = -0.600; socioemotional function: r = -0.486). DATA CONCLUSION 7 T MRI revealed iron dysregulation and microstructure alterations with HD progression, which could precede volume loss, provide added value to HD differentiation, and might be associated with cognitive changes. EVIDENCE LEVEL 2 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Jingwen Yao
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, California, USA
- Department of Radiological Sciences, UCLA, Los Angeles, California, USA
- Department of Bioengineering, UCLA, Los Angeles, California, USA
| | - Melanie A Morrison
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, California, USA
- UCSF/UC Berkeley Graduate Program in Bioengineering, San Francisco and Berkeley, California, USA
| | - Angela Jakary
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, California, USA
| | - Sivakami Avadiappan
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, California, USA
| | - Paul Rowley
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, California, USA
| | - Julia Glueck
- Department of Neurology, UCSF, San Francisco, California, USA
| | | | | | | | | | - Duan Xu
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, California, USA
- UCSF/UC Berkeley Graduate Program in Bioengineering, San Francisco and Berkeley, California, USA
| | - Christopher P Hess
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, California, USA
- Department of Neurology, UCSF, San Francisco, California, USA
| | - Janine M Lupo
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, California, USA
- UCSF/UC Berkeley Graduate Program in Bioengineering, San Francisco and Berkeley, California, USA
| |
Collapse
|
22
|
Kim R, Seong MW, Oh B, Shin HS, Lee JS, Park S, Jang M, Jeon B, Kim HJ, Lee JY. Analysis of HTT CAG repeat expansion among healthy individuals and patients with chorea in Korea. Parkinsonism Relat Disord 2024; 118:105930. [PMID: 37992538 DOI: 10.1016/j.parkreldis.2023.105930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/24/2023]
Abstract
BACKGROUND Although the epidemiology of Huntington's disease (HD) in Korea differs notably from that in Western countries, the genetic disparities between these regions remain unclear. OBJECTIVE To investigate the characteristics and clinical significance of cytosine-adenine-guanine (CAG) repeat size associated with HD in the Korean population. METHODS We analyzed the CAG repeat lengths of the HTT gene in 941 healthy individuals (1,882 alleles) and 954 patients with chorea (1,908 alleles) from two referral hospitals in Korea. We presented normative CAG repeat length data for the Korean population and computed the reduced penetrance (36-39 CAG) and intermediate allele (27-35 CAG) frequencies in the two groups. Furthermore, we investigated the relationship between intermediate alleles and chorea development using logistic regression models in individuals aged ≥55 years. RESULTS The mean (±standard deviation) CAG repeat length in healthy individuals was 17.5 ± 2.0, with a reduced penetrance allele frequency of 0.05 % (1/1882) and intermediate allele frequency of 0.69 % (13/1882). We identified 213 patients with genetically confirmed HD whose CAG repeat length ranged from 39 to 140, with a mean of 45.2 ± 7.9 in the longer allele. Compared with normal CAG repeat alleles, intermediate CAG repeat alleles were significantly related to a higher risk of developing chorea (age of onset range, 63-84 years) in individuals aged ≥55 years. CONCLUSIONS This study provides insights into the specific characteristics of CAG repeat lengths in the HTT gene in the Korean population. The reduced penetrance and intermediate allele frequencies in the Korean general population seem to be lower than those reported in Western populations. The presence of intermediate alleles may increase the risk of chorea in the Korean elderly population, which requires further large-scale investigations.
Collapse
Affiliation(s)
- Ryul Kim
- Department of Neurology, Inha University Hospital, Inha University College of Medicine, Incheon, South Korea
| | - Moon-Woo Seong
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Bumjo Oh
- Department of Familial Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Ho Seop Shin
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Jee-Soo Lee
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Sangmin Park
- Department of Neurology, Daejeon Eulji Medical Center, Eulji University College of Medicine, Daejeon, South Korea
| | - Mihee Jang
- Department of Neurology, JMH Seoul Neurologic Clinic, Seoul, South Korea
| | - Beomseok Jeon
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Han-Joon Kim
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea.
| | - Jee-Young Lee
- Department of Neurology, Seoul Metropolitan Government - Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
23
|
Saft C, Jessen J, Hoffmann R, Lukas C, Skodda S. Speech Biomarkers in Huntington's Disease: A Longitudinal Follow-Up Study in Premanifest Mutation Carriers. J Huntingtons Dis 2024; 13:369-373. [PMID: 38995795 DOI: 10.3233/jhd-240021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Speech alterations have been reported in manifest Huntington's disease (HD) and premanifest mutation carriers (preHD). The aim of our study was to explore these alterations in preHD and whether they can be used as biomarkers. 13 preHD mutation carriers performed reading task, sustained phonation task and syllable repetition tasks at baseline and after 21 months, as well as clinical examination and MRI. Syllable repetition capacity and self-chosen velocity of single syllable repetition differed significantly between time points. There were no changes in clinical ratings or MRI volumetry. Measurements of speech might be sensitive tools for monitoring subclinical changes in preHD.
Collapse
Affiliation(s)
- Carsten Saft
- Department of Neurology, Huntington-Centre NRW, St. Josef Hospital, Ruhr-University of Bochum, Bochum, Germany
| | - Julia Jessen
- Department of Neurology, Huntington-Centre NRW, St. Josef Hospital, Ruhr-University of Bochum, Bochum, Germany
| | - Rainer Hoffmann
- Department of Neurology, Huntington-Centre NRW, St. Josef Hospital, Ruhr-University of Bochum, Bochum, Germany
- Department of Neuropsychiatry, Huntington Centre South, kbo-Isar-Amper-Klinikum, Taufkirchen (Vils), Germany
| | - Carsten Lukas
- Institute of Neuroradiology, St. Josef Hospital, Ruhr-University of Bochum, Bochum, Germany
| | - Sabine Skodda
- Department of Neurology, Knappschaftskrankenhaus, Ruhr-University of Bochum, Bochum, Germany
| |
Collapse
|
24
|
Fan YG, Wu TY, Zhao LX, Jia RJ, Ren H, Hou WJ, Wang ZY. From zinc homeostasis to disease progression: Unveiling the neurodegenerative puzzle. Pharmacol Res 2024; 199:107039. [PMID: 38123108 DOI: 10.1016/j.phrs.2023.107039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/16/2023] [Accepted: 12/10/2023] [Indexed: 12/23/2023]
Abstract
Zinc is a crucial trace element in the human body, playing a role in various physiological processes such as oxidative stress, neurotransmission, protein synthesis, and DNA repair. The zinc transporters (ZnTs) family members are responsible for exporting intracellular zinc, while Zrt- and Irt-like proteins (ZIPs) are involved in importing extracellular zinc. These processes are essential for maintaining cellular zinc homeostasis. Imbalances in zinc metabolism have been linked to the development of neurodegenerative diseases. Disruptions in zinc levels can impact the survival and activity of neurons, thereby contributing to the progression of neurodegenerative diseases through mechanisms like cell apoptosis regulation, protein phase separation, ferroptosis, oxidative stress, and neuroinflammation. Therefore, conducting a systematic review of the regulatory network of zinc and investigating the relationship between zinc dysmetabolism and neurodegenerative diseases can enhance our understanding of the pathogenesis of these diseases. Additionally, it may offer new insights and approaches for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yong-Gang Fan
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China.
| | - Ting-Yao Wu
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - Ling-Xiao Zhao
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Rong-Jun Jia
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Hang Ren
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Wen-Jia Hou
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Zhan-You Wang
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China.
| |
Collapse
|
25
|
Coleman A, Langan MT, Verma G, Knights H, Sturrock A, Leavitt BR, Tabrizi SJ, Scahill RI, Hobbs NZ. Assessment of Perivascular Space Morphometry Across the White Matter in Huntington's Disease Using MRI. J Huntingtons Dis 2024; 13:91-101. [PMID: 38517798 DOI: 10.3233/jhd-231508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2024]
Abstract
Background Perivascular spaces (PVS) are fluid-filled cavities surrounding small cerebral blood vessels. There are limited reports of enlarged PVS across the grey matter in manifest Huntington's disease (HD). Little is known about how PVS morphometry in the white matter may contribute to HD. Enlarged PVS have the potential to both contribute to HD pathology and affect the distribution and success of intraparenchymal and intrathecally administered huntingtin-lowering therapies. Objective To investigate PVS morphometry in the global white matter across the spectrum of HD. Relationships between PVS morphometry and disease burden and severity measures were examined. Methods White matter PVS were segmented on 3T T2 W MRI brain scans of 33 healthy controls, 30 premanifest HD (pre-HD), and 32 early manifest HD (early-HD) participants from the Vancouver site of the TRACK-HD study. PVS count and total PVS volume were measured. Results PVS total count slightly increased in pre-HD (p = 0.004), and early-HD groups (p = 0.005), compared to healthy controls. PVS volume, as a percentage of white matter volume, increased subtly in pre-HD compared to healthy controls (p = 0.044), but not in early-HD. No associations between PVS measures and HD disease burden or severity were found. Conclusions This study reveals relatively preserved PVS morphometry across the global white matter of pre-HD and early-HD. Subtle morphometric abnormalities are implied but require confirmation in a larger cohort. However, in conjunction with previous publications, further investigation of PVS in HD and its potential impact on future treatments, with a focus on subcortical grey matter, is warranted.
