1
|
Hussain Y, Dar MI, Pan X. Circadian Influences on Brain Lipid Metabolism and Neurodegenerative Diseases. Metabolites 2024; 14:723. [PMID: 39728504 DOI: 10.3390/metabo14120723] [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: 11/18/2024] [Revised: 12/09/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024] Open
Abstract
Circadian rhythms are intrinsic, 24 h cycles that regulate key physiological, mental, and behavioral processes, including sleep-wake cycles, hormone secretion, and metabolism. These rhythms are controlled by the brain's suprachiasmatic nucleus, which synchronizes with environmental signals, such as light and temperature, and consequently maintains alignment with the day-night cycle. Molecular feedback loops, driven by core circadian "clock genes", such as Clock, Bmal1, Per, and Cry, are essential for rhythmic gene expression; disruptions in these feedback loops are associated with various health issues. Dysregulated lipid metabolism in the brain has been implicated in the pathogenesis of neurological disorders by contributing to oxidative stress, neuroinflammation, and synaptic dysfunction, as observed in conditions such as Alzheimer's and Parkinson's diseases. Disruptions in circadian gene expression have been shown to perturb lipid regulatory mechanisms in the brain, thereby triggering neuroinflammatory responses and oxidative damage. This review synthesizes current insights into the interconnections between circadian rhythms and lipid metabolism, with a focus on their roles in neurological health and disease. It further examines how the desynchronization of circadian genes affects lipid metabolism and explores the potential mechanisms through which disrupted circadian signaling might contribute to the pathophysiology of neurodegenerative disorders.
Collapse
Affiliation(s)
- Yusuf Hussain
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
- Diabetes and Obesity Research Center, NYU Langone Hospital-Long Island, Mineola, NY 11501, USA
| | - Mohammad Irfan Dar
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
- Diabetes and Obesity Research Center, NYU Langone Hospital-Long Island, Mineola, NY 11501, USA
| | - Xiaoyue Pan
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
- Diabetes and Obesity Research Center, NYU Langone Hospital-Long Island, Mineola, NY 11501, USA
| |
Collapse
|
2
|
Fitzgerald ES, Manousakis JE, Glikmann-Johnston Y, Rankin M, Anderson C, Stout JC, Jackson ML. Sleep fragmentation despite intact rest-activity patterns in premanifest Huntington's disease: An actigraphy study. Sleep Med 2024; 124:16-29. [PMID: 39250876 DOI: 10.1016/j.sleep.2024.08.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 09/11/2024]
Abstract
OBJECTIVE Sleep research in Huntington's disease (HD) has primarily focused on manifest HD, with significantly less attention given to premanifest HD (Pre-HD). Therefore, we investigated sleep and rest-activity patterns in people with Pre-HD versus healthy controls (HC). METHODS We conducted a cross-sectional study including 36 Pre-HD and 48 HC participants. Pre-HD participants were stratified into three groups according to their proximity to estimated diagnosis, using a cytosine-adenine-guanine (CAG) and current age-based predictive model: NEAR (<9 years to diagnosis), MID (9-15 years to diagnosis) and FAR (>15 years to diagnosis). Sleep and rest-activity patterns were assessed using wrist-worn actigraphy, a sleep diary, and sleep questionnaires. RESULTS NEAR and MID groups experienced higher fragmentation index than HC and FAR groups. NEAR and MID groups also exhibited greater WASO than the FAR group. NEAR and MID groups showed lower intra-daily variability (IV) than HC and FAR groups, with the NEAR group also being more active in the most active 10 h (M10). Groups did not differ on subjective sleep measures, inter-daily stability (IS), sleep regularity index, relative amplitude, or amount of activity in the least active 5 h (L5). Considering all Pre-HD participants, fewer years to diagnosis, higher CAG-age-product (CAP) scores (a measure of cumulative exposure to the HD-causing gene mutation) and larger CAG repeat lengths correlated with higher WASO, fragmentation index, L5, IS, and lower sleep efficiency and IV. Higher CAP score correlated with higher M10. CONCLUSIONS Despite intact rest-activity patterns and similar subjective sleep quality to HC, greater sleep fragmentation is a prominent and early feature in Pre-HD. Therefore, reducing sleep fragmentation may be a potential target for sleep intervention in HD. Longitudinal studies using larger samples are needed to assess sleep across the disease spectrum and its impact on clinical outcomes, like cognition.
Collapse
Affiliation(s)
- Emily S Fitzgerald
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Jessica E Manousakis
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Yifat Glikmann-Johnston
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Meg Rankin
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Clare Anderson
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia; Centre for Human Brain Health, School of Psychology, University of Birmingham, Edgbaston, UK
| | - Julie C Stout
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia.
| | - Melinda L Jackson
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| |
Collapse
|
3
|
Scaglione C, Vitiello M, Tonetti L, Giovagnoli S, Barletta G, Calandra-Buonaura G, Guaraldi P, Di Laudo F, Natale V, Provini F. Sleep-wake cycle and 24-h motor activity in early-mid Huntington's disease patients: An actigraphy-based study. J Huntingtons Dis 2024; 13:501-509. [PMID: 39973377 DOI: 10.1177/18796397241287227] [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: 02/21/2025]
Abstract
Background: Disrupted 24-h sleep-wake and rest-activity cycles are known common features in Huntington's disease (HD) patients; however, critical periods during the 24-h cycle have been less studied. Objective: To analyze the differences between early-mid stage HD patients and healthy controls (HC) in sleep patterns and 24-h motor activity by using actigraphic monitoring. Methods: Twenty HD patients (13 females; mean age ± SD 56.45 ± 16.94) at early-mid stage of the disease and 20 HC were actigraphically monitored for a week. We applied the Functional Linear Modeling (FLM) to analyze motor activity from the actigraphic data. We analyzed parameters regarding both the time spent in bed and out of bed; get-up time (GUT); time in bed (TIB); midpoint of sleep (MS); sleep motor activity (SMA); sleep onset latency (SOL); total sleep time (TST); wake after sleep onset (WASO); sleep efficiency (SE); number and duration of awakenings (AWK); diurnal motor activity (DMA) and diurnal total sleep time (DTST). Results: Ten patients were in Stage I, 6 in Stage II and 4 in Stage III. HD patients presented lower SE and higher TIB, SOL, WASO, AWK and AWK > 5 min in comparison to HC. Moreover, higher motor activity was observed in patients with HD, in particular between 2:15 and 4:00 am, from around 40 min prior to bedtime until 20 min after bedtime, and from around 20 min prior to get-up time until 50 min after get-up time. Conclusions: Actigraphy documented a specific 24-h motor pattern in HD, potentially constituting a disease signature.
Collapse
Affiliation(s)
- Cesa Scaglione
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Maria Vitiello
- Department of Neurology and Stroke Unit Cesena (Forlì), Bufalini Hospital, AUSL Romagna, Cesena, Italy
| | - Lorenzo Tonetti
- Department of Psychology "Renzo Canestrari", University of Bologna, Bologna, Italy
| | - Sara Giovagnoli
- Department of Psychology "Renzo Canestrari", University of Bologna, Bologna, Italy
| | - Giorgio Barletta
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Giovanna Calandra-Buonaura
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Pietro Guaraldi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Felice Di Laudo
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Vincenzo Natale
- Department of Psychology "Renzo Canestrari", University of Bologna, Bologna, Italy
| | - Federica Provini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
4
|
Rodrigues L, Bocchetta M, Puonti O, Greve D, Londe AC, França M, Appenzeller S, Rittner L, Iglesias JE. High-resolution segmentations of the hypothalamus and its subregions for training of segmentation models. Sci Data 2024; 11:940. [PMID: 39198456 PMCID: PMC11358401 DOI: 10.1038/s41597-024-03775-2] [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: 01/31/2024] [Accepted: 08/15/2024] [Indexed: 09/01/2024] Open
Abstract
Segmentation of brain structures on magnetic resonance imaging (MRI) is a highly relevant neuroimaging topic, as it is a prerequisite for different analyses such as volumetry or shape analysis. Automated segmentation facilitates the study of brain structures in larger cohorts when compared with manual segmentation, which is time-consuming. However, the development of most automated methods relies on large and manually annotated datasets, which limits the generalizability of these methods. Recently, new techniques using synthetic images have emerged, reducing the need for manual annotation. Here we provide a dataset composed of label maps built from publicly available ultra-high resolution ex vivo MRI from 10 whole hemispheres, which can be used to develop segmentation methods using synthetic data. The label maps are obtained with a combination of manual labels for the hypothalamic regions and automated segmentations for the rest of the brain, and mirrored to simulate entire brains. We also provide the pre-processed ex vivo scans, as this dataset can support future projects to include other structures after these are manually segmented.
Collapse
Affiliation(s)
- Livia Rodrigues
- Massachusetts General Hospital, Harvard Medical School, Boston Campus, USA.
- Universidade Estadual de Campinas, School of Electrical and Computer Engineering, São Paulo, Brazil.
