1
|
Voysey ZJ, Goodman AOG, Rogers L, Holbrook JA, Lazar AS, Barker RA. Sleep abnormalities are associated with greater cognitive deficits and disease activity in Huntington's disease: a 12-year polysomnographic study. Brain Commun 2025; 7:fcaf126. [PMID: 40226381 PMCID: PMC11992570 DOI: 10.1093/braincomms/fcaf126] [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: 07/16/2024] [Revised: 02/10/2025] [Accepted: 03/28/2025] [Indexed: 04/15/2025] Open
Abstract
Increasing evidence suggests that the sleep pathology associated with neurodegenerative diseases can in turn exacerbate both the cognitive deficits and underlying pathobiology of these conditions. Treating sleep may therefore bear significant, even disease-modifying, potential for these conditions, but how best and when to do so remains undetermined. Huntington's disease, by virtue of being an autosomal dominant neurodegenerative disease presenting in mid-life, presents a key 'model' condition through which to advance this field. To date, however, there has been no clinical longitudinal study of sleep abnormalities in Huntington's disease and no robust interrogation of their association with disease onset, cognitive deficits and markers of disease activity. Here, we present the first such study. Huntington's disease gene carriers (n = 28) and age- and sex-matched controls (n = 21) were studied at baseline and 10- and 12-year follow-up. All Huntington's disease gene carriers were premanifest at baseline and were stratified at follow-up into 'prodromal/manifest' versus 'premanifest' groups. Objective sleep abnormalities were assessed through two-night inpatient polysomnography and 2-week domiciliary actigraphy, and their association was explored against Montreal Cognitive Assessment, Trail A/B task, Symbol Digit Modalities Task (SDMT), Hopkins Verbal Learning Task (HVLT) and Montgomery-Asberg Depression Rating Scale (MADRS) scores, plus serum neurofilament light levels. Statistical analysis incorporated cross-sectional ANOVA, longitudinal repeated measures linear models and regressions adjusted for multiple confounders including disease stage. Fifteen Huntington's disease gene carriers phenoconverted to prodromal/early manifest Huntington's disease by study completion. At follow-up, these gene carriers showed more frequent sleep stage changes (P ≤ 0.001, ηp 2 = 0.62) and higher levels of sleep maintenance insomnia (defined by wake after sleep onset, P = 0.002, ηp 2 = 0.52). The latter finding was corroborated by nocturnal motor activity patterns on follow-up actigraphy (P = 0.004, ηp 2 = 0.32). Greater sleep maintenance insomnia was associated with greater cognitive deficits (Trail A P ≤ 0.001, R 2 = 0.78; SDMT P = 0.008, R 2 = 0.63; Trail B P = 0.013, R 2 = 0.60) and higher levels of neurofilament light (P = 0.015, R 2 = 0.39). Longitudinal modelling suggested that sleep stage instability accrues from the early premanifest phase, whereas sleep maintenance insomnia emerges closer to phenoconversion. Baseline sleep stage instability was able to discriminate those who phenoconverted within the study period from those who remained premanifest (area under curve = 0.81, P = 0.024). These results demonstrate that the key sleep abnormalities of premanifest/early Huntington's disease are sleep stage instability and sleep maintenance insomnia and suggest that the former bears value in predicting disease onset, while the latter is associated with greater disease activity and cognitive deficits. Intervention studies to interrogate causation within this association could not only benefit patients with Huntington's disease but also help provide fundamental proof-of-concept findings for the wider sleep-neurodegeneration field.
Collapse
Affiliation(s)
- Zanna J Voysey
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge CB2 0PY, UK
| | - Anna O G Goodman
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge CB2 0PY, UK
| | - Lorraine Rogers
- Royal Papworth Hospital Foundation Trust, Sleep Centre, Cambridge CB2 0AY, UK
| | - Jonathan A Holbrook
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge CB2 0PY, UK
| | - Alpar S Lazar
- Faculty of Medicine and Health Sciences, University of East Anglia, Norwich NR4 7TQ, UK
| | - Roger A Barker
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge CB2 0PY, UK
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| |
Collapse
|
2
|
Scahill RI, Farag M, Murphy MJ, Hobbs NZ, Leocadi M, Langley C, Knights H, Ciosi M, Fayer K, Nakajima M, Thackeray O, Gobom J, Rönnholm J, Weiner S, Hassan YR, Ponraj NKP, Estevez-Fraga C, Parker CS, Malone IB, Hyare H, Long JD, Heslegrave A, Sampaio C, Zhang H, Robbins TW, Zetterberg H, Wild EJ, Rees G, Rowe JB, Sahakian BJ, Monckton DG, Langbehn DR, Tabrizi SJ. Somatic CAG repeat expansion in blood associates with biomarkers of neurodegeneration in Huntington's disease decades before clinical motor diagnosis. Nat Med 2025; 31:807-818. [PMID: 39825149 PMCID: PMC11922752 DOI: 10.1038/s41591-024-03424-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 11/15/2024] [Indexed: 01/20/2025]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease with the age at which characteristic symptoms manifest strongly influenced by inherited HTT CAG length. Somatic CAG expansion occurs throughout life and understanding the impact of somatic expansion on neurodegeneration is key to developing therapeutic targets. In 57 HD gene expanded (HDGE) individuals, ~23 years before their predicted clinical motor diagnosis, no significant decline in clinical, cognitive or neuropsychiatric function was observed over 4.5 years compared with 46 controls (false discovery rate (FDR) > 0.3). However, cerebrospinal fluid (CSF) markers showed very early signs of neurodegeneration in HDGE with elevated neurofilament light (NfL) protein, an indicator of neuroaxonal damage (FDR = 3.2 × 10-12), and reduced proenkephalin (PENK), a surrogate marker for the state of striatal medium spiny neurons (FDR = 2.6 × 10-3), accompanied by brain atrophy, predominantly in the caudate (FDR = 5.5 × 10-10) and putamen (FDR = 1.2 × 10-9). Longitudinal increase in somatic CAG repeat expansion ratio (SER) in blood was a significant predictor of subsequent caudate (FDR = 0.072) and putamen (FDR = 0.148) atrophy. Atypical loss of interruption HTT repeat structures, known to predict earlier age at clinical motor diagnosis, was associated with substantially faster caudate and putamen atrophy. We provide evidence in living humans that the influence of CAG length on HD neuropathology is mediated by somatic CAG repeat expansion. These critical mechanistic insights into the earliest neurodegenerative changes will inform the design of preventative clinical trials aimed at modulating somatic expansion. ClinicalTrials.gov registration: NCT06391619 .
Collapse
Affiliation(s)
- Rachael I Scahill
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Mena Farag
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Michael J Murphy
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Nicola Z Hobbs
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Michela Leocadi
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | | | - Harry Knights
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Marc Ciosi
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Kate Fayer
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Mitsuko Nakajima
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Olivia Thackeray
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - John Rönnholm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
| | - Sophia Weiner
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
| | - Yara R Hassan
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Nehaa K P Ponraj
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Carlos Estevez-Fraga
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Christopher S Parker
- Department of Computer Science and Centre for Medical Image Computing, University College London, London, UK
| | - Ian B Malone
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Harpreet Hyare
- Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Jeffrey D Long
- Department of Psychiatry and Biostatistics, Carver College of Medicine and College of Public Health, University of Iowa, Iowa City, Iowa, USA
| | - Amanda Heslegrave
- Dementia Research Institute, University College London, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Cristina Sampaio
- Faculdade Medicina da Universidade de Lisboa (FMUL), Lisbon, Portugal
- CHDI Management, Inc. Advisors to CHDI Foundation, Princeton, NJ, USA
| | - Hui Zhang
- Department of Computer Science and Centre for Medical Image Computing, University College London, London, UK
| | - Trevor W Robbins
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- Hong Kong Center for Neurodegeneassociated with substantially fasterrative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Edward J Wild
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Geraint Rees
- UCL Institute of Cognitive Neuroscience, University College London, London, UK
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | | | - Darren G Monckton
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Douglas R Langbehn
- Department of Psychiatry and Biostatistics, Carver College of Medicine and College of Public Health, University of Iowa, Iowa City, Iowa, USA
| | - Sarah J Tabrizi
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK.
| |
Collapse
|
3
|
Farag M, Knights H, Scahill RI, McColgan P, Estevez-Fraga C. Neuroimaging Techniques in Huntington's Disease: A Critical Review. Mov Disord Clin Pract 2025. [PMID: 39976324 DOI: 10.1002/mdc3.70010] [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: 08/20/2024] [Revised: 01/24/2025] [Accepted: 02/03/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Huntington's disease (HD) is a hereditary neurodegenerative disorder characterized by cognitive, neuropsychiatric and motor symptoms caused by a CAG trinucleotide repeat expansion in the huntingtin gene. Imaging techniques are crucial for understanding HD pathophysiology and monitoring disease progression. OBJECTIVES This review is targeted at general neurologists and movement disorders specialists with an interest in HD and aims to bring complex imaging, including new experimental techniques, closer to the practicing clinician. METHODS We provide a summary of findings from conventional structural, diffusion and functional imaging in HD studies, together with an update on emerging novel techniques, including multiparametric mapping, multi-shell diffusion techniques, ultra-high field 7-Tesla MRI, positron emission tomography and magnetoencephalography. RESULTS Conventional imaging techniques have deepened our understanding of neuropathological progression in HD, from striatal atrophy to widespread cortical and white matter changes. The integration of novel imaging techniques reviewed has further improved our ability to interrogate, quantify and visualize disease-specific alterations with high precision. CONCLUSIONS Novel imaging techniques have promising roles to further our understanding of HD pathology and as imaging markers for clinical trials, disease staging and therapeutic monitoring. Additionally, the synergistic potential of combining imaging modalities with molecular and genetic data, along with wet biomarkers and clinical data, will help provide a complete and comprehensive view of HD pathology and progression.
Collapse
Affiliation(s)
- Mena Farag
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Harry Knights
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Rachael I Scahill
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Peter McColgan
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- Roche Products Limited, Welwyn Garden City, United Kingdom
| | - Carlos Estevez-Fraga
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- Roche Products Limited, Welwyn Garden City, United Kingdom
| |
Collapse
|
4
|
Handsaker RE, Kashin S, Reed NM, Tan S, Lee WS, McDonald TM, Morris K, Kamitaki N, Mullally CD, Morakabati NR, Goldman M, Lind G, Kohli R, Lawton E, Hogan M, Ichihara K, Berretta S, McCarroll SA. Long somatic DNA-repeat expansion drives neurodegeneration in Huntington's disease. Cell 2025; 188:623-639.e19. [PMID: 39824182 DOI: 10.1016/j.cell.2024.11.038] [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: 06/21/2024] [Revised: 09/15/2024] [Accepted: 11/29/2024] [Indexed: 01/20/2025]
Abstract
In Huntington's disease (HD), striatal projection neurons (SPNs) degenerate during midlife; the core biological question involves how the disease-causing DNA repeat (CAG)n in the huntingtin (HTT) gene leads to neurodegeneration after decades of biological latency. We developed a single-cell method for measuring this repeat's length alongside genome-wide RNA expression. We found that the HTT CAG repeat expands somatically from 40-45 to 100-500+ CAGs in SPNs. Somatic expansion from 40 to 150 CAGs had no apparent cell-autonomous effect, but SPNs with 150-500+ CAGs lost positive and then negative features of neuronal identity, de-repressed senescence/apoptosis genes, and were lost. Our results suggest that somatic repeat expansion beyond 150 CAGs causes SPNs to degenerate quickly and asynchronously. We conclude that in HD, at any one time, most neurons have an innocuous but unstable HTT gene and that HD pathogenesis is a DNA process for almost all of a neuron's life.
Collapse
Affiliation(s)
- Robert E Handsaker
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.
| | - Seva Kashin
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.
| | - Nora M Reed
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Steven Tan
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Won-Seok Lee
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Tara M McDonald
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | | | - Nolan Kamitaki
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher D Mullally
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | | | - Melissa Goldman
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Gabriel Lind
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Rhea Kohli
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | | | - Marina Hogan
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Kiku Ichihara
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Sabina Berretta
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02215, USA.
| | - Steven A McCarroll
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02215, USA; Howard Hughes Medical Institute, Boston, MA 02215, USA.
| |
Collapse
|
5
|
Heim B, Mandler E, Peball M, Carbone F, Schwarzová K, Demjaha R, Tafrali C, Buchmann A, Khalil M, Djamshidian A, Seppi K. Serum neurofilament light chain but not serum glial fibrillary acidic protein is a marker of early Huntington's disease. J Neurol 2025; 272:174. [PMID: 39891767 PMCID: PMC11787204 DOI: 10.1007/s00415-025-12901-y] [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: 09/14/2024] [Revised: 12/31/2024] [Accepted: 01/05/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND Huntington's disease (HD) is caused by CAG trinucleotide expansion on chromosome 4, leading to mutant Huntingtin production. Premanifest carriers show no obvious clinical signs, and early symptoms progress slowly. Fluid biomarkers like neurofilament light (NfL) and glial fibrillary acidic protein (GFAP), measurable in cerebrospinal fluid and serum (sNfL, sGFAP), offer potential predicting HD progression. OBJECTIVE To assess the role of sGFAP and sNfL and clinical biomarkers in different disease stages and correlate with disease progression. METHODS HD mutation carriers were categorized into clinical stages according to their motor symptoms and functional capacities. The Unified HD Rating Scale, cognitive assessments and olfactory tests were used to characterize the patients clinically. Furthermore, sNfL and sGFAP levels were assessed. RESULTS We consecutively included 44 HD mutation carriers (13 premanifest HD (preHD), 18 in early (early HD) and 13 in advanced (advanced HD) disease stages) and 19 healthy controls (HC). Advanced HD patients performed worse on all clinical tasks and had higher sGFAP and sNfL levels compared to other groups (all p values < 0.05). We did not find difference in sGFAP levels between the preHD, early HD and HC group (all p values > 0.05). In contrast, sNfL levels differed significantly between preHD and early HD, and HC (all p values < 0.05). ROC curve analysis revealed that the AUC of sGFAP (0.970) exhibited superior discriminatory accuracy compared to sNfL (0.791) levels in separating advanced from early HD patients. By contrast, ROC curve analysis revealed that the AUC of sNFL (0.988) exhibited superior discriminatory accuracy compared to sGFAP (0.609) levels in separating all HD mutation carriers from HC. CONCLUSIONS Our study indicates that sNfL can detect changes in very early and premanifest HD stages, whereas sGFAP showed differences in more advanced stages only.
Collapse
Affiliation(s)
- Beatrice Heim
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria.
| | - Elias Mandler
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Marina Peball
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Federico Carbone
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Katarína Schwarzová
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Rina Demjaha
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Cansu Tafrali
- Department of Neurology, Medical University of Graz, Graz, Austria
| | | | - Michael Khalil
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Atbin Djamshidian
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Klaus Seppi
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria.
