1
|
Latha Laxmi IP, Tamizhselvi R. Epigenetic events influencing the biological clock: Panacea for neurodegeneration. Heliyon 2024; 10:e38836. [PMID: 39430507 PMCID: PMC11489350 DOI: 10.1016/j.heliyon.2024.e38836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 09/28/2024] [Accepted: 10/01/2024] [Indexed: 10/22/2024] Open
Abstract
The human biological clock is the 24-h internal molecular network of circadian genes in synchronization with other cells in response to external stimuli. The rhythmicity of the clock genes is maintained by positive and negative transcriptional feedback loops coordinating the 24-h oscillation in different tissues. The superchiasmatic nucleus, the central pacemaker of the biological clock diminishes with aging causing alterations in the clock rhythmicity leading to the onset of neurodegenerative diseases mainly Alzheimer's disease, Parkinson's disease, and Huntington's disease. Studies have shown that brain and muscle Arnt -like 1 (Bmal1) and Circadian Locomotor Output Cycles Kaput (Clock) gene expression is altered in the onset of neurodegeneration. One of the major symptoms of neurodegeneration is changes in the sleep/wake cycle. Moreover, variations in circadian clock oscillations can happen due to lifestyle changes, addiction to alcohol, cocaine, drugs, smoking, food habits and most importantly eating and sleep/awake cycle patterns which can significantly impact the expression of circadian genes. Recent studies have focused on the molecular function of clock genes affected due to environmental cues. Epigenetic modifications are influenced by the external environmental factors. This review aims to focus on the principal mechanism of epigenetics influencing circadian rhythm disruption leading to neurodegeneration and as well as targeting the epigenetic modulators could be a novel therapeutic approach to combat neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Ramasamy Tamizhselvi
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| |
Collapse
|
2
|
Zhang W, Liu D, Yuan M, Zhu LQ. The mechanisms of mitochondrial abnormalities that contribute to sleep disorders and related neurodegenerative diseases. Ageing Res Rev 2024; 97:102307. [PMID: 38614368 DOI: 10.1016/j.arr.2024.102307] [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: 03/09/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Sleep is a highly intricate biological phenomenon, and its disorders play a pivotal role in numerous diseases. However, the specific regulatory mechanisms remain elusive. In recent years, the role of mitochondria in sleep disorders has gained considerable attention. Sleep deprivation not only impairs mitochondrial morphology but also decreases the number of mitochondria and triggers mitochondrial dysfunction. Furthermore, mitochondrial dysfunction has been implicated in the onset and progression of various sleep disorder-related neurological diseases, especially neurodegenerative conditions. Therefore, a greater understanding of the impact of sleep disorders on mitochondrial dysfunction may reveal new therapeutic targets for neurodegenerative diseases. In this review, we comprehensively summarize the recent key findings on the mechanisms underlying mitochondrial dysfunction caused by sleep disorders and their role in initiating or exacerbating common neurodegenerative diseases. In addition, we provide fresh insights into the diagnosis and treatment of sleep disorder-related diseases.
Collapse
Affiliation(s)
- Wentao Zhang
- The Second Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Dan Liu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Mei Yuan
- The Second Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Affiliated Nanhua Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| | - Ling-Qiang Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
3
|
Vohra A, Keefe P, Puthanveetil P. Altered Metabolic Signaling and Potential Therapies in Polyglutamine Diseases. Metabolites 2024; 14:320. [PMID: 38921455 PMCID: PMC11205831 DOI: 10.3390/metabo14060320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/17/2024] [Accepted: 05/27/2024] [Indexed: 06/27/2024] Open
Abstract
Polyglutamine diseases comprise a cluster of genetic disorders involving neurodegeneration and movement disabilities. In polyglutamine diseases, the target proteins become aberrated due to polyglutamine repeat formation. These aberrant proteins form the root cause of associated complications. The metabolic regulation during polyglutamine diseases is not well studied and needs more attention. We have brought to light the significance of regulating glutamine metabolism during polyglutamine diseases, which could help in decreasing the neuronal damage associated with excess glutamate and nucleotide generation. Most polyglutamine diseases are accompanied by symptoms that occur due to excess glutamate and nucleotide accumulation. Along with a dysregulated glutamine metabolism, the Nicotinamide adenine dinucleotide (NAD+) levels drop down, and, under these conditions, NAD+ supplementation is the only achievable strategy. NAD+ is a major co-factor in the glutamine metabolic pathway, and it helps in maintaining neuronal homeostasis. Thus, strategies to decrease excess glutamate and nucleotide generation, as well as channelizing glutamine toward the generation of ATP and the maintenance of NAD+ homeostasis, could aid in neuronal health. Along with understanding the metabolic dysregulation that occurs during polyglutamine diseases, we have also focused on potential therapeutic strategies that could provide direct benefits or could restore metabolic homeostasis. Our review will shed light into unique metabolic causes and into ideal therapeutic strategies for treating complications associated with polyglutamine diseases.
Collapse
Affiliation(s)
- Alisha Vohra
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA; (A.V.); (P.K.)
| | - Patrick Keefe
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA; (A.V.); (P.K.)
| | - Prasanth Puthanveetil
- College of Graduate Studies, Department of Pharmacology, Midwestern University, Downers Grove, IL 60515, USA
| |
Collapse
|
4
|
Sharma A, Narasimha K, Manjithaya R, Sheeba V. Restoration of Sleep and Circadian Behavior by Autophagy Modulation in Huntington's Disease. J Neurosci 2023; 43:4907-4925. [PMID: 37268416 PMCID: PMC10312063 DOI: 10.1523/jneurosci.1894-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/25/2023] [Accepted: 02/16/2023] [Indexed: 06/04/2023] Open
Abstract
Circadian and sleep defects are well documented in Huntington's disease (HD). Modulation of the autophagy pathway has been shown to mitigate toxic effects of mutant Huntingtin (HTT) protein. However, it is not clear whether autophagy induction can also rescue circadian and sleep defects. Using a genetic approach, we expressed human mutant HTT protein in a subset of Drosophila circadian neurons and sleep center neurons. In this context, we examined the contribution of autophagy in mitigating toxicity caused by mutant HTT protein. We found that targeted overexpression of an autophagy gene, Atg8a in male flies, induces autophagy pathway and partially rescues several HTT-induced behavioral defects, including sleep fragmentation, a key hallmark of many neurodegenerative disorders. Using cellular markers and genetic approaches, we demonstrate that indeed the autophagy pathway is involved in behavioral rescue. Surprisingly, despite behavioral rescue and evidence for the involvement of the autophagy pathway, the large visible aggregates of mutant HTT protein were not eliminated. We show that the rescue in behavior is associated with increased mutant protein aggregation and possibly enhanced output from the targeted neurons, resulting in the strengthening of downstream circuits. Overall, our study suggests that, in the presence of mutant HTT protein, Atg8a induces autophagy and improves the functioning of circadian and sleep circuits.SIGNIFICANCE STATEMENT Defects in sleep and circadian rhythms are well documented in Huntington's disease. Recent literature suggests that circadian and sleep disturbances can exacerbate neurodegenerative phenotypes. Hence, identifying potential modifiers that can improve the functioning of these circuits could greatly improve disease management. We used a genetic approach to enhance cellular proteostasis and found that overexpression of a crucial autophagy gene, Atg8a, induces the autophagy pathway in the Drosophila circadian and sleep neurons and rescues sleep and activity rhythm. We demonstrate that the Atg8a improves synaptic function of these circuits by possibly enhancing the aggregation of the mutant protein in neurons. Further, our results suggest that differences in basal levels of protein homeostatic pathways is a factor that determines selective susceptibility of neurons.
Collapse
Affiliation(s)
- Ankit Sharma
- Chronobiology and Behavioural Neurogenetics Laboratory, Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India
| | - Kavyashree Narasimha
- Chronobiology and Behavioural Neurogenetics Laboratory, Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India
| | - Ravi Manjithaya
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India
| | - Vasu Sheeba
- Chronobiology and Behavioural Neurogenetics Laboratory, Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India
| |
Collapse
|
5
|
Bains RS, Forrest H, Sillito RR, Armstrong JD, Stewart M, Nolan PM, Wells SE. Longitudinal home-cage automated assessment of climbing behavior shows sexual dimorphism and aging-related decrease in C57BL/6J healthy mice and allows early detection of motor impairment in the N171-82Q mouse model of Huntington's disease. Front Behav Neurosci 2023; 17:1148172. [PMID: 37035623 PMCID: PMC10073658 DOI: 10.3389/fnbeh.2023.1148172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/02/2023] [Indexed: 04/11/2023] Open
Abstract
Monitoring the activity of mice within their home cage is proving to be a powerful tool for revealing subtle and early-onset phenotypes in mouse models. Video-tracking, in particular, lends itself to automated machine-learning technologies that have the potential to improve the manual annotations carried out by humans. This type of recording and analysis is particularly powerful in objective phenotyping, monitoring behaviors with no experimenter intervention. Automated home-cage testing allows the recording of non-evoked voluntary behaviors, which do not require any contact with the animal or exposure to specialist equipment. By avoiding stress deriving from handling, this approach, on the one hand, increases the welfare of experimental animals and, on the other hand, increases the reliability of results excluding confounding effects of stress on behavior. In this study, we show that the monitoring of climbing on the wire cage lid of a standard individually ventilated cage (IVC) yields reproducible data reflecting complex phenotypes of individual mouse inbred strains and of a widely used model of neurodegeneration, the N171-82Q mouse model of Huntington's disease (HD). Measurements in the home-cage environment allowed for the collection of comprehensive motor activity data, which revealed sexual dimorphism, daily biphasic changes, and aging-related decrease in healthy C57BL/6J mice. Furthermore, home-cage recording of climbing allowed early detection of motor impairment in the N171-82Q HD mouse model. Integrating cage-floor activity with cage-lid activity (climbing) has the potential to greatly enhance the characterization of mouse strains, detecting early and subtle signs of disease and increasing reproducibility in preclinical studies.
Collapse
Affiliation(s)
- Rasneer S. Bains
- Mary Lyon Centre at Medical Research Council, Harwell, Oxfordshire, United Kingdom
| | - Hamish Forrest
- Mary Lyon Centre at Medical Research Council, Harwell, Oxfordshire, United Kingdom
| | | | - J. Douglas Armstrong
- Actual Analytics Ltd., Edinburgh, United Kingdom
- School of Informatics, University of Edinburgh, Edinburgh, United Kingdom
| | - Michelle Stewart
- Mary Lyon Centre at Medical Research Council, Harwell, Oxfordshire, United Kingdom
| | - Patrick M. Nolan
- Medical Research Council, Harwell Science Campus, Oxford, United Kingdom
| | - Sara E. Wells
- Mary Lyon Centre at Medical Research Council, Harwell, Oxfordshire, United Kingdom
| |
Collapse
|
6
|
Sleep-Related Changes Prior to Cognitive Dysfunction. Curr Neurol Neurosci Rep 2023; 23:177-183. [PMID: 36881255 DOI: 10.1007/s11910-023-01258-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2023] [Indexed: 03/08/2023]
Abstract
PURPOSE OF REVIEW The aim of this review is to summarize the current evidence on the relationship between sleep and cognition and present available data reporting the impact that sleep alterations may have on cognitive functions. RECENT FINDINGS Research findings support the idea that sleep is involved in cognitive processes and that altered sleep homeostasis or circadian rhythms may lead to clinical and biochemical changes associated with cognitive impairment. Evidence is particularly solid for the association between specific sleep architecture and circadian alterations and Alzheimer's disease. Sleep changes, as early manifestations or possible risk factors for neurodegeneration and cognitive decline, may be appropriate targets for interventions aiming to reduce the likelihood of dementia.