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- Centre for Cognitive and Clinical Neuroscience, Division of Psychology, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, London, UK
| | - Oula Puonti
- Massachusetts General Hospital, Harvard Medical School, Boston Campus, USA
| | - Douglas Greve
- Massachusetts General Hospital, Harvard Medical School, Boston Campus, USA
| | - Ana Carolina Londe
- Universidade Estadual de Campinas - School of Medical Sciences, São Paulo, Brazil
| | - Marcondes França
- Universidade Estadual de Campinas - School of Medical Sciences, São Paulo, Brazil
| | - Simone Appenzeller
- Universidade Estadual de Campinas - School of Medical Sciences, São Paulo, Brazil
| | - Leticia Rittner
- Universidade Estadual de Campinas, School of Electrical and Computer Engineering, São Paulo, Brazil
| | - Juan Eugenio Iglesias
- Massachusetts General Hospital, Harvard Medical School, Boston Campus, USA
- Centre for Medical Image Computing, University College London, London, UK
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, USA
| |
Collapse
|
5
|
Dell’Angelica D, Singh K, Colwell CS, Ghiani CA. Circadian Interventions in Preclinical Models of Huntington's Disease: A Narrative Review. Biomedicines 2024; 12:1777. [PMID: 39200241 PMCID: PMC11351982 DOI: 10.3390/biomedicines12081777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 09/02/2024] Open
Abstract
Huntington's Disease (HD) is a neurodegenerative disorder caused by an autosomal-dominant mutation in the huntingtin gene, which manifests with a triad of motor, cognitive and psychiatric declines. Individuals with HD often present with disturbed sleep/wake cycles, but it is still debated whether altered circadian rhythms are intrinsic to its aetiopathology or a consequence. Conversely, it is well established that sleep/wake disturbances, perhaps acting in concert with other pathophysiological mechanisms, worsen the impact of the disease on cognitive and motor functions and are a burden to the patients and their caretakers. Currently, there is no cure to stop the progression of HD, however, preclinical research is providing cementing evidence that restoring the fluctuation of the circadian rhythms can assist in delaying the onset and slowing progression of HD. Here we highlight the application of circadian-based interventions in preclinical models and provide insights into their potential translation in clinical practice. Interventions aimed at improving sleep/wake cycles' synchronization have shown to improve motor and cognitive deficits in HD models. Therefore, a strong support for their suitability to ameliorate HD symptoms in humans emerges from the literature, albeit with gaps in our knowledge on the underlying mechanisms and possible risks associated with their implementation.
Collapse
Affiliation(s)
- Derek Dell’Angelica
- Department of Psychiatry and Biobehavioural Sciences, Semel Institute for Neuroscience and Human Behaviour, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90024, USA; (D.D.); (K.S.); (C.S.C.)
| | - Karan Singh
- Department of Psychiatry and Biobehavioural Sciences, Semel Institute for Neuroscience and Human Behaviour, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90024, USA; (D.D.); (K.S.); (C.S.C.)
| | - Christopher S. Colwell
- Department of Psychiatry and Biobehavioural Sciences, Semel Institute for Neuroscience and Human Behaviour, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90024, USA; (D.D.); (K.S.); (C.S.C.)
| | - Cristina A. Ghiani
- Department of Psychiatry and Biobehavioural Sciences, Semel Institute for Neuroscience and Human Behaviour, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90024, USA; (D.D.); (K.S.); (C.S.C.)
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90024, USA
| |
Collapse
|
6
|
Canever JB, Queiroz LY, Soares ES, de Avelar NCP, Cimarosti HI. Circadian rhythm alterations affecting the pathology of neurodegenerative diseases. J Neurochem 2024; 168:1475-1489. [PMID: 37358003 DOI: 10.1111/jnc.15883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/27/2023] [Accepted: 06/01/2023] [Indexed: 06/27/2023]
Abstract
The circadian rhythm is a nearly 24-h oscillation found in various physiological processes in the human brain and body that is regulated by environmental and genetic factors. It is responsible for maintaining body homeostasis and it is critical for essential functions, such as metabolic regulation and memory consolidation. Dysregulation in the circadian rhythm can negatively impact human health, resulting in cardiovascular and metabolic diseases, psychiatric disorders, and premature death. Emerging evidence points to a relationship between the dysregulation circadian rhythm and neurodegenerative diseases, suggesting that the alterations in circadian function might play crucial roles in the pathogenesis and progression of neurodegenerative diseases. Better understanding this association is of paramount importance to expand the knowledge on the pathophysiology of neurodegenerative diseases, as well as, to provide potential targets for the development of new interventions based on the dysregulation of circadian rhythm. Here we review the latest findings on dysregulation of circadian rhythm alterations in Parkinson's disease, Alzheimer's disease, Huntington's disease, amyotrophic lateral sclerosis, multiple sclerosis, spinocerebellar ataxia and multiple-system atrophy, focusing on research published in the last 3 years.
Collapse
Affiliation(s)
- Jaquelini Betta Canever
- Postgraduate Program of Neuroscience, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Letícia Yoshitome Queiroz
- Postgraduate Program of Pharmacology, Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Ericks Sousa Soares
- Postgraduate Program of Pharmacology, Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Núbia Carelli Pereira de Avelar
- Laboratory of Aging, Resources and Rheumatology, Department of Health Sciences, Federal University of Santa Catarina, Araranguá, Santa Catarina, Brazil
| | - Helena Iturvides Cimarosti
- Postgraduate Program of Neuroscience, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
- Postgraduate Program of Pharmacology, Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| |
Collapse
|
7
|
Turner M, Griffiths M, Laws M, Vial S, Bartlett D, Cruickshank T. The multidimensional sleep health of individuals with multiple sclerosis and Huntington's disease and healthy controls. J Clin Sleep Med 2024; 20:967-972. [PMID: 38305780 PMCID: PMC11145047 DOI: 10.5664/jcsm.11052] [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: 12/13/2023] [Revised: 01/21/2024] [Accepted: 01/22/2024] [Indexed: 02/03/2024]
Abstract
STUDY OBJECTIVES Sleep issues are common for people with neurodegenerative conditions, yet research has focused on specific aspects of sleep. While important, a more holistic approach to investigating sleep, termed "sleep health," considers sleep's positive and negative aspects. Current studies exploring sleep health have lacked a control group for reference. For the first time, this study investigated the sleep health of people living with multiple sclerosis and Huntington's disease (HD) and compared it with a community sample. METHODS 111 people, including 43 with multiple sclerosis, 19 with HD, and 49 from a community sample, participated in this study. The data, including actigraphy, Pittsburgh Sleep Quality Index, and Epworth Sleepiness Scale, were collected as part of ongoing research studies. Seven sleep health domains were determined from the collected data, and a composite sleep health score was developed. Analysis of variance and independent t tests were performed to identify population and sex differences. RESULTS The HD group had higher sleep regularity and lower sleep rhythmicity than the multiple sclerosis and community sample groups. The HD group had significantly less sleep duration than the multiple sclerosis group. No significant differences between the groups were observed in the sleep health composite score. Males had significantly higher sleep regularity within the HD group but significantly lower sleepiness scores in the community sample. CONCLUSIONS These findings indicate that people with HD may experience greater variance in their wake times, therefore decreasing the consistency of being awake or asleep 24 hours apart. Understanding the mechanisms for this should be explored in people with HD. CITATION Turner M, Griffiths M, Laws M, Vial S, Bartlett D, Cruickshank T. The multidimensional sleep health of individuals with multiple sclerosis and Huntington's disease and healthy controls. J Clin Sleep Med. 2024;20(6):967-972.
Collapse
Affiliation(s)
- Mitchell Turner
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Madeline Griffiths
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Manja Laws
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Shayne Vial
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Danielle Bartlett
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Travis Cruickshank
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
- Perron Institute for Neurological and Translational Sciences, Perth, Western Australia, Australia
| |
Collapse
|
8
|
Ghaderi S, Fatehi F, Kalra S, Mohammadi S, Zemorshidi F, Ramezani M, Hesami O, Pezeshgi S, Batouli SAH. Volume loss in the left anterior-superior subunit of the hypothalamus in amyotrophic lateral sclerosis. CNS Neurosci Ther 2024; 30:e14801. [PMID: 38887187 PMCID: PMC11183167 DOI: 10.1111/cns.14801] [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: 02/26/2024] [Revised: 05/11/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Amyotrophic lateral sclerosis (ALS) causes motor neuron loss and progressive paralysis. While traditionally viewed as motor neuron disease (MND), ALS also affects non-motor regions, such as the hypothalamus. This study aimed to quantify the hypothalamic subregion volumes in patients with ALS versus healthy controls (HCs) and examine their associations with demographic and clinical features. METHODS Forty-eight participants (24 ALS patients and 24 HCs) underwent structural MRI. A deep convolutional neural network was used for the automated segmentation of the hypothalamic subunits, including the anterior-superior (a-sHyp), anterior-inferior (a-iHyp), superior tuberal (supTub), inferior tuberal (infTub), and posterior (posHyp). The neural network was validated using FreeSurfer v7.4.1, with individual head size variations normalized using total intracranial volume (TIV) normalization. Statistical analyses were performed for comparisons using independent sample t-tests. Correlations were calculated using Pearson's and Spearman's tests (p < 0.05). The standard mean difference (SMD) was used to compare the mean differences between parametric variables. RESULTS The volume of the left a-sHyp hypothalamic subunit was significantly lower in ALS patients than in HCs (p = 0.023, SMD = -0.681). No significant correlation was found between the volume of the hypothalamic subunits, body mass index (BMI), and ALSFRS-R in patients with ALS. However, right a-sHyp (r = 0.420, p = 0.041) was correlated with disease duration, whereas right supTub (r = -0.471, p = 0.020) and left postHyp (r = -0.406, p = 0.049) were negatively correlated with age. There was no significant difference in the volume of hypothalamic subunits between males and females, and no significant difference was found between patients with revised ALS Functional Rating Scale (ALSFRS-R) scores ≤41 and >41 and those with a disease duration of 9 months or less. DISCUSSION AND CONCLUSION The main finding suggests atrophy of the left a-sHyp hypothalamic subunit in patients with ALS, which is supported by previous research as an extra-motor neuroimaging finding for ALS.