Collapse
Affiliation(s)
- Annabelle Coleman
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| | - Mackenzie T Langan
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Biomedical Engineering and Imaging Institute at Mount Sinai School of Medicine, New York, NY, USA
| | - Gaurav Verma
- Biomedical Engineering and Imaging Institute at Mount Sinai School of Medicine, New York, NY, USA
| | - Harry Knights
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| | - Aaron Sturrock
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Blair R Leavitt
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Sarah J Tabrizi
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| | - Rachael I Scahill
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| | - Nicola Z Hobbs
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| |
Collapse
|
26
|
Hobbs NZ, Papoutsi M, Delva A, Kinnunen KM, Nakajima M, Van Laere K, Vandenberghe W, Herath P, Scahill RI. Neuroimaging to Facilitate Clinical Trials in Huntington's Disease: Current Opinion from the EHDN Imaging Working Group. J Huntingtons Dis 2024; 13:163-199. [PMID: 38788082 PMCID: PMC11307036 DOI: 10.3233/jhd-240016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2024] [Indexed: 05/26/2024]
Abstract
Neuroimaging is increasingly being included in clinical trials of Huntington's disease (HD) for a wide range of purposes from participant selection and safety monitoring, through to demonstration of disease modification. Selection of the appropriate modality and associated analysis tools requires careful consideration. On behalf of the EHDN Imaging Working Group, we present current opinion on the utility and future prospects for inclusion of neuroimaging in HD trials. Covering the key imaging modalities of structural-, functional- and diffusion- MRI, perfusion imaging, positron emission tomography, magnetic resonance spectroscopy, and magnetoencephalography, we address how neuroimaging can be used in HD trials to: 1) Aid patient selection, enrichment, stratification, and safety monitoring; 2) Demonstrate biodistribution, target engagement, and pharmacodynamics; 3) Provide evidence for disease modification; and 4) Understand brain re-organization following therapy. We also present the challenges of translating research methodology into clinical trial settings, including equipment requirements and cost, standardization of acquisition and analysis, patient burden and invasiveness, and interpretation of results. We conclude, that with appropriate consideration of modality, study design and analysis, imaging has huge potential to facilitate effective clinical trials in HD.
Collapse
Affiliation(s)
- Nicola Z. Hobbs
- HD Research Centre, UCL Institute of Neurology, UCL, London, UK
| | - Marina Papoutsi
- HD Research Centre, UCL Institute of Neurology, UCL, London, UK
- IXICO plc, London, UK
| | - Aline Delva
- Department of Neurosciences, KU Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Belgium
| | | | | | - Koen Van Laere
- Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, KU Leuven, Belgium
- Division of Nuclear Medicine, University Hospitals Leuven, Belgium
| | - Wim Vandenberghe
- Department of Neurosciences, KU Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Belgium
| | | | | |
Collapse
|
27
|
DiFiglia M, Leavitt BR, Macdonald D, Thompson LM. Towards Standardizing Nomenclature in Huntington's Disease Research. J Huntingtons Dis 2024; 13:119-131. [PMID: 38968054 PMCID: PMC11307060 DOI: 10.3233/jhd-240044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2024] [Indexed: 07/07/2024]
Abstract
The field of Huntington's disease research covers many different scientific disciplines, from molecular biology all the way through to clinical practice, and as our understanding of the disease has progressed over the decades, a great deal of different terminology has accrued. The field is also renowned for its collaborative spirit and use of standardized reagents, assays, datasets, models, and clinical measures, so the use of standardized terms is especially important. We have set out to determine, through a consensus exercise involving basic and clinical scientists working in the field, the most appropriate language to use across disciplines. Nominally, this article will serve as the style guide for the Journal of Huntington's Disease (JHD), the only journal devoted exclusively to HD, and we lay out the preferred and standardized terminology and nomenclature for use in JHD publications. However, we hope that this article will also serve as a useful resource to the HD research community at large and that these recommended naming conventions will be adopted widely.
Collapse
Affiliation(s)
- Marian DiFiglia
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Harvard Medical School, Charlestown, MA, USA
| | - Blair R. Leavitt
- Center for Molecular Medicine and Therapeutics and Departments of Medical Genetics and Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Douglas Macdonald
- CHDI Management, Inc., The Company that Manages the Scientific Activities of CHDI Foundation, Inc, Los Angeles, CA, USA
| | - Leslie M. Thompson
- Departments of Psychiatry and Human Behavior and Neurobiology and Behavior, University of California, Irvine, CA, USA
| |
Collapse
|
28
|
Ashner MC, Garcia TP. Understanding the implications of a complete case analysis for regression models with a right-censored covariate. AM STAT 2023; 78:335-344. [PMID: 39070115 PMCID: PMC11281394 DOI: 10.1080/00031305.2023.2282629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 10/28/2023] [Indexed: 07/30/2024]
Abstract
Despite its drawbacks, the complete case analysis is commonly used in regression models with incomplete covariates. Understanding when the complete case analysis will lead to consistent parameter estimation is vital before use. Our aim here is to demonstrate when a complete case analysis is consistent for randomly right-censored covariates and to discuss the implications of its use even when consistent. Across the censored covariate literature, different assumptions are made to ensure a complete case analysis produces a consistent estimator, which leads to confusion in practice. We make several contributions to dispel this confusion. First, we summarize the language surrounding the assumptions that lead to a consistent complete case estimator. Then, we show a unidirectional hierarchical relationship between these assumptions, which leads us to one sufficient assumption to consider before using a complete case analysis. Lastly, we conduct a simulation study to illustrate the performance of a complete case analysis with a right-censored covariate under different censoring mechanism assumptions, and we demonstrate its use with a Huntington disease data example.
Collapse
Affiliation(s)
| | - Tanya P. Garcia
- Department of Biostatistics, University of North Carolina at Chapel Hill
| |
Collapse
|
29
|
Wilton DK, Mastro K, Heller MD, Gergits FW, Willing CR, Fahey JB, Frouin A, Daggett A, Gu X, Kim YA, Faull RLM, Jayadev S, Yednock T, Yang XW, Stevens B. Microglia and complement mediate early corticostriatal synapse loss and cognitive dysfunction in Huntington's disease. Nat Med 2023; 29:2866-2884. [PMID: 37814059 PMCID: PMC10667107 DOI: 10.1038/s41591-023-02566-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 08/24/2023] [Indexed: 10/11/2023]
Abstract
Huntington's disease (HD) is a devastating monogenic neurodegenerative disease characterized by early, selective pathology in the basal ganglia despite the ubiquitous expression of mutant huntingtin. The molecular mechanisms underlying this region-specific neuronal degeneration and how these relate to the development of early cognitive phenotypes are poorly understood. Here we show that there is selective loss of synaptic connections between the cortex and striatum in postmortem tissue from patients with HD that is associated with the increased activation and localization of complement proteins, innate immune molecules, to these synaptic elements. We also found that levels of these secreted innate immune molecules are elevated in the cerebrospinal fluid of premanifest HD patients and correlate with established measures of disease burden.In preclinical genetic models of HD, we show that complement proteins mediate the selective elimination of corticostriatal synapses at an early stage in disease pathogenesis, marking them for removal by microglia, the brain's resident macrophage population. This process requires mutant huntingtin to be expressed in both cortical and striatal neurons. Inhibition of this complement-dependent elimination mechanism through administration of a therapeutically relevant C1q function-blocking antibody or genetic ablation of a complement receptor on microglia prevented synapse loss, increased excitatory input to the striatum and rescued the early development of visual discrimination learning and cognitive flexibility deficits in these models. Together, our findings implicate microglia and the complement cascade in the selective, early degeneration of corticostriatal synapses and the development of cognitive deficits in presymptomatic HD; they also provide new preclinical data to support complement as a therapeutic target for early intervention.
Collapse
Affiliation(s)
- Daniel K Wilton
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US.
| | - Kevin Mastro
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US
| | - Molly D Heller
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US
| | - Frederick W Gergits
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US
| | - Carly Rose Willing
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US
| | - Jaclyn B Fahey
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US
| | - Arnaud Frouin
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US
| | - Anthony Daggett
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Xiaofeng Gu
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Yejin A Kim
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US
| | - Richard L M Faull
- Department of Anatomy with Radiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Suman Jayadev
- Department of Neurology, University of Washington, Seattle, WA, USA
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Ted Yednock
- Annexon Biosciences, South San Francisco, CA, USA
| | - X William Yang
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Beth Stevens
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, US.
- Stanley Center, Broad Institute, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
30
|
Selvadurai LP, Perlman SL, Wilmot GR, Subramony SH, Gomez CM, Ashizawa T, Paulson HL, Onyike CU, Rosenthal LS, Sair HI, Kuo SH, Ratai EM, Zesiewicz TA, Bushara KO, Öz G, Dietiker C, Geschwind MD, Nelson AB, Opal P, Yacoubian TA, Nopoulos PC, Shakkottai VG, Figueroa KP, Pulst SM, Morrison PE, Schmahmann JD. The S-Factor, a New Measure of Disease Severity in Spinocerebellar Ataxia: Findings and Implications. CEREBELLUM (LONDON, ENGLAND) 2023; 22:790-809. [PMID: 35962273 PMCID: PMC10363993 DOI: 10.1007/s12311-022-01424-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/25/2022] [Indexed: 06/15/2023]
Abstract
Spinocerebellar ataxias (SCAs) are progressive neurodegenerative disorders, but there is no metric that predicts disease severity over time. We hypothesized that by developing a new metric, the Severity Factor (S-Factor) using immutable disease parameters, it would be possible to capture disease severity independent of clinical rating scales. Extracting data from the CRC-SCA and READISCA natural history studies, we calculated the S-Factor for 438 participants with symptomatic SCA1, SCA2, SCA3, or SCA6, as follows: ((length of CAG repeat expansion - maximum normal repeat length) /maximum normal repeat length) × (current age - age at disease onset) × 10). Within each SCA type, the S-Factor at the first Scale for the Assessment and Rating of Ataxia (SARA) visit (baseline) was correlated against scores on SARA and other motor and cognitive assessments. In 281 participants with longitudinal data, the slope of the S-Factor over time was correlated against slopes of scores on SARA and other motor rating scales. At baseline, the S-Factor showed moderate-to-strong correlations with SARA and other motor rating scales at the group level, but not with cognitive performance. Longitudinally the S-Factor slope showed no consistent association with the slope of performance on motor scales. Approximately 30% of SARA slopes reflected a trend of non-progression in motor symptoms. The S-Factor is an observer-independent metric of disease burden in SCAs. It may be useful at the group level to compare cohorts at baseline in clinical studies. Derivation and examination of the S-factor highlighted challenges in the use of clinical rating scales in this population.