Collapse
|
7
|
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: 1.0] [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
|
8
|
Morton AJ. Sleep and Circadian Rhythm Dysfunction in Animal Models of Huntington's Disease. J Huntingtons Dis 2023; 12:133-148. [PMID: 37334613 PMCID: PMC10473141 DOI: 10.3233/jhd-230574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2023] [Indexed: 06/20/2023]
Abstract
Sleep and circadian disruption affects most individuals with Huntington's disease (HD) at some stage in their lives. Sleep and circadian dysregulation are also present in many mouse and the sheep models of HD. Here I review evidence for sleep and/or circadian dysfunction in HD transgenic animal models and discuss two key questions: 1) How relevant are such findings to people with HD, and 2) Whether or not therapeutic interventions that ameliorate deficits in animal models of HD might translate to meaningful therapies for people with HD.
Collapse
Affiliation(s)
- A. Jennifer Morton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
9
|
Owen NE, Barker RA, Voysey ZJ. Sleep Dysfunction in Huntington's Disease: Impacts of Current Medications and Prospects for Treatment. J Huntingtons Dis 2023; 12:149-161. [PMID: 37248911 PMCID: PMC10473096 DOI: 10.3233/jhd-230567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2023] [Indexed: 05/31/2023]
Abstract
Sleep dysfunction is highly prevalent in Huntington's disease (HD). Increasing evidence suggests that such dysfunction not only impairs quality of life and exacerbates symptoms but may even accelerate the underlying disease process. Despite this, current HD treatment approaches neither consider the impact of commonly used medications on sleep, nor directly tackle sleep dysfunction. In this review, we discuss approaches to these two areas, evaluating not only literature from clinical studies in HD, but also that from parallel neurodegenerative conditions and preclinical models of HD. We conclude by summarizing a hierarchical framework of current medications with regard to their impact on sleep, and by outlining key emerging sleep therapies.
Collapse
Affiliation(s)
- Natalia E. Owen
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - Roger A. Barker
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
- Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Zanna J. Voysey
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| |
Collapse
|
10
|
Sharma P, Sharma BS, Raval H, Singh V. Endocytosis of GABA receptor: Signaling in nervous system. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 196:125-139. [PMID: 36813355 DOI: 10.1016/bs.pmbts.2022.06.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
GABA (ᵞ-aminobutyric acid), is the principal neurotransmitter known for its inhibitory role in chemical synapses. Being localized primarily in the central nervous system (CNS) it maintains a balance between excitatory (regulated by another neurotransmitter, glutamate) and inhibitory impulses. GABA acts by binding to their specific receptors GABAA and GABAB when released into the post-synaptic nerve terminal. Both of these receptors are responsible for fast and slow inhibition of neurotransmission, respectively. GABAA is a ligand-gated ionopore receptor which opens the Cl- ion channel and decreases the resting potential of the membrane resulting into inhibition of the synapse. On the other hand, GABAB is a metabotropic receptor which increases the K+ ion levels preventing Ca+ ion release inhibiting the release of other neurotransmitters into the presynaptic membrane. The internalization and trafficking of these receptors is also conducted through distinct pathways and mechanism, discussed in detail in the chapter. Without the desired levels of GABA in the body, the psychological and neurological states of brain get hard to maintain. Various neurodegenerative diseases/disorders have been associated to low levels of GABA, such as anxiety, mood disorders, fear, schizophrenia, hungtington's chorea, seizures, epilepsy, etc. The allosteric sites present on GABA receptors have been proved to be potent drug targets to pacify the pathological states of these brain related disorders to an extent. Further in depth studies focussing on the subtypes of GABA receptors and their comprehensive mechanism are required to explore new drug targets and therapeutic avenues for effectual management of GABA related neurological diseases.
Collapse
Affiliation(s)
- Preeti Sharma
- Shree Vipratech Diagnostics, Dehgam, Gujarat, India.
| | - B Sharan Sharma
- Rivaara Labs, KD Hospital, Vaishnodevi Circle, Ahmedabad, Gujarat, India
| | - Hardik Raval
- Shree Vipratech Diagnostics, Dehgam, Gujarat, India
| | - Vijai Singh
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, Gujarat, India
| |
Collapse
|
11
|
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: 0] [Impact Index Per Article: 0] [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
|
12
|
Granado MDJ, Pinato L, Santiago J, Barbalho SM, Parmezzan JEL, Suzuki LM, Cabrini ML, Spressão DRMS, Carneiro de Camargo AL, Guissoni Campos LM. Melatonin receptors and Per1 expression in the inferior olivary nucleus of the Sapajus apella monkey. Front Neurosci 2022; 16:1072772. [PMID: 36605547 PMCID: PMC9809291 DOI: 10.3389/fnins.2022.1072772] [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: 10/17/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
Melatonin is a transducer of photic environmental information and participates in the synchronization of various physiological and behavioral phenomena. Melatonin can act directly in several areas of the central nervous system through its membrane receptors coupled to G protein, called MT1 and MT2 receptors. In some structures, such as the retina, hypothalamus and pars tuberalis, the expression of both melatonin receptors shows circadian variations. Melatonin can act in the synchronization of the clock proteins rhythm in these areas. Using the immunohistochemistry technique, we detected the immunoexpression of the melatonin receptors and clock genes clock protein Per1 in the inferior olivary nucleus (ION) of the Sapajus apella monkey at specific times of the light-dark phase. The mapping performed by immunohistochemistry showed expressive immunoreactivity (IR) Per1 with predominance during daytime. Both melatonin receptors were expressed in the ION without a day/night difference. The presence of both melatonin receptors and the Per1 protein in the inferior olivary nucleus can indicate a functional role not only in physiological, as in sleep, anxiety, and circadian rhythm, but also a chronobiotic role in motor control mechanisms.
Collapse
Affiliation(s)
- Marcos Donizete Junior Granado
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Medical School, University of Marilia (UNIMAR), Marília, Brazil
| | - Luciana Pinato
- Department of Speech, Language and Hearing Sciences, São Paulo State University (UNESP), Marília, Brazil
| | - Jeferson Santiago
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Medical School, University of Marilia (UNIMAR), Marília, Brazil
| | - Sandra Maria Barbalho
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Medical School, University of Marilia (UNIMAR), Marília, Brazil
| | - Jessica Ellen Lima Parmezzan
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Medical School, University of Marilia (UNIMAR), Marília, Brazil
| | - Lenita Mayumi Suzuki
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Medical School, University of Marilia (UNIMAR), Marília, Brazil
| | - Mayara Longui Cabrini
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Medical School, University of Marilia (UNIMAR), Marília, Brazil
| | | | - Ana Letícia Carneiro de Camargo
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Medical School, University of Marilia (UNIMAR), Marília, Brazil
| | - Leila Maria Guissoni Campos
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Medical School, University of Marilia (UNIMAR), Marília, Brazil,*Correspondence: Leila Maria Guissoni Campos,
| |
Collapse
|
13
|
Ravichandran S, Suhasini R, Madheswaran Deepa S, Selvaraj DB, Vergil Andrews JF, Thiagarajan V, Kandasamy M. Intertwining Neuropathogenic Impacts of Aberrant Circadian Rhythm and Impaired Neuroregenerative Plasticity in Huntington’s Disease: Neurotherapeutic Significance of Chemogenetics. JOURNAL OF MOLECULAR PATHOLOGY 2022; 3:355-371. [DOI: 10.3390/jmp3040030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2024] Open
Abstract
Huntington’s disease (HD) is a progressive neurodegenerative disorder characterized by abnormal progressive involuntary movements, cognitive deficits, sleep disturbances, and psychiatric symptoms. The onset and progression of the clinical symptoms have been linked to impaired adult neurogenesis in the brains of subjects with HD, due to the reduced neurogenic potential of neural stem cells (NSCs). Among various pathogenic determinants, an altered clock pathway appears to induce the dysregulation of neurogenesis in neurodegenerative disorders. Notably, gamma-aminobutyric acid (GABA)-ergic neurons that express the vasoactive intestinal peptide (VIP) in the brain play a key role in the regulation of circadian rhythm and neuroplasticity. While an abnormal clock gene pathway has been associated with the inactivation of GABAergic VIP neurons, recent studies suggest the activation of this neuronal population in the brain positively contributes to neuroplasticity. Thus, the activation of GABAergic VIP neurons in the brain might help rectify the irregular circadian rhythm in HD. Chemogenetics refers to the incorporation of genetically engineered receptors or ion channels into a specific cell population followed by its activation using desired chemical ligands. The recent advancement of chemogenetic-based approaches represents a potential scientific tool to rectify the aberrant circadian clock pathways. Considering the facts, the defects in the circadian rhythm can be rectified by the activation of VIP-expressing GABAergic neurons using chemogenetics approaches. Thus, the chemogenetic-based rectification of an abnormal circadian rhythm may facilitate the neurogenic potentials of NSCs to restore the neuroregenerative plasticity in HD. Eventually, the increased neurogenesis in the brain can be expected to mitigate neuronal loss and functional deficits.
Collapse
Affiliation(s)
- Sowbarnika Ravichandran
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India
- School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India
| | - Ramalingam Suhasini
- Photonics and Biophotonics Lab, School of Chemistry, Bharathidasan University, Tiruchirappalli 620024, India
| | - Sudhiksha Madheswaran Deepa
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India
- School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India
| | - Divya Bharathi Selvaraj
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India
| | - Jemi Feiona Vergil Andrews
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India
| | - Viruthachalam Thiagarajan
- Photonics and Biophotonics Lab, School of Chemistry, Bharathidasan University, Tiruchirappalli 620024, India
- Faculty Recharge Programme, University Grants Commission (UGC-FRP), New Delhi 110002, India
| | - Mahesh Kandasamy
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India
- School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India
- Faculty Recharge Programme, University Grants Commission (UGC-FRP), New Delhi 110002, India
| |
Collapse
|
14
|
Whittaker DS, Tamai TK, Bains RS, Villanueva SAM, Luk SHC, Dell’Angelica D, Block GD, Ghiani CA, Colwell CS. Dietary ketosis improves circadian dysfunction as well as motor symptoms in the BACHD mouse model of Huntington's disease. Front Nutr 2022; 9:1034743. [PMID: 36407529 PMCID: PMC9669764 DOI: 10.3389/fnut.2022.1034743] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022] Open
Abstract
Disturbances in sleep/wake cycles are common among patients with neurodegenerative diseases including Huntington's disease (HD) and represent an appealing target for chrono-nutrition-based interventions. In the present work, we sought to determine whether a low-carbohydrate, high-fat diet would ameliorate the symptoms and delay disease progression in the BACHD mouse model of HD. Adult WT and BACHD male mice were fed a normal or a ketogenic diet (KD) for 3 months. The KD evoked a robust rhythm in serum levels of β-hydroxybutyrate and dramatic changes in the microbiome of male WT and BACHD mice. NanoString analysis revealed transcriptional changes driven by the KD in the striatum of both WT and BACHD mice. Disturbances in sleep/wake cycles have been reported in mouse models of HD and are common among HD patients. Having established that the KD had effects on both the WT and mutant mice, we examined its impact on sleep/wake cycles. KD increased daytime sleep and improved the timing of sleep onset, while other sleep parameters were not altered. In addition, KD improved activity rhythms, including rhythmic power, and reduced inappropriate daytime activity and onset variability. Importantly, KD improved motor performance on the rotarod and challenging beam tests. It is worth emphasizing that HD is a genetically caused disease with no known cure. Life-style changes that not only improve the quality of life but also delay disease progression for HD patients are greatly needed. Our study demonstrates the therapeutic potential of diet-based treatment strategies in a pre-clinical model of HD.