Collapse
Affiliation(s)
- Sadegh Ghaderi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in MedicineTehran University of Medical SciencesTehranIran
- Neuromuscular Research Center, Department of Neurology, Shariati HospitalTehran University of Medical SciencesTehranIran
| | - Farzad Fatehi
- Neuromuscular Research Center, Department of Neurology, Shariati HospitalTehran University of Medical SciencesTehranIran
- Neurology DepartmentUniversity Hospitals of Leicester NHS TrustLeicesterUK
| | - Sanjay Kalra
- Neuroscience and Mental Health InstituteUniversity of AlbertaEdmontonAlbertaCanada
- Division of Neurology, Department of MedicineUniversity of AlbertaEdmontonAlbertaCanada
| | - Sana Mohammadi
- Neuromuscular Research Center, Department of Neurology, Shariati HospitalTehran University of Medical SciencesTehranIran
| | - Fariba Zemorshidi
- Neuromuscular Research Center, Department of Neurology, Shariati HospitalTehran University of Medical SciencesTehranIran
- Department of NeurologyMashhad University of Medical SciencesMashhadIran
| | - Mahtab Ramezani
- Neuromuscular Research Center, Department of Neurology, Shariati HospitalTehran University of Medical SciencesTehranIran
| | - Omid Hesami
- Neuromuscular Research Center, Department of Neurology, Shariati HospitalTehran University of Medical SciencesTehranIran
- Department of NeurologyShahid Beheshti University of Medical SciencesTehranIran
| | - Saharnaz Pezeshgi
- Neuromuscular Research Center, Department of Neurology, Shariati HospitalTehran University of Medical SciencesTehranIran
| | - Seyed Amir Hossein Batouli
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in MedicineTehran University of Medical SciencesTehranIran
| |
Collapse
|
9
|
Vernikouskaya I, Müller HP, Roselli F, Ludolph AC, Kassubek J, Rasche V. AI-assisted quantification of hypothalamic atrophy in amyotrophic lateral sclerosis by convolutional neural network-based automatic segmentation. Sci Rep 2023; 13:21505. [PMID: 38057503 PMCID: PMC10700600 DOI: 10.1038/s41598-023-48649-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 11/29/2023] [Indexed: 12/08/2023] Open
Abstract
The hypothalamus is a small structure of the brain with an essential role in metabolic homeostasis, sleep regulation, and body temperature control. Some neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS) and dementia syndromes are reported to be related to hypothalamic volume alterations. Despite its crucial role in human body regulation, neuroimaging studies of this structure are rather scarce due to work-intensive operator-dependent manual delineations from MRI and lack of automated segmentation tools. In this study we present a fully automatic approach based on deep convolutional neural networks (CNN) for hypothalamic segmentation and volume quantification. We applied CNN of U-Net architecture with EfficientNetB0 backbone to allow for accurate automatic hypothalamic segmentation in seconds on a GPU. We further applied our approach for the quantification of the normalized hypothalamic volumes to a large neuroimaging dataset of 432 ALS patients and 112 healthy controls (without the ground truth labels). Using the automated volumetric analysis, we could reproduce hypothalamic atrophy findings associated with ALS by detecting significant volume differences between ALS patients and controls at the group level. In conclusion, a fast and unbiased AI-assisted hypothalamic quantification method is introduced in this study (whose acceptance rate based on the outlier removal strategy was estimated to be above 95%) and made publicly available for researchers interested in the conduction of hypothalamus studies at a large scale.
Collapse
Affiliation(s)
- Ina Vernikouskaya
- Department of Internal Medicine II, Ulm University Medical Center, Albert-Einstein-Allee 23, 89081, Ulm, Germany.
| | | | - Francesco Roselli
- Department of Neurology, University of Ulm, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Albert C Ludolph
- Department of Neurology, University of Ulm, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Jan Kassubek
- Department of Neurology, University of Ulm, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Volker Rasche
- Department of Internal Medicine II, Ulm University Medical Center, Albert-Einstein-Allee 23, 89081, Ulm, Germany
- Core Facility Small Animal MRI, University of Ulm, Ulm, Germany
| |
Collapse
|
10
|
Talwar P, Deantoni M, Van Egroo M, Muto V, Chylinski D, Koshmanova E, Jaspar M, Meyer C, Degueldre C, Berthomier C, Luxen A, Salmon E, Collette F, Dijk DJ, Schmidt C, Phillips C, Maquet P, Sherif S, Vandewalle G. In vivo marker of brainstem myelin is associated to quantitative sleep parameters in healthy young men. Sci Rep 2023; 13:20873. [PMID: 38012207 PMCID: PMC10682495 DOI: 10.1038/s41598-023-47753-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: 08/22/2023] [Accepted: 11/17/2023] [Indexed: 11/29/2023] Open
Abstract
The regional integrity of brain subcortical structures has been implicated in sleep-wake regulation, however, their associations with sleep parameters remain largely unexplored. Here, we assessed association between quantitative Magnetic Resonance Imaging (qMRI)-derived marker of the myelin content of the brainstem and the variability in the sleep electrophysiology in a large sample of 18-to-31 years healthy young men (N = 321; ~ 22 years). Separate Generalized Additive Model for Location, Scale and Shape (GAMLSS) revealed that sleep onset latency and slow wave energy were significantly associated with MTsat estimates in the brainstem (pcorrected ≤ 0.03), with overall higher MTsat value associated with values reflecting better sleep quality. The association changed with age, however (MTsat-by-age interaction-pcorrected ≤ 0.03), with higher MTsat value linked to better values in the two sleep metrics in the younger individuals of our sample aged ~ 18 to 20 years. Similar associations were detected across different parts of the brainstem (pcorrected ≤ 0.03), suggesting that the overall maturation and integrity of the brainstem was associated with both sleep metrics. Our results suggest that myelination of the brainstem nuclei essential to regulation of sleep is associated with inter-individual differences in sleep characteristics during early adulthood. They may have implications for sleep disorders or neurological diseases related to myelin.
Collapse
Affiliation(s)
- Puneet Talwar
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
| | - Michele Deantoni
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
| | - Maxime Van Egroo
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
| | - Vincenzo Muto
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium
- Psychology and Cognitive Neuroscience Research Unit, University of Liège, Liège, Belgium
| | - Daphne Chylinski
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
| | - Ekaterina Koshmanova
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
| | - Mathieu Jaspar
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium
| | - Christelle Meyer
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium
| | - Christian Degueldre
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
| | | | - André Luxen
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
| | - Eric Salmon
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
- Psychology and Cognitive Neuroscience Research Unit, University of Liège, Liège, Belgium
- Department of Neurology, CHU of Liège, Liège, Belgium
| | - Fabienne Collette
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
- Psychology and Cognitive Neuroscience Research Unit, University of Liège, Liège, Belgium
| | - D-J Dijk
- Sleep Research Centre, University of Surrey, Guildford, UK
- UK Dementia Research Institute, University of Surrey, Guildford, UK
| | - Christina Schmidt
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
- Psychology and Cognitive Neuroscience Research Unit, University of Liège, Liège, Belgium
| | - Christophe Phillips
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
- In Silico Medicine Unit, GIGA-Institute, University of Liège, Liège, Belgium
| | - Pierre Maquet
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium
- Department of Neurology, CHU of Liège, Liège, Belgium
| | - Siya Sherif
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
| | - Gilles Vandewalle
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium.
| |
Collapse
|
11
|
Xie H, Shih CH, Aldoohan SD, Wall JT, Wang X. Hypothalamus volume mediates the association between adverse childhood experience and PTSD development after adulthood trauma. Transl Psychiatry 2023; 13:274. [PMID: 37542036 PMCID: PMC10403516 DOI: 10.1038/s41398-023-02576-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/06/2023] Open
Abstract
The hypothalamus is critical for regulation of the hypothalamic-pituitary-adrenal (HPA) axis and response to stress. Adverse childhood experience (ACE) can affect brain structure, which may contribute to development of posttraumatic stress disorder (PTSD) after subsequent adult trauma. It is unclear, however, if ACE history is particularly associated with aspects of hypothalamic structure which contribute to development of PTSD. To address this issue, the present study longitudinally assessed hypothalamic volumes and their associations with ACE and early post-trauma stress symptoms in subjects who did or did not develop PTSD during 12 months after adult trauma. 109 subjects (18-60 years, F/M = 75/34) completed the PTSD Checklist (PCL) questionnaire for post-trauma stress symptoms, the Childhood Trauma Questionnaire (CTQ) for ACE assessment, and an initial MRI brain scan for hypothalamic volume measurement, within 2 weeks after adult trauma. At post-trauma 12 months, subjects underwent a subsequent PTSD diagnosis interview using the Clinician-Administered PTSD Scale (CAPS), and a follow-up MRI scan. Left and right hypothalamus volumes at 2 weeks after adult trauma negatively correlated with CTQ scores. Right hypothalamus volume at this early time mediated an association between ACE and PTSD symptoms 12 months later. Right hypothalamus volumes also remained persistently smaller from 2 weeks to 12 months after trauma in survivors who developed PTSD. These results suggest that smaller right hypothalamus volume may be related to ACE history in ways that contribute to PTSD development after trauma in adulthood.