- Department of Neurology, Hospital Kufstein, Kufstein, Austria.
| |
Collapse
|
6
|
Aqel S, Ahmad J, Saleh I, Fathima A, Al Thani AA, Mohamed WMY, Shaito AA. Advances in Huntington's Disease Biomarkers: A 10-Year Bibliometric Analysis and a Comprehensive Review. BIOLOGY 2025; 14:129. [PMID: 40001897 PMCID: PMC11852324 DOI: 10.3390/biology14020129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 02/27/2025]
Abstract
Neurodegenerative disorders (NDs) cause progressive neuronal loss and are a significant public health concern, with NDs projected to become the second leading global cause of death within two decades. Huntington's disease (HD) is a rare, progressive ND caused by an autosomal-dominant mutation in the huntingtin (HTT) gene, leading to severe neuronal loss in the brain and resulting in debilitating motor, cognitive, and psychiatric symptoms. Given the complex pathology of HD, biomarkers are essential for performing early diagnosis, monitoring disease progression, and evaluating treatment efficacy. However, the identification of consistent HD biomarkers is challenging due to the prolonged premanifest HD stage, HD's heterogeneous presentation, and its multiple underlying biological pathways. This study involves a 10-year bibliometric analysis of HD biomarker research, revealing key research trends and gaps. The study also features a comprehensive literature review of emerging HD biomarkers, concluding the need for better stratification of HD patients and well-designed longitudinal studies to validate HD biomarkers. Promising candidate wet HD biomarkers- including neurofilament light chain protein (NfL), microRNAs, the mutant HTT protein, and specific metabolic and inflammatory markers- are discussed, with emphasis on their potential utility in the premanifest HD stage. Additionally, biomarkers reflecting brain structural deficits and motor or behavioral impairments, such as neurophysiological (e.g., motor tapping, speech, EEG, and event-related potentials) and imaging (e.g., MRI, PET, and diffusion tensor imaging) biomarkers, are evaluated. The findings underscore that the discovery and validation of reliable HD biomarkers urgently require improved patient stratification and well-designed longitudinal studies. Reliable biomarkers, particularly in the premanifest HD stage, are crucial for optimizing HD clinical management strategies, enabling personalized treatment approaches, and advancing clinical trials of HD-modifying therapies.
Collapse
Affiliation(s)
- Sarah Aqel
- Medical Research Center, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar;
| | - Jamil Ahmad
- Medical Education, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar;
| | - Iman Saleh
- Biological Science Program, Department of Biological and Environmental Sciences, College of Art and Science, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Aseela Fathima
- Biomedical Research Center (BRC), QU Health Sector, Qatar University, Doha P.O. Box 2713, Qatar; (A.F.); (A.A.A.T.)
- Department of Biomedical Sciences, College of Health Sciences, QU Health Sector, Qatar University, Doha P.O. Box 2713, Qatar
| | - Asmaa A. Al Thani
- Biomedical Research Center (BRC), QU Health Sector, Qatar University, Doha P.O. Box 2713, Qatar; (A.F.); (A.A.A.T.)
- Department of Biomedical Sciences, College of Health Sciences, QU Health Sector, Qatar University, Doha P.O. Box 2713, Qatar
| | - Wael M. Y. Mohamed
- Department of Basic Medical Sciences, Kulliyyah of Medicine, International Islamic University Malaysia (IIUM), Kuantan 50728, Malaysia;
- Clinical Pharmacology Department, Menoufia Medical School, Menoufia University, Shebin El-Kom 32511, Egypt
| | - Abdullah A. Shaito
- Biomedical Research Center (BRC), QU Health Sector, Qatar University, Doha P.O. Box 2713, Qatar; (A.F.); (A.A.A.T.)
- Department of Biomedical Sciences, College of Health Sciences, QU Health Sector, Qatar University, Doha P.O. Box 2713, Qatar
- College of Medicine, QU Health Sector, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
7
|
Bamford AR, Parkin GM, Corey-Bloom J, Thomas EA. Comparisons of neurodegenerative disease biomarkers across different biological fluids from patients with Huntington's disease. J Neurol 2025; 272:158. [PMID: 39849121 PMCID: PMC11759467 DOI: 10.1007/s00415-024-12785-4] [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: 09/25/2024] [Accepted: 10/11/2024] [Indexed: 01/25/2025]
Abstract
Fluid biomarkers play important roles in many aspects of neurodegenerative diseases, such as Huntington's disease (HD). However, a main question relates to how well levels of biomarkers measured in CSF are correlated with those measured in peripheral fluids, such as blood or saliva. In this study, we quantified levels of four neurodegenerative disease-related proteins, neurofilament light (NfL), total tau (t-tau), glial fibrillary acidic protein (GFAP) and YKL-40 in matched CSF, plasma and saliva samples from Huntingtin (HTT) gene-positive individuals (n = 21) using electrochemiluminescence assays. In addition, salivary levels of NfL, t-tau, and GFAP were quantified from a larger cohort (n = 95). We found both positive and negative correlations in the levels of these biomarkers among different biofluids. Most notably, in contrast to the significant positive correlations observed between CSF and plasma levels for NfL and GFAP, we detected significant negative correlations between the CSF and saliva levels of NfL and GFAP. With regard to clinical measures, both plasma and CSF levels of NfL were significantly positively correlated with Total Motor Score and chorea, whereas saliva levels of NfL showed significant correlations in the opposite direction. Additional correlations between salivary biomarkers with clinical data, adjusting for age, sex and CAG repeat length, confirmed that salivary NfL was significantly negatively associated with chorea scores in manifest HD, but not premanifest (PM), individuals. In contrast, salivary t-tau was positively associated with measures of cognition in PM participants. These findings suggest that salivary levels of NfL and t-tau proteins may exemplify non-invasive biomarkers for disease symptoms at different stages of illness. Further, these findings highlight the notion that different forms of disease proteins exist in different biological fluids.
Collapse
Affiliation(s)
- Alison R Bamford
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
- Institute for Interdisciplinary Salivary Bioscience Research, University of California Irvine, Irvine, CA, USA
| | - Georgia M Parkin
- Phoenix Australia - Centre for Posttraumatic Mental Health, Department of Psychiatry, University of Melbourne, Parkville, VIC, Australia
| | - Jody Corey-Bloom
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA
| | - Elizabeth A Thomas
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA.
- Institute for Interdisciplinary Salivary Bioscience Research, University of California Irvine, Irvine, CA, USA.
- Department of Neurosciences, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
8
|
Moon J, Oh E, Kim M, Kim R, Yoo D, Shin C, Lee JY, Kim JM, Koh SB, Kim M, Jeon B. A Practical Guide for Diagnostic Investigations and Special Considerations in Patients With Huntington's Disease in Korea. J Mov Disord 2025; 18:17-30. [PMID: 39725405 PMCID: PMC11824489 DOI: 10.14802/jmd.24232] [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: 11/15/2024] [Revised: 12/21/2024] [Accepted: 12/24/2024] [Indexed: 12/28/2024] Open
Abstract
This review provides a comprehensive framework for the diagnostic approach and management of Huntington's disease (HD) tailored to the Korean population. Key topics include genetic counseling, predictive testing, and reproductive options like preimplantation genetic testing. Strategies for assessing disease progression in premanifest HD through laboratory investigations, biofluid, and imaging biomarkers are highlighted. Special considerations for juvenile and late-onset HD, along with associated comorbidities like diabetes mellitus, hypertension, and cardiovascular abnormalities, are discussed. The guide emphasizes personalized symptom management, including pharmacotherapy, physical therapy, and nutritional support, while exploring emerging disease-modifying treatments. A multidisciplinary care model is advocated to improve outcomes for HD patients and caregivers in Korea.
Collapse
Affiliation(s)
- Jangsup Moon
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Korea
| | - Eungseok Oh
- Department of Neurology, Chungnam National University Hospital, Daejeon, Korea
- Department of Neurology, Chungnam National University College of Medicine, Daejeon, Korea
| | - Minkyeong Kim
- Department of Neurology, Gyeongsang National University Hospital, Jinju, Korea
| | - Ryul Kim
- Department of Neurology, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Dallah Yoo
- Department of Neurology, Kyung Hee University Hospital, Kyung Hee University School of Medicine, Seoul, Korea
| | - Chaewon Shin
- Department of Neurology, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Neurology, Chungnam National University Sejong Hospital, Sejong, Korea
| | - Jee-Young Lee
- Department of Neurology, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Jong-Min Kim
- Department of Neurology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Seong-Beom Koh
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Manho Kim
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Beomseok Jeon
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - on behalf of the Korean Huntington’s Disease Society
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Neurology, Chungnam National University Hospital, Daejeon, Korea
- Department of Neurology, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Neurology, Gyeongsang National University Hospital, Jinju, Korea
- Department of Neurology, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
- Department of Neurology, Kyung Hee University Hospital, Kyung Hee University School of Medicine, Seoul, Korea
- Department of Neurology, Chungnam National University Sejong Hospital, Sejong, Korea
- Department of Neurology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
9
|
Voysey ZJ, Owen NE, Holbrook JA, Malpetti M, Le Draoulec C, Spindler LRB, Goodman AOG, Lazar AS, Barker RA. A 14-year longitudinal study of neurofilament light chain dynamics in premanifest and transitional Huntington's disease. J Neurol 2024; 271:7572-7582. [PMID: 39361164 PMCID: PMC11588772 DOI: 10.1007/s00415-024-12700-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: 06/05/2024] [Revised: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND Growing evidence supports the value of neurofilament light (NfL) as a prognostic biomarker in premanifest Huntington's disease (HD). To date, however, there has been no longitudinal study exceeding 3 years examining either its serial dynamics or predictive power in HD. We aimed to conduct the first such study. METHODS Serum NfL was sampled using ultrasensitive immunoassay at four timepoints across a 14-year period in a cohort of HD gene carriers (n = 21) and controls (n = 14). Gene carriers were premanifest at baseline. Clinical features of HD were evaluated by Unified Huntington's Disease Rating Scale (UHDRS TMS), Montreal Cognitive Assessment (MoCA), Trail A/B task, Symbol Digit Modalities Task and semantic/phonemic fluency tasks. RESULTS 14/21 HD gene carriers converted to prodromal or manifest disease by the final timepoint ("converters"). At baseline and each subsequent timepoint, NfL levels were higher in converters than in non-converters and controls (p = < 0.001-0.03, ηp2 = 0.25-0.66). The estimated rate of change in NfL was higher in converters than in non-converters (p = 0.03) and controls (p = 0.001). Baseline NfL was able to discriminate converters from non-converters (area under curve = 1.000, p = 0.003). A higher rate of change in NfL was predictive of more severe motor (UHDRS-TMS p = 0.007, β = 0.711, R2 = 0.468) and cognitive deficits (MoCA p = 0.007, β = - 0.798, R2 = 0.604; Trail B, p = 0.007, β = 0.772, R2 = 0.567; phonemic fluency p = 0.035, β = - 0.632, R2 = 0.345). CONCLUSIONS Our data suggest that (1) NfL longitudinal dynamics in premanifest/transitional HD are non-constant; rising faster in those closer to disease onset, and (2) NfL can identify individuals at risk of conversion to manifest disease and predict clinical trajectory, > 10 years from disease onset.
Collapse
Affiliation(s)
- Z J Voysey
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK.
| | - N E Owen
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - J A Holbrook
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - M Malpetti
- Department of Clinical Neurosciences, Cambridge Centre for Frontotemporal Dementia and Related Disorders, University of Cambridge, Cambridge, UK
| | - C Le Draoulec
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - L R B Spindler
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - A O G Goodman
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - A S Lazar
- Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK
| | - R A Barker
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
10
|
Anderson DG, Ferreira-Correia A, Rodrigues FB, Byrne LM, Wild EJ, Krause A. Comparative analysis of neurofilament light chain in Huntington's disease like 2 and Huntington's disease. J Huntingtons Dis 2024:18796397241300141. [PMID: 39973395 DOI: 10.1177/18796397241300141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Huntington's disease-like 2 (HDL2) closely resembles Huntington's disease (HD) in clinical and pathological features. Neurofilament light chain (NfL) is an important biomarker in HD research and holds potential in HDL2. To evaluate NfL's utility in HDL2, a comparative analysis among HDL2 (n = 12), HD (n = 9), and unaffected controls (n = 9) was conducted. Employing a cross-sectional design, NfL levels were assessed in blood plasma. Concentrations were notably elevated in both HD and HDL2 groups compared to controls. HD patients displayed higher NfL levels than HDL2, possibly reflecting disease duration differences. NfL effectively distinguished HDL2 from controls, highlighting its promise as a possible biomarker in HDL2 research.
Collapse
Affiliation(s)
- David G Anderson
- Department of Neurology, Institute of Neurological Sciences, Queen Elizabeth University Hospital and University of Glasgow, Glasgow, UK
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Aline Ferreira-Correia
- Department of Psychology, School of Human and Community Development, University of the Witwatersrand, Johannesburg, South Africa
| | - Filipe B Rodrigues
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
- Laboratory of Clinical Pharmacology and Therapeutics, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Lisbon, Portugal
| | - Lauren M Byrne
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Edward J Wild
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Amanda Krause
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
11
|
Li X, Tong H, Xu S, Zhou G, Yang T, Yin S, Yang S, Li X, Li S. Neuroinflammatory Proteins in Huntington's Disease: Insights into Mechanisms, Diagnosis, and Therapeutic Implications. Int J Mol Sci 2024; 25:11787. [PMID: 39519337 PMCID: PMC11546928 DOI: 10.3390/ijms252111787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Huntington's disease (HD) is a hereditary neurodegenerative disorder caused by a CAG tract expansion in the huntingtin gene (HTT). HD is characterized by involuntary movements, cognitive decline, and behavioral changes. Pathologically, patients with HD show selective striatal neuronal vulnerability at the early disease stage, although the mutant protein is ubiquitously expressed. Activation of the immune system and glial cell-mediated neuroinflammatory responses are early pathological features and have been found in all neurodegenerative diseases (NDDs), including HD. However, the role of inflammation in HD, as well as its therapeutic significance, has been less extensively studied compared to other NDDs. This review highlights the significantly elevated levels of inflammatory proteins and cellular markers observed in various HD animal models and HD patient tissues, emphasizing the critical roles of microglia, astrocytes, and oligodendrocytes in mediating neuroinflammation in HD. Moreover, it expands on recent discoveries related to the peripheral immune system's involvement in HD. Although current immunomodulatory treatments and inflammatory biomarkers for adjunctive diagnosis in HD are limited, targeting inflammation in combination with other therapies, along with comprehensive personalized treatment approaches, shows promising therapeutic potential.