Collapse
Affiliation(s)
- Daniel S. Whittaker
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - T. Katherine Tamai
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Raj S. Bains
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Sophia Anne Marie Villanueva
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Shu Hon Christopher Luk
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Derek Dell’Angelica
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Gene D. Block
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Cristina A. Ghiani
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Christopher S. Colwell
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
15
|
Mauri S, Favaro M, Bernardo G, Mazzotta GM, Ziviani E. Mitochondrial autophagy in the sleeping brain. Front Cell Dev Biol 2022; 10:956394. [PMID: 36092697 PMCID: PMC9449320 DOI: 10.3389/fcell.2022.956394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
A significant percentage of the mitochondrial mass is replaced on a daily basis via mechanisms of mitochondrial quality control. Through mitophagy (a selective type of autophagy that promotes mitochondrial proteostasis) cells keep a healthy pool of mitochondria, and prevent oxidative stress and inflammation. Furthermore, mitophagy helps adapting to the metabolic demand of the cells, which changes on a daily basis.Core components of the mitophagy process are PINK1 and Parkin, which mutations are linked to Parkinson’s Disease. The crucial role of PINK1/Parkin pathway during stress-induced mitophagy has been extensively studied in vitro in different cell types. However, recent advances in the field allowed discovering that mitophagy seems to be only slightly affected in PINK1 KO mice and flies, putting into question the physiological relevance of this pathway in vivo in the whole organism. Indeed, several cell-specific PINK1/Parkin-independent mitophagy pathways have been recently discovered, which appear to be activated under physiological conditions such as those that promote mitochondrial proteome remodeling during differentiation or in response to specific physiological stimuli.In this Mini Review we want to summarize the recent advances in the field, and add another level of complexity by focusing attention on a potentially important aspect of mitophagy regulation: the implication of the circadian clock. Recent works showed that the circadian clock controls many aspects of mitochondrial physiology, including mitochondrial morphology and dynamic, respiratory activity, and ATP synthesis. Furthermore, one of the essential functions of sleep, which is controlled by the clock, is the clearance of toxic metabolic compounds from the brain, including ROS, via mechanisms of proteostasis. Very little is known about a potential role of the clock in the quality control mechanisms that maintain the mitochondrial repertoire healthy during sleep/wake cycles. More importantly, it remains completely unexplored whether (dys)function of mitochondrial proteostasis feedbacks to the circadian clockwork.
Collapse
Affiliation(s)
| | | | | | | | - Elena Ziviani
- *Correspondence: Gabriella M. Mazzotta, Elena Ziviani,
| |
Collapse
|
16
|
Cheng WY, Ho YS, Chang RCC. Linking circadian rhythms to microbiome-gut-brain axis in aging-associated neurodegenerative diseases. Ageing Res Rev 2022; 78:101620. [PMID: 35405323 DOI: 10.1016/j.arr.2022.101620] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/13/2022] [Accepted: 04/06/2022] [Indexed: 12/12/2022]
Abstract
Emerging evidence suggests that both disruption of circadian rhythms and gut dysbiosis are closely related to aging-associated neurodegenerative diseases. Over the last decade, the microbiota-gut-brain axis has been an emerging field and revolutionized studies in pathology, diagnosis, and treatment of neurological disorders. Crosstalk between the brain and gut microbiota can be accomplished via the endocrine, immune, and nervous system. Recent studies have shown that the composition and diurnal oscillation of gut microbiota are influenced by host circadian rhythms. This provides a new perspective for investigating the microbiome-gut-brain axis. We aim to review current understanding and research on the dynamic interaction between circadian rhythms and the microbiome-gut-brain axis. Furthermore, we will address the possible neurodegenerative disease contribution through circadian rhythms and microbiome-gut-brain axis crosstalk.
Collapse
Affiliation(s)
- Wai-Yin Cheng
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Yuen-Shan Ho
- School of Nursing, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region.
| | - Raymond Chuen-Chung Chang
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
17
|
Prakash P, Pradhan AK, Sheeba V. Hsp40 overexpression in pacemaker neurons delays circadian dysfunction in a Drosophila model of Huntington's disease. Dis Model Mech 2022; 15:275556. [PMID: 35645202 PMCID: PMC9254228 DOI: 10.1242/dmm.049447] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 05/24/2022] [Indexed: 12/13/2022] Open
Abstract
Circadian disturbances are early features of neurodegenerative diseases, including Huntington's disease (HD). Emerging evidence suggests that circadian decline feeds into neurodegenerative symptoms, exacerbating them. Therefore, we asked whether known neurotoxic modifiers can suppress circadian dysfunction. We performed a screen of neurotoxicity-modifier genes to suppress circadian behavioural arrhythmicity in a Drosophila circadian HD model. The molecular chaperones Hsp40 and HSP70 emerged as significant suppressors in the circadian context, with Hsp40 being the more potent mitigator. Upon Hsp40 overexpression in the Drosophila circadian ventrolateral neurons (LNv), the behavioural rescue was associated with neuronal rescue of loss of circadian proteins from small LNv soma. Specifically, there was a restoration of the molecular clock protein Period and its oscillations in young flies and a long-lasting rescue of the output neuropeptide Pigment dispersing factor. Significantly, there was a reduction in the expanded Huntingtin inclusion load, concomitant with the appearance of a spot-like Huntingtin form. Thus, we provide evidence implicating the neuroprotective chaperone Hsp40 in circadian rehabilitation. The involvement of molecular chaperones in circadian maintenance has broader therapeutic implications for neurodegenerative diseases. This article has an associated First Person interview with the first author of the paper. Summary: This study shows, for the first time, a neuroprotective role of chaperone Hsp40 in suppressing circadian dysfunction associated with Huntington's disease in a Drosophila model.
Collapse
Affiliation(s)
- Pavitra Prakash
- Evolutionary and Integrative Biology Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, India
| | - Arpit Kumar Pradhan
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, India
| | - Vasu Sheeba
- Evolutionary and Integrative Biology Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, India.,Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, India
| |
Collapse
|
18
|
Gómez-Jaramillo L, Cano-Cano F, González-Montelongo MDC, Campos-Caro A, Aguilar-Diosdado M, Arroba AI. A New Perspective on Huntington's Disease: How a Neurological Disorder Influences the Peripheral Tissues. Int J Mol Sci 2022; 23:6089. [PMID: 35682773 PMCID: PMC9181740 DOI: 10.3390/ijms23116089] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 11/22/2022] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by a toxic, aggregation-prone expansion of CAG repeats in the HTT gene with an age-dependent progression that leads to behavioral, cognitive and motor symptoms. Principally affecting the frontal cortex and the striatum, mHTT disrupts many cellular functions. In fact, increasing evidence shows that peripheral tissues are affected by neurodegenerative diseases. It establishes an active crosstalk between peripheral tissues and the brain in different neurodegenerative diseases. This review focuses on the current knowledge of peripheral tissue effects in HD animal and cell experimental models and identifies biomarkers and mechanisms involved or affected in the progression of the disease as new therapeutic or early diagnostic options. The particular changes in serum/plasma, blood cells such as lymphocytes, immune blood cells, the pancreas, the heart, the retina, the liver, the kidney and pericytes as a part of the blood-brain barrier are described. It is important to note that several changes in different mouse models of HD present differences between them and between the different ages analyzed. The understanding of the impact of peripheral organ inflammation in HD may open new avenues for the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Laura Gómez-Jaramillo
- Undad de Investigación, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), 11002 Cádiz, Spain; (L.G.-J.); (F.C.-C.); (M.d.C.G.-M.); (A.C.-C.); (M.A.-D.)
| | - Fátima Cano-Cano
- Undad de Investigación, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), 11002 Cádiz, Spain; (L.G.-J.); (F.C.-C.); (M.d.C.G.-M.); (A.C.-C.); (M.A.-D.)
| | - María del Carmen González-Montelongo
- Undad de Investigación, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), 11002 Cádiz, Spain; (L.G.-J.); (F.C.-C.); (M.d.C.G.-M.); (A.C.-C.); (M.A.-D.)
| | - Antonio Campos-Caro
- Undad de Investigación, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), 11002 Cádiz, Spain; (L.G.-J.); (F.C.-C.); (M.d.C.G.-M.); (A.C.-C.); (M.A.-D.)
- Área de Genética, Departamento de Biomedicina, Biotecnología y Salud Pública, Universidad de Cádiz, 11002 Cádiz, Spain
| | - Manuel Aguilar-Diosdado
- Undad de Investigación, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), 11002 Cádiz, Spain; (L.G.-J.); (F.C.-C.); (M.d.C.G.-M.); (A.C.-C.); (M.A.-D.)
- Departamento de Endocrinología y Nutrición, Hospital Universitario Puerta del Mar, Universidad de Cádiz, 11002 Cádiz, Spain
| | - Ana I. Arroba
- Undad de Investigación, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), 11002 Cádiz, Spain; (L.G.-J.); (F.C.-C.); (M.d.C.G.-M.); (A.C.-C.); (M.A.-D.)
- Área de Genética, Departamento de Biomedicina, Biotecnología y Salud Pública, Universidad de Cádiz, 11002 Cádiz, Spain
| |
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
|
Wang Q, Cao F, Wu Y. Orexinergic System in Neurodegenerative Diseases. Front Aging Neurosci 2021; 13:713201. [PMID: 34483883 PMCID: PMC8416170 DOI: 10.3389/fnagi.2021.713201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 07/20/2021] [Indexed: 01/16/2023] Open
Abstract
Orexinergic system consisting of orexins and orexin receptors plays an essential role in regulating sleep–wake states, whereas sleep disruption is a common symptom of a number of neurodegenerative diseases. Emerging evidence reveals that the orexinergic system is disturbed in various neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), and multiple sclerosis (MS), whereas the dysregulation of orexins and/or orexin receptors contributes to the pathogenesis of these diseases. In this review, we summarized advanced knowledge of the orexinergic system and its role in sleep, and reviewed the dysregulation of the orexinergic system and its role in the pathogenesis of AD, PD, HD, and MS. Moreover, the therapeutic potential of targeting the orexinergic system for the treatment of these diseases was discussed.