Collapse
Affiliation(s)
- Louisa P Selvadurai
- Ataxia Center, Cognitive Behavioral Neurology Unit, Laboratory for Neuroanatomy and Cerebellar Neurobiology, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Susan L Perlman
- Department of Neurology, University of California, Los Angeles, Los Angeles, CA, USA
| | - George R Wilmot
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sub H Subramony
- Department of Neurology, University of Florida College of Medicine, McKnight Brain Institute, Gainesville, FL, USA
| | | | - Tetsuo Ashizawa
- Department of Neurology, Houston Methodist Research Institute, Houston, TX, USA
| | - Henry L Paulson
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Chiadi U Onyike
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Liana S Rosenthal
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Haris I Sair
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sheng-Han Kuo
- Department of Neurology, Columbia University, New York, NY, USA
| | - Eva-Maria Ratai
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Theresa A Zesiewicz
- Department of Neurology, Ataxia Research Center, University of South Florida, Tampa, FL, USA
| | - Khalaf O Bushara
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
| | - Gülin Öz
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, USA
| | - Cameron Dietiker
- Department of Neurology, University of California, San Francisco, CA, USA
| | | | - Alexandra B Nelson
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Puneet Opal
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Talene A Yacoubian
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Peggy C Nopoulos
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Vikram G Shakkottai
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Karla P Figueroa
- Department of Neurology, University of Utah, Salt Lake City, UT, USA
| | - Stefan M Pulst
- Department of Neurology, University of Utah, Salt Lake City, UT, USA
| | - Peter E Morrison
- Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | - Jeremy D Schmahmann
- Ataxia Center, Cognitive Behavioral Neurology Unit, Laboratory for Neuroanatomy and Cerebellar Neurobiology, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
31
|
Sharma G, Biswas SS, Mishra J, Navik U, Kandimalla R, Reddy PH, Bhatti GK, Bhatti JS. Gut microbiota dysbiosis and Huntington's disease: Exploring the gut-brain axis and novel microbiota-based interventions. Life Sci 2023; 328:121882. [PMID: 37356750 DOI: 10.1016/j.lfs.2023.121882] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/17/2023] [Accepted: 06/22/2023] [Indexed: 06/27/2023]
Abstract
Huntington's disease (HD) is a complex progressive neurodegenerative disorder affected by genetic, environmental, and metabolic factors contributing to its pathogenesis. Gut dysbiosis is termed as the alterations of intestinal microbial profile. Emerging research has highlighted the pivotal role of gut dysbiosis in HD, focusing on the gut-brain axis as a novel research parameter in science. This review article provides a comprehensive overview of gut microbiota dysbiosis and its relationship with HD and its pathogenesis along with the future challenges and opportunities. The focuses on the essential mechanisms which link gut dysbiosis to HD pathophysiology including neuroinflammation, immune system dysregulation, altered metabolites composition, and neurotransmitter imbalances. We also explored the impacts of gut dysbiosis on HD onset, severity, and symptoms such as cognitive decline, motor dysfunction, and psychiatric symptoms. Furthermore, we highlight recent advances in therapeutics including microbiota-based therapeutic approaches, including dietary interventions, prebiotics, probiotics, fecal microbiota transplantation, and combination therapies with conventional HD treatments and their applications in managing HD. The future challenges are also highlighted as the heterogeneity of gut microbiota, interindividual variability, establishing causality between gut dysbiosis and HD, identifying optimal therapeutic targets and strategies, and ensuring the long-term safety and efficacy of microbiota-based interventions. This review provides a better understanding of the potential role of gut microbiota in HD pathogenesis and guides the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Garvita Sharma
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Shristi Saroj Biswas
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Jayapriya Mishra
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Umashanker Navik
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, India.
| | - Ramesh Kandimalla
- CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience and Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX 79409, USA.
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, India
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India.
| |
Collapse
|
32
|
Wibawa P, Walterfang M, Malpas CB, Glikmann‐Johnston Y, Poudel G, Razi A, Hannan AJ, Velakoulis D, Georgiou‐Karistianis N. Selective perforant-pathway atrophy in Huntington disease: MRI analysis of hippocampal subfields. Eur J Neurol 2023; 30:2650-2660. [PMID: 37306313 PMCID: PMC10946817 DOI: 10.1111/ene.15918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/17/2023] [Accepted: 06/08/2023] [Indexed: 06/13/2023]
Abstract
INTRODUCTION While individuals with Huntington disease (HD) show memory impairment that indicates hippocampal dysfunction, the available literature does not consistently identify structural evidence for involvement of the whole hippocampus but rather suggests that hippocampal atrophy may be confined to certain hippocampal subregions. METHODS We processed T1-weighted MRI from IMAGE-HD study using FreeSurfer 7.0 and compared the volumes of the hippocampal subfields among 36 early motor symptomatic (symp-HD), 40 pre-symptomatic (pre-HD), and 36 healthy control individuals across three timepoints over 36 months. RESULTS Mixed-model analyses revealed significantly lower subfield volumes in symp-HD, compared with pre-HD and control groups, in the subicular regions of the perforant-pathway: presubiculum, subiculum, dentate gyrus, tail, and right molecular layer. These adjoining subfields aggregated into a single principal component, which demonstrated an accelerated rate of atrophy in the symp-HD. Volumes between pre-HD and controls did not show any significant difference. In the combined HD groups, CAG repeat length and disease burden score were associated with presubiculum, molecular layer, tail, and perforant-pathway subfield volumes. Hippocampal left tail and perforant-pathway subfields were associated with motor onset in the pre-HD group. CONCLUSIONS Hippocampal subfields atrophy in early symptomatic HD affects key regions of the perforant-pathway, which may implicate the distinctive memory impairment at this stage of illness. Their volumetric associations with genetic and clinical markers suggest the selective susceptibility of these subfields to mutant Huntingtin and disease progression.
Collapse
Affiliation(s)
- Pierre Wibawa
- NeuropsychiatryRoyal Melbourne HospitalParkvilleVictoriaAustralia
- Melbourne Neuropsychiatry CenterUniversity of MelbourneParkvilleVictoriaAustralia
- School of Psychological Sciences and Turner Institute for Brain and Mental HealthMonash UniversityClaytonVictoriaAustralia
| | - Mark Walterfang
- NeuropsychiatryRoyal Melbourne HospitalParkvilleVictoriaAustralia
- Melbourne Neuropsychiatry CenterUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Charles B. Malpas
- NeuropsychiatryRoyal Melbourne HospitalParkvilleVictoriaAustralia
- Melbourne Neuropsychiatry CenterUniversity of MelbourneParkvilleVictoriaAustralia
| | - Yifat Glikmann‐Johnston
- School of Psychological Sciences and Turner Institute for Brain and Mental HealthMonash UniversityClaytonVictoriaAustralia
| | - Govinda Poudel
- Mary Mackillop Institute for Health ResearchAustralian Catholic UniversityFitzroyVictoriaAustralia
| | - Adeel Razi
- School of Psychological Sciences and Turner Institute for Brain and Mental HealthMonash UniversityClaytonVictoriaAustralia
| | - Anthony J. Hannan
- Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Dennis Velakoulis
- NeuropsychiatryRoyal Melbourne HospitalParkvilleVictoriaAustralia
- Melbourne Neuropsychiatry CenterUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Nellie Georgiou‐Karistianis
- School of Psychological Sciences and Turner Institute for Brain and Mental HealthMonash UniversityClaytonVictoriaAustralia
| |
Collapse
|
33
|
Davis MC, Hill AT, Fitzgerald PB, Bailey NW, Stout JC, Hoy KE. Neurophysiological correlates of non-motor symptoms in late premanifest and early-stage manifest huntington's disease. Clin Neurophysiol 2023; 153:166-176. [PMID: 37506604 DOI: 10.1016/j.clinph.2023.06.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 05/22/2023] [Accepted: 06/18/2023] [Indexed: 07/30/2023]
Abstract
OBJECTIVE To find sensitive neurophysiological correlates of non-motor symptoms in Huntington's disease (HD), which are essential for the development and assessment of novel treatments. METHODS We used resting state EEG to examine differences in oscillatory activity (analysing the isolated periodic as well as the complete EEG signal) and functional connectivity in 22 late premanifest and early stage people with HD and 20 neurotypical controls. We then assessed the correlations between these neurophysiological markers and clinical measures of apathy and processing speed. RESULTS Significantly lower theta and greater delta resting state power was seen in the HD group, as well as significantly greater delta connectivity. There was a significant positive correlation between theta power and processing speed, however there were no associations between the neurophysiological and apathy measures. CONCLUSIONS We speculate that these changes in oscillatory power and connectivity reflect ongoing, frontally concentrated degenerative and compensatory processes associated with HD. SIGNIFICANCE Our findings support the potential utility of quantitative EEG as a proximate marker of processing speed, but not apathy in HD.