Collapse
Affiliation(s)
- Xinhui Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Huichun Tong
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Shuying Xu
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Gongke Zhou
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Tianqi Yang
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Shurui Yin
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Sitong Yang
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Xiaojiang Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Shihua Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| |
Collapse
|
12
|
Horne K, Carmichael A, Mercieca EC, Glikmann-Johnston Y, Stout JC, Irish M. Delineating the neural substrates of autobiographical memory impairment in Huntington's disease. Eur J Neurosci 2024; 60:6509-6524. [PMID: 39419578 DOI: 10.1111/ejn.16576] [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: 07/22/2024] [Revised: 09/12/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024]
Abstract
Emerging evidence suggests that autobiographical memory (ABM) is altered in Huntington's disease (HD). While these impairments are typically attributed to frontostriatal dysfunction, the neural substrates of ABM impairment in HD remain unexplored. To this end, we assessed ABM in 30 participants with genetically confirmed HD (18 premanifest, 12 manifest) and 24 age-matched healthy controls. Participants completed the Autobiographical Interview to assess free and probed ABM recall and underwent structural brain imaging. Whole-brain voxel-based morphometry (VBM) was used to explore voxel-wise associations between ABM performance and grey matter intensity (False Discovery Rate corrected at q = 0.05). Relative to controls, HD participants displayed significantly less detailed ABM retrieval across free and probed recall conditions, irrespective of disease stage. Recall performance did not differ significantly between manifest and premanifest HD groups. VBM analyses indicated that poorer ABM performance was associated with atrophy of a distributed cortico-subcortical network. Key regions implicated irrespective of ABM condition included the bilateral occipital cortex, left precuneus, right parahippocampal gyrus and right caudate nucleus. In addition, probed ABM recall was associated with the superior and inferior frontal gyri, frontal pole, right hippocampus, nucleus accumbens, paracingulate gyrus and cerebellum. Overall, our findings indicate that ABM impairments in HD reflect the progressive degeneration of a distributed cortico-subcortical brain network comprising medial temporal, frontal, striatal and posterior parietal cortices. Our findings advance our understanding of the neurocognitive profile of HD, providing an important foundation for future interventions to support memory function in this population.
Collapse
Affiliation(s)
- Kristina Horne
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Psychology, The University of Sydney, Sydney, New South Wales, Australia
| | - Anna Carmichael
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Clayton, Australia
| | - Emily-Clare Mercieca
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Clayton, Australia
| | - Yifat Glikmann-Johnston
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Clayton, Australia
| | - Julie C Stout
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Clayton, Australia
| | - Muireann Irish
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Psychology, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
13
|
Caron NS, Byrne LM, Lemarié FL, Bone JN, Aly AEE, Ko S, Anderson C, Casal LL, Hill AM, Hawellek DJ, McColgan P, Wild EJ, Leavitt BR, Hayden MR. Elevated plasma and CSF neurofilament light chain concentrations are stabilized in response to mutant huntingtin lowering in the brains of Huntington's disease mice. Transl Neurodegener 2024; 13:50. [PMID: 39380076 PMCID: PMC11460072 DOI: 10.1186/s40035-024-00443-8] [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: 05/06/2024] [Accepted: 09/11/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Therapeutic approaches aimed at lowering toxic mutant huntingtin (mHTT) levels in the brain can reverse disease phenotypes in animal models of Huntington's disease (HD) and are currently being evaluated in clinical trials. Sensitive and dynamic response biomarkers are needed to assess the efficacy of such candidate therapies. Neurofilament light chain (NfL) is a biomarker of neurodegeneration that increases in cerebrospinal fluid (CSF) and blood with progression of HD. However, it remains unknown whether NfL in biofluids could serve as a response biomarker for assessing the efficacy of disease-modifying therapies for HD. METHODS Longitudinal plasma and cross-sectional CSF samples were collected from the YAC128 transgenic mouse model of HD and wild-type (WT) littermate control mice throughout the natural history of disease. Additionally, biofluids were collected from YAC128 mice following intracerebroventricular administration of an antisense oligonucleotide (ASO) targeting the mutant HTT transgene (HTT ASO), at ages both before and after the onset of disease phenotypes. NfL concentrations in plasma and CSF were quantified using ultrasensitive single-molecule array technology. RESULTS Plasma and CSF NfL concentrations were significantly elevated in YAC128 compared to WT littermate control mice from 9 months of age. Treatment of YAC128 mice with either 15 or 50 µg HTT ASO resulted in a dose-dependent, allele-selective reduction of mHTT throughout the brain at a 3-month interval, which was sustained with high-dose HTT ASO treatment for up to 6 months. Lowering of brain mHTT prior to the onset of regional brain atrophy and HD-like motor deficits in this model had minimal effect on plasma NfL at either dose, but led to a dose-dependent reduction of CSF NfL. In contrast, initiating mHTT lowering in the brain after the onset of neuropathological and behavioural phenotypes in YAC128 mice resulted in a dose-dependent stabilization of NfL increases in both plasma and CSF. CONCLUSIONS Our data provide evidence that the response of NfL in biofluids is influenced by the magnitude of mHTT lowering in the brain and the timing of intervention, suggesting that NfL may serve as a promising exploratory response biomarker for HD.
Collapse
Affiliation(s)
- Nicholas S Caron
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Lauren M Byrne
- UCL Huntington's Disease Centre, University College London Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Fanny L Lemarié
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Jeffrey N Bone
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Department of Statistics, University of British Columbia, Vancouver, BC, V6T 1Z2, Canada
| | - Amirah E-E Aly
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Seunghyun Ko
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada
| | - Christine Anderson
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada
| | - Lorenzo L Casal
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada
| | - Austin M Hill
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada
| | - David J Hawellek
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Peter McColgan
- Roche Products Ltd., Welwyn Garden City, AL7 1TW, United Kingdom
| | - Edward J Wild
- UCL Huntington's Disease Centre, University College London Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Blair R Leavitt
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Michael R Hayden
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada.
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
14
|
Herrero‐Lorenzo M, Pérez‐Pérez J, Escaramís G, Martínez‐Horta S, Pérez‐González R, Rivas‐Asensio E, Kulisevsky J, Gámez‐Valero A, Martí E. Small RNAs in plasma extracellular vesicles define biomarkers of premanifest changes in Huntington's disease. J Extracell Vesicles 2024; 13:e12522. [PMID: 39377487 PMCID: PMC11633361 DOI: 10.1002/jev2.12522] [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: 01/30/2024] [Revised: 08/06/2024] [Accepted: 09/10/2024] [Indexed: 10/09/2024] Open
Abstract
Despite the advances in the understanding of Huntington's disease (HD), there is a need for molecular biomarkers to categorize mutation carriers during the preclinical stage of the disease preceding functional decline. Small RNAs (sRNAs) are a promising source of biomarkers since their expression levels are highly sensitive to pathobiological processes. Here, using an optimized method for plasma extracellular vesicles (EVs) purification and an exhaustive analysis pipeline of sRNA sequencing data, we show that EV-sRNAs are downregulated early in mutation carriers and that this deregulation is associated with premanifest cognitive performance. Seven candidate sRNAs (tRF-Glu-CTC, tRF-Gly-GCC, miR-451a, miR-21-5p, miR-26a-5p, miR-27a-3p and let7a-5p) were validated in additional subjects, showing a significant diagnostic accuracy at premanifest stages. Of these, miR-21-5p was significantly decreased over time in a longitudinal study; and miR-21-5p and miR-26a-5p levels correlated with cognitive changes in the premanifest cohort. In summary, the present results suggest that deregulated plasma EV-sRNAs define an early biosignature in mutation carriers with specific species highlighting the progression and cognitive changes occurring at the premanifest stage.
Collapse
Affiliation(s)
- Marina Herrero‐Lorenzo
- Department of BiomedicineFaculty of Medicine, Institute of NeurosciencesUniversity of BarcelonaBarcelonaCatalunyaSpain
| | - Jesús Pérez‐Pérez
- Movement Disorders UnitNeurology DepartmentSant Pau HospitalBarcelonaCatalunyaSpain
- Biomedical Research Institute (IIB‐Sant Pau)BarcelonaCatalunyaSpain
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED)MadridSpain
| | - Georgia Escaramís
- Department of BiomedicineFaculty of Medicine, Institute of NeurosciencesUniversity of BarcelonaBarcelonaCatalunyaSpain
- Biomedical Research Networking Center for Epidemiology and Public Health (CIBERESP)Spanish Ministry of Science and InnovationMadridSpain
| | - Saül Martínez‐Horta
- Movement Disorders UnitNeurology DepartmentSant Pau HospitalBarcelonaCatalunyaSpain
- Biomedical Research Institute (IIB‐Sant Pau)BarcelonaCatalunyaSpain
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED)MadridSpain
| | - Rocío Pérez‐González
- Movement Disorders UnitNeurology DepartmentSant Pau HospitalBarcelonaCatalunyaSpain
- Biomedical Research Institute (IIB‐Sant Pau)BarcelonaCatalunyaSpain
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED)MadridSpain
- Alicante Institute for Health and Biomedical Research (ISABIAL) and Neuroscience InstituteAlicanteSpain
| | - Elisa Rivas‐Asensio
- Movement Disorders UnitNeurology DepartmentSant Pau HospitalBarcelonaCatalunyaSpain
- Biomedical Research Institute (IIB‐Sant Pau)BarcelonaCatalunyaSpain
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED)MadridSpain
| | - Jaime Kulisevsky
- Movement Disorders UnitNeurology DepartmentSant Pau HospitalBarcelonaCatalunyaSpain
- Biomedical Research Institute (IIB‐Sant Pau)BarcelonaCatalunyaSpain
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED)MadridSpain
| | - Ana Gámez‐Valero
- Department of BiomedicineFaculty of Medicine, Institute of NeurosciencesUniversity of BarcelonaBarcelonaCatalunyaSpain
- Biomedical Research Networking Center for Epidemiology and Public Health (CIBERESP)Spanish Ministry of Science and InnovationMadridSpain
| | - Eulàlia Martí
- Department of BiomedicineFaculty of Medicine, Institute of NeurosciencesUniversity of BarcelonaBarcelonaCatalunyaSpain
- Biomedical Research Networking Center for Epidemiology and Public Health (CIBERESP)Spanish Ministry of Science and InnovationMadridSpain
- August Pi i Sunyer Biomedical research Institute (IDIBAPS), BarcelonaCatalunyaSpain
| |
Collapse
|
15
|
Ghofrani-Jahromi M, Poudel GR, Razi A, Abeyasinghe PM, Paulsen JS, Tabrizi SJ, Saha S, Georgiou-Karistianis N. Prognostic enrichment for early-stage Huntington's disease: An explainable machine learning approach for clinical trial. Neuroimage Clin 2024; 43:103650. [PMID: 39142216 PMCID: PMC11367643 DOI: 10.1016/j.nicl.2024.103650] [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: 04/18/2024] [Revised: 07/11/2024] [Accepted: 07/31/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND In Huntington's disease clinical trials, recruitment and stratification approaches primarily rely on genetic load, cognitive and motor assessment scores. They focus less on in vivo brain imaging markers, which reflect neuropathology well before clinical diagnosis. Machine learning methods offer a degree of sophistication which could significantly improve prognosis and stratification by leveraging multimodal biomarkers from large datasets. Such models specifically tailored to HD gene expansion carriers could further enhance the efficacy of the stratification process. OBJECTIVES To improve stratification of Huntington's disease individuals for clinical trials. METHODS We used data from 451 gene positive individuals with Huntington's disease (both premanifest and diagnosed) from previously published cohorts (PREDICT, TRACK, TrackON, and IMAGE). We applied whole-brain parcellation to longitudinal brain scans and measured the rate of lateral ventricular enlargement, over 3 years, which was used as the target variable for our prognostic random forest regression models. The models were trained on various combinations of features at baseline, including genetic load, cognitive and motor assessment score biomarkers, as well as brain imaging-derived features. Furthermore, a simplified stratification model was developed to classify individuals into two homogenous groups (low risk and high risk) based on their anticipated rate of ventricular enlargement. RESULTS The predictive accuracy of the prognostic models substantially improved by integrating brain imaging features alongside genetic load, cognitive and motor biomarkers: a 24 % reduction in the cross-validated mean absolute error, yielding an error of 530 mm3/year. The stratification model had a cross-validated accuracy of 81 % in differentiating between moderate and fast progressors (precision = 83 %, recall = 80 %). CONCLUSIONS This study validated the effectiveness of machine learning in differentiating between low- and high-risk individuals based on the rate of ventricular enlargement. The models were exclusively trained using features from HD individuals, which offers a more disease-specific, simplified, and accurate approach for prognostic enrichment compared to relying on features extracted from healthy control groups, as done in previous studies. The proposed method has the potential to enhance clinical utility by: i) enabling more targeted recruitment of individuals for clinical trials, ii) improving post-hoc evaluation of individuals, and iii) ultimately leading to better outcomes for individuals through personalized treatment selection.
Collapse
Affiliation(s)
| | - Govinda R Poudel
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne VIC3000, Australia
| | - Adeel Razi
- Turner Institute for Brain and Mental Health, Monash University, Clayton VIC3800, Australia
| | - Pubu M Abeyasinghe
- Turner Institute for Brain and Mental Health, Monash University, Clayton VIC3800, Australia
| | - Jane S Paulsen
- Department of Neurology, University of Wisconsin-Madison, 1685 Highland Avenue, Madison, WI, USA
| | - Sarah J Tabrizi
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology, UK Dementia Research Institute, Department of Neurodegenerative Diseases, University College London, London, UK
| | - Susmita Saha
- Turner Institute for Brain and Mental Health, Monash University, Clayton VIC3800, Australia
| | | |
Collapse
|
16
|
Machacek M, Garcia-Montoya E, McColgan P, Sanwald-Ducray P, Mazer NA. NfL concentration in CSF is a quantitative marker of the rate of neurodegeneration in aging and Huntington's disease: a semi-mechanistic model-based analysis. Front Neurosci 2024; 18:1420198. [PMID: 39022122 PMCID: PMC11253127 DOI: 10.3389/fnins.2024.1420198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/10/2024] [Indexed: 07/20/2024] Open
Abstract
The concentrations of neurofilament light chain (NfL) in cerebrospinal fluid (CSF) and plasma have become key biomarkers of many neurodegenerative diseases, including Huntington's Disease (HD). However, the relationship between the dynamics of NfL concentrations in CSF and the time-course of neurodegeneration (whole brain atrophy) has not yet been described in a quantitative and mechanistic manner. Here, we present a novel semi-mechanistic model, which postulates that the amount of NfL entering the CSF corresponds to the amount of NfL released from damaged neurons, whose degeneration results in a decrease in brain volume. In mathematical terms, the model expresses the NfL concentration in CSF in terms of the NfL concentration in brain tissue, the rate of change of whole brain volume and the CSF flow rate. To test our model, we used a non-linear mixed effects approach to analyze NfL and brain volume data from the HD-CSF study, a 24-month prospective study of individuals with premanifest HD, manifest HD and healthy controls. The time-course of whole brain volume, obtained from MRI, was represented empirically by a 2nd order polynomial, from which its rate of change was computed. CSF flow rates in healthy and HD populations were taken from recent literature data. By estimating the NfL concentration in brain tissue, the model successfully described the time-course of the NfL concentration in CSF in both HD subjects and healthy controls. Furthermore, the model-derived estimate of NfL concentration in brain agreed well with recent direct experimental measurements. The consistency of our model with the NfL and brain volume data suggests that the NfL concentration in CSF reflects the rate, rather than the extent, of neurodegeneration and that the increase in NfL concentration over time is a measure of the accelerating rate of neurodegeneration associated with aging and HD. For HD subjects, the degree of acceleration was found to increase markedly with the number of CAG repeats on their HTT gene. The application of our semi-mechanistic NfL model to other neurodegenerative diseases is discussed.