Collapse
Affiliation(s)
- Hong Xie
- Department of Neurosciences, University of Toledo, Toledo, OH, USA.
| | - Chia-Hao Shih
- Department of Emergency Medicine, University of Toledo, Toledo, OH, USA
| | | | - John T Wall
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | - Xin Wang
- Department of Psychiatry, University of Toledo, Toledo, OH, USA
| |
Collapse
|
12
|
Morrone CD, Raghuraman R, Hussaini SA, Yu WH. Proteostasis failure exacerbates neuronal circuit dysfunction and sleep impairments in Alzheimer's disease. Mol Neurodegener 2023; 18:27. [PMID: 37085942 PMCID: PMC10119020 DOI: 10.1186/s13024-023-00617-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/29/2023] [Indexed: 04/23/2023] Open
Abstract
Failed proteostasis is a well-documented feature of Alzheimer's disease, particularly, reduced protein degradation and clearance. However, the contribution of failed proteostasis to neuronal circuit dysfunction is an emerging concept in neurodegenerative research and will prove critical in understanding cognitive decline. Our objective is to convey Alzheimer's disease progression with the growing evidence for a bidirectional relationship of sleep disruption and proteostasis failure. Proteostasis dysfunction and tauopathy in Alzheimer's disease disrupts neurons that regulate the sleep-wake cycle, which presents behavior as impaired slow wave and rapid eye movement sleep patterns. Subsequent sleep loss further impairs protein clearance. Sleep loss is a defined feature seen early in many neurodegenerative disorders and contributes to memory impairments in Alzheimer's disease. Canonical pathological hallmarks, β-amyloid, and tau, directly disrupt sleep, and neurodegeneration of locus coeruleus, hippocampal and hypothalamic neurons from tau proteinopathy causes disruption of the neuronal circuitry of sleep. Acting in a positive-feedback-loop, sleep loss and circadian rhythm disruption then increase spread of β-amyloid and tau, through impairments of proteasome, autophagy, unfolded protein response and glymphatic clearance. This phenomenon extends beyond β-amyloid and tau, with interactions of sleep impairment with the homeostasis of TDP-43, α-synuclein, FUS, and huntingtin proteins, implicating sleep loss as an important consideration in an array of neurodegenerative diseases and in cases of mixed neuropathology. Critically, the dynamics of this interaction in the neurodegenerative environment are not fully elucidated and are deserving of further discussion and research. Finally, we propose sleep-enhancing therapeutics as potential interventions for promoting healthy proteostasis, including β-amyloid and tau clearance, mechanistically linking these processes. With further clinical and preclinical research, we propose this dynamic interaction as a diagnostic and therapeutic framework, informing precise single- and combinatorial-treatments for Alzheimer's disease and other brain disorders.
Collapse
Affiliation(s)
- Christopher Daniel Morrone
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON, M5T 1R8, Canada.
| | - Radha Raghuraman
- Taub Institute, Columbia University Irving Medical Center, 630W 168th Street, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 630W 168th Street, New York, NY, 10032, USA
| | - S Abid Hussaini
- Taub Institute, Columbia University Irving Medical Center, 630W 168th Street, New York, NY, 10032, USA.
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 630W 168th Street, New York, NY, 10032, USA.
| | - Wai Haung Yu
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON, M5T 1R8, Canada.
- Geriatric Mental Health Research Services, Centre for Addiction and Mental Health, 250 College St., Toronto, ON, M5T 1R8, Canada.
- Department of Pharmacology and Toxicology, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
13
|
Saade-Lemus S, Videnovic A. Sleep Disorders and Circadian Disruption in Huntington's Disease. J Huntingtons Dis 2023; 12:121-131. [PMID: 37424473 PMCID: PMC10473087 DOI: 10.3233/jhd-230576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2023] [Indexed: 07/11/2023]
Abstract
Sleep and circadian alterations are common in patients with Huntington's disease (HD). Understanding the pathophysiology of these alterations and their association with disease progression and morbidity can guide HD management. We provide a narrative review of the clinical and basic-science studies centered on sleep and circadian function on HD. Sleep/wake disturbances among HD patients share many similarities with other neurodegenerative diseases. Overall, HD patients and animal models of the disease present with sleep changes early in the clinical course of the disease, including difficulties with sleep initiation and maintenance leading to decreased sleep efficiency, and progressive deterioration of normal sleep architecture. Despite this, sleep alterations remain frequently under-reported by patients and under-recognized by health professionals. The degree of sleep and circadian alterations has not consistently shown to be CAG dose-dependent. Evidence based treatment recommendations are insufficient due to lack of well-designed intervention trials. Approaches aimed at improving circadian entrainment, such as including light therapy, and time-restricted feeding have demonstrated a potential to delay symptom progression in some basic HD investigations. Larger study cohorts, comprehensive assessment of sleep and circadian function, and reproducibility of findings are needed in future in order to better understand sleep and circadian function in HD and to develop effective treatments.
Collapse
Affiliation(s)
- Sandra Saade-Lemus
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Aleksandar Videnovic
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
14
|
Bailey GA, Matthews C, Szewczyk-krolikowski K, Moore P, Komarzynski S, Davies EH, Peall KJ. Use of remote monitoring and integrated platform for the evaluation of sleep quality in adult-onset idiopathic cervical dystonia. J Neurol 2023; 270:1759-1769. [PMID: 36414751 PMCID: PMC9971061 DOI: 10.1007/s00415-022-11490-4] [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] [Received: 09/02/2022] [Revised: 11/08/2022] [Accepted: 11/11/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND Up to 70% of individuals diagnosed with adult-onset idiopathic focal cervical dystonia (AOIFCD) report difficulties with sleep. Larger cohort studies using wrist-worn accelerometer devices have emerged as an alternative to smaller polysomnography studies, in order to evaluate sleep architecture. METHODS To measure activity during the sleep/wake cycle, individuals wore a consumer-grade wrist device (Garmin vivosmart 4) continuously over 7 days on their non-dominant wrist, while completing a daily sleep diary and standardised sleep and non-motor questionnaires via a dedicated app. Sleep measures were derived from the captured raw triaxial acceleration and heart rate values using previously published validated algorithms. RESULTS Data were collected from 50 individuals diagnosed with AOIFCD and 47 age- and sex-matched controls. Those with AOIFCD self-reported significantly higher levels of excessive daytime sleepiness (p = 0.04) and impaired sleep quality (p = 0.03), while accelerometer measurements found the AOIFCD cohort to have significantly longer total sleep times (p = 0.004) and time spent in NREM sleep (p = 0.009), compared to controls. Overall, there was limited agreement between wearable-derived sleep parameters, and self-reported sleep diary and visual analogue scale records. DISCUSSION This study shows the potential feasibility of using consumer-grade wearable devices in estimating sleep measures at scale in dystonia cohorts. Those diagnosed with AOIFCD were observed to have altered sleep architecture, notably longer total sleep time and NREM sleep, compared to controls. These findings suggest that previously reported disruptions to brainstem circuitry and serotonin neurotransmission may contribute to both motor and sleep pathophysiology.
Collapse
Affiliation(s)
- Grace A. Bailey
- grid.5600.30000 0001 0807 5670Neuroscience and Mental Health Research Institute, Cardiff University School of Medicine, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ UK
| | | | | | - Peter Moore
- grid.416928.00000 0004 0496 3293The Walton Centre NHS Foundation Trust, Liverpool, UK
| | | | | | - Kathryn J. Peall
- grid.5600.30000 0001 0807 5670Neuroscience and Mental Health Research Institute, Cardiff University School of Medicine, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ UK
| |
Collapse
|
15
|
Fitzgerald ES, Stout JC, Glikmann-Johnston Y, Anderson C, Jackson ML. Sleep, Circadian Rhythms, and Cognitive Dysfunction in Huntington's Disease. J Huntingtons Dis 2023; 12:293-304. [PMID: 37599535 DOI: 10.3233/jhd-230578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
BACKGROUND In healthy people, sleep and circadian disruption are linked to cognitive deficits. People with Huntington's disease (HD), who have compromised brain function and sleep and circadian disturbances, may be even more susceptible to these cognitive effects. OBJECTIVE To conduct a comprehensive review and synthesis of the literature in HD on the associations of cognitive dysfunction with disturbed sleep and circadian rhythms. METHODS We searched MEDLINE via OVID, CINAHL Plus, EMBASE via OVID, and PubMed in May 2023. The first author then screened by title and abstract and conducted a full review of remaining articles. RESULTS Eight studies investigating the influence of sleep and/or circadian rhythms on cognitive function in HD were found. In manifest HD, poorer sleep was associated with worse cognitive function. For behavioral 24-hour (circadian) rhythms, two studies indicated that later wake times correlated with poorer cognitive function. No reported studies in HD examined altered physiological 24-hour (circadian) rhythms and cognitive impairment. CONCLUSION Some associations exist between poor sleep and cognitive dysfunction in manifest HD, yet whether these associations are present before clinical diagnosis is unknown. Whether circadian disturbances relate to cognitive impairment in HD also remains undetermined. To inform sleep and circadian interventions aimed at improving cognitive symptoms in HD, future research should include a range of disease stages, control for external factors, and utilize robust cognitive batteries targeted to the aspects of cognitive function known to be adversely affected in HD.
Collapse
Affiliation(s)
- Emily S Fitzgerald
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Julie C Stout
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Yifat Glikmann-Johnston
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Clare Anderson
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Melinda L Jackson
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| |
Collapse
|
16
|
Rodrigues L, Rezende TJR, Wertheimer G, Santos Y, França M, Rittner L. A benchmark for hypothalamus segmentation on T1-weighted MR images. Neuroimage 2022; 264:119741. [PMID: 36368499 DOI: 10.1016/j.neuroimage.2022.119741] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 09/23/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022] Open
Abstract
The hypothalamus is a small brain structure that plays essential roles in sleep regulation, body temperature control, and metabolic homeostasis. Hypothalamic structural abnormalities have been reported in neuropsychiatric disorders, such as schizophrenia, amyotrophic lateral sclerosis, and Alzheimer's disease. Although mag- netic resonance (MR) imaging is the standard examination method for evaluating this region, hypothalamic morphological landmarks are unclear, leading to subjec- tivity and high variability during manual segmentation. Due to these limitations, it is common to find contradicting results in the literature regarding hypothalamic volumetry. To the best of our knowledge, only two automated methods are available in the literature for hypothalamus segmentation, the first of which is our previous method based on U-Net. However, both methods present performance losses when predicting images from different datasets than those used in training. Therefore, this project presents a benchmark consisting of a diverse T1-weighted MR image dataset comprising 1381 subjects from IXI, CC359, OASIS, and MiLI (the latter created specifically for this benchmark). All data were provided using automatically generated hypothalamic masks and a subset containing manually annotated masks. As a baseline, a method for fully automated segmentation of the hypothalamus on T1-weighted MR images with a greater generalization ability is presented. The pro- posed method is a teacher-student-based model with two blocks: segmentation and correction, where the second corrects the imperfections of the first block. After using three datasets for training (MiLI, IXI, and CC359), the prediction performance of the model was measured on two test sets: the first was composed of data from IXI, CC359, and MiLI, achieving a Dice coefficient of 0.83; the second was from OASIS, a dataset not used for training, achieving a Dice coefficient of 0.74. The dataset, the baseline model, and all necessary codes to reproduce the experiments are available at https://github.com/MICLab-Unicamp/HypAST and https://sites.google.com/ view/calgary-campinas-dataset/hypothalamus-benchmarking. In addition, a leaderboard will be maintained with predictions for the test set submitted by anyone working on the same task.