Collapse
Affiliation(s)
- Marie-Claire Davis
- Central Clinical School, Department of Psychiatry, Monash University, Victoria, Australia; Statewide Progressive Neurological Disease Service, Calvary Health Care Bethlehem, Victoria, Australia.
| | - Aron T Hill
- Central Clinical School, Department of Psychiatry, Monash University, Victoria, Australia; Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Victoria, Australia.
| | - Paul B Fitzgerald
- Central Clinical School, Department of Psychiatry, Monash University, Victoria, Australia; School of Medicine and Psychology, Australian National University, Canberra, ACT, Australia.
| | - Neil W Bailey
- Central Clinical School, Department of Psychiatry, Monash University, Victoria, Australia; School of Medicine and Psychology, Australian National University, Canberra, ACT, Australia; Monarch Research Institute Monarch Mental Health Group, 225 Clarence Street, Sydney, NSW 2000, Australia.
| | - Julie C Stout
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, 18 Innovation Walk, Clayton Campus, Wellington Road, Clayton, VIC 3800, Australia.
| | - Kate E Hoy
- Central Clinical School, Department of Psychiatry, Monash University, Victoria, Australia; The Bionics Institute of Australia, 384-388 Albert St, East Melbourne, VIC 3002, Australia.
| |
Collapse
|
34
|
Le Stanc L, Youssov K, Giavazzi M, Sliwinski A, Bachoud-Lévi AC, Jacquemot C. Language disorders in patients with striatal lesions: Deciphering the role of the striatum in language performance. Cortex 2023; 166:91-106. [PMID: 37354871 DOI: 10.1016/j.cortex.2023.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/22/2023] [Accepted: 04/13/2023] [Indexed: 06/26/2023]
Abstract
The classical neural model of language refers to a cortical network involving frontal, parietal and temporal regions. However, patients with subcortical lesions of the striatum have language difficulties. We investigated whether the striatum is directly involved in language or whether its role in decision-making has an indirect effect on language performance, by testing carriers of Huntington's disease (HD) mutations and controls. HD is a genetic neurodegenerative disease primarily affecting the striatum and causing language disorders. We asked carriers of the HD mutation in the premanifest (before clinical diagnosis) and early disease stages, and controls to perform two discrimination tasks, one involving linguistic and the other non-linguistic stimuli. We used the hierarchical drift diffusion model (HDDM) to analyze the participants' responses and to assess the decision and non-decision parameters separately. We hypothesized that any language deficits related to decision-making impairments would be reflected in the decision parameters of linguistic and non-linguistic tasks. We also assessed the relative contributions of both HDDM decision and non-decision parameters to the participants' behavioral data (response time and discriminability). Finally, we investigated whether the decision and non-decision parameters of the HDDM were correlated with brain atrophy. The HDDM analysis showed that patients with early HD have impaired decision parameters relative to controls, regardless of the task. In both tasks, decision parameters better explained the variance of response time and discriminability performance than non-decision parameters. In the linguistic task, decision parameters were positively correlated with gray matter volume in the ventral striatum and putamen, whereas non-decision parameters were not. Language impairment in patients with striatal atrophy is better explained by a deficit of decision-making than by a deficit of core linguistic processing. These results suggest that the striatum is involved in language through the modulation of decision-making, presumably by regulating the process of choice between linguistic alternatives.
Collapse
Affiliation(s)
- Lorna Le Stanc
- Département d'Études Cognitives, École Normale Supérieure-PSL, Paris, France; Institut Mondor de Recherche Biomédicale, Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France; Université Paris-Est Créteil, Faculté de Médecine, Créteil, France; Université Paris Cité, LaPsyDÉ, CNRS, Paris, France
| | - Katia Youssov
- Département d'Études Cognitives, École Normale Supérieure-PSL, Paris, France; Institut Mondor de Recherche Biomédicale, Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France; Université Paris-Est Créteil, Faculté de Médecine, Créteil, France; AP-HP, Centre de Référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, Créteil, France
| | - Maria Giavazzi
- Département d'Études Cognitives, École Normale Supérieure-PSL, Paris, France; Institut Mondor de Recherche Biomédicale, Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France; Université Paris-Est Créteil, Faculté de Médecine, Créteil, France
| | - Agnès Sliwinski
- Département d'Études Cognitives, École Normale Supérieure-PSL, Paris, France; Institut Mondor de Recherche Biomédicale, Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France; Université Paris-Est Créteil, Faculté de Médecine, Créteil, France; AP-HP, Centre de Référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, Créteil, France
| | - Anne-Catherine Bachoud-Lévi
- Département d'Études Cognitives, École Normale Supérieure-PSL, Paris, France; Institut Mondor de Recherche Biomédicale, Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France; Université Paris-Est Créteil, Faculté de Médecine, Créteil, France; AP-HP, Centre de Référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, Créteil, France
| | - Charlotte Jacquemot
- Département d'Études Cognitives, École Normale Supérieure-PSL, Paris, France; Institut Mondor de Recherche Biomédicale, Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France; Université Paris-Est Créteil, Faculté de Médecine, Créteil, France.
| |
Collapse
|
35
|
Hu B, Younes L, Bu X, Liu CF, Ratnanather JT, Paulsen J, Georgiou-Karistianis N, Miller MI, Ross C, Faria AV. Mixed longitudinal and cross-sectional analyses of deep gray matter and white matter using diffusion weighted images in premanifest and manifest Huntington's disease. Neuroimage Clin 2023; 39:103493. [PMID: 37582307 PMCID: PMC10448214 DOI: 10.1016/j.nicl.2023.103493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/29/2023] [Accepted: 08/07/2023] [Indexed: 08/17/2023]
Abstract
Changes in the brain of patients with Huntington's disease (HD) begin years before clinical onset, so it remains critical to identify biomarkers to track these early changes. Metrics derived from tensor modeling of diffusion-weighted MRIs (DTI), that indicate the microscopic brain structure, can add important information to regional volumetric measurements. This study uses two large-scale longitudinal, multicenter datasets, PREDICT-HD and IMAGE-HD, to trace changes in DTI of HD participants with a broad range of CAP scores (a product of CAG repeat expansion and age), including those with pre-manifest disease (i.e., prior to clinical onset). Utilizing a fully automated data-driven approach to study the whole brain divided in regions of interest, we traced changes in DTI metrics (diffusivity and fractional anisotropy) versus CAP scores, using sigmoidal and linear regression models. We identified points of inflection in the sigmoidal regression using change-point analysis. The deep gray matter showed more evident and earlier changes in DTI metrics over CAP scores, compared to the deep white matter. In the deep white matter, these changes were more evident and occurred earlier in superior and posterior areas, compared to anterior and inferior areas. The curves of mean diffusivity vs. age of HD participants within a fixed CAP score were different from those of controls, indicating that the disease has an additional effect to age on the microscopic brain structure. These results show the regional and temporal vulnerability of the white matter and deep gray matter in HD, with potential implications for experimental therapeutics.
Collapse
Affiliation(s)
- Beini Hu
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Laurent Younes
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA
| | - Xuan Bu
- Department of Radiology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Chin-Fu Liu
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - J Tilak Ratnanather
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jane Paulsen
- Department of Psychiatry, Neurology, Psychological Brain Sciences, University of Iowa, USA; Department Neurology, University of Wisconsin-Madison, USA
| | - Nellie Georgiou-Karistianis
- School of Psychological Sciences and Turner Institute of Brain and Mental Health, Monash University, Australia
| | - Michael I Miller
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Christopher Ross
- Department of Psychiatry, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Andreia V Faria
- Department of Radiology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
36
|
Ouwerkerk J, Feleus S, van der Zwaan KF, Li Y, Roos M, van Roon-Mom WMC, de Bot ST, Wolstencroft KJ, Mina E. Machine learning in Huntington's disease: exploring the Enroll-HD dataset for prognosis and driving capability prediction. Orphanet J Rare Dis 2023; 18:218. [PMID: 37501188 PMCID: PMC10375780 DOI: 10.1186/s13023-023-02785-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/18/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND In biomedicine, machine learning (ML) has proven beneficial for the prognosis and diagnosis of different diseases, including cancer and neurodegenerative disorders. For rare diseases, however, the requirement for large datasets often prevents this approach. Huntington's disease (HD) is a rare neurodegenerative disorder caused by a CAG repeat expansion in the coding region of the huntingtin gene. The world's largest observational study for HD, Enroll-HD, describes over 21,000 participants. As such, Enroll-HD is amenable to ML methods. In this study, we pre-processed and imputed Enroll-HD with ML methods to maximise the inclusion of participants and variables. With this dataset we developed models to improve the prediction of the age at onset (AAO) and compared it to the well-established Langbehn formula. In addition, we used recurrent neural networks (RNNs) to demonstrate the utility of ML methods for longitudinal datasets, assessing driving capabilities by learning from previous participant assessments. RESULTS Simple pre-processing imputed around 42% of missing values in Enroll-HD. Also, 167 variables were retained as a result of imputing with ML. We found that multiple ML models were able to outperform the Langbehn formula. The best ML model (light gradient boosting machine) improved the prognosis of AAO compared to the Langbehn formula by 9.2%, based on root mean squared error in the test set. In addition, our ML model provides more accurate prognosis for a wider CAG repeat range compared to the Langbehn formula. Driving capability was predicted with an accuracy of 85.2%. The resulting pre-processing workflow and code to train the ML models are available to be used for related HD predictions at: https://github.com/JasperO98/hdml/tree/main . CONCLUSIONS Our pre-processing workflow made it possible to resolve the missing values and include most participants and variables in Enroll-HD. We show the added value of a ML approach, which improved AAO predictions and allowed for the development of an advisory model that can assist clinicians and participants in estimating future driving capability.