Collapse
Affiliation(s)
| | | | - Peter McColgan
- Roche Products Limited, Welwyn Garden City, United Kingdom
| | - Patricia Sanwald-Ducray
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Norman Alan Mazer
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
- NAM Consulting, Pfeffingen, Switzerland
| |
Collapse
|
17
|
Mehanna R, Jankovic J. Systemic Symptoms in Huntington's Disease: A Comprehensive Review. Mov Disord Clin Pract 2024; 11:453-464. [PMID: 38529740 PMCID: PMC11078495 DOI: 10.1002/mdc3.14029] [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: 12/02/2023] [Revised: 02/26/2024] [Accepted: 03/12/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Although Huntington's disease (HD) is usually thought of as a triad of motor, cognitive, and psychiatric symptoms, there is growing appreciation of HD as a systemic illness affecting the entire body. OBJECTIVES This review aims to draw attention to these systemic non-motor symptoms in HD. METHODS We identified relevant studies published in English by searching MEDLINE (from 1966 to September 2023), using the following subject headings: Huntington disease, autonomic, systemic, cardiovascular, respiratory, gastrointestinal, urinary, sexual and cutaneous, and additional specific symptoms. RESULTS Data from 123 articles were critically reviewed with focus on systemic features associated with HD, such as cardiovascular, respiratory, gastrointestinal, urinary, sexual and sweating. CONCLUSION This systematic review draws attention to a variety of systemic and autonomic co-morbidities in patients with HD. Not all of them correlate with the severity of the primary HD symptoms or CAG repeats. More research is needed to better understand the pathophysiology and treatment of systemic and autonomic dysfunction in HD.
Collapse
Affiliation(s)
- Raja Mehanna
- Department of NeurologyUniversity of Texas Health Science Center at Houston, McGovern Medical SchoolHoustonTXUSA
| | - Joseph Jankovic
- Parkinson's Disease Center and Movement Disorders Clinic, Department of NeurologyBaylor College of MedicineHoustonTXUSA
| |
Collapse
|
18
|
Khalil M, Teunissen CE, Lehmann S, Otto M, Piehl F, Ziemssen T, Bittner S, Sormani MP, Gattringer T, Abu-Rumeileh S, Thebault S, Abdelhak A, Green A, Benkert P, Kappos L, Comabella M, Tumani H, Freedman MS, Petzold A, Blennow K, Zetterberg H, Leppert D, Kuhle J. Neurofilaments as biomarkers in neurological disorders - towards clinical application. Nat Rev Neurol 2024; 20:269-287. [PMID: 38609644 DOI: 10.1038/s41582-024-00955-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2024] [Indexed: 04/14/2024]
Abstract
Neurofilament proteins have been validated as specific body fluid biomarkers of neuro-axonal injury. The advent of highly sensitive analytical platforms that enable reliable quantification of neurofilaments in blood samples and simplify longitudinal follow-up has paved the way for the development of neurofilaments as a biomarker in clinical practice. Potential applications include assessment of disease activity, monitoring of treatment responses, and determining prognosis in many acute and chronic neurological disorders as well as their use as an outcome measure in trials of novel therapies. Progress has now moved the measurement of neurofilaments to the doorstep of routine clinical practice for the evaluation of individuals. In this Review, we first outline current knowledge on the structure and function of neurofilaments. We then discuss analytical and statistical approaches and challenges in determining neurofilament levels in different clinical contexts and assess the implications of neurofilament light chain (NfL) levels in normal ageing and the confounding factors that need to be considered when interpreting NfL measures. In addition, we summarize the current value and potential clinical applications of neurofilaments as a biomarker of neuro-axonal damage in a range of neurological disorders, including multiple sclerosis, Alzheimer disease, frontotemporal dementia, amyotrophic lateral sclerosis, stroke and cerebrovascular disease, traumatic brain injury, and Parkinson disease. We also consider the steps needed to complete the translation of neurofilaments from the laboratory to the management of neurological diseases in clinical practice.
Collapse
Affiliation(s)
- Michael Khalil
- Department of Neurology, Medical University of Graz, Graz, Austria.
| | - Charlotte E Teunissen
- Neurochemistry Laboratory Department of Laboratory Medicine, Amsterdam Neuroscience, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, Netherlands
| | - Sylvain Lehmann
- LBPC-PPC, Université de Montpellier, INM INSERM, IRMB CHU de Montpellier, Montpellier, France
| | - Markus Otto
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Tjalf Ziemssen
- Center of Clinical Neuroscience, Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience (FTN), and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Maria Pia Sormani
- Department of Health Sciences, University of Genova, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Thomas Gattringer
- Department of Neurology, Medical University of Graz, Graz, Austria
- Division of Neuroradiology, Vascular and Interventional Radiology, Department of Radiology, Medical University of Graz, Graz, Austria
| | - Samir Abu-Rumeileh
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Simon Thebault
- Multiple Sclerosis Division, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmed Abdelhak
- Weill Institute for Neurosciences, Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| | - Ari Green
- Weill Institute for Neurosciences, Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| | - Pascal Benkert
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital and University of Basel, Basel, Switzerland
| | - Ludwig Kappos
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital and University of Basel, Basel, Switzerland
| | - Manuel Comabella
- Neurology Department, Multiple Sclerosis Centre of Catalonia, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Hayrettin Tumani
- Department of Neurology, CSF Laboratory, Ulm University Hospital, Ulm, Germany
| | - Mark S Freedman
- Department of Medicine, University of Ottawa, The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Axel Petzold
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Neurology, MS Centre and Neuro-ophthalmology Expertise Centre Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
- Moorfields Eye Hospital, The National Hospital for Neurology and Neurosurgery and the Queen Square Institute of Neurology, UCL, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, P. R. China
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - David Leppert
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital and University of Basel, Basel, Switzerland
| | - Jens Kuhle
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland.
- Department of Neurology, University Hospital and University of Basel, Basel, Switzerland.
| |
Collapse
|
19
|
Field SE, Curle AJ, Barker RA. Inflammation and Huntington's disease - a neglected therapeutic target? Expert Opin Investig Drugs 2024; 33:451-467. [PMID: 38758356 DOI: 10.1080/13543784.2024.2348738] [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: 12/19/2023] [Accepted: 04/24/2024] [Indexed: 05/18/2024]
Abstract
INTRODUCTION Huntington's Disease (HD) is a genetic neurodegenerative disease for which there is currently no disease-modifying treatment. One of several underlying mechanisms proposed to be involved in HD pathogenesis is inflammation; there is now accumulating evidence that the immune system may play an integral role in disease pathology and progression. As such, modulation of the immune system could be a potential therapeutic target for HD. AREAS COVERED To date, the number of trials targeting immune aspects of HD has been limited. However, targeting it, may have great advantages over other therapeutic areas, given that many drugs already exist that have actions in this system coupled to the fact that inflammation can be measured both peripherally and, to some extent, centrally using CSF and PET imaging. In this review, we look at evidence that the immune system and the newly emerging area of the microbiome are altered in HD patients, and then present and discuss clinical trials that have targeted different parts of the immune system. EXPERT OPINION We then conclude by discussing how this field might develop going forward, focusing on the role of imaging and other biomarkers to monitor central immune activation and response to novel treatments in HD.
Collapse
Affiliation(s)
- Sophie E Field
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, and MRC-WT Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Annabel J Curle
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, and MRC-WT Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Roger A Barker
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, and MRC-WT Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
20
|
Jellinger KA. Mild cognitive impairment in Huntington's disease: challenges and outlooks. J Neural Transm (Vienna) 2024; 131:289-304. [PMID: 38265518 DOI: 10.1007/s00702-024-02744-8] [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: 11/30/2023] [Accepted: 01/14/2024] [Indexed: 01/25/2024]
Abstract
Although Huntington's disease (HD) has classically been viewed as an autosomal-dominant inherited neurodegenerative motor disorder, cognitive and/or behavioral changes are predominant and often an early manifestation of disease. About 40% of individuals in the presymptomatic period of HD meet the criteria for mild cognitive impairment, later progressing to dementia. The heterogenous spectrum of cognitive decline is characterized by deficits across multiple domains, particularly executive dysfunctions, but the underlying pathogenic mechanisms are still poorly understood. Investigating the pathophysiology of cognitive changes may give insight into important and early neurodegenerative events. Multimodal imaging revealed circuit-wide gray and white matter degenerative processes in several key brain regions, affecting prefronto-striatal/cortico-basal ganglia circuits and many other functional brain networks. Studies in transgenic animal models indicated early synaptic dysfunction, deficient neurotrophic transport and other molecular changes contributing to neuronal death. Synaptopathy within the cerebral cortex, striatum and hippocampus may be particularly important in mediating cognitive and neuropsychiatric manifestations of HD, although many other neuronal systems are involved. The interaction of mutant huntingtin protein (mHTT) with tau and its implication for cognitive impairment in HD is a matter of discussion. Further neuroimaging and neuropathological studies are warranted to better elucidate early pathophysiological mechanisms and to develop validated biomarkers to detect patients' cognitive status during the early stages of the condition significantly to implement effective preventing or management strategies.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, 1150, Vienna, Austria.
| |
Collapse
|
21
|
Lotspeich SC, Ashner MC, Vazquez JE, Richardson BD, Grosser KF, Bodek BE, Garcia TP. Making Sense of Censored Covariates: Statistical Methods for Studies of Huntington's Disease. ANNUAL REVIEW OF STATISTICS AND ITS APPLICATION 2024; 11:255-277. [PMID: 38962579 PMCID: PMC11220439 DOI: 10.1146/annurev-statistics-040522-095944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
The landscape of survival analysis is constantly being revolutionized to answer biomedical challenges, most recently the statistical challenge of censored covariates rather than outcomes. There are many promising strategies to tackle censored covariates, including weighting, imputation, maximum likelihood, and Bayesian methods. Still, this is a relatively fresh area of research, different from the areas of censored outcomes (i.e., survival analysis) or missing covariates. In this review, we discuss the unique statistical challenges encountered when handling censored covariates and provide an in-depth review of existing methods designed to address those challenges. We emphasize each method's relative strengths and weaknesses, providing recommendations to help investigators pinpoint the best approach to handling censored covariates in their data.
Collapse
Affiliation(s)
- Sarah C Lotspeich
- Department of Statistical Sciences, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Marissa C Ashner
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jesus E Vazquez
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Brian D Richardson
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kyle F Grosser
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Benjamin E Bodek
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Tanya P Garcia
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
22
|
Migliore S, Bianco SD, Scocchia M, Maffi S, Busi LC, Ceccarelli C, Curcio G, Mazza T, Squitieri F. Prodromal Cognitive Changes as a Prognostic Indicator of Forthcoming Huntington's Disease Severity: A Retrospective Longitudinal Study. Mov Disord Clin Pract 2024; 11:363-372. [PMID: 38264920 PMCID: PMC10982604 DOI: 10.1002/mdc3.13975] [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: 06/06/2023] [Revised: 11/30/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND Cognitive changes in Huntington's disease (HD) precede motor manifestations. ENROLL-HD platform includes four cognitive measures of information processing speed (IPS). Our group is eager to seek clinical markers in the life stage that is as close as possible to the age of onset (ie, the so called prodromal HD phase) because this is the best time for therapeutic interventions. OBJECTIVES Our study aimed to test whether cognitive scores in prodromal ENROLL-HD mutation carriers show the potential to predict the severity of motor and behavioral changes once HD became fully manifested. METHODS From the global ENROLL-HD cohort of 21,343 participants, we first selected a premanifest Cohort#1 (ie, subjects with Total Motor Score (TMS) <10 and Diagnostic Confidence Level (DCL) <4, N = 1.222). From this cohort, we then focused on a prodromal Cohort#2 of subjects who were ascertained to phenoconvert into manifest HD at follow-up visits (ie, subjects from 6 ≤ TMS≤9 and DCL <4 to TMS≥10 and DCL = 4, n = 206). RESULTS The main results of our study showed that low IPS before phenoconversion in Cohort#2 predicted the severity of motor and behavioral manifestations. By combining the four IPS cognitive measures (eg, the Categorical Verbal Fluency Test; Stroop Color Naming Test; Stroop Word Reading; Symbol Digit Modalities Test), we generated a Composite Cognition Score (CCS). The lower the CCS score the higher the TMS and the apathy scores in the same longitudinally followed-up patients after phenoconversion. CONCLUSIONS CCS might represent a clinical instrument to predict the prognosis of mutation carriers who are close to manifesting HD.
Collapse
Affiliation(s)
- Simone Migliore
- Huntington and Rare Diseases Unit, Fondazione IRCCS Casa Sollievo Della Sofferenza HospitalSan Giovanni RotondoItaly
| | | | - Marta Scocchia
- Rare Neurological Diseases Centre (CMNR)Fondazione Italian League for Research on Huntington (LIRH)RomeItaly
| | - Sabrina Maffi
- Huntington and Rare Diseases Unit, Fondazione IRCCS Casa Sollievo Della Sofferenza HospitalSan Giovanni RotondoItaly
| | - Ludovica Camilla Busi
- Rare Neurological Diseases Centre (CMNR)Fondazione Italian League for Research on Huntington (LIRH)RomeItaly
| | - Consuelo Ceccarelli
- Rare Neurological Diseases Centre (CMNR)Fondazione Italian League for Research on Huntington (LIRH)RomeItaly
| | - Giuseppe Curcio
- Department of Biotechnological and Applied Clinical SciencesUniversity of L'AquilaL'AquilaItaly
| | - Tommaso Mazza
- Bioinformatics Unit, Fondazione IRCCS "Casa Sollievo della Sofferenza"San Giovanni RotondoItaly
| | - Ferdinando Squitieri
- Huntington and Rare Diseases Unit, Fondazione IRCCS Casa Sollievo Della Sofferenza HospitalSan Giovanni RotondoItaly
- Rare Neurological Diseases Centre (CMNR)Fondazione Italian League for Research on Huntington (LIRH)RomeItaly
| |
Collapse
|
23
|
Vidas-Guscic N, van Rijswijk J, Van Audekerke J, Jeurissen B, Nnah I, Tang H, Muñoz-Sanjuan I, Pustina D, Cachope R, Van der Linden A, Bertoglio D, Verhoye M. Diffusion MRI marks progressive alterations in fiber integrity in the zQ175DN mouse model of Huntington's disease. Neurobiol Dis 2024; 193:106438. [PMID: 38365045 DOI: 10.1016/j.nbd.2024.106438] [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: 12/04/2023] [Revised: 01/24/2024] [Accepted: 02/13/2024] [Indexed: 02/18/2024] Open
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disease affecting motor and cognitive abilities. Multiple studies have found white matter anomalies in HD-affected humans and animal models of HD. The identification of sensitive white-matter-based biomarkers in HD animal models will be important in understanding disease mechanisms and testing the efficacy of therapeutic interventions. Here we investigated the progression of white matter deficits in the knock-in zQ175DN heterozygous (HET) mouse model of HD at 3, 6 and 11 months of age (M), reflecting different states of phenotypic progression. We compared findings from traditional diffusion tensor imaging (DTI) and advanced fixel-based analysis (FBA) diffusion metrics for their sensitivity in detecting white matter anomalies in the striatum, motor cortex, and segments of the corpus callosum. FBA metrics revealed progressive and widespread reductions of fiber cross-section and fiber density in myelinated bundles of HET mice. The corpus callosum genu was the most affected structure in HET mice at 6 and 11 M based on the DTI and FBA metrics, while the striatum showed the earliest progressive differences starting at 3 M based on the FBA metrics. Overall, FBA metrics detected earlier and more prominent alterations in myelinated fiber bundles compared to the DTI metrics. Luxol fast blue staining showed no loss in myelin density, indicating that diffusion anomalies could not be explained by myelin reduction but diffusion anomalies in HET mice were accompanied by increased levels of neurofilament light chain protein at 11 M. Altogether, our findings reveal progressive alterations in myelinated fiber bundles that can be measured using diffusion MRI, representing a candidate noninvasive imaging biomarker to study phenotype progression and the efficacy of therapeutic interventions in zQ175DN mice. Moreover, our study exposed higher sensitivity of FBA than DTI metrics, suggesting a potential benefit of adopting these advanced metrics in other contexts, including biomarker development in humans.