Collapse
Affiliation(s)
- Qinqin Wang
- Shandong Collaborative Innovation Center for Diagnosis, Treatment & Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Fei Cao
- Shandong Collaborative Innovation Center for Diagnosis, Treatment & Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China
| | - Yili Wu
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, School of Mental Health and The Affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou, China.,Oujiang Laboratory, Wenzhou, China
| |
Collapse
|
21
|
Falck RS, Davis JC, Best JR, Chan PCY, Li LC, Wyrough AB, Bennett KJ, Backhouse D, Liu-Ambrose T. Effect of a Multimodal Lifestyle Intervention on Sleep and Cognitive Function in Older Adults with Probable Mild Cognitive Impairment and Poor Sleep: A Randomized Clinical Trial. J Alzheimers Dis 2021; 76:179-193. [PMID: 32444553 DOI: 10.3233/jad-200383] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Poor sleep is common among older adults with mild cognitive impairment (MCI) and may contribute to further cognitive decline. Whether multimodal lifestyle intervention that combines bright light therapy (BLT), physical activity (PA), and good sleep hygiene can improve sleep in older adults with MCI and poor sleep is unknown. OBJECTIVE To assess the effect of a multimodal lifestyle intervention on sleep in older adults with probable MCI and poor sleep. METHODS This was a 24-week proof-of-concept randomized trial of 96 community-dwelling older adults aged 65-85 years with probable MCI (<26/30 on the Montreal Cognitive Assessment) and poor sleep (>5 on the Pittsburgh Sleep Quality Index [PSQI]). Participants were allocated to either a multimodal lifestyle intervention (INT); or 2) education + attentional control (CON). INT participants received four once-weekly general sleep hygiene education classes, followed by 20-weeks of: 1) individually-timed BLT; and 2) individually-tailored PA promotion. Our primary outcome was sleep efficiency measured using the MotionWatch8© (MW8). Secondary outcomes were MW8-measured sleep duration, fragmentation index, wake-after-sleep-onset, latency, and PSQI-measured subjective sleep quality. RESULTS There were no significant between-group differences in MW8 measured sleep efficiency at 24-weeks (estimated mean difference [INT -CON]: 1.18%; 95% CI [-0.99, 3.34]), or any other objective-estimate of sleep. However, INT participants reported significantly better subjective sleep quality at 24-weeks (estimated mean difference: -1.39; 95% CI [-2.72, -0.06]) compared to CON. CONCLUSION Among individuals with probable MCI and poor sleep, a multimodal lifestyle intervention improves subjective sleep quality, but not objectively estimated sleep.
Collapse
Affiliation(s)
- Ryan S Falck
- Aging, Mobility, and Cognitive Neuroscience Lab, Department of Physical Therapy, University of British Columbia, Vancouver, British Columbia, Canada.,Djavad Mowafaghian Centre for Brain Health, Vancouver Coastal Health Research Institute. University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jennifer C Davis
- Faculty of Management, University of British Columbia-Okanagan Campus, Kelowna, British Columbia, Canada
| | - John R Best
- Aging, Mobility, and Cognitive Neuroscience Lab, Department of Physical Therapy, University of British Columbia, Vancouver, British Columbia, Canada.,Djavad Mowafaghian Centre for Brain Health, Vancouver Coastal Health Research Institute. University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Patrick C Y Chan
- Aging, Mobility, and Cognitive Neuroscience Lab, Department of Physical Therapy, University of British Columbia, Vancouver, British Columbia, Canada.,Djavad Mowafaghian Centre for Brain Health, Vancouver Coastal Health Research Institute. University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Linda C Li
- Arthritis Research Canada, University of British Columbia, Vancouver, British Columbia, Canada
| | - Anne B Wyrough
- Aging, Mobility, and Cognitive Neuroscience Lab, Department of Physical Therapy, University of British Columbia, Vancouver, British Columbia, Canada.,Djavad Mowafaghian Centre for Brain Health, Vancouver Coastal Health Research Institute. University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kimberly J Bennett
- Aging, Mobility, and Cognitive Neuroscience Lab, Department of Physical Therapy, University of British Columbia, Vancouver, British Columbia, Canada.,Djavad Mowafaghian Centre for Brain Health, Vancouver Coastal Health Research Institute. University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Daniel Backhouse
- Aging, Mobility, and Cognitive Neuroscience Lab, Department of Physical Therapy, University of British Columbia, Vancouver, British Columbia, Canada.,Djavad Mowafaghian Centre for Brain Health, Vancouver Coastal Health Research Institute. University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Teresa Liu-Ambrose
- Aging, Mobility, and Cognitive Neuroscience Lab, Department of Physical Therapy, University of British Columbia, Vancouver, British Columbia, Canada.,Djavad Mowafaghian Centre for Brain Health, Vancouver Coastal Health Research Institute. University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
22
|
Carter B, Justin HS, Gulick D, Gamsby JJ. The Molecular Clock and Neurodegenerative Disease: A Stressful Time. Front Mol Biosci 2021; 8:644747. [PMID: 33889597 PMCID: PMC8056266 DOI: 10.3389/fmolb.2021.644747] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
Circadian rhythm dysfunction occurs in both common and rare neurodegenerative diseases. This dysfunction manifests as sleep cycle mistiming, alterations in body temperature rhythms, and an increase in symptomatology during the early evening hours known as Sundown Syndrome. Disruption of circadian rhythm homeostasis has also been implicated in the etiology of neurodegenerative disease. Indeed, individuals exposed to a shifting schedule of sleep and activity, such as health care workers, are at a higher risk. Thus, a bidirectional relationship exists between the circadian system and neurodegeneration. At the heart of this crosstalk is the molecular circadian clock, which functions to regulate circadian rhythm homeostasis. Over the past decade, this connection has become a focal point of investigation as the molecular clock offers an attractive target to combat both neurodegenerative disease pathogenesis and circadian rhythm dysfunction, and a pivotal role for neuroinflammation and stress has been established. This review summarizes the contributions of molecular clock dysfunction to neurodegenerative disease etiology, as well as the mechanisms by which neurodegenerative diseases affect the molecular clock.
Collapse
Affiliation(s)
- Bethany Carter
- Gamsby Laboratory, USF Health Byrd Alzheimer's Center and Research Institute, University of South Florida Health, Tampa, FL, United States
| | - Hannah S Justin
- Gamsby Laboratory, USF Health Byrd Alzheimer's Center and Research Institute, University of South Florida Health, Tampa, FL, United States
| | - Danielle Gulick
- Gamsby Laboratory, USF Health Byrd Alzheimer's Center and Research Institute, University of South Florida Health, Tampa, FL, United States.,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Joshua J Gamsby
- Gamsby Laboratory, USF Health Byrd Alzheimer's Center and Research Institute, University of South Florida Health, Tampa, FL, United States.,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
23
|
Ruan W, Yuan X, Eltzschig HK. Circadian rhythm as a therapeutic target. Nat Rev Drug Discov 2021; 20:287-307. [PMID: 33589815 DOI: 10.1038/s41573-020-00109-w] [Citation(s) in RCA: 179] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2020] [Indexed: 12/20/2022]
Abstract
The circadian clock evolved in diverse organisms to integrate external environmental changes and internal physiology. The clock endows the host with temporal precision and robust adaptation to the surrounding environment. When circadian rhythms are perturbed or misaligned, as a result of jet lag, shiftwork or other lifestyle factors, adverse health consequences arise, and the risks of diseases such as cancer, cardiovascular diseases or metabolic disorders increase. Although the negative impact of circadian rhythm disruption is now well established, it remains underappreciated how to take advantage of biological timing, or correct it, for health benefits. In this Review, we provide an updated account of the circadian system and highlight several key disease areas with altered circadian signalling. We discuss environmental and lifestyle modifications of circadian rhythm and clock-based therapeutic strategies, including chronotherapy, in which dosing time is deliberately optimized for maximum therapeutic index, and pharmacological agents that target core clock components and proximal regulators. Promising progress in research, disease models and clinical applications should encourage a concerted effort towards a new era of circadian medicine.
Collapse
Affiliation(s)
- Wei Ruan
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA.,Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyi Yuan
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Holger K Eltzschig
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
24
|
van Wamelen DJ, Aziz NA. Hypothalamic pathology in Huntington disease. HANDBOOK OF CLINICAL NEUROLOGY 2021; 182:245-255. [PMID: 34266596 DOI: 10.1016/b978-0-12-819973-2.00017-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Huntington's disease (HD), an autosomal dominant hereditary disorder associated with the accumulation of mutant huntingtin, is classically associated with cognitive decline and motor symptoms, notably chorea. However, growing evidence suggests that nonmotor symptoms are equally prevalent and debilitating. Some of these symptoms may be linked to hypothalamic pathology, demonstrated by findings in HD animal models and HD patients showing specific changes in hypothalamic neuropeptidergic populations and their associated functions. At least some of these alterations are likely due to local mutant huntingtin expression and toxicity, while others are likely caused by disturbed hypothalamic circuitry. Common problems include circadian rhythm disorders, including desynchronization of daily hormone excretion patterns, which could be targeted by novel therapeutic interventions, such as timed circadian interventions with light therapy or melatonin. However, translation of these findings from bench-to-bedside is hampered by differences in murine HD models and HD patients, including mutant huntingtin trinucleotide repeat length, which is highly heterogeneous across the various models. In this chapter, we summarize the current knowledge regarding hypothalamic alterations in HD patients and animal models, and the potential for these findings to be translated into clinical practice and management.
Collapse
Affiliation(s)
- Daniel J van Wamelen
- Department of Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom; Parkinson's Foundation Centre of Excellence, King's College Hospital, London, United Kingdom; Department of Neurology, Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands.
| | - N Ahmad Aziz
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany; Department of Neurology, Faculty of Medicine, University of Bonn, Bonn, Germany
| |
Collapse
|
25
|
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: 4.8] [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
|
26
|
Hua J, Sun H, Shen Y. Improvement in sleep duration was associated with higher cognitive function: a new association. Aging (Albany NY) 2020; 12:20623-20644. [PMID: 33082298 PMCID: PMC7655193 DOI: 10.18632/aging.103948] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/08/2020] [Indexed: 02/06/2023]
Abstract
Objectives: Previous studies have indicated that improvement in sleep duration might correlate with better cognition. We aimed to examine the associations between changes in sleep duration and cognitive function. Results: A change from short sleep duration (SSD) to moderate sleep duration (MSD) was associated with better global cognition scores (β=0.54, P <0.01). A change from SSD to long sleep duration (LSD) (β=-0.94, P <0.001) or a change from LSD to SSD (β=-1.38, P <0.01) was associated with lower global cognition. For individuals with MSD, a≥2 h increase (β=-0.89, P <0.001) or decrease (β=-0.70, P <0.001) in sleep duration was associated with lower global cognition. Conclusions: For short sleepers, improvement in sleep duration correlated with better cognition. For long sleepers, there was no need to reduce sleep duration. Excessive changes or deviation from the moderate duration was associated with lower cognition. Methods: A total of 10325 individuals aged 45 and older from the China Health and Retirement Longitudinal Study (CHARLS) were included. Self-reported nocturnal sleep duration and cognitive function were assessed in the three waves of CHARLS from 2011 to 2015. Cognitive function was assessed by a global cognition score, which included episodic memory, visuospatial abilities, calculation, orientation and attention.
Collapse
Affiliation(s)
- Jianian Hua
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, PR China.,Medical College of Soochow University, Suzhou 215123, PR China
| | - Hongpeng Sun
- Department of Child Health, School of Public Health, Medical College of Soochow University, Suzhou 215123, PR China
| | - Yueping Shen
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou 215123, PR China
| |
Collapse
|
27
|
Impact of circadian and diurnal rhythms on cellular metabolic function and neurodegenerative diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 154:393-412. [PMID: 32739012 DOI: 10.1016/bs.irn.2020.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
The 24-h rotational period of the earth has driven evolution of biological systems that serve to synchronize organismal physiology and behavior to this predictable environmental event. In mammals, the circadian (circa, "about" and dia, "a day") clock keeps 24-h time at the organismal and cellular level, optimizing biological function for a given time of day. The most obvious circadian output is the sleep-wake cycle, though countless bodily functions, ranging from hormone levels to cognitive function, are influenced by the circadian clock. Here we discuss the regulation of metabolic pathways by the circadian clock, discuss the evidence implicating circadian and sleep disruption in neurodegenerative diseases, and suggest some possible connections between the clock, metabolism, and neurodegenerative disease.