Collapse
Affiliation(s)
- Livia Rodrigues
- Medical Image Computing Lab, School of Electrical and Computer Engineering (FEEC), University of Campinas, Albert Einstein Street, 400, Campinas, SP 13083-887, Brazil.
| | - Thiago Junqueira Ribeiro Rezende
- Department of Neurology, School of Medical Sciences, University of Campinas, Tessalia Vieira de Camargo Street, 126, Campinas, SP 13083-887, Brazil
| | - Guilherme Wertheimer
- Department of Neurology, School of Medical Sciences, University of Campinas, Tessalia Vieira de Camargo Street, 126, Campinas, SP 13083-887, Brazil
| | - Yves Santos
- Department of Neurology, School of Medical Sciences, University of Campinas, Tessalia Vieira de Camargo Street, 126, Campinas, SP 13083-887, Brazil
| | - Marcondes França
- Department of Neurology, School of Medical Sciences, University of Campinas, Tessalia Vieira de Camargo Street, 126, Campinas, SP 13083-887, Brazil
| | - Leticia Rittner
- Medical Image Computing Lab, School of Electrical and Computer Engineering (FEEC), University of Campinas, Albert Einstein Street, 400, Campinas, SP 13083-887, Brazil
| |
Collapse
|
17
|
Tabrizi SJ, Estevez-Fraga C, van Roon-Mom WMC, Flower MD, Scahill RI, Wild EJ, Muñoz-Sanjuan I, Sampaio C, Rosser AE, Leavitt BR. Potential disease-modifying therapies for Huntington's disease: lessons learned and future opportunities. Lancet Neurol 2022; 21:645-658. [PMID: 35716694 PMCID: PMC7613206 DOI: 10.1016/s1474-4422(22)00121-1] [Citation(s) in RCA: 153] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 02/18/2022] [Accepted: 03/04/2022] [Indexed: 01/03/2023]
Abstract
Huntington's disease is the most frequent autosomal dominant neurodegenerative disorder; however, no disease-modifying interventions are available for patients with this disease. The molecular pathogenesis of Huntington's disease is complex, with toxicity that arises from full-length expanded huntingtin and N-terminal fragments of huntingtin, which are both prone to misfolding due to proteolysis; aberrant intron-1 splicing of the HTT gene; and somatic expansion of the CAG repeat in the HTT gene. Potential interventions for Huntington's disease include therapies targeting huntingtin DNA and RNA, clearance of huntingtin protein, DNA repair pathways, and other treatment strategies targeting inflammation and cell replacement. The early termination of trials of the antisense oligonucleotide tominersen suggest that it is time to reflect on lessons learned, where the field stands now, and the challenges and opportunities for the future.
Collapse
Affiliation(s)
- Sarah J Tabrizi
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK.
| | - Carlos Estevez-Fraga
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | | | - Michael D Flower
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Rachael I Scahill
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Edward J Wild
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | | | - Cristina Sampaio
- CHDI Management, CHDI Foundation Los Angeles, CA, USA; Laboratory of Clinical Pharmacology, Faculdade de Medicina de Lisboa, Lisbon, Portugal
| | - Anne E Rosser
- BRAIN unit, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Blair R Leavitt
- Centre for Huntington's disease, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
18
|
Guo J, Jiang Z, Biswal BB, Zhou B, Xie D, Gao Q, Sheng W, Chen H, Zhang Y, Fan Y, Wang J, Liu C, Chen H. Hypothalamic Atrophy, Expanded
CAG
Repeat, and Low Body Mass Index in Spinocerebellar Ataxia Type 3. Mov Disord 2022; 37:1541-1546. [PMID: 35426475 DOI: 10.1002/mds.29029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 11/08/2022] Open
Affiliation(s)
- Jing Guo
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital University of Electronic Science and Technology of China Chengdu China
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology University of Electronic Science and Technology of China Chengdu China
- Department of Radiology, Southwest Hospital Army Medical University (Third Military Medical University) Chongqing China
| | - Zhouyu Jiang
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology University of Electronic Science and Technology of China Chengdu China
- MOE Key Lab for Neuroinformation, High‐Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province University of Electronic Science and Technology of China Chengdu China
| | - Bharat B. Biswal
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology University of Electronic Science and Technology of China Chengdu China
- Department of Biomedical Engineering New Jersey Institute of Technology Newark New Jersey USA
| | - Bo Zhou
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital University of Electronic Science and Technology of China Chengdu China
| | - Dongjing Xie
- Department of Neurology, Xinqiao Hospital and The Second Affiliated Hospital Army Medical University (Third Military Medical University) Chongqing China
| | - Qing Gao
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology University of Electronic Science and Technology of China Chengdu China
- MOE Key Lab for Neuroinformation, High‐Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province University of Electronic Science and Technology of China Chengdu China
| | - Wei Sheng
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology University of Electronic Science and Technology of China Chengdu China
- MOE Key Lab for Neuroinformation, High‐Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province University of Electronic Science and Technology of China Chengdu China
| | - Hui Chen
- Department of Radiology, Southwest Hospital Army Medical University (Third Military Medical University) Chongqing China
| | - Yuhan Zhang
- Department of Radiology, Southwest Hospital Army Medical University (Third Military Medical University) Chongqing China
| | - Yunshuang Fan
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology University of Electronic Science and Technology of China Chengdu China
- MOE Key Lab for Neuroinformation, High‐Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province University of Electronic Science and Technology of China Chengdu China
| | - Jian Wang
- Department of Radiology, Southwest Hospital Army Medical University (Third Military Medical University) Chongqing China
| | - Chen Liu
- Department of Radiology, Southwest Hospital Army Medical University (Third Military Medical University) Chongqing China
| | - Huafu Chen
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital University of Electronic Science and Technology of China Chengdu China
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology University of Electronic Science and Technology of China Chengdu China
- Department of Radiology, Southwest Hospital Army Medical University (Third Military Medical University) Chongqing China
- MOE Key Lab for Neuroinformation, High‐Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province University of Electronic Science and Technology of China Chengdu China
| |
Collapse
|
19
|
Abstract
Endogenous biological clocks, orchestrated by the suprachiasmatic nucleus, time the circadian rhythms that synchronize physiological and behavioural functions in humans. The circadian system influences most physiological processes, including sleep, alertness and cognitive performance. Disruption of circadian homeostasis has deleterious effects on human health. Neurodegenerative disorders involve a wide range of symptoms, many of which exhibit diurnal variations in frequency and intensity. These disorders also disrupt circadian homeostasis, which in turn has negative effects on symptoms and quality of life. Emerging evidence points to a bidirectional relationship between circadian homeostasis and neurodegeneration, suggesting that circadian function might have an important role in the progression of neurodegenerative disorders. Therefore, the circadian system has become an attractive target for research and clinical care innovations. Studying circadian disruption in neurodegenerative disorders could expand our understanding of the pathophysiology of neurodegeneration and facilitate the development of novel, circadian-based interventions for these disabling disorders. In this Review, we discuss the alterations to the circadian system that occur in movement (Parkinson disease and Huntington disease) and cognitive (Alzheimer disease and frontotemporal dementia) neurodegenerative disorders and provide directions for future investigations in this field.
Collapse
|
20
|
Gabery S, Kwa JE, Cheong RY, Baldo B, Ferrari Bardile C, Tan B, McLean C, Georgiou-Karistianis N, Poudel GR, Halliday G, Pouladi MA, Petersén Å. Early white matter pathology in the fornix of the limbic system in Huntington disease. Acta Neuropathol 2021; 142:791-806. [PMID: 34448021 PMCID: PMC8500909 DOI: 10.1007/s00401-021-02362-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/09/2021] [Accepted: 08/18/2021] [Indexed: 12/13/2022]
Abstract
Huntington disease (HD) is a fatal neurodegenerative disorder caused by an expanded CAG repeat in the huntingtin (HTT) gene. The typical motor symptoms have been associated with basal ganglia pathology. However, psychiatric and cognitive symptoms often precede the motor component and may be due to changes in the limbic system. Recent work has indicated pathology in the hypothalamus in HD but other parts of the limbic system have not been extensively studied. Emerging evidence suggests that changes in HD also include white matter pathology. Here we investigated if the main white matter tract of the limbic system, the fornix, is affected in HD. We demonstrate that the fornix is 34% smaller already in prodromal HD and 41% smaller in manifest HD compared to controls using volumetric analyses of MRI of the IMAGE-HD study. In post-mortem fornix tissue from HD cases, we confirm the smaller fornix volume in HD which is accompanied by signs of myelin breakdown and reduced levels of the transcription factor myelin regulating factor but detect no loss of oligodendrocytes. Further analyses using RNA-sequencing demonstrate downregulation of oligodendrocyte identity markers in the fornix of HD cases. Analysis of differentially expressed genes based on transcription-factor/target-gene interactions also revealed enrichment for binding sites of SUZ12 and EZH2, components of the Polycomb Repressive Complex 2, as well as RE1 Regulation Transcription Factor. Taken together, our data show that there is early white matter pathology of the fornix in the limbic system in HD likely due to a combination of reduction in oligodendrocyte genes and myelin break down.