Collapse
Affiliation(s)
- Nicholas Vidas-Guscic
- Bio-Imaging Lab, University of Antwerp, Antwerp, Belgium; μNeuro Center for Excellence, University of Antwerp, Antwerp, Belgium.
| | - Joëlle van Rijswijk
- Bio-Imaging Lab, University of Antwerp, Antwerp, Belgium; μNeuro Center for Excellence, University of Antwerp, Antwerp, Belgium
| | - Johan Van Audekerke
- Bio-Imaging Lab, University of Antwerp, Antwerp, Belgium; μNeuro Center for Excellence, University of Antwerp, Antwerp, Belgium
| | - Ben Jeurissen
- μNeuro Center for Excellence, University of Antwerp, Antwerp, Belgium; Vision Lab, University of Antwerp, Antwerp, Belgium; Lab for Equilibrium Investigations and Aerospace, University of Antwerp, Antwerp, Belgium
| | - Israel Nnah
- Charles River Laboratories, Shrewsbury, MA, United states
| | - Haiying Tang
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Inc., Princeton, NJ, United States
| | - Ignacio Muñoz-Sanjuan
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Inc., Princeton, NJ, United States
| | - Dorian Pustina
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Inc., Princeton, NJ, United States
| | - Roger Cachope
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Inc., Princeton, NJ, United States
| | - Annemie Van der Linden
- Bio-Imaging Lab, University of Antwerp, Antwerp, Belgium; μNeuro Center for Excellence, University of Antwerp, Antwerp, Belgium
| | - Daniele Bertoglio
- Bio-Imaging Lab, University of Antwerp, Antwerp, Belgium; μNeuro Center for Excellence, University of Antwerp, Antwerp, Belgium
| | - Marleen Verhoye
- Bio-Imaging Lab, University of Antwerp, Antwerp, Belgium; μNeuro Center for Excellence, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
24
|
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
|
25
|
Cheng Y, Zhang S, Shang H. Latest advances on new promising molecular-based therapeutic approaches for Huntington's disease. J Transl Int Med 2024; 12:134-147. [PMID: 38779119 PMCID: PMC11107186 DOI: 10.2478/jtim-2023-0142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024] Open
Abstract
Huntington's disease (HD) is a devastating, autosomal-dominant inherited, neurodegenerative disorder characterized by progressive motor deficits, cognitive impairments, and neuropsychiatric symptoms. It is caused by excessive cytosine-adenine-guanine (CAG) trinucleotide repeats within the huntingtin gene (HTT). Presently, therapeutic interventions capable of altering the trajectory of HD are lacking, while medications for abnormal movement and psychiatric symptoms are limited. Numerous pre-clinical and clinical studies have been conducted and are currently underway to test the efficacy of therapeutic approaches targeting some of these mechanisms with varying degrees of success. In this review, we update the latest advances on new promising molecular-based therapeutic strategies for this disorder, including DNA-targeting techniques such as zinc-finger proteins, transcription activator-like effector nucleases, and CRISPR/Cas9; post-transcriptional huntingtin-lowering approaches such as RNAi, antisense oligonucleotides, and small-molecule splicing modulators; and novel methods to clear the mHTT protein, such as proteolysis-targeting chimeras. We mainly focus on the ongoing clinical trials and the latest pre-clinical studies to explore the progress of emerging potential HD therapeutics.
Collapse
Affiliation(s)
- Yangfan Cheng
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare disease center, West China Hospital, Sichuan University, Chengdu610041, Sichuan Province, China
| | - Sirui Zhang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare disease center, West China Hospital, Sichuan University, Chengdu610041, Sichuan Province, China
| | - Huifang Shang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare disease center, West China Hospital, Sichuan University, Chengdu610041, Sichuan Province, China
| |
Collapse
|
26
|
Reilmann R, Anderson KE, Feigin A, Tabrizi SJ, Leavitt BR, Stout JC, Piccini P, Schubert R, Loupe P, Wickenberg A, Borowsky B, Rynkowski G, Volkinshtein R, Li T, Savola JM, Hayden M, Gordon MF. Safety and efficacy of laquinimod for Huntington's disease (LEGATO-HD): a multicentre, randomised, double-blind, placebo-controlled, phase 2 study. Lancet Neurol 2024; 23:243-255. [PMID: 38280392 DOI: 10.1016/s1474-4422(23)00454-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 11/02/2023] [Accepted: 11/17/2023] [Indexed: 01/29/2024]
Abstract
BACKGROUND Laquinimod modulates CNS inflammatory pathways thought to be involved in the pathology of Huntington's disease. Studies with laquinimod in transgenic rodent models of Huntington's disease suggested improvements in motor function, reduction of brain volume loss, and prolonged survival. We aimed to evaluate the safety and efficacy of laquinimod in improving motor function and reducing caudate volume loss in patients with Huntington's disease. METHODS LEGATO-HD was a multicentre, double-blind, placebo-controlled, phase 2 study done at 48 sites across ten countries (Canada, Czech Republic, Germany, Italy, Netherlands, Portugal, Russia, Spain, UK, and USA). Patients aged 21-55 years with a cytosine-adenosine-guanine (CAG) repeat length of between 36 and 49 who had symptomatic Huntington's disease with a Unified Huntington's Disease Rating Scale-Total Motor Score (UHDRS-TMS) of higher than 5 and a Total Functional Capacity score of 8 or higher were randomly assigned (1:1:1:1) by centralised interactive response technology to laquinimod 0·5 mg, 1·0 mg, or 1·5 mg, or to matching placebo, administered orally once daily over 52 weeks; people involved in the randomisation had no other role in the study. Participants, investigators, and study personnel were masked to treatment assignment. The 1·5 mg group was discontinued before recruitment was finished because of cardiovascular safety concerns in multiple sclerosis studies. The primary endpoint was change from baseline in the UHDRS-TMS and the secondary endpoint was percent change in caudate volume, both comparing the 1·0 mg group with the placebo group at week 52. Primary and secondary endpoints were assessed in the full analysis set (ie, all randomised patients who received at least one dose of study drug and had at least one post-baseline UHDRS-TMS assessment). Safety measures included adverse event frequency and severity, and clinical and laboratory examinations, and were assessed in the safety analysis set (ie, all randomised patients who received at least one dose of study drug). This trial is registered with ClinicalTrials.gov, NCT02215616, and EudraCT, 2014-000418-75, and is now complete. FINDINGS Between Oct 28, 2014, and June 19, 2018, 352 adults with Huntington's disease (179 [51%] men and 173 [49%] women; mean age 43·9 [SD 7·6] years and 340 [97%] White) were randomly assigned: 107 to laquinimod 0·5 mg, 107 to laquinimod 1·0 mg, 30 to laquinimod 1·5 mg, and 108 to matching placebo. Least squares mean change from baseline in UHDRS-TMS at week 52 was 1·98 (SE 0·83) in the laquinimod 1·0 mg group and 1·2 (0·82) in the placebo group (least squares mean difference 0·78 [95% CI -1·42 to 2·98], p=0·4853). Least squares mean change in caudate volume was 3·10% (SE 0·38) in the 1·0 mg group and 4·86% (0·38) in the placebo group (least squares mean difference -1·76% [95% CI -2·67 to -0·85]; p=0·0002). Laquinimod was well tolerated and there were no new safety findings. Serious adverse events were reported by eight (7%) patients on placebo, seven (7%) on laquinimod 0·5 mg, five (5%) on laquinimod 1·0 mg, and one (3%) on laquinimod 1·5 mg. There was one death, which occurred in the placebo group and was unrelated to treatment. The most frequent adverse events in all laquinimod dosed groups (0·5 mg, 1·0 mg, and 1·5 mg) were headache (38 [16%]), diarrhoea (24 [10%]), fall (18 [7%]), nasopharyngitis (20 [8%]), influenza (15 [6%]), vomiting (13 [5%]), arthralgia (11 [5%]), irritability (ten [4%]), fatigue (eight [3%]), and insomnia (eight [3%]). INTERPRETATION Laquinimod did not show a significant effect on motor symptoms assessed by the UHDRS-TMS, but significantly reduced caudate volume loss compared with placebo at week 52. Huntington's disease has a chronic and slowly progressive course, and this study does not address whether a longer duration of laquinimod treatment could have produced detectable and meaningful changes in the clinical assessments. FUNDING Teva Pharmaceutical Industries.
Collapse
Affiliation(s)
- Ralf Reilmann
- George Huntington Institute, Münster, Germany; Department of Clinical Radiology, University of Münster, Münster, Germany; Department of Neurodegenerative Diseases and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.
| | - Karen E Anderson
- Department of Psychiatry and Department of Neurology, Georgetown University School of Medicine, Washington, DC, USA
| | - Andrew Feigin
- New York University Langone Health, New York, NY, USA
| | - Sarah J Tabrizi
- University College London Queen Square Institute of Neurology, London, UK
| | - Blair R Leavitt
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Julie C Stout
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Paola Piccini
- Department of Brain Sciences, Imperial College London, London, UK
| | | | - Pippa Loupe
- Research and Development, Teva Pharmaceutical Industries, Petah Tikva, Israel
| | | | | | - Gail Rynkowski
- Research and Development, Teva Pharmaceutical Industries, Petah Tikva, Israel
| | - Rita Volkinshtein
- Research and Development, Teva Pharmaceutical Industries, Petah Tikva, Israel
| | - Thomas Li
- Research and Development, Teva Pharmaceutical Industries, Petah Tikva, Israel
| | | | - Michael Hayden
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada; Prilenia Therapeutics, Herzliya, Israel
| | - Mark Forrest Gordon
- Research and Development, Teva Pharmaceutical Industries, Petah Tikva, Israel
| |
Collapse
|
27
|
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
|
28
|
Knights H, Coleman A, Hobbs NZ, Tabrizi SJ, Scahill RI. Freesurfer Software Update Significantly Impacts Striatal Volumes in the Huntington's Disease Young Adult Study and Will Influence HD-ISS Staging. J Huntingtons Dis 2024; 13:77-90. [PMID: 38489194 DOI: 10.3233/jhd-231512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
Background The Huntington's Disease Integrated Staging System (HD-ISS) defined disease onset using volumetric cut-offs for caudate and putamen derived from FreeSurfer 6 (FS6). The impact of the latest software update (FS7) on volumes remains unknown. The Huntington's Disease Young Adult Study (HD-YAS) is appropriately positioned to explore differences in FS bias when detecting early atrophy. Objective Explore the relationships and differences between raw caudate and putamen volumes, calculated total intracranial volumes (cTICV), and adjusted caudate and putamen volumes, derived from FS6 and FS7, in HD-YAS. Methods Images from 123 participants were segmented and quality controlled. Relationships and differences between volumes were explored using intraclass correlation (ICC) and Bland-Altman analysis. Results Across the whole cohort, ICC for raw caudate and putamen was 0.99, cTICV 0.93, adjusted caudate 0.87, and adjusted putamen 0.86 (all p < 0.0005). Compared to FS6, FS7 calculated: i) larger raw caudate (+0.8%, p < 0.00005) and putamen (+1.9%, p < 0.00005), with greater difference for larger volumes; and ii) smaller cTICV (-5.1%, p < 0.00005), with greater difference for smaller volumes. The systematic and proportional difference in cTICV was greater than raw volumes. When raw volumes were adjusted for cTICV, these effects compounded (adjusted caudate +7.0%, p < 0.00005; adjusted putamen +8.2%, p < 0.00005), with greater difference for larger volumes. Conclusions As new software is released, it is critical that biases are explored since differences have the potential to significantly alter the findings of HD trials. Until conversion factors are defined, the HD-ISS must be applied using FS6. This should be incorporated into the HD-ISS online calculator.
Collapse
Affiliation(s)
- Harry Knights
- Department of Neurodegenerative Disease, Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Annabelle Coleman
- Department of Neurodegenerative Disease, Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Nicola Z Hobbs
- Department of Neurodegenerative Disease, Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Sarah J Tabrizi
- Department of Neurodegenerative Disease, Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Rachael I Scahill
- Department of Neurodegenerative Disease, Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
29
|
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
|
30
|
Hamilton J, Farag M, Tabrizi SJ. Complementary insights into corticostriatal synapse loss and cognition in Huntington's disease. Cell Rep Med 2023; 4:101314. [PMID: 38118416 PMCID: PMC10772369 DOI: 10.1016/j.xcrm.2023.101314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 12/22/2023]
Abstract
In a recent study, Wilton and colleagues link activation of the classical complement pathway with corticostriatal synapse loss and cognitive decline in Huntington's disease.1.
Collapse
Affiliation(s)
- Joseph Hamilton
- UCL Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Mena Farag
- UCL Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Sarah J Tabrizi
- UCL Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK; UK Dementia Research Institute, University College London, London WC1N 3BG, UK.
| |
Collapse
|
31
|
Rajagopal S, Donaldson J, Flower M, Hensman Moss DJ, Tabrizi SJ. Genetic modifiers of repeat expansion disorders. Emerg Top Life Sci 2023; 7:325-337. [PMID: 37861103 PMCID: PMC10754329 DOI: 10.1042/etls20230015] [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: 05/19/2023] [Revised: 09/20/2023] [Accepted: 10/09/2023] [Indexed: 10/21/2023]
Abstract
Repeat expansion disorders (REDs) are monogenic diseases caused by a sequence of repetitive DNA expanding above a pathogenic threshold. A common feature of the REDs is a strong genotype-phenotype correlation in which a major determinant of age at onset (AAO) and disease progression is the length of the inherited repeat tract. Over a disease-gene carrier's life, the length of the repeat can expand in somatic cells, through the process of somatic expansion which is hypothesised to drive disease progression. Despite being monogenic, individual REDs are phenotypically variable, and exploring what genetic modifying factors drive this phenotypic variability has illuminated key pathogenic mechanisms that are common to this group of diseases. Disease phenotypes are affected by the cognate gene in which the expansion is found, the location of the repeat sequence in coding or non-coding regions and by the presence of repeat sequence interruptions. Human genetic data, mouse models and in vitro models have implicated the disease-modifying effect of DNA repair pathways via the mechanisms of somatic mutation of the repeat tract. As such, developing an understanding of these pathways in the context of expanded repeats could lead to future disease-modifying therapies for REDs.