Collapse
Affiliation(s)
- Jasper Ouwerkerk
- Department of Pathology and Clinical Bioinformatics, Erasmus Medical Center (EMC), Wytemaweg, 3015 CN, Rotterdam, The Netherlands
| | - Stephanie Feleus
- Department of Neurology, Leiden University Medical Center (LUMC), PO Box 9600, 2300 RC, Leiden, The Netherlands
- Department of Clinical Epidemiology, Leiden University Medical Center (LUMC), PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Kasper F van der Zwaan
- Department of Neurology, Leiden University Medical Center (LUMC), PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Yunlei Li
- Department of Pathology and Clinical Bioinformatics, Erasmus Medical Center (EMC), Wytemaweg, 3015 CN, Rotterdam, The Netherlands
| | - Marco Roos
- Department of Human Genetics, Leiden University Medical Center (LUMC), PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Willeke M C van Roon-Mom
- Department of Human Genetics, Leiden University Medical Center (LUMC), PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Susanne T de Bot
- Department of Neurology, Leiden University Medical Center (LUMC), PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Katherine J Wolstencroft
- Leiden Institute of Advanced Computer Science (LIACS), Leiden University, Niels Bohrweg 1, 2333 CA, Leiden, The Netherlands
| | - Eleni Mina
- Department of Human Genetics, Leiden University Medical Center (LUMC), PO Box 9600, 2300 RC, Leiden, The Netherlands.
| |
Collapse
|
37
|
Bao YF, Li XY, Dong Y, Wu ZY. Loss of CAA interruption and intergenerational CAG instability in Chinese patients with Huntington's disease. J Mol Med (Berl) 2023; 101:869-876. [PMID: 37231148 DOI: 10.1007/s00109-023-02329-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 04/06/2023] [Accepted: 04/28/2023] [Indexed: 05/27/2023]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by CAG expansions in huntingtin (HTT) gene, involving motor, cognitive, and neuropsychiatric symptoms. However, genetic modifiers and CAG repeat instability may lead to variations of clinical manifestations, making diagnosis of HD difficult. In this study, we recruited 229 HD individuals from 164 families carrying expanded CAG repeats of HTT, and analyzed loss of CAA interruption (LOI) on the expanded allele and CAG instability during germline transmission. Sanger sequencing and TA cloning were used to determine CAG repeat length and identify LOI variants. Detailed clinical features and genetic testing results were collected. We identified 6 individuals with LOI variants from 3 families, and all probands presented with earlier motor onset age than predicted onset age. In addition, we also presented 2 families with extreme CAG instability during germline transmission. One family showed an expansion from 35 to 66 CAG repeats, while the other family showed both CAG expansion and contraction in lineal three generations. In conclusion, we present the first document of Asian HD population with LOI variant, and we suggest that for symptomatic individuals with intermediate or reduced penetrance allele or negative family history, HTT gene sequencing should be considered in the clinical practice. KEY MESSAGES : We screened the loss of CAA interruption (LOI) variant in a Chinese HD cohort and presented the first document of Asian patients with Huntington's disease carrying LOI variant. We identified 6 individuals with LOI variants from 3 families, and all probands presented with earlier motor onset age than predicted onset age. We presented 2 families with extreme CAG instability during germline transmission. One family showed an expansion from 35 to 66 CAG repeats, while the other family showed both CAG expansion and contraction in lineal three generations. We suggest that for symptomatic individuals with intermediate or reduced penetrance allele or negative family history, HTT gene sequencing should be considered in the clinical practice.
Collapse
Affiliation(s)
- Yu-Feng Bao
- Department of Medical Genetics and Center for Rare Diseases, Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China
- Department of Neurology and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China
| | - Xiao-Yan Li
- Department of Medical Genetics and Center for Rare Diseases, Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China
- Department of Neurology and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China
| | - Yi Dong
- Department of Medical Genetics and Center for Rare Diseases, Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China
- Department of Neurology and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China
| | - Zhi-Ying Wu
- Department of Medical Genetics and Center for Rare Diseases, Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China.
- Department of Neurology and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China.
| |
Collapse
|
38
|
Hatano Y, Ishihara T, Hirokawa S, Onodera O. Machine Learning Approach for the Prediction of Age-Specific Probability of SCA3 and DRPLA by Survival Curve Analysis. Neurol Genet 2023; 9:e200075. [PMID: 37152445 PMCID: PMC10159758 DOI: 10.1212/nxg.0000000000200075] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 03/23/2023] [Indexed: 05/09/2023]
Abstract
Background and Objectives As the number of repeats in the expansion increases, polyglutamine diseases tend to show at a younger age. From this relationship, attempts have been made to predict age at onset by parametric survival analysis. However, a method for a more accurate prediction has been desirable. In this study, we examined 2 methods for survival analysis using machine learning and 6 conventional methods for parametric survival analysis of spinocerebellar ataxia (SCA)3 and dentatorubral-pallidoluysian atrophy (DRPLA). Methods We compared the performance of 2 machine learning methods of survival analysis (random survival forest [RSF] and DeepSurv) and 6 methods of parametric survival analysis (Weibull, exponential, Gaussian, logistic, loglogistic, and log Gaussian). Training and evaluation were performed using the leave-one-out cross-validation method, and evaluation criteria included root mean squared error (RMSE), mean absolute error (MAE), and the integrated Brier score. The latter was used as the primary end point, and the survival analysis model yielding the best result was used to predict the asymptomatic probability. Results Among the models examined, the RSF and DeepSurv machine learning methods had a higher prediction accuracy than the parametric methods of survival analysis. For both SCA3 and DRPLA, RSF had a higher accuracy than DeepSurv for the assessment of RMSE (SCA3: 7.37, DRPLA: 10.78), MAE (SCA3: 5.52, DRPLA: 8.17), and the integrated Brier score (SCA3: 0.05, DRPLA: 0.077). Using RSF, we determined the age-specific probability distribution of age at onset based on CAG repeat size and current age. Discussion In this study, we have demonstrated the superiority of machine learning methods for predicting age at onset of SCA3 and DRPLA using survival analysis. Such accurate prediction of onset will be useful for genetic counseling of carriers and for devising methods to verify the effects of interventions for unaffected individuals.
Collapse
Affiliation(s)
- Yuya Hatano
- Department of Neurology, Brain Research Institute, Niigata University, Niigata-shi, Japan
| | - Tomohiko Ishihara
- Department of Neurology, Brain Research Institute, Niigata University, Niigata-shi, Japan
| | - Sachiko Hirokawa
- Department of Neurology, Brain Research Institute, Niigata University, Niigata-shi, Japan
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, Niigata-shi, Japan
| |
Collapse
|
39
|
Zhu Y, Li M, Bai J, Wang H, Huang X. Hypertension, antihypertensive drugs, and age at onset of Huntington's disease. Orphanet J Rare Dis 2023; 18:125. [PMID: 37226269 DOI: 10.1186/s13023-023-02734-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 05/14/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Associations between blood pressure (BP) with age at onset of Huntington's disease (HD) have reported inconsistent findings. We used Mendelian randomization (MR) to assess effects of BP and lowering systolic BP (SBP) via the genes encoding targets of antihypertensive drugs on age at onset of HD. METHODS Genetic variants from genome-wide association studies(GWAS) of BP traits and BP-lowering variants in genes encoding antihypertensive drugs targets were extracted. Summary statistics for age at onset of HD were retrieved from the GWAS meta-analysis of HD residual age at onset from the GEM-HD Consortium included 9064 HD patients of European ancestry (4417 males and 4,647 females). MR estimates were calculated using the inverse variance weighted method, supplemented by MR-Egger, weighted median, and MR-PRESSO methods. RESULTS Genetically predicted SBP or diastolic BP increase was associated with a later age at onset of HD. However, after SBP/DBP was present as a covariate using multivariable MR method, no significant causal association was suggested. A 10-mm Hg reduction in SBP through variants in genes encoding targets of calcium channel blockers (CCB) was associated with an earlier age at onset of HD (β=-0.220 years, 95% CI =-0.337 to -0.102, P = 2.42 × 10- 4). We did not find a causal association between angiotensin converting enzyme inhibitors and β-blockers with the earlier HD onset. No heterogeneity and horizontal pleiotropy were identified. CONCLUSIONS This MR analysis provided evidence that genetically determined SBP lowering through antihypertensive drugs might be associated with an earlier age at onset of HD. The results may have a potential impact on management of hypertension in the pre-motor-manifest HD population.
Collapse
Affiliation(s)
- Yahui Zhu
- Medical School of Chinese PLA, Beijing, China
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Mao Li
- Medical School of Chinese PLA, Beijing, China
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Jiongming Bai
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
- College of Medicine, Nankai University, Tianjin, China
| | - Haoran Wang
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
- College of Medicine, Nankai University, Tianjin, China
| | - Xusheng Huang
- Medical School of Chinese PLA, Beijing, China.