Collapse
Affiliation(s)
- Sanaz Gabery
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, BMC D11, 22184, Lund, Sweden
| | - Jing Eugene Kwa
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
| | - Rachel Y Cheong
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, BMC D11, 22184, Lund, Sweden
| | - Barbara Baldo
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, BMC D11, 22184, Lund, Sweden
- Evotec SE, HD Research and Translational Sciences, 22419, Hamburg, Germany
| | - Costanza Ferrari Bardile
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
- Department of Medical Genetics, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, V5Z 4H4, Canada
| | - Brendan Tan
- School of Psychological Sciences, Monash University, Clayton, VIC, 3180, Australia
| | - Catriona McLean
- Department of Pathology, Alfred Hospital, Melbourne, VIC, Australia
| | | | - Govinda R Poudel
- School of Psychological Sciences, Monash University, Clayton, VIC, 3180, Australia
| | - Glenda Halliday
- The Brain and Mind Centre and Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Mahmoud A Pouladi
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
- Department of Medical Genetics, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, V5Z 4H4, Canada
- Department of Physiology, National University of Singapore (NUS), Singapore, 117597, Singapore
| | - Åsa Petersén
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, BMC D11, 22184, Lund, Sweden.
| |
Collapse
|
21
|
Abstract
Neurodegenerative diseases encompass a large group of conditions that are clinically and pathologically diverse yet are linked by a shared pathology of misfolded proteins. The accumulation of insoluble aggregates is accompanied by a progressive loss of vulnerable neurons. For some patients, the symptoms are motor focused (ataxias), while others experience cognitive and psychiatric symptoms (dementias). Among the shared symptoms of neurodegenerative diseases is a disruption of the sleep/wake cycle that occurs early in the trajectory of the disease and may be a risk factor for disease development. In many cases, the disruption in the timing of sleep and other rhythmic physiological markers immediately raises the possibility of neurodegeneration-driven disruption of the circadian timing system. The aim of this Review is to summarize the evidence supporting the hypothesis that circadian disruption is a core symptom within neurodegenerative diseases, including Alzheimer's disease, Huntington's disease, and Parkinson's disease, and to discuss the latest progress in this field. The Review discusses evidence that neurodegenerative processes may disrupt the structure and function of the circadian system and describes circadian-based interventions as well as timed drug treatments that may improve a wide range of symptoms associated with neurodegenerative disorders. It also identifies key gaps in our knowledge.
Collapse
|
22
|
Ogilvie AC, Nopoulos PC, Schultz JL. Sleep disturbances by disease type and stage in Huntington's disease. Parkinsonism Relat Disord 2021; 91:13-18. [PMID: 34450461 DOI: 10.1016/j.parkreldis.2021.08.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Sleep disturbances are a common symptom in patients with Huntington's disease (HD). However, it is unclear when in the disease course of HD sleep disturbances become more frequent compared to the general population. This study investigated the frequency and odds of developing sleep disturbances between adults with HD or at-risk for HD and non-HD controls. METHODS Participants from the Enroll-HD study were split by both disease type and disease severity using CAG length, diagnostic confidence level, and total functional capacity score. Multivariate logistic regression was used to calculate odds ratios adjusted for age, sex, tobacco and alcohol use, depression and psychosis scores, and cognition to compare HD groups to non-HD controls. Cox proportional hazards models and Kaplan Meier curves were used to determine differences in probabilities of developing sleep disturbances and how sleep disturbances are related to age at motor onset. RESULTS There were significant differences between HD participants and non-HD controls in both the disease type and disease stage analyses (p < 0.001). The odds of a sleep disturbance increased with worsening disease stage and was highest in those with juvenile HD. The development of a sleep disorder in manifest HD participants was observed to be around the time of disease onset. CONCLUSIONS Sleep disturbances are more frequent in HD patients than those without HD. There are also differences based on disease type and stage. This is supplemented by the finding that the onset of sleep disturbances occurs near the time of motor onset of HD.
Collapse
Affiliation(s)
- Amy C Ogilvie
- Department of Psychiatry, The Carver College of Medicine at the University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA; Department of Epidemiology, The College of Public Health at the University of Iowa, 145 N Riverside Drive, Iowa City, IA, 52242, USA.
| | - Peg C Nopoulos
- Department of Psychiatry, The Carver College of Medicine at the University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA; Department of Neurology, The Carver College of Medicine at the University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA; Stead Family Department of Pediatrics, The University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA, 52242, USA.
| | - Jordan L Schultz
- Department of Psychiatry, The Carver College of Medicine at the University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA; Department of Neurology, The Carver College of Medicine at the University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA; Division of Pharmacy Practice and Sciences, The College of Pharmacy at the University of Iowa, 180 South Grand Avenue, Iowa City, IA, 52242, USA.
| |
Collapse
|
23
|
Chuang CL, Demontis F. Systemic manifestation and contribution of peripheral tissues to Huntington's disease pathogenesis. Ageing Res Rev 2021; 69:101358. [PMID: 33979693 DOI: 10.1016/j.arr.2021.101358] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 03/23/2021] [Accepted: 05/07/2021] [Indexed: 12/11/2022]
Abstract
Huntington disease (HD) is an autosomal dominant neurodegenerative disease that is caused by expansion of cytosine/adenosine/guanine repeats in the huntingtin (HTT) gene, which leads to a toxic, aggregation-prone, mutant HTT-polyQ protein. Beyond the well-established mechanisms of HD progression in the central nervous system, growing evidence indicates that also peripheral tissues are affected in HD and that systemic signaling originating from peripheral tissues can influence the progression of HD in the brain. Herein, we review the systemic manifestation of HD in peripheral tissues, and the impact of systemic signaling on HD pathogenesis. Mutant HTT induces a body wasting syndrome (cachexia) primarily via its activity in skeletal muscle, bone, adipose tissue, and heart. Additional whole-organism effects induced by mutant HTT include decline in systemic metabolic homeostasis, which stems from derangement of pancreas, liver, gut, hypothalamic-pituitary-adrenal axis, and circadian functions. In addition to spreading via the bloodstream and a leaky blood brain barrier, HTT-polyQ may travel long distance via its uptake by neurons and its axonal transport from the peripheral to the central nervous system. Lastly, signaling factors that are produced and/or secreted in response to therapeutic interventions such as exercise or in response to mutant HTT activity in peripheral tissues may impact HD. In summary, these studies indicate that HD is a systemic disease that is influenced by intertissue signaling and by the action of pathogenic HTT in peripheral tissues. We propose that treatment strategies for HD should include the amelioration of HD symptoms in peripheral tissues. Moreover, harnessing signaling between peripheral tissues and the brain may provide a means for reducing HD progression in the central nervous system.
Collapse
|
24
|
Wu X, Bai F, Wang Y, Zhang L, Liu L, Chen Y, Li H, Zhang T. Circadian Rhythm Disorders and Corresponding Functional Brain Abnormalities in Young Female Nurses: A Preliminary Study. Front Neurol 2021; 12:664610. [PMID: 33995261 PMCID: PMC8120025 DOI: 10.3389/fneur.2021.664610] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/08/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: Shift work is associated with a decrease in melatonin level and perturbation of the circadian rhythm; however, it is unknown if these lead to functional brain changes. In this study, we investigated whether circadian rhythm disorders caused by shift work are related to changes in brain functional connectivity (FC) and regional homogeneity (ReHo) using whole-brain resting-state functional magnetic resonance imaging (fMRI). Methods: This prospective case-control study included nine female night shift nurses and nine age-matched female day work nurses with normal sleep rhythms. To assess sleep quality and mood, participants were asked to complete questionnaires. Serum melatonin and cortisol levels were measured. ReHo of whole-brain resting-state function and seed-based FC of the bilateral hypothalamus were compared between groups. Variables that differed significantly between groups were used to examine the association between questionnaire scores and hormone levels and fMRI data. Results: The night shift nurses had significantly lower sleep quality and melatonin levels; lower ReHo activation in the bilateral cerebellar hemisphere and higher ReHo in the bilateral occipital lobe and left parietal lobe; and higher FC from the hypothalamus to the right cingulate gyrus, right putamen, and vermis than did the day shift nurses. Activation of the right cerebellar hemisphere left superior parietal gyrus, and the right superior occipital gyrus was correlated with sleep quality scores. Moreover, activation of the right cerebellar hemisphere (r = 0.583, P = 0.011) was correlated with melatonin levels, and higher sleepiness scores were associated with stronger FC between the hypothalamus and vermis (r = 0.501, P = 0.034). Conclusions: Circadian rhythm disorder caused by night shift work can lead to a decrease in sleep quality and melatonin level, as well as a series of changes in brain FC and ReHo.
Collapse
Affiliation(s)
- Xiaoli Wu
- Department of Neurorehabilitation, Rehabilitation Medicine of Capital Medical University, China Rehabilitation Research Centre, Beijing, China
| | - Fan Bai
- China Rehabilitation Science Institute of China Rehabilitation Research Centre, Beijing, China.,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Yunlei Wang
- China Rehabilitation Science Institute of China Rehabilitation Research Centre, Beijing, China.,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Lu Zhang
- Department of Neurorehabilitation, Rehabilitation Medicine of Capital Medical University, China Rehabilitation Research Centre, Beijing, China
| | - Lixu Liu
- Department of Neurorehabilitation, Rehabilitation Medicine of Capital Medical University, China Rehabilitation Research Centre, Beijing, China
| | - Yudong Chen
- Department of Neurorehabilitation, Rehabilitation Medicine of Capital Medical University, China Rehabilitation Research Centre, Beijing, China
| | - Hanzhi Li
- Department of Neurorehabilitation, Rehabilitation Medicine of Capital Medical University, China Rehabilitation Research Centre, Beijing, China
| | - Tong Zhang
- Department of Neurorehabilitation, Rehabilitation Medicine of Capital Medical University, China Rehabilitation Research Centre, Beijing, China.,China Rehabilitation Science Institute of China Rehabilitation Research Centre, Beijing, China.,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| |
Collapse
|
25
|
Hair and salivary cortisol and their relationship with lifestyle, mood and cognitive outcomes in premanifest Huntington's disease. Sci Rep 2021; 11:5464. [PMID: 33750863 PMCID: PMC7943576 DOI: 10.1038/s41598-021-84726-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 02/19/2021] [Indexed: 11/09/2022] Open
Abstract
Salivary cortisol dysrhythmias have been reported in some, but not all studies assessing hypothalamic-pituitary-adrenal (HPA) axis function in Huntington's disease (HD). These differences are presumed to be due to environmental influences on temporal salivary cortisol measurement. Further exploration of HPA-axis function using a more stable and longer-term measure, such as hair cortisol, is needed to confirm earlier findings. This study aimed to evaluate hair and salivary cortisol concentrations and their associations with clinical and lifestyle outcomes in individuals with premanifest HD (n = 26) compared to healthy controls (n = 14). Participants provided saliva and hair samples and data were collected on clinical disease outcomes, mood, cognition, physical activity, cognitive reserve, sleep quality and social network size to investigate relationships between clinical and lifestyle outcomes and cortisol concentrations. Hair and salivary cortisol concentrations did not significantly differ between the premanifest HD and control groups. No significant associations were observed between hair or salivary cortisol concentrations and cognitive, mood or lifestyle outcomes. However, hair cortisol concentrations were significantly associated with disease outcomes in individuals with premanifest HD. Significant associations between hair cortisol concentrations and measures of disease burden and onset may suggest a potential disease marker and should be explored longitudinally in a larger sample of individuals with HD.