Collapse
|
28
|
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: 5.5] [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
|
29
|
Xu F, Kula-Eversole E, Iwanaszko M, Hutchison AL, Dinner A, Allada R. Circadian Clocks Function in Concert with Heat Shock Organizing Protein to Modulate Mutant Huntingtin Aggregation and Toxicity. Cell Rep 2020; 27:59-70.e4. [PMID: 30943415 PMCID: PMC7237104 DOI: 10.1016/j.celrep.2019.03.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/24/2019] [Accepted: 03/02/2019] [Indexed: 01/08/2023] Open
Abstract
Neurodegenerative diseases commonly involve the disruption of circadian rhythms. Studies indicate that mutant Huntingtin (mHtt), the cause of Huntington’s disease (HD), disrupts circadian rhythms often before motor symptoms are evident. Yet little is known about the molecular mechanisms by which mHtt impairs circadian rhythmicity and whether circadian clocks can modulate HD pathogenesis. To address this question, we used a Drosophila HD model. We found that both environmental and genetic perturbations of the circadian clock alter mHtt-mediated neurodegeneration. To identify potential genetic pathways that mediate these effects, we applied a behavioral platform to screen for clock-regulated HD suppressors, identifying a role for Heat Shock Protein 70/90 Organizing Protein (Hop). Hop knockdown paradoxically reduces mHtt aggregation and toxicity. These studies demonstrate a role for the circadian clock in a neurodegenerative disease model and reveal a clock-regulated molecular and cellular pathway that links clock function to neurodegenerative disease. Disruption of circadian rhythms is frequently observed across a range of neurodegenerative diseases. Here, Xu et al. demonstrate that perturbation of circadian clocks alters the toxicity of the mutant Huntingtin protein, the cause of Huntington’s disease (HD). Moreover, they reveal a key mechanistic link between the clock and HD.
Collapse
Affiliation(s)
- Fangke Xu
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | | | - Marta Iwanaszko
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Alan L Hutchison
- Medical Scientist Training Program, University of Chicago, Chicago, IL, USA
| | - Aaron Dinner
- James Franck Institute, University of Chicago, Chicago, IL, USA
| | - Ravi Allada
- Department of Neurobiology, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
30
|
Sanchez REA, Bussi IL, Ben-Hamo M, Caldart CS, Catterall WA, De La Iglesia HO. Circadian regulation of sleep in a pre-clinical model of Dravet syndrome: dynamics of sleep stage and siesta re-entrainment. Sleep 2020; 42:5539047. [PMID: 31346614 DOI: 10.1093/sleep/zsz173] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 05/24/2019] [Indexed: 12/11/2022] Open
Abstract
STUDY OBJECTIVES Sleep disturbances are common co-morbidities of epileptic disorders. Dravet syndrome (DS) is an intractable epilepsy accompanied by disturbed sleep. While there is evidence that daily sleep timing is disrupted in DS, the difficulty of chronically recording polysomnographic sleep from patients has left our understanding of the effect of DS on circadian sleep regulation incomplete. We aim to characterize circadian sleep regulation in a mouse model of DS. METHODS Here we exploit long-term electrocorticographic recordings of sleep in a mouse model of DS in which one copy of the Scn1a gene is deleted. This model both genocopies and phenocopies the disease in humans. We test the hypothesis that the deletion of Scn1a in DS mice is associated with impaired circadian regulation of sleep. RESULTS We find that DS mice show impairments in circadian sleep regulation, including a fragmented rhythm of non-rapid eye movement (NREM) sleep and an elongated circadian period of sleep. Next, we characterize re-entrainment of sleep stages and siesta following jet lag in the mouse. Strikingly, we find that re-entrainment of sleep following jet lag is normal in DS mice, in contrast to previous demonstrations of slowed re-entrainment of wheel-running activity. Finally, we report that DS mice are more likely to have an absent or altered daily "siesta". CONCLUSIONS Our findings support the hypothesis that the circadian regulation of sleep is altered in DS and highlight the value of long-term chronic polysomnographic recording in studying the role of the circadian clock on sleep/wake cycles in pre-clinical models of disease.
Collapse
Affiliation(s)
- Raymond E A Sanchez
- Department of Biology, University of Washington, Seattle, WA.,Graduate Program in Neuroscience, University of Washington, Seattle WA
| | - Ivana L Bussi
- Department of Biology, University of Washington, Seattle, WA
| | - Miriam Ben-Hamo
- Department of Biology, University of Washington, Seattle, WA
| | | | - William A Catterall
- Graduate Program in Neuroscience, University of Washington, Seattle WA.,Department of Pharmacology, University of Washington, Seattle WA
| | - Horacio O De La Iglesia
- Department of Biology, University of Washington, Seattle, WA.,Graduate Program in Neuroscience, University of Washington, Seattle WA
| |
Collapse
|
31
|
De Nobrega AK, Luz KV, Lyons LC. Resetting the Aging Clock: Implications for Managing Age-Related Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1260:193-265. [PMID: 32304036 DOI: 10.1007/978-3-030-42667-5_9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Worldwide, individuals are living longer due to medical and scientific advances, increased availability of medical care and changes in public health policies. Consequently, increasing attention has been focused on managing chronic conditions and age-related diseases to ensure healthy aging. The endogenous circadian system regulates molecular, physiological and behavioral rhythms orchestrating functional coordination and processes across tissues and organs. Circadian disruption or desynchronization of circadian oscillators increases disease risk and appears to accelerate aging. Reciprocally, aging weakens circadian function aggravating age-related diseases and pathologies. In this review, we summarize the molecular composition and structural organization of the circadian system in mammals and humans, and evaluate the technological and societal factors contributing to the increasing incidence of circadian disorders. Furthermore, we discuss the adverse effects of circadian dysfunction on aging and longevity and the bidirectional interactions through which aging affects circadian function using examples from mammalian research models and humans. Additionally, we review promising methods for managing healthy aging through behavioral and pharmacological reinforcement of the circadian system. Understanding age-related changes in the circadian clock and minimizing circadian dysfunction may be crucial components to promote healthy aging.
Collapse
Affiliation(s)
- Aliza K De Nobrega
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Kristine V Luz
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Lisa C Lyons
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
32
|
Abnormal Photic Entrainment to Phase-Delaying Stimuli in the R6/2 Mouse Model of Huntington's Disease, despite Retinal Responsiveness to Light. eNeuro 2019; 6:ENEURO.0088-19.2019. [PMID: 31744839 PMCID: PMC6905640 DOI: 10.1523/eneuro.0088-19.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 10/22/2019] [Accepted: 10/28/2019] [Indexed: 12/13/2022] Open
Abstract
The circadian clock located in the suprachiasmatic nucleus (SCN) in mammals entrains to ambient light via the retinal photoreceptors. This allows behavioral rhythms to change in synchrony with seasonal and daily changes in light period. Circadian rhythmicity is progressively disrupted in Huntington's disease (HD) and in HD mouse models such as the transgenic R6/2 line. Although retinal afferent inputs to the SCN are disrupted in R6/2 mice at late stages, they can respond to changes in light/dark cycles, as seen in jet lag and 23 h/d paradigms. To investigate photic entrainment and SCN function in R6/2 mice at different stages of disease, we first assessed the effect on locomotor activity of exposure to a 15 min light pulse given at different times of the day. We then placed the mice under five non-standard light conditions. These were light cycle regimes (T-cycles) of T21 (10.5 h light/dark), T22 (11 h light/dark), T26 (13 h light/dark), constant light, or constant dark. We found a progressive impairment in photic synchronization in R6/2 mice when the stimuli required the SCN to lengthen rhythms (phase-delaying light pulse, T26, or constant light), but normal synchronization to stimuli that required the SCN to shorten rhythms (phase-advancing light pulse and T22). Despite the behavioral abnormalities, we found that Per1 and c-fos gene expression remained photo-inducible in SCN of R6/2 mice. Both the endogenous drift of the R6/2 mouse SCN to shorter periods and its inability to adapt to phase-delaying changes will contribute to the HD circadian dysfunction.
Collapse
|
33
|
Xu F, Kula-Eversole E, Iwanaszko M, Lim C, Allada R. Ataxin2 functions via CrebA to mediate Huntingtin toxicity in circadian clock neurons. PLoS Genet 2019; 15:e1008356. [PMID: 31593562 PMCID: PMC6782096 DOI: 10.1371/journal.pgen.1008356] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 08/07/2019] [Indexed: 12/12/2022] Open
Abstract
Disrupted circadian rhythms is a prominent and early feature of neurodegenerative diseases including Huntington’s disease (HD). In HD patients and animal models, striatal and hypothalamic neurons expressing molecular circadian clocks are targets of mutant Huntingtin (mHtt) pathogenicity. Yet how mHtt disrupts circadian rhythms remains unclear. In a genetic screen for modifiers of mHtt effects on circadian behavior in Drosophila, we discovered a role for the neurodegenerative disease gene Ataxin2 (Atx2). Genetic manipulations of Atx2 modify the impact of mHtt on circadian behavior as well as mHtt aggregation and demonstrate a role for Atx2 in promoting mHtt aggregation as well as mHtt-mediated neuronal dysfunction. RNAi knockdown of the Fragile X mental retardation gene, dfmr1, an Atx2 partner, also partially suppresses mHtt effects and Atx2 effects depend on dfmr1. Atx2 knockdown reduces the cAMP response binding protein A (CrebA) transcript at dawn. CrebA transcript level shows a prominent diurnal regulation in clock neurons. Loss of CrebA also partially suppresses mHtt effects on behavior and cell loss and restoration of CrebA can suppress Atx2 effects. Our results indicate a prominent role of Atx2 in mediating mHtt pathology, specifically via its regulation of CrebA, defining a novel molecular pathway in HD pathogenesis. Circadian clocks evolved to anticipate 24 h environmental rhythms driven by the earth’s daily rotation and regulate nearly all aspects of behavior, physiology and the genome. Disruptions of the circadian clock have been associated with a wide range of human diseases, including neurodegenerative diseases such as Huntington’s disease (HD). Using an HD animal model in which a mutant Huntingtin (mHtt) protein is expressed, we identify a role for the RNA binding protein and neurodegenerative disease gene Ataxin-2 (Atx2) in mediating mHtt effects on circadian behavioral rhythms. Using transcriptomics, we identify the transcription factor CrebA as a potential target of both Atx2 and the circadian clock. Finally, we demonstrate a role for CrebA in mediating mHtt effects on circadian behavior, defining a novel Atx2-CrebA pathway in a neurodegenerative disease model. These studies define the molecular mechanisms by which mHtt can disrupt circadian rhythms identifying potential novel therapeutic targets for this uniformly fatal disease.