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| |
Collapse
|
40
|
Davis MC, Fitzgerald PB, Bailey NW, Sullivan C, Stout JC, Hill AT, Hoy KE. Effects of medial prefrontal transcranial alternating current stimulation on neural activity and connectivity in people with Huntington's disease and neurotypical controls. Brain Res 2023; 1811:148379. [PMID: 37121424 DOI: 10.1016/j.brainres.2023.148379] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 03/30/2023] [Accepted: 04/25/2023] [Indexed: 05/02/2023]
Abstract
We investigated the effects of transcranial alternating current stimulation (tACS) targeted to the medial prefrontal cortex (mPFC) on resting electroencephalographic (EEG) indices of oscillatory power, aperiodic exponent and offset, and functional connectivity in 22 late premanifest and early manifest stage individuals with HD and 20 neurotypical controls. Participants underwent three 20-minute sessions of tACS at least 72 hours apart; one session at alpha frequency (either each participant's Individualised Alpha Frequency (IAF), or 10Hz when an IAF was not detected); one session at delta frequency (2Hz); and a session of sham tACS. Session order was randomised and counterbalanced across participants. EEG recordings revealed a reduction of the spectral exponent ('flattening' of the 1/f slope) of the eyes-open aperiodic signal in participants with HD following alpha-tACS, suggestive of an enhancement in excitatory tone. Contrary to expectation, there were no changes in oscillatory power or functional connectivity in response to any of the tACS conditions in the participants with HD. By contrast, alpha-tACS increased delta power in neurotypical controls, who further demonstrated significant increases in theta power and theta functional connectivity in response to delta-tACS. This study contributes to the rapidly growing literature on the potential experimental and therapeutic applications of tACS by examining neurophysiological outcome measures in people with HD as well as neurotypical controls.
Collapse
Affiliation(s)
- Marie-Claire Davis
- Central Clinical School, Department of Psychiatry, Monash University, Victoria, Australia; Statewide Progressive Neurological Disease Service, Calvary Health Care Bethlehem, Victoria Australia.
| | - Paul B Fitzgerald
- School of Medicine and Psychology, Australian National University, Canberra, ACT, Australia
| | - Neil W Bailey
- Central Clinical School, Department of Psychiatry, Monash University, Victoria, Australia; School of Medicine and Psychology, Australian National University, Canberra, ACT, Australia; Monarch Research Institute Monarch Mental Health Group, Sydney, NSW, Australia
| | - Caley Sullivan
- Central Clinical School, Department of Psychiatry, Monash University, Victoria, Australia
| | - Julie C Stout
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Melbourne, Victoria, Australia
| | - Aron T Hill
- Central Clinical School, Department of Psychiatry, Monash University, Victoria, Australia; Cognitive Neuroscience Unit, School of Psychology, Deakin University, Melbourne, Australia
| | - Kate E Hoy
- Central Clinical School, Department of Psychiatry, Monash University, Victoria, Australia; The Bionics Institute of Australia, 384-388 Albert St, East Melbourne, VIC, 3002, Australia
| |
Collapse
|
41
|
Davis MC, Hill AT, Fitzgerald PB, Bailey NW, Sullivan C, Stout JC, Hoy KE. Medial prefrontal transcranial alternating current stimulation for apathy in Huntington's disease. Prog Neuropsychopharmacol Biol Psychiatry 2023; 126:110776. [PMID: 37120005 DOI: 10.1016/j.pnpbp.2023.110776] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/01/2023]
Abstract
We investigated the effects of transcranial alternating current stimulation (tACS) targeted to the bilateral medial prefrontal cortex (mPFC) and administered at either delta or alpha frequencies, on brain activity and apathy in people with Huntington's disease (HD) (n = 17). Given the novelty of the protocol, neurotypical controls (n = 20) were also recruited. All participants underwent three 20-min sessions of tACS; one session at alpha frequency (Individualised Alpha Frequency (IAF), or 10 Hz when an IAF was not detected); one session at delta frequency (2 Hz); and a session of sham tACS. Participants completed the Monetary Incentive Delay (MID) task with simultaneous recording of EEG immediately before and after each tACS condition. The MID task presents participants with cues signalling potential monetary gains or losses that increase activity in key regions of the cortico-basal ganglia-thalamocortical networks, with dysfunction of the latter network being implicated in the pathophysiology of apathy. We used the P300 and Contingent Negative Variation (CNV) event-related potentials elicited during the MID task as markers of mPFC engagement. HD participants' CNV amplitude significantly increased in response to alpha-tACS, but not delta-tACS or sham. Neurotypical controls' P300 and CNV were not modulated by any of the tACS conditions, but they did demonstrate a significant decrease in post-target response times following alpha-tACS. We present this as preliminary evidence of the ability of alpha-tACS to modulate brain activity associated with apathy in HD.
Collapse
Affiliation(s)
- Marie-Claire Davis
- Central Clinical School, Department of Psychiatry, Monash University, Victoria, Australia; Statewide Progressive Neurological Disease Service, Calvary Health Care Bethlehem, Victoria, Australia.
| | - Aron T Hill
- Central Clinical School, Department of Psychiatry, Monash University, Victoria, Australia; Cognitive Neuroscience Unit, School of Psychology, Deakin University, Melbourne, Australia
| | - Paul B Fitzgerald
- Central Clinical School, Department of Psychiatry, Monash University, Victoria, Australia; School of Medicine and Psychology, Australian National University, Canberra, ACT, Australia
| | - Neil W Bailey
- Central Clinical School, Department of Psychiatry, Monash University, Victoria, Australia; School of Medicine and Psychology, Australian National University, Canberra, ACT, Australia; Monarch Research Institute Monarch Mental Health Group, Sydney, NSW, Australia
| | - Caley Sullivan
- Central Clinical School, Department of Psychiatry, Monash University, Victoria, Australia
| | - Julie C Stout
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Melbourne, Victoria, Australia
| | - Kate E Hoy
- Central Clinical School, Department of Psychiatry, Monash University, Victoria, Australia; The Bionics Institute of Australia, 384-388 Albert St, East Melbourne, VIC 3002, Australia
| |
Collapse
|
42
|
El Haj M, Caillaud M, Moustafa A, Prundean A, Scherer C, Verny C, Allain P. "Ten euros now" temporal discounting in Huntington disease. Neurol Sci 2023:10.1007/s10072-023-06775-z. [PMID: 36964316 DOI: 10.1007/s10072-023-06775-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/21/2023] [Indexed: 03/26/2023]
Abstract
BACKGROUND When making decisions, one often faces a trade-off between immediate and long-term rewards. In these situations, people may prefer immediate over later rewards, even if immediate rewards are smaller than later ones; a phenomenon known as temporal discounting. In this study, we, for the first time, assessed temporal discounting in three populations: participants with manifest Huntington disease (HD), participants with premanifest HD, and control participants. METHODS Using the temporal discounting task, we invited participants to choose between small immediate amount of money vs. delayed, but larger amount of money (e.g., "Which do you prefer: you get 10 euros right now or 50 euros in a month?"). We also measured inhibition in order to test if it impacts discounting performance. RESULTS Analysis demonstrated higher temporal discounting (i.e., a preference for the immediate rewards) in participants with manifest HD compared to those with premanifest HD or control participants, but no significant differences were observed in participants with premanifest HD and control participants. Analysis also demonstrated significant correlations between temporal discounting and scores on an inhibition test in participants with manifest HD, but not in those with premanifest HD or in control participants. DISCUSSION We suggest that, when making decisions, patients with manifest HD may have difficulties with suppressing the temptation of smaller, but immediate, rewards.
Collapse
Affiliation(s)
- Mohamad El Haj
- Nantes Université, Univ Angers, Laboratoire de Psychologie des Pays de la Loire (LPPL - EA 4638), F-44000, Nantes, France.
- CHU Nantes, Clinical Gerontology Department, 41 rue Pierre et Marie Curie, 44093, Nantes, France.
- Institut Universitaire de France, Paris, France.
- Faculté de Psychologie, LPPL - Laboratoire de Psychologie des Pays de la Loire, Université Nantes, Chemin de la Censive du Tertre, BP 81227, 44312, Nantes Cedex 3, France.
| | - Marie Caillaud
- University of Texas, Clinical Neuroscience Lab, 108 East Dean Keeton St., Austin, TX, 78712, USA
| | - Ahmed Moustafa
- School of Psychology, Faculty of Society and Design, Bond University, Gold Coast, Queensland, Australia
- Department of Human Anatomy and Physiology, the Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | | | | | | | - Philippe Allain
- Département de Neurologie, CHU Angers, Angers, France
- Laboratoire de Psychologie des Pays de la Loire, LPPL EA 4638 SFR Confluences, UNIV Angers, Nantes Université, Maison de la recherche Germaine Tillion, 5 bis Boulevard Lavoisier, 49045, Angers Cedex 01, France
| |
Collapse
|
43
|
Nguyen NH, Shin SJ, Dodd-Eaton EB, Ning J, Wang W. Personalized Risk Prediction for Cancer Survivors: A Bayesian Semi-parametric Recurrent Event Model with Competing Outcomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.28.530537. [PMID: 36909464 PMCID: PMC10002693 DOI: 10.1101/2023.02.28.530537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Multiple primary cancers are increasingly more frequent due to improved survival of cancer patients. Characteristics of the first primary cancer largely impact the risk of developing subsequent primary cancers. Hence, model-based risk characterization of cancer survivors that captures patient-specific variables is needed for healthcare policy making. We propose a Bayesian semi-parametric framework, where the occurrence processes of the competing cancer types follow independent non-homogeneous Poisson processes and adjust for covariates including the type and age at diagnosis of the first primary. Applying this framework to a historically collected cohort with families presenting a highly enriched history of multiple primary tumors and diverse cancer types, we have derived a suite of age-to-onset penetrance curves for cancer survivors. This includes penetrance estimates for second primary lung cancer, potentially impactful to ongoing cancer screening decisions. Using Receiver Operating Characteristic (ROC) curves, we have validated the good predictive performance of our models in predicting second primary lung cancer, sarcoma, breast cancer, and all other cancers combined, with areas under the curves (AUCs) at 0.89, 0.91, 0.76 and 0.68, respectively. In conclusion, our framework provides covariate-adjusted quantitative risk assessment for cancer survivors, hence moving a step closer to personalized health management for this unique population.