Collapse
|
26
|
Tortelli R, Rodrigues FB, Wild EJ. The use of wearable/portable digital sensors in Huntington's disease: A systematic review. Parkinsonism Relat Disord 2021; 83:93-104. [PMID: 33493786 PMCID: PMC7957324 DOI: 10.1016/j.parkreldis.2021.01.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/13/2020] [Accepted: 01/08/2021] [Indexed: 01/26/2023]
Abstract
In chronic neurological conditions, wearable/portable devices have potential as innovative tools to detect subtle early disease manifestations and disease fluctuations for the purpose of clinical diagnosis, care and therapeutic development. Huntington's disease (HD) has a unique combination of motor and non-motor features which, combined with recent and anticipated therapeutic progress, gives great potential for such devices to prove useful. The present work aims to provide a comprehensive account of the use of wearable/portable devices in HD and of what they have contributed so far. We conducted a systematic review searching MEDLINE, Embase, and IEEE Xplore. Thirty references were identified. Our results revealed large variability in the types of sensors used, study design, and the measured outcomes. Digital technologies show considerable promise for therapeutic research and clinical management of HD. However, more studies with standardized devices and harmonized protocols are needed to optimize the potential applicability of wearable/portable devices in HD. Wearable/portable sensors have been proposed to detect and quantify manifestations of many neurodegenerative diseases. No systematic review so far has examined their use in Huntington's disease (HD). This work draws a broad picture of the digital wearable-based landscape in HD. The utility of wearables in clinical practice and therapeutic research still needs to be proved. Collaborative efforts are needed to further investigate their clinical use in HD.
Collapse
Affiliation(s)
- Rosanna Tortelli
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Filipe B Rodrigues
- 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.
| |
Collapse
|
27
|
Voysey Z, Fazal SV, Lazar AS, Barker RA. The sleep and circadian problems of Huntington's disease: when, why and their importance. J Neurol 2020; 268:2275-2283. [PMID: 33355880 PMCID: PMC8179890 DOI: 10.1007/s00415-020-10334-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/15/2020] [Accepted: 11/19/2020] [Indexed: 11/30/2022]
Abstract
Introduction Mounting evidence supports the existence of an important feedforward cycle between sleep and neurodegeneration, wherein neurodegenerative diseases cause sleep and circadian abnormalities, which in turn exacerbate and accelerate neurodegeneration. If so, sleep therapies bear important potential to slow progression in these diseases. Findings This cycle is challenging to study, as its bidirectional nature renders cause difficult to disentangle from effect. Likewise, well-controlled intervention studies are often impractical in the setting of established neurodegenerative disease. It is this that makes understanding sleep and circadian abnormalities in Huntington’s disease (HD) important: as a monogenic fully penetrant neurodegenerative condition presenting in midlife, it provides a rare opportunity to study sleep and circadian abnormalities longitudinally, prior to and throughout disease manifestation, and in the absence of confounds rendered by age and comorbidities. It also provides potential to trial sleep therapies at a preclinical or early disease stage. Moreover, its monogenic nature facilitates the development of transgenic animal models through which to run parallel pre-clinical studies. HD, therefore, provides a key model condition through which to gain new insights into the sleep-neurodegeneration interface. Conclusions Here, we begin by summarising contemporary knowledge of sleep abnormalities in HD, and consider how well these parallel those of Alzheimer’s and Parkinson’s as more common neurodegenerative conditions. We then discuss what is currently known of the sleep-neurodegeneration cyclical relationship in HD. We conclude by outlining key directions of current and future investigation by which to advance the sleep-neurodegeneration field via studies in HD.
Collapse
Affiliation(s)
- Z Voysey
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK.
| | - S V Fazal
- 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, WT-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
28
|
Disruption of zinc transporter ZnT3 transcriptional activity and synaptic vesicular zinc in the brain of Huntington's disease transgenic mouse. Cell Biosci 2020; 10:106. [PMID: 32944220 PMCID: PMC7488477 DOI: 10.1186/s13578-020-00459-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 08/05/2020] [Indexed: 12/27/2022] Open
Abstract
Background Huntington’s disease (HD) is a neurodegenerative disease that involves a complex combination of psychiatric, cognitive and motor impairments. Synaptic dysfunction has been implicated in HD pathogenesis. However, the mechanisms have not been clearly delineated. Synaptic vesicular zinc is closely linked to modulating synaptic transmission and maintaining cognitive ability. It is significant to assess zinc homeostasis for further revealing the pathogenesis of synaptic dysfunction and cognitive impairment in HD. Results Histochemical staining by autometallography indicated that synaptic vesicular zinc was decreased in the hippocampus, cortex and striatum of N171-82Q HD transgenic mice. Analyses by immunohistochemistry, Western blot and RT-PCR found that the expression of zinc transporter 3 (ZnT3) required for transport of zinc into synaptic vesicles was obviously reduced in these three brain regions of the HD mice aged from 14 to 20 weeks and BHK cells expressing mutant huntingtin. Significantly, dual-luciferase reporter gene and chromatin immunoprecipitation assays demonstrated that transcription factor Sp1 could activate ZnT3 transcription via its binding to the GC boxes in ZnT3 promoter. Moreover, mutant huntingtin was found to inhibit the binding of Sp1 to the promoter of ZnT3 and down-regulate ZnT3 expression, and the decline in ZnT3 expression could be ameliorated through overexpression of Sp1. Conclusions This is first study to reveal a significant loss of synaptic vesicular zinc and a decline in ZnT3 transcriptional activity in the HD transgenic mice. Our work sheds a novel mechanistic insight into pathogenesis of HD that mutant huntingtin down-regulates expression of ZnT3 through inhibiting binding of Sp1 to the promoter of ZnT3 gene, causing disruption of synaptic vesicular zinc homeostasis. Disrupted vesicular zinc ultimately leads to early synaptic dysfunction and cognitive deficits in HD. It is also suggested that maintaining normal synaptic vesicular zinc concentration is a potential therapeutic strategy for HD.
Collapse
|
29
|
Cheong RY, Tonetto S, von Hörsten S, Petersén Å. Imbalance of the oxytocin-vasopressin system contributes to the neuropsychiatric phenotype in the BACHD mouse model of Huntington disease. Psychoneuroendocrinology 2020; 119:104773. [PMID: 32590293 DOI: 10.1016/j.psyneuen.2020.104773] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/30/2020] [Accepted: 06/08/2020] [Indexed: 12/12/2022]
Abstract
Neuropsychiatric disturbances with altered social cognition, depression and anxiety are among the most debilitating early features in the fatal neurodegenerative disorder Huntington disease (HD) which is caused by an expanded CAG repeat in the huntingtin gene. The underlying neurobiological mechanisms are not known. Neuropathological analyses of postmortem human HD hypothalamic tissue have demonstrated loss of the neuropeptides oxytocin and vasopressin. The dynamic interplay between these neuropeptides is crucial for modulating emotional and social behavior but its role in HD is unclear. In the present study, we have investigated the effect of expressing the mutant huntingtin gene on the development of behavioral changes using the transgenic BACHD mouse model at different ages. We show for the first time that BACHD mice exhibit deficits in social behavior with parallel aberrations in the balance of the oxytocin-vasopressin system. Importantly, our data also show that restoration of the interplay within the system with an acute dose of intranasal oxytocin immediately prior to behavioral testing can rescue the depressive-like phenotype but not anxiety-like behavior in this transgenic model. These findings demonstrate that imbalances in the oxytocin-vasopressin interplay contribute to the neuropsychiatric component of HD and suggest that interventions aimed at restoring the blunted levels of oxytocin may confer therapeutic benefits for this disease.