Collapse
Affiliation(s)
- Sangeerthana Rajagopal
- UCL Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, U.K
- UK Dementia Research Institute, University College London, London WCC1N 3BG, U.K
| | - Jasmine Donaldson
- UCL Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, U.K
- UK Dementia Research Institute, University College London, London WCC1N 3BG, U.K
| | - Michael Flower
- UCL Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, U.K
- UK Dementia Research Institute, University College London, London WCC1N 3BG, U.K
| | - Davina J Hensman Moss
- UCL Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, U.K
- UK Dementia Research Institute, University College London, London WCC1N 3BG, U.K
- St George's University of London, London SW17 0RE, U.K
| | - Sarah J Tabrizi
- UCL Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, U.K
- UK Dementia Research Institute, University College London, London WCC1N 3BG, U.K
| |
Collapse
|
32
|
Thompson LM, Orr HT. HD and SCA1: Tales from two 30-year journeys since gene discovery. Neuron 2023; 111:3517-3530. [PMID: 37863037 PMCID: PMC10842341 DOI: 10.1016/j.neuron.2023.09.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 07/21/2023] [Accepted: 09/26/2023] [Indexed: 10/22/2023]
Abstract
One of the more transformative findings in human genetics was the discovery that the expansion of unstable nucleotide repeats underlies a group of inherited neurological diseases. A subset of these unstable repeat neurodegenerative diseases is due to the expansion of a CAG trinucleotide repeat encoding a stretch of glutamines, i.e., the polyglutamine (polyQ) repeat neurodegenerative diseases. Among the CAG/polyQ repeat diseases are Huntington's disease (HD) and spinocerebellar ataxia type 1 (SCA1), in which the expansions are within widely expressed proteins. Although both HD and SCA1 are autosomal dominantly inherited, and both typically cause mid- to late-life-onset movement disorders with cognitive decline, they each are characterized by distinct clinical characteristics and predominant sites of neuropathology. Importantly, the respective affected proteins, Huntingtin (HTT, HD) and Ataxin 1 (ATXN1, SCA1), have unique functions and biological properties. Here, we review HD and SCA1 with a focus on how their disease-specific and shared features may provide informative insights.
Collapse
Affiliation(s)
- Leslie M Thompson
- Department of Psychiatry and Human Behavior, Department of Neurobiology and Behavior, Department of Biological Chemistry, Institute of Memory Impairments and Neurological Disorders, Sue and Bill Gross Stem Cell Center, University of California Irvine, Irvine, CA 92697, USA
| | - Harry T Orr
- Department of Laboratory Medicine and Pathology, Institute for Translational Neuroscience, University of Minnesota, Minneapolis and Saint Paul, MN 55455, USA.
| |
Collapse
|
33
|
Estevez-Fraga C, Altmann A, Parker CS, Scahill RI, Costa B, Chen Z, Manzoni C, Zarkali A, Durr A, Roos RAC, Landwehrmeyer B, Leavitt BR, Rees G, Tabrizi SJ, McColgan P. Genetic topography and cortical cell loss in Huntington's disease link development and neurodegeneration. Brain 2023; 146:4532-4546. [PMID: 37587097 PMCID: PMC10629790 DOI: 10.1093/brain/awad275] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 07/12/2023] [Accepted: 07/28/2023] [Indexed: 08/18/2023] Open
Abstract
Cortical cell loss is a core feature of Huntington's disease (HD), beginning many years before clinical motor diagnosis, during the premanifest stage. However, it is unclear how genetic topography relates to cortical cell loss. Here, we explore the biological processes and cell types underlying this relationship and validate these using cell-specific post-mortem data. Eighty premanifest participants on average 15 years from disease onset and 71 controls were included. Using volumetric and diffusion MRI we extracted HD-specific whole brain maps where lower grey matter volume and higher grey matter mean diffusivity, relative to controls, were used as proxies of cortical cell loss. These maps were combined with gene expression data from the Allen Human Brain Atlas (AHBA) to investigate the biological processes relating genetic topography and cortical cell loss. Cortical cell loss was positively correlated with the expression of developmental genes (i.e. higher expression correlated with greater atrophy and increased diffusivity) and negatively correlated with the expression of synaptic and metabolic genes that have been implicated in neurodegeneration. These findings were consistent for diffusion MRI and volumetric HD-specific brain maps. As wild-type huntingtin is known to play a role in neurodevelopment, we explored the association between wild-type huntingtin (HTT) expression and developmental gene expression across the AHBA. Co-expression network analyses in 134 human brains free of neurodegenerative disorders were also performed. HTT expression was correlated with the expression of genes involved in neurodevelopment while co-expression network analyses also revealed that HTT expression was associated with developmental biological processes. Expression weighted cell-type enrichment (EWCE) analyses were used to explore which specific cell types were associated with HD cortical cell loss and these associations were validated using cell specific single nucleus RNAseq (snRNAseq) data from post-mortem HD brains. The developmental transcriptomic profile of cortical cell loss in preHD was enriched in astrocytes and endothelial cells, while the neurodegenerative transcriptomic profile was enriched for neuronal and microglial cells. Astrocyte-specific genes differentially expressed in HD post-mortem brains relative to controls using snRNAseq were enriched in the developmental transcriptomic profile, while neuronal and microglial-specific genes were enriched in the neurodegenerative transcriptomic profile. Our findings suggest that cortical cell loss in preHD may arise from dual pathological processes, emerging as a consequence of neurodevelopmental changes, at the beginning of life, followed by neurodegeneration in adulthood, targeting areas with reduced expression of synaptic and metabolic genes. These events result in age-related cell death across multiple brain cell types.
Collapse
Affiliation(s)
- Carlos Estevez-Fraga
- Department of Neurodegenerative Disease, University College London, London WC1B 5EH, UK
| | - Andre Altmann
- Centre for Medical Image Computing, University College London, London WC1V 6LJ, UK
| | - Christopher S Parker
- Centre for Medical Image Computing, University College London, London WC1V 6LJ, UK
| | - Rachael I Scahill
- Department of Neurodegenerative Disease, University College London, London WC1B 5EH, UK
| | - Beatrice Costa
- Department of Neurodegenerative Disease, University College London, London WC1B 5EH, UK
- Gladstone Institutes, San Francisco, CA 94158, USA
| | - Zhongbo Chen
- Department of Neurodegenerative Disease, University College London, London WC1B 5EH, UK
| | - Claudia Manzoni
- School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Angeliki Zarkali
- Dementia Research Centre, University College London, London WC1N 3AR, UK
| | - Alexandra Durr
- Sorbonne Université, Paris Brain Institute (ICM), AP-HP, Inserm, CNRS, Paris 75013, France
| | - Raymund A C Roos
- Department of Neurology, Leiden University Medical Centre, Leiden 2333, The Netherlands
| | | | - Blair R Leavitt
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, University of British Columbia, Vancouver BC V5Z 4H4Canada
- Division of Neurology, Department of Medicine, University of British Columbia Hospital, Vancouver BC V6T 2B5, Canada
| | - Geraint Rees
- Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of Neurology, University College London, London WC1N 3AR, UK
| | - Sarah J Tabrizi
- Department of Neurodegenerative Disease, University College London, London WC1B 5EH, UK
| | - Peter McColgan
- Department of Neurodegenerative Disease, University College London, London WC1B 5EH, UK
| |
Collapse
|
34
|
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: 34] [Impact Index Per Article: 17.0] [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
|
35
|
Mühlbäck A, Mana J, Wallner M, Frank W, Lindenberg KS, Hoffmann R, Klempířová O, Klempíř J, Landwehrmeyer GB, Bezdicek O. Establishing normative data for the evaluation of cognitive performance in Huntington's disease considering the impact of gender, age, language, and education. J Neurol 2023; 270:4903-4913. [PMID: 37347292 PMCID: PMC10511566 DOI: 10.1007/s00415-023-11823-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: 02/03/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/23/2023]
Abstract
BACKGROUND A declining cognitive performance is a hallmark of Huntington's disease (HD). The neuropsychological battery of the Unified HD Rating Scale (UHDRS'99) is commonly used for assessing cognition. However, there is a need to identify and minimize the impact of confounding factors, such as language, gender, age, and education level on cognitive decline. OBJECTIVES Aim is to provide appropriate, normative data to allow clinicians to identify disease-associated cognitive decline in diverse HD populations by compensating for the impact of confounding factors METHODS: Sample data, N = 3267 (60.5% females; mean age of 46.9 years (SD = 14.61, range 18-86) of healthy controls were used to create a normative dataset. For each neuropsychological test, a Bayesian generalized additive model with age, education, gender, and language as predictors was constructed to appropriately stratify the normative dataset. RESULTS With advancing age, there was a non-linear decline in cognitive performance. In addition, performance was dependent on educational levels and language in all tests. Gender had a more limited impact. Standardized scores have been calculated to ease the interpretation of an individual's test outcome. A web-based online tool has been created to provide free access to normative data. CONCLUSION For defined neuropsychological tests, the impact of gender, age, education, and language as factors confounding disease-associated cognitive decline can be minimized at the level of a single patient examination.
Collapse
Affiliation(s)
- Alžbeta Mühlbäck
- Department of Neurology, Ulm University, Oberer Eselsberg 45, 89081, Ulm, Germany.
- Huntington Center South, kbo-Isar-Amper-Klinikum, Taufkirchen, Germany.
- Department of Neurology and Center of Clinical Neuroscience, 1st Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia.
| | - Josef Mana
- Department of Neurology and Center of Clinical Neuroscience, 1st Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | | | - Wiebke Frank
- Department of Neurology, Ulm University, Oberer Eselsberg 45, 89081, Ulm, Germany
| | - Katrin S Lindenberg
- Department of Neurology, Ulm University, Oberer Eselsberg 45, 89081, Ulm, Germany
| | - Rainer Hoffmann
- Huntington Center South, kbo-Isar-Amper-Klinikum, Taufkirchen, Germany
| | - Olga Klempířová
- Department of Neurology and Center of Clinical Neuroscience, 1st Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Jiří Klempíř
- Department of Neurology and Center of Clinical Neuroscience, 1st Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | | | - Ondrej Bezdicek
- Department of Neurology and Center of Clinical Neuroscience, 1st Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| |
Collapse
|
36
|
Vishweswaraiah S, Yilmaz A, Saiyed N, Khalid A, Koladiya PR, Pan X, Macias S, Robinson AC, Mann D, Green BD, Kerševičiūte I, Gordevičius J, Radhakrishna U, Graham SF. Integrative Analysis Unveils the Correlation of Aminoacyl-tRNA Biosynthesis Metabolites with the Methylation of the SEPSECS Gene in Huntington's Disease Brain Tissue. Genes (Basel) 2023; 14:1752. [PMID: 37761892 PMCID: PMC10530570 DOI: 10.3390/genes14091752] [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: 08/02/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
The impact of environmental factors on epigenetic changes is well established, and cellular function is determined not only by the genome but also by interacting partners such as metabolites. Given the significant impact of metabolism on disease progression, exploring the interaction between the metabolome and epigenome may offer new insights into Huntington's disease (HD) diagnosis and treatment. Using fourteen post-mortem HD cases and fourteen control subjects, we performed metabolomic profiling of human postmortem brain tissue (striatum and frontal lobe), and we performed DNA methylome profiling using the same frontal lobe tissue. Along with finding several perturbed metabolites and differentially methylated loci, Aminoacyl-tRNA biosynthesis (adj p-value = 0.0098) was the most significantly perturbed metabolic pathway with which two CpGs of the SEPSECS gene were correlated. This study improves our understanding of molecular biomarker connections and, importantly, increases our knowledge of metabolic alterations driving HD progression.
Collapse
Affiliation(s)
- Sangeetha Vishweswaraiah
- Department of Obstetrics and Gynecology, Corewell Health William Beaumont University Hospital, 3601 W. 13 Mile Road, Royal Oak, MI 48073, USA; (S.V.); (U.R.)
| | - Ali Yilmaz
- Metabolomics Department, Corewell Health Research Institute, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA; (A.Y.); (N.S.); (A.K.); (P.R.K.)
| | - Nazia Saiyed
- Metabolomics Department, Corewell Health Research Institute, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA; (A.Y.); (N.S.); (A.K.); (P.R.K.)
| | - Abdullah Khalid
- Metabolomics Department, Corewell Health Research Institute, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA; (A.Y.); (N.S.); (A.K.); (P.R.K.)
| | - Purvesh R. Koladiya
- Metabolomics Department, Corewell Health Research Institute, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA; (A.Y.); (N.S.); (A.K.); (P.R.K.)
| | - Xiaobei Pan
- Advanced Asset Technology Centre, Institute for Global Food Security, Queen’s University Belfast, Belfast BT9 5DL, UK; (X.P.); (S.M.); (B.D.G.)
| | - Shirin Macias
- Advanced Asset Technology Centre, Institute for Global Food Security, Queen’s University Belfast, Belfast BT9 5DL, UK; (X.P.); (S.M.); (B.D.G.)
| | - Andrew C. Robinson
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Division of Neuroscience, The University of Manchester, Salford Royal Hospital, Salford M6 8HD, UK; (A.C.R.); (D.M.)
| | - David Mann
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Division of Neuroscience, The University of Manchester, Salford Royal Hospital, Salford M6 8HD, UK; (A.C.R.); (D.M.)
| | - Brian D. Green
- Advanced Asset Technology Centre, Institute for Global Food Security, Queen’s University Belfast, Belfast BT9 5DL, UK; (X.P.); (S.M.); (B.D.G.)
| | - Ieva Kerševičiūte
- VUGENE, LLC, 625 Kenmoor Ave Suite 301 PMB 96578, Grand Rapids, MI 49546, USA; (I.K.); (J.G.)
| | - Juozas Gordevičius
- VUGENE, LLC, 625 Kenmoor Ave Suite 301 PMB 96578, Grand Rapids, MI 49546, USA; (I.K.); (J.G.)
| | - Uppala Radhakrishna
- Department of Obstetrics and Gynecology, Corewell Health William Beaumont University Hospital, 3601 W. 13 Mile Road, Royal Oak, MI 48073, USA; (S.V.); (U.R.)
| | - Stewart F. Graham
- Department of Obstetrics and Gynecology, Corewell Health William Beaumont University Hospital, 3601 W. 13 Mile Road, Royal Oak, MI 48073, USA; (S.V.); (U.R.)
