Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neurobiol Dis ; 145: 105064, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32889171

RESUMO

Nighttime light pollution is linked to metabolic and cognitive dysfunction. Many patients with autism spectrum disorders (ASD) show disturbances in their sleep/wake cycle, and may be particularly vulnerable to the impact of circadian disruptors. In this study, we examined the impact of exposure to dim light at night (DLaN, 5 lx) in a model of ASD: the contactin associated protein-like 2 knock out (Cntnap2 KO) mice. DLaN was sufficient to disrupt locomotor activity rhythms, exacerbate the excessive grooming and diminish the social preference in Cntnap2 mutant mice. On a molecular level, DLaN altered the phase and amplitude of PER2:LUC rhythms in a tissue-specific manner in vitro. Daily treatment with melatonin reduced the excessive grooming of the mutant mice to wild-type levels and improved activity rhythms. Our findings suggest that common circadian disruptors such as light at night should be considered in the management of ASD.


Assuntos
Transtorno do Espectro Autista , Depressores do Sistema Nervoso Central/farmacologia , Ritmo Circadiano/efeitos dos fármacos , Iluminação/efeitos adversos , Melatonina/farmacologia , Animais , Transtorno do Espectro Autista/genética , Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética
2.
J Neurosci Res ; 97(12): 1606-1623, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31359503

RESUMO

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.


Assuntos
Ritmo Circadiano/fisiologia , Proteína Huntingtina/fisiologia , Doença de Huntington/fisiopatologia , Doença de Huntington/psicologia , Locomoção/fisiologia , Animais , Ritmo Circadiano/genética , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Frequência Cardíaca/genética , Frequência Cardíaca/fisiologia , Proteína Huntingtina/genética , Doença de Huntington/genética , Locomoção/genética , Masculino , Camundongos Transgênicos , Atividade Motora/genética , Atividade Motora/fisiologia , Neurônios/fisiologia , Sono/genética , Sono/fisiologia , Núcleo Supraquiasmático/fisiologia
3.
J Biol Rhythms ; 33(5): 535-554, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30084274

RESUMO

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.


Assuntos
Ritmo Circadiano , Jejum , Doença de Huntington/terapia , Animais , Estudos de Coortes , Modelos Animais de Doenças , Frequência Cardíaca , Masculino , Camundongos , Camundongos Transgênicos , Atividade Motora , Fotoperíodo , Qualidade de Vida
4.
eNeuro ; 5(1)2018.
Artigo em Inglês | MEDLINE | ID: mdl-29302618

RESUMO

Huntington's disease (HD) patients suffer from a 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 fatigue during the day. The heterozygous Q175 mouse model of HD has been shown to phenocopy many HD core symptoms including circadian dysfunctions. Because circadian dysfunction manifests early in the disease in both patients and mouse models, we sought to determine if early intervention that improve circadian rhythmicity can benefit HD and delay disease progression. We determined the effects of time-restricted feeding (TRF) on the Q175 mouse model. At six months of age, the animals were divided into two groups: ad libitum (ad lib) and TRF. The TRF-treated Q175 mice were exposed to a 6-h feeding/18-h fasting regimen that was designed to be aligned with the middle of the time when mice are normally active. After three months of treatment (when mice reached the early disease stage), the TRF-treated Q175 mice showed improvements in their locomotor activity rhythm and sleep awakening time. Furthermore, we found improved heart rate variability (HRV), suggesting that their autonomic nervous system dysfunction was improved. Importantly, treated Q175 mice exhibited improved motor performance compared to untreated Q175 controls, and the motor improvements were correlated with improved circadian output. Finally, we found that the expression of several HD-relevant markers was restored to WT levels in the striatum of the treated mice using NanoString gene expression assays.


Assuntos
Ritmo Circadiano , Doença de Huntington/dietoterapia , Atividade Motora , Animais , Sistema Nervoso Autônomo/fisiopatologia , Ritmo Circadiano/fisiologia , Modelos Animais de Doenças , Ingestão de Alimentos/fisiologia , Jejum/fisiologia , Frequência Cardíaca/fisiologia , Doença de Huntington/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/fisiologia , Sono/fisiologia , Fatores de Tempo
5.
Artigo em Inglês | MEDLINE | ID: mdl-28971617

RESUMO

Huntington's disease (HD) is an autosomal dominant, neurodegenerative disorder characterized by motor as well as nonmotor symptoms for which there is currently no cure. The Q175 mouse model of HD recapitulates many of the symptoms identified in HD patients including disruptions of the sleep/wake cycle. In this study, we sought to determine if the daily administration of the histamine-3 receptor (H3R) antagonist/inverse agonist 6-[(3-cyclobutyl-2,3,4,5-tetrahydro-1H-3-benzazepin-7-yl)oxy]-N-methyl-3-pyridinecarboxamide hydrochloride (GSK189254) would improve nonmotor symptoms in the Q175 line. This class of drugs acts on autoreceptors found at histaminergic synapses and results in increased levels of histamine (HA). HA is a neuromodulator whose levels vary with a daily rhythm with peak release during the active cycle and relatively lower levels during sleep. H3Rs are widely expressed in brain regions involved in cognitive processes and activation of these receptors promotes wakefulness. We administered GSK189254 nightly to homozygote and heterozygote Q175 mice for 4 weeks and confirmed that the plasma levels of the drug were elevated to a therapeutic range. We demonstrate that daily treatment with GSK189254 improved several behavioral measures in the Q175 mice including strengthening activity rhythms, cognitive performance and mood as measured by the tail suspension test. The treatment also reduced inappropriate activity during the normal sleep time. The drug treatment did not alter motor performance and coordination as measured by the challenging beam test. Our findings suggest that drugs targeting the H3R system may show benefits as cognitive enhancers in the management of HD.