Collapse
Affiliation(s)
- Fangke Xu
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Elzbieta Kula-Eversole
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Marta Iwanaszko
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Chunghun Lim
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Ravi Allada
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
- * E-mail:
| |
Collapse
|
34
|
|
35
|
Abstract
Fundamental aspects of neurobiology are time-of-day regulated. Therefore, it is not surprising that neurodegenerative and psychiatric diseases are accompanied by sleep and circadian rhythm disruption. Although the direction of causation remains unclear, abnormal sleep-wake patterns often occur early in disease, exacerbate progression, and are a common primary complaint from patients. Circadian medicine incorporates knowledge of 24-hour biological rhythms to improve treatment. This article highlights how research and technologic advances in circadian biology might translate to improved patient care.
Collapse
|
36
|
Smarr B, Cutler T, Loh DH, Kudo T, Kuljis D, Kriegsfeld L, Ghiani CA, Colwell CS. Circadian dysfunction in the Q175 model of Huntington's disease: Network analysis. J Neurosci Res 2019; 97:1606-1623. [PMID: 31359503 DOI: 10.1002/jnr.24505] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/29/2019] [Accepted: 07/16/2019] [Indexed: 12/19/2022]
Abstract
Disturbances in sleep/wake cycle are a common complaint of individuals with Huntington's disease (HD) and are displayed by HD mouse models. The underlying mechanisms, including the possible role of the circadian timing system, have been the topic of a number of recent studies. The (z)Q175 mouse is a knock-in model in which the human exon 1 sequence of the huntingtin gene is inserted into the mouse DNA with approximately 190 CAG repeats. Among the numerous models available, the heterozygous Q175 offers strong construct validity with a single copy of the mutation, genetic precision of the insertion and control of mutation copy number. In this review, we will summarize the evidence that this model exhibits disrupted diurnal and circadian rhythms in locomotor activity. We found overwhelming evidence for autonomic dysfunction including blunted daily rhythms in heart rate and core body temperature (CBT), reduced heart rate variability, and almost a complete failure of the sympathetic arm of the autonomic nervous system to function during the baroreceptor reflex. Mechanistically, the Q175 mouse model exhibits deficits in the neural output of the central circadian clock, the suprachiasmatic nucleus along with an enhancement of at least one type of potassium current in these neurons. Finally, we report a novel network analysis examining the phase coherence between activity, CBT, and cardiovascular measures. Such analyses found that even young Q175 mutants (heterozygous or homozygous) show coherence degradation, and suggests that loss of phase coherence is a variable that should be considered as a possible biomarker for HD.
Collapse
Affiliation(s)
- Benjamin Smarr
- Department of Psychology, University of California Berkeley, Berkeley, California.,Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California
| | - Tamara Cutler
- Laboratory of Circadian and Sleep Medicine, Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Dawn H Loh
- Laboratory of Circadian and Sleep Medicine, Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Takashi Kudo
- Laboratory of Circadian and Sleep Medicine, Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Dika Kuljis
- Laboratory of Circadian and Sleep Medicine, Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Lance Kriegsfeld
- Department of Psychology, University of California Berkeley, Berkeley, California.,Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California
| | - Cristina A Ghiani
- Laboratory of Circadian and Sleep Medicine, Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Department of Pathology, University of California Los Angeles, Los Angeles, California.,Laboratory Medicine, University of California Los Angeles, Los Angeles, California
| | - Christopher S Colwell
- Laboratory of Circadian and Sleep Medicine, Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
37
|
Cabanas M, Pistono C, Puygrenier L, Rakesh D, Jeantet Y, Garret M, Cho YH. Neurophysiological and Behavioral Effects of Anti-Orexinergic Treatments in a Mouse Model of Huntington's Disease. Neurotherapeutics 2019; 16:784-796. [PMID: 30915710 PMCID: PMC6694444 DOI: 10.1007/s13311-019-00726-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Huntington's disease (HD) is associated with sleep and circadian disturbances in addition to hallmark motor and cognitive impairments. Electrophysiological studies on HD mouse models have revealed an aberrant oscillatory activity at the beta frequency, during sleep, that is associated with HD pathology. Moreover, HD animal models display an abnormal sleep-wake cycle and sleep fragmentation. In this study, we investigated a potential involvement of the orexinergic system dysfunctioning in sleep-wake and circadian disturbances and abnormal network (i.e., beta) activity in the R6/1 mouse model. We found that the age at which orexin activity starts to deviate from normal activity pattern coincides with that of sleep disturbances as well as the beta activity. We also found that acute administration of Suvorexant, an orexin 1 and orexin 2 receptor antagonist, was sufficient to decrease the beta power significantly and to improve sleep in R6/1 mice. In addition, a 5-day treatment paradigm alleviated cognitive deficits and induced a gain of body weight in female HD mice. These results suggest that restoring normal activity of the orexinergic system could be an efficient therapeutic solution for sleep and behavioral disturbances in HD.
Collapse
Affiliation(s)
- Magali Cabanas
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, CNRS UMR 5287, Allee Geoffroy St Hilaire, CS 50023, 33615, Pessac Cedex, France
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, University of Bordeaux, Bordeaux, France
| | - Cristiana Pistono
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, CNRS UMR 5287, Allee Geoffroy St Hilaire, CS 50023, 33615, Pessac Cedex, France
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, University of Bordeaux, Bordeaux, France
| | - Laura Puygrenier
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, CNRS UMR 5287, Allee Geoffroy St Hilaire, CS 50023, 33615, Pessac Cedex, France
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, University of Bordeaux, Bordeaux, France
| | - Divyangana Rakesh
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, CNRS UMR 5287, Allee Geoffroy St Hilaire, CS 50023, 33615, Pessac Cedex, France
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, University of Bordeaux, Bordeaux, France
| | - Yannick Jeantet
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, CNRS UMR 5287, Allee Geoffroy St Hilaire, CS 50023, 33615, Pessac Cedex, France
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, University of Bordeaux, Bordeaux, France
| | - Maurice Garret
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, CNRS UMR 5287, Allee Geoffroy St Hilaire, CS 50023, 33615, Pessac Cedex, France
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, University of Bordeaux, Bordeaux, France
| | - Yoon H Cho
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, CNRS UMR 5287, Allee Geoffroy St Hilaire, CS 50023, 33615, Pessac Cedex, France.
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, University of Bordeaux, Bordeaux, France.
| |
Collapse
|
38
|
Rudenko O, Springer C, Skov LJ, Madsen AN, Hasholt L, Nørremølle A, Holst B. Ghrelin-mediated improvements in the metabolic phenotype in the R6/2 mouse model of Huntington's disease. J Neuroendocrinol 2019; 31:e12699. [PMID: 30776164 DOI: 10.1111/jne.12699] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/17/2019] [Accepted: 02/14/2019] [Indexed: 12/18/2022]
Abstract
Huntington's disease (HD) is a heritable neurodegenerative disorder, characterised by metabolic disturbances, along with cognitive and psychiatric impairments. Targeting metabolic HD dysfunction via the maintenance of body weight and fat mass and restoration of peripheral energy metabolism can improve the progression of neurological symptoms. In this respect, we focused on the therapeutic potential of the orexigenic peptide hormone ghrelin, which plays an important role in promoting a positive energy balance. In the present study, we found a significant disruption of circadian metabolic regulation in a R6/2 mouse HD model in the late stage of disease. Daily circadian rhythms of activity, energy expenditure, respiratory exchange ratio and feeding were strongly attenuated in R6/2 mice. During the rest phase, R6/2 mice had a higher total activity, elevated energy expenditure and excessive water consumption compared to control mice. We also found that, in the late stage of disease, R6/2 mice had ghrelin axis deficiency as a result of low circulating ghrelin levels, in addition to down-regulation of the ghrelin receptor and several key signalling molecules in the hypothalamus, as well as a reduced responsiveness to exogenous peripheral ghrelin. We demonstrated that, in pre-symptomatic mice, responsiveness to ghrelin is preserved. Chronic ghrelin treatment efficiently increased lean body mass and decreased the energy expenditure and fat utilisation of R6/2 mice in the early stage of disease. In addition, ghrelin treatment was also effective in the normalisation of drinking behaviour and the rest activity of these mice. Ghrelin treatment could provide a novel therapeutic possibility for delaying disease progression; however, deficiency in ghrelin receptor expression could limit its therapeutic potential in the late stage of disease.
Collapse
Affiliation(s)
- Olga Rudenko
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Cecilie Springer
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Louisa J Skov
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Andreas N Madsen
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Lis Hasholt
- Medical Genetics Program, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Nørremølle
- Medical Genetics Program, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte Holst
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
39
|
Park S, Colwell CS. Do Disruptions in the Circadian Timing System Contribute to Autonomic Dysfunction in Huntington's Disease? THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2019; 92:291-303. [PMID: 31249490 PMCID: PMC6585531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Huntington's disease (HD) patients suffer from a progressive neurodegenerative disorder that inflicts both motor and non-motor symptoms. HD is caused by a CAG repeat expansion within the first exon of the huntingtin (HTT) gene that produces a polyglutamine repeat that leads to protein misfolding, soluble aggregates, and inclusion bodies detected throughout the body. Both clinical and preclinical research indicate that cardiovascular dysfunction should be considered a core symptom in at least a subset of HD patients. There is strong evidence for dysautonomia (dysfunctional autonomic nervous system, ANS) in HD patients that can be detected early in the disease progression. The temporal patterning of ANS function is controlled by the circadian timing system based in the anterior hypothalamus. Patients with neurodegenerative diseases including HD exhibit disrupted sleep/wake cycle and, in preclinical models, there is compelling evidence that the circadian timing system is compromised early in the disease process. Here we review data from preclinical models of HD that explore the intersection between disruption of circadian rhythms and dysautonomia. This work will lead to new therapeutic strategies and standards of care for HD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Saemi Park
- Molecular, Cellular and Integrative Physiology graduate program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA,Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Christopher S. Colwell
- Molecular, Cellular and Integrative Physiology graduate program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA,Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA,Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA,To whom all correspondence should be addressed: Christopher S. Colwell, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095; . http://orcid.org/0000-0002-1059-184X
| |
Collapse
|
40
|
Buhl E, Higham JP, Hodge JJL. Alzheimer's disease-associated tau alters Drosophila circadian activity, sleep and clock neuron electrophysiology. Neurobiol Dis 2019; 130:104507. [PMID: 31207389 DOI: 10.1016/j.nbd.2019.104507] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/24/2019] [Accepted: 06/13/2019] [Indexed: 01/06/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, which is associated with an enormous personal, social and economic burden worldwide. However, there are few current treatments with none of them targeting the underlying causes of the disease. Sleep and circadian rhythm defects are not only distressing symptoms of AD and other tauopathies and are a leading cause of care home admission but are also thought to accelerate AD pathology. Despite this, little is understood about the underlying causes of these behavioural changes. Expression of the 0N4R isoform of tau has been associated with AD pathology and we show that expressing it in the Drosophila clock network gives rise to circadian and sleep phenotypes which closely match the behavioural changes seen in human AD patients. Tauopathic flies exhibited greater locomotor activity throughout the day and night and displayed a loss of sleep, particularly at night. Under constant darkness, the locomotor behaviour of tau-expressing flies was less rhythmic than controls indicating a defect in their intrinsic circadian rhythm. Current clamp recordings from wake-promoting, pigment dispersing factor (PDF)-positive large lateral ventral clock neurons (l-LNvs) revealed elevated spontaneous firing throughout the day and night which likely underlies the observed hyperactive circadian phenotype. Interestingly, expression of tau in only the PDF-positive pacemaker neurons, which are thought to be the most important for behaviour under constant conditions, was not sufficient or even necessary to affect circadian rhythmicity. This work establishes Drosophila as a model to investigate interactions between human pathological versions of tau and the machinery that controls neuronal excitability, allowing the identification of underlying mechanisms of disease that may reveal new therapeutic targets.