- Metabolomics Department, Corewell Health Research Institute, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA; (A.Y.); (N.S.); (A.K.); (P.R.K.)
- Department of Obstetrics and Gynecology, Oakland University-William Beaumont School of Medicine, Rochester, MI 48309, USA
| |
Collapse
|
37
|
Liu CF, Younes L, Tong XJ, Hinkle JT, Wang M, Phatak S, Xu X, Bu X, Looi V, Bang J, Tabrizi SJ, Scahill RI, Paulsen JS, Georgiou-Karistianis N, Faria AV, Miller MI, Ratnanather JT, Ross CA. Longitudinal imaging highlights preferential basal ganglia circuit atrophy in Huntington's disease. Brain Commun 2023; 5:fcad214. [PMID: 37744022 PMCID: PMC10516592 DOI: 10.1093/braincomms/fcad214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/09/2023] [Accepted: 08/17/2023] [Indexed: 09/26/2023] Open
Abstract
Huntington's disease is caused by a CAG repeat expansion in the Huntingtin gene (HTT), coding for polyglutamine in the Huntingtin protein, with longer CAG repeats causing earlier age of onset. The variable 'Age' × ('CAG'-L), where 'Age' is the current age of the individual, 'CAG' is the repeat length and L is a constant (reflecting an approximation of the threshold), termed the 'CAG Age Product' (CAP) enables the consideration of many individuals with different CAG repeat expansions at the same time for analysis of any variable and graphing using the CAG Age Product score as the X axis. Structural MRI studies have showed that progressive striatal atrophy begins many years prior to the onset of diagnosable motor Huntington's disease, confirmed by longitudinal multicentre studies on three continents, including PREDICT-HD, TRACK-HD and IMAGE-HD. However, previous studies have not clarified the relationship between striatal atrophy, atrophy of other basal ganglia structures, and atrophy of other brain regions. The present study has analysed all three longitudinal datasets together using a single image segmentation algorithm and combining data from a large number of subjects across a range of CAG Age Product score. In addition, we have used a strategy of normalizing regional atrophy to atrophy of the whole brain, in order to determine which regions may undergo preferential degeneration. This made possible the detailed characterization of regional brain atrophy in relation to CAG Age Product score. There is dramatic selective atrophy of regions involved in the basal ganglia circuit-caudate, putamen, nucleus accumbens, globus pallidus and substantia nigra. Most other regions of the brain appear to have slower but steady degeneration. These results support (but certainly do not prove) the hypothesis of circuit-based spread of pathology in Huntington's disease, possibly due to spread of mutant Htt protein, though other connection-based mechanisms are possible. Therapeutic targets related to prion-like spread of pathology or other mechanisms may be suggested. In addition, they have implications for current neurosurgical therapeutic approaches, since delivery of therapeutic agents solely to the caudate and putamen may miss other structures affected early, such as nucleus accumbens and output nuclei of the striatum, the substantia nigra and the globus pallidus.
Collapse
Affiliation(s)
- Chin-Fu Liu
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Laurent Younes
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Xiao J Tong
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore MD 21287, USA
| | - Jared T Hinkle
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Medical Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Maggie Wang
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sanika Phatak
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Xin Xu
- Division of Magnetic Resonance, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Xuan Bu
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Vivian Looi
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jee Bang
- Division of Neurobiology, Department of Psychiatry, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sarah J Tabrizi
- HD Research Centre, University College London Queen Square Institute of Neurology, UCL, London, UK
| | - Rachael I Scahill
- HD Research Centre, University College London Queen Square Institute of Neurology, UCL, London, UK
| | - Jane S Paulsen
- Department of Neurology, University of Wisconsin, Madison, WI 53705, USA
| | - Nellie Georgiou-Karistianis
- School of Psychological Sciences and The Turner Institute for Brain and Mental Health, Monash University, Melbourne, Victoria 3800, Australia
| | - Andreia V Faria
- Division of Magnetic Resonance, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Michael I Miller
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - J Tilak Ratnanather
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Christopher A Ross
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore MD 21287, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Division of Neurobiology, Department of Psychiatry, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
38
|
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
|
39
|
Wijeratne PA, Eshaghi A, Scotton WJ, Kohli M, Aksman L, Oxtoby NP, Pustina D, Warner JH, Paulsen JS, Scahill RI, Sampaio C, Tabrizi SJ, Alexander DC. The temporal event-based model: Learning event timelines in progressive diseases. IMAGING NEUROSCIENCE (CAMBRIDGE, MASS.) 2023; 1:1-19. [PMID: 37719837 PMCID: PMC10503481 DOI: 10.1162/imag_a_00010] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 07/15/2023] [Indexed: 09/19/2023]
Abstract
Timelines of events, such as symptom appearance or a change in biomarker value, provide powerful signatures that characterise progressive diseases. Understanding and predicting the timing of events is important for clinical trials targeting individuals early in the disease course when putative treatments are likely to have the strongest effect. However, previous models of disease progression cannot estimate the time between events and provide only an ordering in which they change. Here, we introduce the temporal event-based model (TEBM), a new probabilistic model for inferring timelines of biomarker events from sparse and irregularly sampled datasets. We demonstrate the power of the TEBM in two neurodegenerative conditions: Alzheimer's disease (AD) and Huntington's disease (HD). In both diseases, the TEBM not only recapitulates current understanding of event orderings but also provides unique new ranges of timescales between consecutive events. We reproduce and validate these findings using external datasets in both diseases. We also demonstrate that the TEBM improves over current models; provides unique stratification capabilities; and enriches simulated clinical trials to achieve a power of 80 % with less than half the cohort size compared with random selection. The application of the TEBM naturally extends to a wide range of progressive conditions.
Collapse
Affiliation(s)
- Peter A. Wijeratne
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
- Department of Informatics, University of Sussex, Brighton, United Kingdom
| | - Arman Eshaghi
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, University College London, London, United Kingdom
| | - William J. Scotton
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London, London, United Kingdom
| | - Maitrei Kohli
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
| | - Leon Aksman
- Keck School of Medicine, University of Southern California, Los Angeles, California, United States
| | - Neil P. Oxtoby
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
| | - Dorian Pustina
- CHDI Management/CHDI Foundation, Princeton, New Jersey, United States
| | - John H. Warner
- CHDI Management/CHDI Foundation, Princeton, New Jersey, United States
| | - Jane S. Paulsen
- Departments of Neurology and Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Rachael I. Scahill
- Huntington’s Disease Centre, Department of Neurodegenerative Disease, University College London, Queen Square, London, United Kingdom
| | - Cristina Sampaio
- CHDI Management/CHDI Foundation, Princeton, New Jersey, United States
| | - Sarah J. Tabrizi
- Huntington’s Disease Centre, Department of Neurodegenerative Disease, University College London, Queen Square, London, United Kingdom
| | - Daniel C. Alexander
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
| |
Collapse
|
40
|
van de Zande NA, Bulk M, Najac C, van der Weerd L, de Bresser J, Lewerenz J, Ronen I, de Bot ST. Study protocol of IMAGINE-HD: Imaging iron accumulation and neuroinflammation with 7T-MRI + CSF in Huntington's disease. Neuroimage Clin 2023; 39:103450. [PMID: 37327706 PMCID: PMC10509525 DOI: 10.1016/j.nicl.2023.103450] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/18/2023]
Abstract
INTRODUCTION Strong evidence suggests a significant role for iron accumulation in the brain in addition to the well-documented neurodegenerative aspects of Huntington's disease (HD). The putative mechanisms by which iron is linked to the HD pathogenesis are multiple, including oxidative stress, ferroptosis and neuroinflammation. However, no previous study in a neurodegenerative disease has linked the observed increase of brain iron accumulation as measured by MRI with well-established cerebrospinal fluid (CSF) and blood biomarkers for iron accumulation, or with associated processes such as neuroinflammation. This study is designed to link quantitative data from iron levels and neuroinflammation metabolites obtained from 7T MRI of HD patients, with specific and well-known clinical biofluid markers for iron accumulation, neurodegeneration and neuroinflammation. Biofluid markers will provide quantitative measures of overall iron accumulation, neurodegeneration and neuroinflammation, while MRI measurements on the other hand will provide quantitative spatial information on brain pathology, neuroinflammation and brain iron accumulation, which will be linked to clinical outcome measures. METHODS This is an observational cross-sectional study, IMAGINE-HD, in HD gene expansion carriers and healthy controls. We include premanifest HD gene expansion carriers and patients with manifest HD in an early or moderate stage. The study includes a 7T MRI scan of the brain, clinical evaluation, motor, functional, and neuropsychological assessments, and sampling of CSF and blood for the detection of iron, neurodegenerative and inflammatory markers. Quantitative Susceptibility Maps will be reconstructed using T2* weighted images to quantify brain iron levels and Magnetic Resonance Spectroscopy will be used to obtain information about neuroinflammation by measuring cell-specific intracellular metabolites' level and diffusion. Age and sex matched healthy subjects are included as a control group. DISCUSSION Results from this study will provide an important basis for the evaluation of brain iron levels and neuroinflammation metabolites as an imaging biomarker for disease stage in HD and their relationship with the salient pathomechanisms of the disease on the one hand, and with clinical outcome on the other.
Collapse
Affiliation(s)
| | - Marjolein Bulk
- C.J. Gorter MRI Center, Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands.
| | - Chloé Najac
- C.J. Gorter MRI Center, Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands.
| | - Louise van der Weerd
- Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands; Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands.
| | - Jeroen de Bresser
- Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands.
| | - Jan Lewerenz
- Department of Neurology, University of Ulm, Ulm, Baden-Württemberg, Germany.
| | - Itamar Ronen
- Clinical Imaging Sciences Centre, Brighton and Sussex Medical School, Brighton, United Kingdom.
| | | |
Collapse
|
41
|
Minikel EV, Vallabh SM. Where have prions been all our lives? Brain 2023; 146:2206-2207. [PMID: 37161596 PMCID: PMC10232229 DOI: 10.1093/brain/awad143] [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: 04/26/2023] [Accepted: 04/26/2023] [Indexed: 05/11/2023] Open
Abstract
This scientific commentary refers to ‘Seed amplification and neurodegeneration marker trajectories in individuals at risk of prion disease’ by Mok et al. (https://doi.org/10.1093/brain/awad101).
Collapse
Affiliation(s)
- Eric Vallabh Minikel
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Prion Alliance, Cambridge, MA 02139, USA
- Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Sonia M Vallabh
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Prion Alliance, Cambridge, MA 02139, USA
- Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
42
|
Li XY, Xie JJ, Wang JH, Bao YF, Dong Y, Gao B, Shen T, Huang PY, Ying HC, Xu H, Roe AW, Lai HY, Wu ZY. Perivascular spaces relate to the course and cognition of Huntington's disease. Transl Neurodegener 2023; 12:30. [PMID: 37287074 DOI: 10.1186/s40035-023-00359-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/25/2023] [Indexed: 06/09/2023] Open
Affiliation(s)
- Xiao-Yan Li
- Department of Medical Genetics and Center for Rare Diseases, Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Juan-Juan Xie
- Department of Medical Genetics and Center for Rare Diseases, Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
- Interdisciplinary Institute of Neuroscience and Technology, College of Biomedical Engineering and Instrument Science, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China
| | - Jin-Hong Wang
- College of Computer Science and Technology, Zhejiang University, Hangzhou, China
| | - Yu-Feng Bao
- Department of Medical Genetics and Center for Rare Diseases, Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Dong
- Department of Medical Genetics and Center for Rare Diseases, Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Bin Gao
- Department of Medical Genetics and Center for Rare Diseases, Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Ting Shen
- Interdisciplinary Institute of Neuroscience and Technology, College of Biomedical Engineering and Instrument Science, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China
| | - Pei-Yu Huang
- Department of Radiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hao-Chao Ying
- School of Public Health, Zhejiang University, Hangzhou, China
| | - Han Xu
- Department of Medical Genetics and Center for Rare Diseases, Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Anna Wang Roe
- Interdisciplinary Institute of Neuroscience and Technology, College of Biomedical Engineering and Instrument Science, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China.
- MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China.
| | - Hsin-Yi Lai
- Department of Medical Genetics and Center for Rare Diseases, Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China.
- Interdisciplinary Institute of Neuroscience and Technology, College of Biomedical Engineering and Instrument Science, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China.
- MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China.
| | - Zhi-Ying Wu
- Department of Medical Genetics and Center for Rare Diseases, Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China.
- MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China.
- CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai, China.
| |
Collapse
|
43
|
Li XY, Bao YF, Xie JJ, Gao B, Qian SX, Dong Y, Wu ZY. Application Value of Serum Neurofilament Light Protein for Disease Staging in Huntington's Disease. Mov Disord 2023. [PMID: 37148558 DOI: 10.1002/mds.29430] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/06/2023] [Accepted: 04/18/2023] [Indexed: 05/08/2023] Open
Abstract
BACKGROUND Neurofilament light protein (NfL) has been proven to be a sensitive biomarker for Huntington's disease (HD). However, these studies did not include HD patients at advanced stages or with larger CAG repeats (>50), leading to a knowledge gap of the characteristics of NfL. METHODS Serum NfL (sNfL) levels were quantified using an ultrasensitive immunoassay. Participants were assessed by clinical scales and 7.0 T magnetic resonance imaging. Longitudinal samples and clinical data were obtained. RESULTS Baseline samples were available from 110 controls, 90 premanifest HD (pre-HD) and 137 HD individuals. We found levels of sNfL significantly increased in HD compared to pre-HD and controls (both P < 0.0001). The increase rates of sNfL were differed by CAG repeat lengths. However, there was no difference in sNfL levels in manifest HD from early to late stages. In addition, sNfL levels were associated with cognitive measures in pre-HD and manifest HD group, respectively. The increased levels of sNfL were also closely related to microstructural changes in white matter. In the longitudinal analysis, baseline sNfL did not correlate with subsequent clinical function decline. Random forest analysis revealed that sNfL had good power for predicting disease onset. CONCLUSIONS Although sNfL levels are independent of disease stages in manifest HD, it is still an optimal indicator for predicting disease onset and has potential use as a surrogate biomarker of treatment effect in clinical trials. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Xiao-Yan Li
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu-Feng Bao
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Juan-Juan Xie
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Bin Gao
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Shu-Xia Qian
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Dong
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhi-Ying Wu
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai, China
| |
Collapse
|
44
|
Bräuer S, Falkenburger B. [Gene Therapy for Huntington Disease]. FORTSCHRITTE DER NEUROLOGIE-PSYCHIATRIE 2023; 91:141-146. [PMID: 37040787 PMCID: PMC10089766 DOI: 10.1055/a-2042-2338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Englisch: Being one of the most common genetic neurodegenerative disease, Huntington's disease has been a model disease - also for gene therapy. Among the various options, the development of antisense oligonucleotides is the most advanced. Further options at the RNA level include micro-RNAs and modulators of RNA processing (splicing), at the DNA level zinc finger proteins. Several products are in clinical trials. These differ in their mode of application and in the extent of systemic availability. Another important difference between therapeutic strategies could be whether all forms of the huntingtin protein are targeted in the same extent, or whether a therapy preferentially targets particular toxic forms such as the exon1 protein. The results of the recently terminated GENERATION HD1 trial were somewhat sobering, most likely due to the side effect-related hydrocephalus. Therefore they represent just one step towards the development of an effective gene therapy against Huntington's disease.