Collapse
Affiliation(s)
- Nam H Nguyen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Statistics, Rice University, Houston, TX
| | - Seung Jun Shin
- Department of Statistics, Korea University, Seoul, Korea
| | - Elissa B Dodd-Eaton
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jing Ning
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Wenyi Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
44
|
PerezGrovas-Saltijeral A, Ochoa-Morales A, Jara-Prado A, Velázquez-Cruz R, Rivera-Paredez B, Dávila-OrtizdeMontellano D, Benítez-Alonso EO, Santamaría-Olmedo M, Sevilla-Montoya R, Marfil-Marín E, Valdés-Flores M, Martínez-Ruano L, Camacho-Molina A, Hidalgo-Bravo A. Unraveling the role of relative telomere length and CAG expansion on initial symptoms of juvenile Huntington disease. Eur J Neurol 2023; 30:612-621. [PMID: 36421025 DOI: 10.1111/ene.15644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND PURPOSE Juvenile-onset Huntington disease (JHD) is defined when symptoms initiate before 20 years of age. Mechanisms explaining differences between juvenile and adult onset are not fully understood. Our aim was to analyze the distribution of initial symptoms in a cohort of JHD patients and to explore its relationship with CAG expansion and relative telomere length (RTL). METHODS A total of 84 JHD patients and 54 neurologically healthy age and sex matched individuals were recruited. CAG length was measured by southern blot or triplet repeat primed polymerase chain reaction. RTL was measured using the Cawthon method. RESULTS Psychiatric symptoms were most frequent when considering the entire cohort. When divided into onset before or after 10 years, cognitive symptoms were more frequent in the youngest, whilst in the older group psychiatric symptoms prevailed. Motor symptoms were rare in the youngest and epilepsy was observed only in this group as well as a larger CAG expansion. RTL analysis revealed shorter telomeres in JHD patients compared to controls. This difference is not influenced by age, initial symptoms, time of disease or CAG expansion. CONCLUSIONS To the best of our knowledge this is the largest cohort of JHD patients reported. Psychiatric manifestations deserve special attention when JHD is suspected and epilepsy is especially important in the youngest patients. Initial symptoms seem to be influenced by CAG expansion and therefore age of onset. RTL is significantly reduced in JHD patients which can influence the characteristic neurodegeneration of JHD and contribute to the clinical discrepancy between adult and juvenile forms of Huntington disease.
Collapse
Affiliation(s)
| | - Adriana Ochoa-Morales
- Department of Neurogenetics, National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| | - Aurelio Jara-Prado
- Department of Neurogenetics, National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| | - Rafael Velázquez-Cruz
- Genomics of Bone Metabolism Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City, Mexico
| | - Berenice Rivera-Paredez
- Research Center in Policies, Population and Health, School of Medicine, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | | | - Edmar O Benítez-Alonso
- Department of Neurogenetics, National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| | | | - Rosalba Sevilla-Montoya
- Department of Genetics and Human Genomics, National Institute of Perinatology, Mexico City, Mexico
| | | | | | - Leticia Martínez-Ruano
- Department of Neurogenetics, National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| | - Alejandra Camacho-Molina
- Department of Neurogenetics, National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| | | |
Collapse
|
45
|
Olufunmilayo EO, Gerke-Duncan MB, Holsinger RMD. Oxidative Stress and Antioxidants in Neurodegenerative Disorders. Antioxidants (Basel) 2023; 12:antiox12020517. [PMID: 36830075 PMCID: PMC9952099 DOI: 10.3390/antiox12020517] [Citation(s) in RCA: 85] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Neurodegenerative disorders constitute a substantial proportion of neurological diseases with significant public health importance. The pathophysiology of neurodegenerative diseases is characterized by a complex interplay of various general and disease-specific factors that lead to the end point of neuronal degeneration and loss, and the eventual clinical manifestations. Oxidative stress is the result of an imbalance between pro-oxidant species and antioxidant systems, characterized by an elevation in the levels of reactive oxygen and reactive nitrogen species, and a reduction in the levels of endogenous antioxidants. Recent studies have increasingly highlighted oxidative stress and associated mitochondrial dysfunction to be important players in the pathophysiologic processes involved in neurodegenerative conditions. In this article, we review the current knowledge of the general effects of oxidative stress on the central nervous system, the different specific routes by which oxidative stress influences the pathophysiologic processes involved in Alzheimer's disease, Parkinson's disease, Amyotrophic Lateral Sclerosis and Huntington's disease, and how oxidative stress may be therapeutically reversed/mitigated in order to stall the pathological progression of these neurodegenerative disorders to bring about clinical benefits.
Collapse
Affiliation(s)
- Edward O. Olufunmilayo
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Department of Medicine, University College Hospital, Queen Elizabeth Road, Oritamefa, Ibadan 5116, PMB, Nigeria
| | - Michelle B. Gerke-Duncan
- Education Innovation, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - R. M. Damian Holsinger
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Correspondence:
| |
Collapse
|
46
|
Lemoine L, Lunven M, Fraisse N, Youssov K, Bapst B, Morgado G, Reilmann R, Busse M, Craufurd D, Rosser A, de Gardelle V, Bachoud-Lévi AC. The striatum in time production: The model of Huntington's disease in longitudinal study. Neuropsychologia 2023; 179:108459. [PMID: 36567007 DOI: 10.1016/j.neuropsychologia.2022.108459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 12/16/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
The unified model of time processing suggests that the striatum is a central structure involved in all tasks that require the processing of temporal durations. Patients with Huntington's disease exhibit striatal degeneration and a deficit in time perception in interval timing tasks (i.e. for duration ranging from hundreds of milliseconds to minutes), but whether this deficit extends to time production remains unclear. In this study, we investigated whether symptomatic patients (HD, N = 101) or presymptomatic gene carriers (Pre-HD, N = 31) of Huntington's disease had a deficit in time production for durations between 4 and 10 s compared to healthy controls and whether this deficit developed over a year for patients. We found a clear deficit in temporal production for HD patients, whereas Pre-HD performed similarly to Controls. For HD patients and Pre-HD participants, task performance was correlated with grey matter volume in the amygdala and caudate, bilaterally. These results confirm that the striatum is involved in interval timing not only in perception but also in production, in accordance with the unified model of time processing. Furthermore, exploratory factor analyses on our data indicated that temporal production was associated with clinical assessments of psychomotor and executive functions. Finally, when retested twelve months later, the deficit of HD patients remained stable, although striatal degeneration was more pronounced. Thus, the simple, short and language-independent temporal production task may be a useful clinical tool to detect striatal degeneration in patients in early stages of Huntington's disease. However, its usefulness to detect presymptomatic stages or for monitoring the evolution of HD over a year seems limited.
Collapse
Affiliation(s)
- Laurie Lemoine
- Département d'Etudes Cognitives, Ecole Normale Supérieure, PSL University, Paris, France; Université Paris Est, Faculté de Médecine, Créteil, France; Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France; AP-HP, Centre de référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, Créteil, France
| | - Marine Lunven
- Département d'Etudes Cognitives, Ecole Normale Supérieure, PSL University, Paris, France; Université Paris Est, Faculté de Médecine, Créteil, France; Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France; AP-HP, Centre de référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, Créteil, France
| | - Nicolas Fraisse
- Département d'Etudes Cognitives, Ecole Normale Supérieure, PSL University, Paris, France; Université Paris Est, Faculté de Médecine, Créteil, France; Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France; AP-HP, Centre de référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, Créteil, France
| | - Katia Youssov
- Département d'Etudes Cognitives, Ecole Normale Supérieure, PSL University, Paris, France; Université Paris Est, Faculté de Médecine, Créteil, France; Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France; AP-HP, Centre de référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, Créteil, France
| | - Blanche Bapst
- Université Paris Est, Faculté de Médecine, Créteil, France; Service de Neuroradiologie, Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Graça Morgado
- Inserm, Centre d'Investigation Clinique 1430, Hôpital Henri Mondor, Créteil, France
| | - Ralf Reilmann
- George-Huntington-Institute, Technology-Park, Muenster, Germany; Department of Clinical Radiology University of Muenster, Muenster, Germany; Dept. of Neurodegeneration and Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Monica Busse
- Centre for Trials Research, Cardiff University, United Kingdom; NMHRI, School of Medicine, And Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - David Craufurd
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom; 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
| | - Anne Rosser
- NMHRI, School of Medicine, And Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, United Kingdom; Wales Brain Research and Intracranial Neurotherapeutics (BRAIN) Unit, Wales, United Kingdom
| | | | - Anne-Catherine Bachoud-Lévi
- Département d'Etudes Cognitives, Ecole Normale Supérieure, PSL University, Paris, France; Université Paris Est, Faculté de Médecine, Créteil, France; Inserm U955, Equipe E01 Neuropsychologie Interventionnelle, Créteil, France; AP-HP, Centre de référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, Créteil, France.