Collapse
Affiliation(s)
- Rachel Y Cheong
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden.
| | - Simone Tonetto
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden
| | - Stephan von Hörsten
- Department of Experimental Therapy and Preclinical Center, Friedrich-Alexander-University, 91054 Erlangen, Germany
| | - Åsa Petersén
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden
| |
Collapse
|
30
|
Kennaway DJ. Measuring melatonin by immunoassay. J Pineal Res 2020; 69:e12657. [PMID: 32281677 DOI: 10.1111/jpi.12657] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/29/2020] [Accepted: 03/31/2020] [Indexed: 12/11/2022]
Abstract
The pineal gland hormone melatonin continues to be of considerable interest to biomedical researchers. Of particular interest is the pattern of secretion of melatonin in relation to sleep timing as well as its potential role in certain diseases. Measuring melatonin in biological fluids such as blood and saliva presents particular methodological challenges since the production and secretion of the hormone are known to be extremely low during the light phase in almost all situations. Active secretion only occurs around the time of lights out in a wide range of species. The challenge then is to develop practical high-throughput assays that are sufficiently sensitive and accurate enough to detect levels of melatonin less than 1 pg/mL in biological fluids. Mass spectrometry assays have been developed that achieve the required sensitivity, but are really not practical or even widely available to most researchers. Melatonin radioimmunoassays and ELISA have been developed and are commercially available. But the quality of the results that are being published is very variable, partly not only because of poor experimental designs, but also because of poor assays. In this review, I discuss issues around the design of studies involving melatonin measurement. I then provide a critical assessment of 21 immunoassay kits marketed by 11 different companies with respect to validation, specificity and sensitivity. Technical managers of the companies were contacted in an attempt to obtain information not available online or in kit inserts. A search of the literature was also conducted to uncover papers that have reported the use of these assays, and where possible, both daytime and night-time plasma or saliva melatonin concentrations were extracted and tabulated. The results of the evaluations are disturbing, with many kits lacking any validation studies or using inadequate validation methods. Few assays have been properly assessed for specificity, while others report cross-reaction profiles that can be expected to result in over estimation of the melatonin levels. Some assays are not fit for purpose because they are not sensitive enough to determine plasma or saliva DLMO of 10 and 3 pg/mL, respectively. Finally, some assays produce unrealistically high daytime melatonin levels in humans and laboratory animals in the order of hundreds of pg/mL. In summary, this review provides a comprehensive and unique assessment of the current commercial melatonin immunoassays and their use in publications. It provides researchers new to the field with the information they need to design valid melatonin studies from both the perspective of experimental/clinical trial design and the best assay methodologies. It will also hopefully help journal editors and reviewers who may not be fully aware of the pitfalls of melatonin measurement make better informed decisions on publication acceptability.
Collapse
Affiliation(s)
- David J Kennaway
- Robinson Research Institute and Adelaide School of Medicine, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
31
|
Circadian alterations in patients with neurodegenerative diseases: Neuropathological basis of underlying network mechanisms. Neurobiol Dis 2020; 144:105029. [PMID: 32736083 DOI: 10.1016/j.nbd.2020.105029] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/19/2020] [Accepted: 07/23/2020] [Indexed: 01/16/2023] Open
Abstract
Circadian organization of physiology and behavior is an important biological process that allows organisms to anticipate and prepare for daily changes and demands. Disruptions in this system precipitates a wide range of health issues. In patients with neurodegenerative diseases, alterations of circadian rhythms are among the most common and debilitating symptoms. Although a growing awareness of these symptoms has occurred during the last decade, their underlying neuropathophysiological circuitry remains poorly understood and consequently no effective therapeutic strategies are available to alleviate these health issues. Recent studies have examined the neuropathological status of the different neural components of the circuitry governing the generation of circadian rhythms in neurodegenerative diseases. In this review, we will dissect the potential contribution of dysfunctions in the different nodes of this circuitry to circadian alterations in patients with neurodegenerative diseases. A deeper understanding of these mechanisms will provide not only a better understanding of disease neuro-pathophysiology, but also hold the promise for developing effective and mechanisms-based therapies.
Collapse
|
32
|
Cheong RY, Gabery S, Petersén Å. The Role of Hypothalamic Pathology for Non-Motor Features of Huntington's Disease. J Huntingtons Dis 2020; 8:375-391. [PMID: 31594240 PMCID: PMC6839491 DOI: 10.3233/jhd-190372] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Huntington’s disease (HD) is a fatal genetic neurodegenerative disorder. It has mainly been considered a movement disorder with cognitive symptoms and these features have been associated with pathology of the striatum and cerebral cortex. Importantly, individuals with the mutant huntingtin gene suffer from a spectrum of non-motor features often decades before the motor disorder manifests. These symptoms and signs include a range of psychiatric symptoms, sleep problems and metabolic changes with weight loss particularly in later stages. A higher body mass index at diagnosis is associated with slower disease progression. The common psychiatric symptom of apathy progresses with the disease. The fact that non-motor features are present early in the disease and that they show an association to disease progression suggest that unravelling the underlying neurobiological mechanisms may uncover novel targets for early disease intervention and better symptomatic treatment. The hypothalamus and the limbic system are important brain regions that regulate emotion, social cognition, sleep and metabolism. A number of studies using neuroimaging, postmortem human tissue and genetic manipulation in animal models of the disease has collectively shown that the hypothalamus and the limbic system are affected in HD. These findings include the loss of neuropeptide-expressing neurons such as orexin (hypocretin), oxytocin, vasopressin, somatostatin and VIP, and increased levels of SIRT1 in distinct nuclei of the hypothalamus. This review provides a summary of the results obtained so far and highlights the potential importance of these changes for the understanding of non-motor features in HD.
Collapse
Affiliation(s)
- Rachel Y Cheong
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Sanaz Gabery
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Åsa Petersén
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
33
|
Memon AA, Coleman JJ, Amara AW. Effects of exercise on sleep in neurodegenerative disease. Neurobiol Dis 2020; 140:104859. [PMID: 32243913 PMCID: PMC7497904 DOI: 10.1016/j.nbd.2020.104859] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/22/2020] [Accepted: 03/30/2020] [Indexed: 02/07/2023] Open
Abstract
As the population ages, the incidence and prevalence of neurodegenerative disorders will continue to increase. Persons with neurodegenerative disease frequently experience sleep disorders, which not only affect quality of life, but potentially accelerate progression of the disease. Unfortunately, pharmacological interventions are often futile or have adverse effects. Therefore, investigation of non-pharmacological interventions has the potential to expand the treatment landscape for these disorders. The last decade has observed increasing recognition of the beneficial role of exercise in brain diseases, and neurodegenerative disorders in particular. In this review, we will focus on the therapeutic role of exercise for sleep dysfunction in four neurodegenerative diseases, namely Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. Available data suggest that exercise may have the potential to improve sleep disorders and attenuate neurodegeneration, particularly in Alzheimer's disease and Parkinson's disease. However, additional research is required in order to understand the most effective exercise therapy for these indications; the best way to monitor the response to interventions; the influence of exercise on sleep dysfunction in Huntington's disease and amyotrophic lateral sclerosis; and the mechanisms underlying exercise-induced sleep modifications.
Collapse
Affiliation(s)
- Adeel A Memon
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294, United States of America
| | - Juliana J Coleman
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294, United States of America
| | - Amy W Amara
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294, United States of America; UAB Center for Exercise Medicine, Birmingham, AL 35205, United States of America; UAB Sleep and Circadian Research Core, United States of America.
| |
Collapse
|
34
|
Bartlett DM, Dominguez D JF, Lazar AS, Kordsachia CC, Rankin TJ, Lo J, Govus AD, Power BD, Lampit A, Eastwood PR, Ziman MR, Cruickshank TM. Multidisciplinary rehabilitation reduces hypothalamic grey matter volume loss in individuals with preclinical Huntington's disease: A nine-month pilot study. J Neurol Sci 2019; 408:116522. [PMID: 31665619 DOI: 10.1016/j.jns.2019.116522] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/04/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Hypothalamic pathology is a well-documented feature of Huntington's disease (HD) and is believed to contribute to circadian rhythm and habitual sleep disturbances. Currently, no therapies exist to combat hypothalamic changes, nor circadian rhythm and habitual sleep disturbances in HD. OBJECTIVE To evaluate the effects of multidisciplinary rehabilitation on hypothalamic volume, brain-derived neurotrophic factor (BDNF), circadian rhythm and habitual sleep in individuals with preclinical HD. METHODS Eighteen individuals with HD (ten premanifest and eight prodromal) undertook a nine-month multidisciplinary rehabilitation intervention (intervention group), which included exercise, cognitive and dual task training and social events, and were compared to a community sample of eleven individuals with premanifest HD receiving no intervention (control group). Hypothalamic volume, serum BDNF, salivary cortisol and melatonin concentrations, subjective sleep quality, daytime somnolence, habitual sleep-wake patterns, stress and anxiety and depression symptomatology were evaluated. RESULTS Hypothalamus grey matter volume loss was significantly attenuated in the intervention group compared to the control group after controlling for age, gender, Unified Huntington's Disease Rating Scale-Total Motor Score and number of cytosine-adenine-guanine repeats. Serum BDNF levels were maintained in the intervention group, but decreased in the control group following the study period. Both groups exhibited decreases in cortisol and melatonin concentrations. No changes were observed in sleep or mood outcomes. CONCLUSIONS This exploratory study provides evidence that multidisciplinary rehabilitation can reduce hypothalamic volume loss and maintain peripheral BDNF levels in individuals with preclinical HD but may not impact on circadian rhythm. Larger, randomised controlled trials are required to confirm these findings.
Collapse
Affiliation(s)
- Danielle M Bartlett
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.
| | - Juan F Dominguez D
- Cognition and Emotion Research Centre & Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, Australia
| | - Alpar S Lazar
- Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, Norfolk, United Kingdom
| | - Catarina C Kordsachia
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
| | - Tim J Rankin
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Johnny Lo
- School of Science, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Andrew D Govus
- School of Allied Health, Department of Human Services, Nutrition and Sport, La Trobe University, Melbourne, Victoria, Australia
| | - Brian D Power
- School of Medicine, The University of Notre Dame, Fremantle, Western Australia, Australia
| | - Amit Lampit
- Department of Psychiatry, University of Melbourne, Victoria, Australia; Department of Neurology, Charité - Universitätsmedizin Berlin, Germany
| | - Peter R Eastwood
- Centre for Sleep Science, School of Human Sciences, Faculty of Science, University of Western Australia, Crawley, Western Australia, Australia
| | - Mel R Ziman
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia; School of Biomedical Science, University of Western Australia, Crawley, Western Australia, Australia
| | - Travis M Cruickshank
- Exercise Medicine Research Institute, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia; Perron Institute for Neurological and Translational Science, Perth, Western Australia, Australia
| |
Collapse
|