Collapse
Affiliation(s)
- Stefan Bräuer
- Klinik und Poliklinik für Neurologie, Universitätsklinikum an der TU Dresden, Dresden, Germany
| | - Björn Falkenburger
- Klinik und Poliklinik für Neurologie, Universitätsklinikum an der TU Dresden, Dresden, Germany
| |
Collapse
|
45
|
Lin TW, Chang JK, Wu YR, Sun TH, Cheng YY, Ren CT, Pan MH, Wu JL, Chang KH, Yang HI, Chen CM, Wu CY, Chen YR. Ganglioside-focused Glycan Array Reveals Abnormal Anti-GD1b Auto-antibody in Plasma of Preclinical Huntington's Disease. Mol Neurobiol 2023; 60:3873-3882. [PMID: 36976478 DOI: 10.1007/s12035-023-03307-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 03/08/2023] [Indexed: 03/29/2023]
Abstract
Huntington's disease (HD) is a progressive and devastating neurodegenerative disease marked by inheritable CAG nucleotide expansion. For offspring of HD patients carrying abnormal CAG expansion, biomarkers that predict disease onset are crucially important but still lacking. Alteration of brain ganglioside patterns has been observed in the pathology of patients carrying HD. Here, by using a novel and sensitive ganglioside-focused glycan array, we examined the potential of anti-glycan auto-antibodies for HD. In this study, we collected plasma from 97 participants including 42 control (NC), 16 pre-manifest HD (pre-HD), and 39 HD cases and measured the anti-glycan auto-antibodies by a novel ganglioside-focused glycan array. The association between plasma anti-glycan auto-antibodies and disease progression was analyzed using univariate and multivariate logistic regression. The disease-predictive capacity of anti-glycan auto-antibodies was further investigated by receiver operating characteristic (ROC) analysis. We found that anti-glycan auto-antibodies were generally higher in the pre-HD group when compared to the NC and HD groups. Specifically, anti-GD1b auto-antibody demonstrated the potential for distinguishing between pre-HD and control groups. Moreover, in combination with age and the number of CAG repeat, the level of anti-GD1b antibody showed excellent predictability with an area under the ROC curve (AUC) of 0.95 to discriminate between pre-HD carriers and HD patients. With glycan array technology, this study demonstrated abnormal auto-antibody responses that showed temporal changes from pre-HD to HD.
Collapse
Affiliation(s)
- Tien-Wei Lin
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Jung-Kai Chang
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Yih-Ru Wu
- Department of Neurology, Linkou Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tsung-Hsien Sun
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Yang-Yu Cheng
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Chien-Tai Ren
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Mei-Hung Pan
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Jin-Lin Wu
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
- Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung, Taiwan
| | - Kuo-Hsuan Chang
- Department of Neurology, Linkou Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hwai-I Yang
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Linkou Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Chung-Yi Wu
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan.
| | - Yun-Ru Chen
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan.
| |
Collapse
|
46
|
Stoebner ZA, Hett K, Lyu I, Johnson H, Paulsen JS, Long JD, Oguz I. Comprehensive shape analysis of the cortex in Huntington's disease. Hum Brain Mapp 2023; 44:1417-1431. [PMID: 36409662 PMCID: PMC9921229 DOI: 10.1002/hbm.26125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 09/15/2022] [Accepted: 09/28/2022] [Indexed: 11/22/2022] Open
Abstract
The striatum has traditionally been the focus of Huntington's disease research due to the primary insult to this region and its central role in motor symptoms. Beyond the striatum, evidence of cortical alterations caused by Huntington's disease has surfaced. However, findings are not coherent between studies which have used cortical thickness for Huntington's disease since it is the well-established cortical metric of interest in other diseases. In this study, we propose a more comprehensive approach to cortical morphology in Huntington's disease using cortical thickness, sulcal depth, and local gyrification index. Our results show consistency with prior findings in cortical thickness, including its limitations. Our comparison between cortical thickness and local gyrification index underscores the complementary nature of these two measures-cortical thickness detects changes in the sensorimotor and posterior areas while local gyrification index identifies insular differences. Since local gyrification index and cortical thickness measures detect changes in different regions, the two used in tandem could provide a clinically relevant measure of disease progression. Our findings suggest that differences in insular regions may correspond to earlier neurodegeneration and may provide a complementary cortical measure for detection of subtle early cortical changes due to Huntington's disease.
Collapse
Affiliation(s)
- Zachary A Stoebner
- Department of Computer Science, Vanderbilt University, Nashville, Tennessee, USA.,University of Texas at Austin, Austin, Texas, USA
| | - Kilian Hett
- Department of Computer Science, Vanderbilt University, Nashville, Tennessee, USA
| | - Ilwoo Lyu
- Department of Computer Science, Vanderbilt University, Nashville, Tennessee, USA.,Department of Computer Science and Engineering, UNIST, Ulsan, South Korea
| | - Hans Johnson
- Department of Electrical and Computer Engineering, University of Iowa, Iowa City, Iowa, USA
| | - Jane S Paulsen
- Department of Neurology, University of Wisconsin, Madison, Wisconsin, USA
| | - Jeffrey D Long
- Department of Psychiatry, University of Iowa, Iowa City, Iowa, USA.,Department of Biostatistics, University of Iowa, Iowa City, Iowa, USA
| | - Ipek Oguz
- Department of Computer Science, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
47
|
Sturchio A, Duker AP, Muñoz-Sanjuan I, Espay AJ. Subtyping monogenic disorders: Huntington disease. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:171-184. [PMID: 36803810 DOI: 10.1016/b978-0-323-85555-6.00003-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Huntington disease is a highly disabling neurodegenerative disease characterized by psychiatric, cognitive, and motor deficits. The causal genetic mutation in huntingtin (Htt, also known as IT15), located on chromosome 4p16.3, leads to an expansion of a triplet coding for polyglutamine. The expansion is invariably associated with the disease when >39 repeats. Htt encodes for the protein huntingtin (HTT), which carries out many essential biological functions in the cell, in particular in the nervous system. The precise mechanism of toxicity is not known. Based on a one-gene-one-disease framework, the prevailing hypothesis ascribes toxicity to the universal aggregation of HTT. However, the aggregation process into mutant huntingtin (mHTT) is associated with a reduction of the levels of wild-type HTT. A loss of wild-type HTT may plausibly be pathogenic, contributing to the disease onset and progressive neurodegeneration. Moreover, many other biological pathways are altered in Huntington disease, such as in the autophagic system, mitochondria, and essential proteins beyond HTT, potentially explaining biological and clinical differences among affected individuals. As one gene does not mean one disease, future efforts at identifying specific Huntington subtypes are important to design biologically tailored therapeutic approaches that correct the corresponding biological pathways-rather than continuing to exclusively target the common denominator of HTT aggregation for elimination.
Collapse
Affiliation(s)
- Andrea Sturchio
- James J. and Joan A. Gardner Family Center for Parkinson's disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, United States; Department of Clinical Neuroscience, Neuro Svenningsson, Karolinska Institutet, Stockholm, Sweden.
| | - Andrew P Duker
- James J. and Joan A. Gardner Family Center for Parkinson's disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| | | | - Alberto J Espay
- James J. and Joan A. Gardner Family Center for Parkinson's disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, United States.
| |
Collapse
|
48
|
Kouba T, Frank W, Tykalova T, Mühlbäck A, Klempíř J, Lindenberg KS, Landwehrmeyer GB, Rusz J. Speech biomarkers in Huntington's disease: A cross-sectional study in pre-symptomatic, prodromal and early manifest stages. Eur J Neurol 2023; 30:1262-1271. [PMID: 36732902 DOI: 10.1111/ene.15726] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/25/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND PURPOSE Motor speech alterations are a prominent feature of clinically manifest Huntington's disease (HD). Objective acoustic analysis of speech can quantify speech alterations. It is currently unknown, however, at what stage of HD speech alterations can be reliably detected. We aimed to explore the patterns and extent of speech alterations using objective acoustic analysis in HD and to assess correlations with both rater-assessed phenotypical features and biological determinants of HD. METHODS Speech samples were acquired from 44 premanifest (29 pre-symptomatic and 15 prodromal) and 25 manifest HD gene expansion carriers, and 25 matched healthy controls. A quantitative automated acoustic analysis of 10 speech dimensions was performed. RESULTS Automated speech analysis allowed us to differentiate between participants with HD and controls, with areas under the curve of 0.74 for pre-symptomatic, 0.92 for prodromal, and 0.97 for manifest stages. In addition to irregular alternating motion rates and prolonged pauses seen only in manifest HD, both prodromal and manifest HD displayed slowed articulation rate, slowed alternating motion rates, increased loudness variability, and unstable steady-state position of articulators. In participants with premanifest HD, speech alteration severity was associated with cognitive slowing (r = -0.52, p < 0.001) and the extent of bradykinesia (r = 0.43, p = 0.004). Speech alterations correlated with a measure of exposure to mutant gene products (CAG-age-product score; r = 0.60, p < 0.001). CONCLUSION Speech abnormalities in HD are associated with other motor and cognitive deficits and are measurable already in premanifest stages of HD. Therefore, automated speech analysis might represent a quantitative HD biomarker with potential for assessing disease progression.
Collapse
Affiliation(s)
- Tomas Kouba
- Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Wiebke Frank
- Department of Neurology, University Ulm, Ulm, Germany
| | - Tereza Tykalova
- Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Alzbeta Mühlbäck
- Department of Neurology, University Ulm, Ulm, Germany.,Department of Neuropsychiatry, Huntington Center South, kbo-Isar-Amper-Klinikum Taufkirchen (Vils), Taufkirchen, Germany.,Department of Neurology and Center of Clinical Neuroscience, 1st Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Jiří Klempíř
- Department of Neurology and Center of Clinical Neuroscience, 1st Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | | | | | - Jan Rusz
- Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic.,Department of Neurology and Center of Clinical Neuroscience, 1st Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic.,Department of Neurology & ARTORG Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
49
|
Pfalzer AC, Watson KH, Ciriegio AE, Hale L, Diehl S, McDonell KE, Vnencak-Jones C, Huitz E, Snow A, Roth MC, Guthrie CS, Riordan H, Long JD, Compas BE, Claassen DO. Impairments to executive function in emerging adults with Huntington disease. J Neurol Neurosurg Psychiatry 2023; 94:130-135. [PMID: 36450478 DOI: 10.1136/jnnp-2022-329812] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/11/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND AND OBJECTIVES The clinical diagnosis of Huntington disease (HD) is typically made once motor symptoms and chorea are evident. Recent reports highlight the onset of cognitive and psychiatric symptoms before motor manifestations. These findings support further investigations of cognitive function across the lifespan of HD sufferers. METHODS To assess cognitive symptoms in the developing brain, we administered assessments from the National Institutes of Health Toolbox Cognitive Battery, an age-appropriate cognitive assessment with population norms, to a cohort of children, adolescents and young adults with (gene-expanded; GE) and without (gene-not-expanded; GNE) the trinucleotide cytosine, adenine, guanine (CAG) expansion in the Huntingtin gene. These five assessments that focus on executive function are well validated and form a composite score, with population norms. We modelled these scores across age, and CAP score to estimate the slope of progression, comparing these results to motor symptoms. RESULTS We find significant deficits in the composite measure of executive function in GE compared with GNE participants. GE participant performance on working memory was significantly lower compared with GNE participants. Modelling these results over age suggests that these deficits occur as early as 18 years of age, long before motor manifestations of HD. CONCLUSIONS This work provides strong evidence that impairments in executive function occur as early as the second decade of life, well before anticipated motor onset. Future investigations should delineate whether these impairments in executive function are due to abnormalities in neurodevelopment or early sequelae of a neurodegenerative process.
Collapse
Affiliation(s)
- Anna C Pfalzer
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kelly H Watson
- Psychology and Human Development, Vanderbilt University Peabody College of Education and Human Development, Nashville, Tennessee, USA
| | - Abagail E Ciriegio
- Psychology and Human Development, Vanderbilt University Peabody College of Education and Human Development, Nashville, Tennessee, USA
| | - Lisa Hale
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Spencer Diehl
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Katherine E McDonell
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Cindy Vnencak-Jones
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Elizabeth Huitz
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Abigail Snow
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Marissa C Roth
- Department of Psychology and Human Development, Vanderbilt University, Nashville, Tennessee, USA
| | - Cara S Guthrie
- Department of Psychology and Human Development, Vanderbilt University, Nashville, Tennessee, USA
| | - Heather Riordan
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jeffrey D Long
- Department of Psychiatry, University of Iowa, Iowa City, Iowa, USA
| | - Bruce E Compas
- Psychology and Human Development, Vanderbilt University Peabody College of Education and Human Development, Nashville, Tennessee, USA
| | - Daniel O Claassen
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
50
|
Zhang S, Cheng Y, Shang H. The updated development of blood-based biomarkers for Huntington's disease. J Neurol 2023; 270:2483-2503. [PMID: 36692635 PMCID: PMC9873222 DOI: 10.1007/s00415-023-11572-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/25/2023]
Abstract
Huntington's disease is a progressive neurodegenerative disease caused by mutation of the huntingtin (HTT) gene. The identification of mutation carriers before symptom onset provides an opportunity to intervene in the early stage of the disease course. Optimal biomarkers are of great value to reflect neuropathological and clinical progression and are sensitive to potential disease-modifying treatments. Blood-based biomarkers have the merits of minimal invasiveness, low cost, easy accessibility and safety. In this review, we summarized the updated development of blood-based biomarkers for HD from six aspects, including neuronal injuries, oxidative stress, endocrine functions, immune reactions, metabolism and differentially expressed miRNAs. The blood-based biomarkers presented and discussed in this review were close to clinical applicability and might facilitate clinical design as surrogate endpoints. Exploration and validation of robust blood-based biomarkers require further standard and systemic study design in the future.
Collapse
Affiliation(s)
- Sirui Zhang
- grid.412901.f0000 0004 1770 1022Laboratory of Neurodegenerative Disorders, Department of Neurology, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan China ,grid.412901.f0000 0004 1770 1022National Clinical Research Center for Geriatric, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, 610041 China ,grid.412901.f0000 0004 1770 1022West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041 China
| | - Yangfan Cheng
- grid.412901.f0000 0004 1770 1022Laboratory of Neurodegenerative Disorders, Department of Neurology, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan China ,grid.412901.f0000 0004 1770 1022National Clinical Research Center for Geriatric, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, 610041 China
| | - Huifang Shang
- grid.412901.f0000 0004 1770 1022Laboratory of Neurodegenerative Disorders, Department of Neurology, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan China ,grid.412901.f0000 0004 1770 1022National Clinical Research Center for Geriatric, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, 610041 China
| |
Collapse
|