| |
Collapse
|
47
|
Xie J, Xie L, Wei H, Li XJ, Lin L. Dynamic Regulation of DNA Methylation and Brain Functions. BIOLOGY 2023; 12:152. [PMID: 36829430 PMCID: PMC9952911 DOI: 10.3390/biology12020152] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/10/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023]
Abstract
DNA cytosine methylation is a principal epigenetic mechanism underlying transcription during development and aging. Growing evidence suggests that DNA methylation plays a critical role in brain function, including neurogenesis, neuronal differentiation, synaptogenesis, learning, and memory. However, the mechanisms underlying aberrant DNA methylation in neurodegenerative diseases remain unclear. In this review, we provide an overview of the contribution of 5-methycytosine (5mC) and 5-hydroxylcytosine (5hmC) to brain development and aging, with a focus on the roles of dynamic 5mC and 5hmC changes in the pathogenesis of neurodegenerative diseases, particularly Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD). Identification of aberrant DNA methylation sites could provide potential candidates for epigenetic-based diagnostic and therapeutic strategies for neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | | | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Li Lin
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| |
Collapse
|
48
|
Schultz JL, Langbehn DR, Al-Kaylani HM, van der Plas E, Koscik TR, Epping EA, Espe-Pfeifer PB, Martin EP, Moser DJ, Magnotta VA, Nopoulos PC. Longitudinal Clinical and Biological Characteristics in Juvenile-Onset Huntington's Disease. Mov Disord 2023; 38:113-122. [PMID: 36318082 PMCID: PMC9851979 DOI: 10.1002/mds.29251] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/26/2022] [Accepted: 09/29/2022] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Juvenile-onset Huntington's disease (JOHD) is a rare form of Huntington's disease (HD) characterized by symptom onset before the age of 21 years. Observational data in this cohort is lacking. OBJECTIVES Quantify measures of disease progression for use in clinical trials of patients with JOHD. METHODS Participants who received a motor diagnosis of HD before the age of 21 were included in the Kids-JOHD study. The comparator group consisted of children and young adults who were at-risk for inheriting the genetic mutation that causes HD, but who were found to have a CAG repeat in the non-expanded range (gene non-expanded [GNE]). RESULTS Data were obtained between March 17, 2006, and February 13, 2020. There were 26 JOHD participants and 78 GNE participants who were comparable on age (16.03 vs. 14.43, respectively) and sex (53.8% female vs. 57.7% female, respectively). The mean annualized decrease in striatal volume in the JOHD group was -3.99% compared to -0.06% in the GNE (mean difference [MD], -3.93%; 95% confidence intervals [CI], [-4.98 to -2.80], FDR < 0.0001). The mean increase in the Unified Huntington's Disease Rating Scale Total Motor Score per year in the JOHD group was 7.29 points compared to a mean decrease of -0.21 point in the GNE (MD, 7.5; 95% CI, [5.71-9.28], FDR < 0·0001). CONCLUSIONS These findings demonstrate that structural brain imaging and clinical measures in JOHD may be potential biomarkers of disease progression for use in clinical trials. Collaborative efforts are required to validate these results in a larger cohort of patients with JOHD. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Jordan L. Schultz
- Carver College of Medicine at the University of Iowa, Department of Psychiatry, 200 Hawkins Drive, Iowa City, IA
- Carver College of Medicine at the University of Iowa, Department of Neurology, 200 Hawkins Drive, Iowa City, IA
- University of Iowa College of Pharmacy, Division of Pharmacy Practice and Sciences, 200 Hawkins Drive, Iowa City, IA
| | - Douglas R. Langbehn
- Carver College of Medicine at the University of Iowa, Department of Psychiatry, 200 Hawkins Drive, Iowa City, IA
| | - Hend M. Al-Kaylani
- Carver College of Medicine at the University of Iowa, Department of Psychiatry, 200 Hawkins Drive, Iowa City, IA
| | - Ellen van der Plas
- Carver College of Medicine at the University of Iowa, Department of Psychiatry, 200 Hawkins Drive, Iowa City, IA
| | - Timothy R. Koscik
- Carver College of Medicine at the University of Iowa, Department of Psychiatry, 200 Hawkins Drive, Iowa City, IA
| | - Eric A. Epping
- Carver College of Medicine at the University of Iowa, Department of Psychiatry, 200 Hawkins Drive, Iowa City, IA
| | - Patricia B. Espe-Pfeifer
- Carver College of Medicine at the University of Iowa, Department of Psychiatry, 200 Hawkins Drive, Iowa City, IA
| | - Erin P. Martin
- Carver College of Medicine at the University of Iowa, Department of Psychiatry, 200 Hawkins Drive, Iowa City, IA
| | - David J. Moser
- Carver College of Medicine at the University of Iowa, Department of Psychiatry, 200 Hawkins Drive, Iowa City, IA
| | - Vincent A. Magnotta
- Carver College of Medicine at the University of Iowa, Department of Psychiatry, 200 Hawkins Drive, Iowa City, IA
- Carver College of Medicine at the University of Iowa, Department of Radiology, 200 Hawkins Drive, Iowa City, IA
| | - Peggy C. Nopoulos
- Carver College of Medicine at the University of Iowa, Department of Psychiatry, 200 Hawkins Drive, Iowa City, IA
- Carver College of Medicine at the University of Iowa, Department of Neurology, 200 Hawkins Drive, Iowa City, IA
- Stead Family Children’s Hospital at the University of Iowa, 200 Hawkins Drive, Iowa City, IA
| |
Collapse
|
49
|
Jeyakumar N, Hilmer SN, Teixeira-Pinto A, Loy CT. Frailty and Associated Environmental Factors Only Have Small Effects on Age of Onset in Huntington's Disease. J Huntingtons Dis 2023; 12:355-361. [PMID: 38007671 DOI: 10.3233/jhd-230572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
BACKGROUND Over one third of age of onset variation in Huntington's disease is unexplained by CAG repeat length. In Alzheimer's disease, frailty partly modulates the relationship between neuropathology and dementia. OBJECTIVE We investigated whether a multi-domain frailty index, reflecting non-genetic factors in Huntington's disease, similarly modulates the relationship between CAG repeat length and age of onset. METHODS We created a frailty index assessing comorbidities, substance abuse, polypharmacy, and education. We applied multiple linear regression models to 2,741 subjects with manifest Huntington's disease from the Enroll-HD cohort study, including 729 subjects with late-onset (post-60 years) disease, using frailty index or constituent item scores and CAG repeat length as independent variables. We used actual and "residual" ages of onset (difference between actual and CAG-based predicted onset) as dependent variables, the latter offsetting the increased time available to accumulate comorbidities in older subjects. RESULTS Higher frailty index scores were associated with significantly lower residual ages of onset in the late-onset subgroup (p = 0.03), though the effect was small (R2 = 0.27 with frailty as a predictor vs. 0.26 without). Number of comorbidities was also associated with significantly lower residual ages of onset in the late-onset subgroup (p = 0.04). Drug abuse and smoking were associated with significantly earlier ages of onset in the whole cohort (p < 0.01, p = 0.02) and late-onset subgroup (p < 0.01, p = 0.03). CONCLUSIONS The impact of non-genetic factors on age of onset, assessed using a frailty index or separately, in Huntington's disease is limited.
Collapse
Affiliation(s)
| | - Sarah N Hilmer
- The Kolling Institute, The University of Sydney, Sydney, Australia
| | | | - Clement T Loy
- Macquarie Medical School, Macquarie University, Sydney, Australia
| |
Collapse
|
50
|
Geijtenbeek KW, Janzen J, Bury AE, Sanz-Sanz A, Hoebe RA, Bondulich MK, Bates GP, Reits EAJ, Schipper-Krom S. Reduction in PA28αβ activation in HD mouse brain correlates to increased mHTT aggregation in cell models. PLoS One 2022; 17:e0278130. [PMID: 36574405 PMCID: PMC9794069 DOI: 10.1371/journal.pone.0278130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 11/09/2022] [Indexed: 12/29/2022] Open
Abstract
Huntington's disease is an autosomal dominant heritable disorder caused by an expanded CAG trinucleotide repeat at the N-terminus of the Huntingtin (HTT) gene. Lowering the levels of soluble mutant HTT protein prior to aggregation through increased degradation by the proteasome would be a therapeutic strategy to prevent or delay the onset of disease. Native PAGE experiments in HdhQ150 mice and R6/2 mice showed that PA28αβ disassembles from the 20S proteasome during disease progression in the affected cortex, striatum and hippocampus but not in cerebellum and brainstem. Modulating PA28αβ activated proteasomes in various in vitro models showed that PA28αβ improved polyQ degradation, but decreased the turnover of mutant HTT. Silencing of PA28αβ in cells lead to an increase in mutant HTT aggregates, suggesting that PA28αβ is critical for overall proteostasis, but only indirectly affects mutant HTT aggregation.
Collapse
Affiliation(s)
| | - Jolien Janzen
- Amsterdam UMC Location University of Amsterdam, Medical Biology, Amsterdam, The Netherlands
| | - Aleksandra E. Bury
- Amsterdam UMC Location University of Amsterdam, Medical Biology, Amsterdam, The Netherlands
| | - Alicia Sanz-Sanz
- Amsterdam UMC Location University of Amsterdam, Medical Biology, Amsterdam, The Netherlands
| | - Ron A. Hoebe
- Amsterdam UMC Location University of Amsterdam, Medical Biology, Amsterdam, The Netherlands
| | - Marie K. Bondulich
- Department of Neurodegenerative Disease, Huntington’s Disease Centre and UK Dementia Research Institute at UCL, Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Gillian P. Bates
- Department of Neurodegenerative Disease, Huntington’s Disease Centre and UK Dementia Research Institute at UCL, Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Eric A. J. Reits
- Amsterdam UMC Location University of Amsterdam, Medical Biology, Amsterdam, The Netherlands
- * E-mail:
| | - Sabine Schipper-Krom
- Amsterdam UMC Location University of Amsterdam, Medical Biology, Amsterdam, The Netherlands
| |
Collapse
|