Collapse
Affiliation(s)
- Edgar Buhl
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol BS8 1TD, UK.
| | - James P Higham
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - James J L Hodge
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol BS8 1TD, UK.
| |
Collapse
|
41
|
Herzog–Krzywoszanska R, Krzywoszanski L. Sleep Disorders in Huntington's Disease. Front Psychiatry 2019; 10:221. [PMID: 31031659 PMCID: PMC6474183 DOI: 10.3389/fpsyt.2019.00221] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 03/26/2019] [Indexed: 12/13/2022] Open
Abstract
Huntington's chorea (Huntington's disease, HD) is a genetic disorder caused by autosomal dominant mutation, leading to progressive neurodegenerative changes in the central nervous system. Involuntary movements such as chorea occur typically in HD patients, accompanied by progressive cognitive and psychiatric disturbances. Other common symptoms of HD are circadian and sleep abnormalities, which are observed from the earliest stages of the disease or even before the occurrence of clinical symptoms. The most common sleep problems reported by HD patients include insomnia, difficulties in falling asleep, frequent nocturnal awakenings, and excessive daytime sleepiness. Also, specific changes in sleep architecture have been identified in HD. In this paper, we review studies on sleep and circadian rhythm disorders in HD. We outline findings concerning sleep patterns and disturbances of circadian rhythms in HD patients, as well as the role of psychiatric disorders and motor disorders in HD patients' sleep problems. We also discuss problems related to the different methods of diagnosing sleep disorders in HD. Furthermore, the adverse effects of medication used for the treatment of core HD symptoms as one of the sources of sleep disturbances in HD are emphasized. In conclusion, the diversity and complexity of the determinants of sleep and circadian rhythm disorders in HD are highlighted. Finally, the relevance of effective treatment to improve patients' functioning and quality of life as well as the potential relief of their cognitive and emotional symptoms is addressed.
Collapse
Affiliation(s)
| | - Lukasz Krzywoszanski
- Neurocognitive Psychology Unit, Chair of Psychology, Faculty of Pedagogy, Pedagogical University of Krakow, Krakow, Poland
| |
Collapse
|
42
|
Fifel K, Videnovic A. Chronotherapies for Parkinson's disease. Prog Neurobiol 2019; 174:16-27. [PMID: 30658126 PMCID: PMC6377295 DOI: 10.1016/j.pneurobio.2019.01.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 11/18/2018] [Accepted: 01/14/2019] [Indexed: 02/08/2023]
Abstract
Parkinson's disease (PD) is the second-most common progressive neurodegenerative disorder. Although the clinical diagnosis of PD is still based on its cardinal motor dysfunctions, several non-motor symptoms (NMS) have been established as integral part of the disease. Unlike motor disorders, development of therapies against NMS are still challenging and remain a critical unmet clinical need. During the last decade, several studies have characterised the molecular, physiological and behavioural alterations of the circadian system in PD patients. As a consequence, and given the ubiquitous nature of circadian rhythms in the entire organism, the biological clock has emerged as a potential therapeutic target to ease suffering from both motor and NMS in PD patients. Here we discuss the emerging field of using bright light, physical exercise and melatonin as chronotherapeutic tools to alleviate motor disorders, sleep/wake alterations, anxiety and depression in PD patients. We also highlight the potential of these readily available therapies to improve the general quality of life and wellbeing of PD patients. Finally, we provide specific data- and mechanisms-driven recommendations that might help improve the therapeutic benefit of light and physical exercise in PD patients.
Collapse
Affiliation(s)
- Karim Fifel
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan; Department of Molecular Cell Biology, Neurophysiology unit, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, the Netherlands; Stem Cell and Brain Research Institute, Department of Chronobiology, 18 Avenue du Doyen Lépine, 69500, Bron, France; Laboratory of Pharmacology, Neurobiology and Behavior, Associated CNRST Unit (URAC-37), Cadi Ayyad University, Marrakech, Morocco.
| | - Aleksandar Videnovic
- Movement Disorders Unit and Division of Sleep Medicine, Massachusetts General Hospital, Harvard Medical School, 165 Cambridge Street, Suite 600, Boston, MA, 02446, USA
| |
Collapse
|
43
|
Lin MS, Liao PY, Chen HM, Chang CP, Chen SK, Chern Y. Degeneration of ipRGCs in Mouse Models of Huntington's Disease Disrupts Non-Image-Forming Behaviors Before Motor Impairment. J Neurosci 2019; 39:1505-1524. [PMID: 30587542 PMCID: PMC6381252 DOI: 10.1523/jneurosci.0571-18.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 11/22/2018] [Accepted: 12/17/2018] [Indexed: 12/20/2022] Open
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs), which express the photopigment melanopsin, are photosensitive neurons in the retina and are essential for non-image-forming functions, circadian photoentrainment, and pupillary light reflexes. Five subtypes of ipRGCs (M1-M5) have been identified in mice. Although ipRGCs are spared in several forms of inherited blindness, they are affected in Alzheimer's disease and aging, which are associated with impaired circadian rhythms. Huntington's disease (HD) is an autosomal neurodegenerative disease caused by the expansion of a CAG repeat in the huntingtin gene. In addition to motor function impairment, HD mice also show impaired circadian rhythms and loss of ipRGC. Here, we found that, in HD mouse models (R6/2 and N171-82Q male mice), the expression of melanopsin was reduced before the onset of motor deficits. The expression of retinal T-box brain 2, a transcription factor essential for ipRGCs, was associated with the survival of ipRGCs. The number of M1 ipRGCs in R6/2 male mice was reduced due to apoptosis, whereas non-M1 ipRGCs were relatively resilient to HD progression. Most importantly, the reduced innervations of M1 ipRGCs, which was assessed by X-gal staining in R6/2-OPN4Lacz/+ male mice, contributed to the diminished light-induced c-fos and vasoactive intestinal peptide in the suprachiasmatic nuclei (SCN), which may explain the impaired circadian photoentrainment in HD mice. Collectively, our results show that M1 ipRGCs were susceptible to the toxicity caused by mutant Huntingtin. The resultant impairment of M1 ipRGCs contributed to the early degeneration of the ipRGC-SCN pathway and disrupted circadian regulation during HD progression.SIGNIFICANCE STATEMENT Circadian disruption is a common nonmotor symptom of Huntington's disease (HD). In addition to the molecular defects in the suprachiasmatic nuclei (SCN), the cause of circadian disruption in HD remains to be further explored. We hypothesized that ipRGCs, by integrating light input to the SCN, participate in the circadian regulation in HD mice. We report early reductions in melanopsin in two mouse models of HD, R6/2, and N171-82Q. Suppression of retinal T-box brain 2, a transcription factor essential for ipRGCs, by mutant Huntingtin might mediate the reduced number of ipRGCs. Importantly, M1 ipRGCs showed higher susceptibility than non-M1 ipRGCs in R6/2 mice. The resultant impairment of M1 ipRGCs contributed to the early degeneration of the ipRGC-SCN pathway and the circadian abnormality during HD progression.
Collapse
Affiliation(s)
- Meng-Syuan Lin
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei 115, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan, and
| | - Po-Yu Liao
- Department of Life Science, National Taiwan University, Taipei 106, Taiwan
| | - Hui-Mei Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan, and
| | - Ching-Pang Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan, and
| | - Shih-Kuo Chen
- Department of Life Science, National Taiwan University, Taipei 106, Taiwan
| | - Yijuang Chern
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan, and
| |
Collapse
|
44
|
Seifalian A, Hart A. Circadian Rhythms: Will It Revolutionise the Management of Diseases? J Lifestyle Med 2019; 9:1-11. [PMID: 30918828 PMCID: PMC6425903 DOI: 10.15280/jlm.2019.9.1.1] [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: 05/25/2018] [Accepted: 09/26/2018] [Indexed: 11/22/2022] Open
Abstract
The Nobel Prize for Medicine in 2017 was awarded to Michael Young, Michael Rosbash and Jeffrey Hall for their discoveries into the molecular mechanisms controlling circadian rhythms (CR). The aims of this paper were to present the mechanisms behind the CRs and discuss the impact this could have on human health. We argued that further research in this field has the potential to revolutionise healthcare through understanding the influence on the pathogenesis of disease, including in cardiovascular, mental and neurological health, as well as influence on cognitive function. The research has shown that intrinsic CRs have physiological and biochemical influences on the body, which may affect the efficiency of drug absorption due to the altered activity of enzymes. There is strong data to suggest CR disturbances, due to either shift work, sleep disorders or frequent travel between time zones, has negative impact on health. This article aims to summarise the extent of this impact and analyse CRs as a potential therapeutic target, as well as describing the pathophysiology and mechanisms driving the course of disease among people with CR disorders. These new discoveries may revolutionise the way in which treatment is provided in the future with more focus on lifestyle changes to provide treatment and more optimal precision medicine. Pharmaceutical companies and healthcare staff must consider the significant message provided from this data and use the information to optimise drug delivery and treatment provision. The facts of CRs role in healthcare can no longer be ignored.
Collapse
Affiliation(s)
- Amelia Seifalian
- University College London Medical School, London, United Kingdom
| | - Ashley Hart
- University College London Medical School, London, United Kingdom
| |
Collapse
|
45
|
Paul JR, Davis JA, Goode LK, Becker BK, Fusilier A, Meador-Woodruff A, Gamble KL. Circadian regulation of membrane physiology in neural oscillators throughout the brain. Eur J Neurosci 2019; 51:109-138. [PMID: 30633846 DOI: 10.1111/ejn.14343] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 12/19/2018] [Accepted: 12/21/2018] [Indexed: 12/21/2022]
Abstract
Twenty-four-hour rhythmicity in physiology and behavior are driven by changes in neurophysiological activity that vary across the light-dark and rest-activity cycle. Although this neural code is most prominent in neurons of the primary circadian pacemaker in the suprachiasmatic nucleus (SCN) of the hypothalamus, there are many other regions in the brain where region-specific function and behavioral rhythmicity may be encoded by changes in electrical properties of those neurons. In this review, we explore the existing evidence for molecular clocks and/or neurophysiological rhythms (i.e., 24 hr) in brain regions outside the SCN. In addition, we highlight the brain regions that are ripe for future investigation into the critical role of circadian rhythmicity for local oscillators. For example, the cerebellum expresses rhythmicity in over 2,000 gene transcripts, and yet we know very little about how circadian regulation drives 24-hr changes in the neural coding responsible for motor coordination. Finally, we conclude with a discussion of how our understanding of circadian regulation of electrical properties may yield insight into disease mechanisms which may lead to novel chronotherapeutic strategies in the future.
Collapse
Affiliation(s)
- Jodi R Paul
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jennifer A Davis
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Lacy K Goode
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Bryan K Becker
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Allison Fusilier
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Aidan Meador-Woodruff
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Karen L Gamble
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
46
|
Rieke L, Fels M, Schubert R, Habbel B, Matheis T, Schuldenzucker V, Kemper N, Reilmann R. Activity Behaviour of Minipigs Transgenic for the Huntington Gene. J Huntingtons Dis 2019; 8:23-31. [PMID: 30689591 DOI: 10.3233/jhd-180325] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND To increase the reliability of translating preclinical findings to humans, large animal models, such as the transgenic (tg) Libechov minipig, were established. As minipigs possess high genetic homology with humans and have similarities in anatomy, physiology and metabolism to humans, they are considered for studying neurodegenerative diseases longitudinally. Recently, sleep abnormalities and changes in circadian rhythm in Huntington's disease (HD) patients were acknowledged to present one of the early symptoms in HD. OBJECTIVE The aim of the present study was to explore the activity behaviour of Libechov minipigs and to investigate whether tgHD and wildtype (wt) minipigs exhibit differences in activity behaviour. Furthermore, it was investigated whether activity assessments may serve as reliable endpoints for phenotyping minipigs transgenic for the Huntington gene. METHODS Activity behaviour of minipigs was studied by video recording the stables twice a week over a total study period of five weeks for a cohort of five tgHD minipigs and five wt minipigs. Statistical analysis was performed using the linear mixed model. Once a week, the distances covered by two minipigs in focus (tgHD, wt) were measured using the VideoMotionTracker® software. RESULTS Libechov minipigs showed a biphasic pattern of activity, spending most of the time inactive or grubbing in litter. Differences in activity behaviour (rooting, resting and standing) were detected between wt and tgHD minipigs. The influence of the genotype on behavioural patterns was observed during circadian monitoring. TgHD minipigs covered longer distances on average and during every 24 h observation period than wt minipigs. CONCLUSION Activity behaviour may be a viable marker for phenotyping minipigs transgenic for the Huntington gene. Video recordings of behavioural patterns provide a non-invasive opportunity to capture potential disease signs. Phenotypic progression including the age of disease manifestation may be explored by documentation of circadian characteristics.
Collapse
Affiliation(s)
- Lorena Rieke
- George-Huntington-Institute, Technology Park Muenster, Muenster, Germany.,Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Michaela Fels
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Robin Schubert
- George-Huntington-Institute, Technology Park Muenster, Muenster, Germany
| | - Benjamin Habbel
- George-Huntington-Institute, Technology Park Muenster, Muenster, Germany
| | - Tamara Matheis
- George-Huntington-Institute, Technology Park Muenster, Muenster, Germany.,Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | | | - Nicole Kemper
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Ralf Reilmann
- George-Huntington-Institute, Technology Park Muenster, Muenster, Germany.,Department of Radiology, Universitaetsklinikum Muenster, Muenster, Germany.,Department of Neurodegenerative Diseases and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
47
|
Hsu YT, Chang YG, Chern Y. Insights into GABA Aergic system alteration in Huntington's disease. Open Biol 2018; 8:rsob.180165. [PMID: 30518638 PMCID: PMC6303784 DOI: 10.1098/rsob.180165] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/30/2018] [Indexed: 12/15/2022] Open
Abstract
Huntington's disease (HD) is an autosomal dominant progressive neurodegenerative disease that is characterized by a triad of motor, psychiatric and cognitive impairments. There is still no effective therapy to delay or halt the disease progress. The striatum and cortex are two particularly affected brain regions that exhibit dense reciprocal excitatory glutamate and inhibitory gamma-amino butyric acid (GABA) connections. Imbalance between excitatory and inhibitory signalling is known to greatly affect motor and cognitive processes. Emerging evidence supports the hypothesis that disrupted GABAergic circuits underlie HD pathogenesis. In the present review, we focused on the multiple defects recently found in the GABAergic inhibitory system, including altered GABA level and synthesis, abnormal subunit composition and distribution of GABAA receptors and aberrant GABAA receptor-mediated signalling. In particular, the important role of cation–chloride cotransporters (i.e. NKCC1 and KCC2) is discussed. Recent studies also suggest that neuroinflammation contributes significantly to the abnormal GABAergic inhibition in HD. Thus, GABAA receptors and cation–chloride cotransporters are potential therapeutic targets for HD. Given the limited availability of therapeutic treatments for HD, a better understanding of GABAergic dysfunction in HD could provide novel therapeutic opportunities.
Collapse
Affiliation(s)
- Yi-Ting Hsu
- PhD Program for Translational Medicine, China Medical University and Academia Sinica, Taiwan, Republic of China.,Department of Neurology, China Medical University Hospital, Taichung, Taiwan, Republic of China
| | - Ya-Gin Chang
- Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan, Republic of China.,Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei, Taiwan, Republic of China
| | - Yijuang Chern
- PhD Program for Translational Medicine, China Medical University and Academia Sinica, Taiwan, Republic of China .,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, Republic of China
| |
Collapse
|
48
|
Gjerstad MD, Alves G, Maple-Grødem J. Excessive Daytime Sleepiness and REM Sleep Behavior Disorders in Parkinson's Disease: A Narrative Review on Early Intervention With Implications to Neuroprotection. Front Neurol 2018; 9:961. [PMID: 30487775 PMCID: PMC6246656 DOI: 10.3389/fneur.2018.00961] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 10/25/2018] [Indexed: 11/13/2022] Open
Abstract
Sleep contributes to the consolidation of our memory and facilitates learning. Short term sleep deprivation temporarily reduces mnestic capacity, whereas long lasting sleep deprivation is associated with structural changes in the hippocampus and cortical areas. However, it is unknown whether early intervention and treatment of sleep disorders could have a neuroprotective effect. In neurodegenerative diseases sleep disorders can occur at preclinical stages and are frequently observed in patients with established Parkinson's disease (PD) and other α-synucleinopathies. REM sleep behavior disorder (RBD) is recognized as a hallmark for the development of α-synucleinopathies and may predict early cognitive decline, while excessive daytime sleepiness (EDS) is present in 12% of patients with PD before treatment initiation and increases continuously over time, causing substantial restrictions for the patients' social life. In more advanced disease, EDS is associated with dementia. Even though well recognized, limited attention has been given to genetics or the treatment of RBD and EDS in early PD. Systematic screening and early intervention can be expected to increase the patients' quality of life, but it remains unclear if this will also impact disease progression. Intervention studies in preclinical and early stages of α-synucleinopathies are needed to increase our understanding of the underlying pathomechanisms and may also provide important inroads to help clarify whether sleep disturbances are secondary to the neurodegenerative process or also contribute to disease exacerbation.
Collapse
Affiliation(s)
- Michaela D Gjerstad
- The Norwegian Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway.,Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway.,Department of Neurology, Stavanger University Hospital, Stavanger, Norway
| | - Guido Alves
- The Norwegian Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway.,Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway.,Department of Neurology, Stavanger University Hospital, Stavanger, Norway
| | - Jodi Maple-Grødem
- The Norwegian Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway.,Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway
| |
Collapse
|
49
|
Kuljis D, Kudo T, Tahara Y, Ghiani CA, Colwell CS. Pathophysiology in the suprachiasmatic nucleus in mouse models of Huntington's disease. J Neurosci Res 2018; 96:1862-1875. [PMID: 30168855 DOI: 10.1002/jnr.24320] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/04/2018] [Accepted: 08/07/2018] [Indexed: 12/30/2022]
Abstract
Disturbances in sleep/wake cycle are a common complaint of individuals with Huntington's disease (HD) and are displayed by HD mouse models. The underlying mechanisms, including the possible role of the circadian timing system, are not well established. The BACHD mouse model of HD exhibits disrupted behavioral and physiological rhythms, including decreased electrical activity in the central circadian clock (suprachiasmatic nucleus, SCN). In this study, electrophysiological techniques were used to explore the ionic underpinning of the reduced spontaneous neural activity in male mice. We found that SCN neural activity rhythms were lost early in the disease progression and was accompanied by loss of the normal daily variation in resting membrane potential in the mutant SCN neurons. The low neural activity could be transiently reversed by direct current injection or application of exogenous N-methyl-d-aspartate (NMDA) thus demonstrating that the neurons have the capacity to discharge at WT levels. Exploring the potassium currents known to regulate the electrical activity of SCN neurons, our most striking finding was that these cells in the mutants exhibited an enhancement in the large-conductance calcium activated K+ (BK) currents. The expression of the pore forming subunit (Kcnma1) of the BK channel was higher in the mutant SCN. We found a similar decrease in daytime electrical activity and enhancement in the magnitude of the BK currents early in disease in another HD mouse model (Q175). These findings suggest that SCN neurons of both HD models exhibit early pathophysiology and that dysregulation of BK current may be responsible.
Collapse
Affiliation(s)
- Dika Kuljis
- Department of Neurobiology, University of California Los Angeles, Los Angeles, California.,Department of Biological Sciences, Mellon Institute, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Takashi Kudo
- Department of Psychiatry & Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California.,Okinawa Institute of Science and Technology Graduate University, Onna-son, Japan
| | - Yu Tahara
- Department of Psychiatry & Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California
| | - Cristina A Ghiani
- Department of Psychiatry & Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California.,Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California
| | - Christopher S Colwell
- Department of Psychiatry & Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
50
|
Whittaker DS, Loh DH, Wang HB, Tahara Y, Kuljis D, Cutler T, Ghiani CA, Shibata S, Block GD, Colwell CS. Circadian-based Treatment Strategy Effective in the BACHD Mouse Model of Huntington's Disease. J Biol Rhythms 2018; 33:535-554. [PMID: 30084274 DOI: 10.1177/0748730418790401] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Huntington's disease (HD) patients suffer from progressive neurodegeneration that results in cognitive, psychiatric, cardiovascular, and motor dysfunction. Disturbances in sleep-wake cycles are common among HD patients with reports of delayed sleep onset, frequent bedtime awakenings, and excessive fatigue. The BACHD mouse model exhibits many HD core symptoms including circadian dysfunction. Because circadian dysfunction manifests early in the disease in both patients and mouse models, we sought to determine if early interventions that improve circadian rhythmicity could benefit HD symptoms and delay disease progression. We evaluated the effects of time-restricted feeding (TRF) on the BACHD mouse model. At 3 months of age, the animals were divided into 2 groups: ad lib and TRF. The TRF-treated BACHD mice were exposed to a 6-h feeding/18-h fasting regimen that was designed to be aligned with the middle (ZT 15-21) of the period when mice are normally active (ZT 12-24). Following 3 months of treatment (when mice reached the early disease stage), the TRF-treated BACHD mice showed improvements in their locomotor activity and sleep behavioral rhythms. Furthermore, we found improved heart rate variability, suggesting that their autonomic nervous system dysfunction was improved. On a molecular level, TRF altered the phase but not the amplitude of the PER2::LUC rhythms measured in vivo and in vitro. Importantly, treated BACHD mice exhibited improved motor performance compared with untreated BACHD controls, and the motor improvements were correlated with improved circadian output. It is worth emphasizing that HD is a genetically caused disease with no known cure. Lifestyle changes that not only improve the quality of life but also delay disease progression for HD patients are greatly needed. Our study demonstrates the therapeutic potential of circadian-based treatment strategies in a preclinical model of HD.
Collapse
Affiliation(s)
- Daniel S Whittaker
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California, USA
| | - Dawn H Loh
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California, USA
| | - Huei-Bin Wang
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California, USA
| | - Yu Tahara
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California, USA
| | - Dika Kuljis
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California, USA
| | - Tamara Cutler
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California, USA
| | - Cristina A Ghiani
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, California, USA
| | - Shigenobu Shibata
- Waseda Institute for Advanced Study, Waseda University, Shinjuku-ku, Tokyo, Japan
| | - Gene D Block
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California, USA
| | - Christopher S Colwell
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|