Assuntos
Benzazepinas/administração & dosagem , Cognição/efeitos dos fármacos , Antagonistas dos Receptores Histamínicos H3/administração & dosagem , Doença de Huntington/tratamento farmacológico , Niacinamida/análogos & derivados , Animais , Benzazepinas/farmacologia , Modelos Animais de Doenças , Esquema de Medicação , Antagonistas dos Receptores Histamínicos H3/farmacologia , Humanos , Doença de Huntington/metabolismo , Doença de Huntington/psicologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Atividade Motora , Niacinamida/administração & dosagem , Niacinamida/farmacologia , Receptores Histamínicos H3
6.
Physiol Rep ; 5(11)2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28576852

RESUMO

Cardiovascular dysautonomia as well as the deterioration of circadian rhythms are among the earliest detectable pathophysiological changes in individuals with Huntington's disease (HD). Preclinical research requires mouse models that recapitulate disease symptoms and the Q175 knock-in model offers a number of advantages but potential autonomic dysfunction has not been explored. In this study, we sought to test the dual hypotheses that cardiovascular dysautonomia can be detected early in disease progression in the Q175 model and that this dysfunction varies with the daily cycle. Using radiotelemetry implants, we observed a significant reduction in the diurnal and circadian activity rhythms in the Q175 mutants at the youngest ages. By middle age, the autonomically driven rhythms in core body temperature were highly compromised, and the Q175 mutants exhibited striking episodes of hypothermia that increased in frequency with mutant huntingtin gene dosage. In addition, Q175 mutants showed higher resting heart rate (HR) during sleep and greatly reduced correlation between activity and HR HR variability was reduced in the mutants in both time and frequency domains, providing more evidence of autonomic dysfunction. Measurement of the baroreceptor reflex revealed that the Q175 mutant could not appropriately increase HR in response to a pharmacologically induced decrease in blood pressure. Echocardiograms showed reduced ventricular mass and ejection fraction in mutant hearts. Finally, cardiac histopathology revealed localized points of fibrosis resembling those caused by myocardial infarction. Thus, the Q175 mouse model of HD exhibits cardiovascular dysautonomia similar to that seen in HD patients with prominent sympathetic dysfunction during the resting phase of the activity rhythm.


Assuntos
Sistema Nervoso Autônomo/fisiopatologia , Coração/fisiopatologia , Proteína Huntingtina/genética , Doença de Huntington/fisiopatologia , Animais , Barorreflexo , Pressão Sanguínea , Temperatura Corporal , Ritmo Circadiano , Coração/inervação , Frequência Cardíaca , Doença de Huntington/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Volume Sistólico
7.
Artigo em Inglês | MEDLINE | ID: mdl-31236494

RESUMO

Patients with Huntington's disease (HD) exhibit movement disorders, psychiatric disturbance and cognitive impairments as the disease progresses. Abnormal sleep/wake cycles are common among HD patients with reports of delayed sleep onset, fatigue during the day, and a delayed pattern of melatonin secretion all of which suggest circadian dysfunction. Mouse models of HD confirm disrupted circadian rhythms with pathophysiology found in the central circadian clock (suprachiasmatic nucleus). Importantly, circadian dysfunction manifests early in disease, even before the classic motor symptoms, in both patients and mouse models. Therefore, we hypothesize that the circadian dysfunction may interact with the disease pathology and exacerbate the HD symptoms. If correct, early intervention may benefit patients and delay disease progression. One test of this hypothesis is to determine whether light therapy designed to strengthen this intrinsic timing system can delay the disease progression in mouse models. Therefore, we determined the impact of blue wavelength-enriched light on two HD models: the BACHD and Q175 mice. Both models received 6 h of blue-light at the beginning of their daily light cycle for 3 months. After treatment, both genotypes showed improvements in their locomotor activity rhythm without significant change to their sleep behavior. Critically, treated mice of both lines exhibited improved motor performance compared to untreated controls. Focusing on the Q175 genotype, we sought to determine whether the treatment altered signaling pathways in brain regions known to be impacted by HD using NanoString gene expression assays. We found that the expression of several HD relevant markers was altered in the striatum and cortex of the treated mice. Our study demonstrates that strengthening the circadian system can delay the progression of HD in pre-clinical models. This work suggests that lighting conditions should be considered when managing treatment of HD and other neurodegenerative disorders.

8.
PLoS One ; 11(2): e0147583, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26871695

RESUMO

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder that affects men and women in equal numbers, but some epidemiological studies indicate there may be sex differences in disease progression. One of the early symptoms of HD is disruptions in the circadian timing system, but it is currently unknown whether sex is a factor in these alterations. Since sex differences in HD could provide important insights to understand cellular and molecular mechanism(s) and designing early intervention strategies, we used the bacterial artificial chromosome transgenic mouse model of HD (BACHD) to examine whether sex differences in circadian behavioral rhythms are detectable in an animal model of the disease. Similar to BACHD males, BACHD females display circadian disruptions at both 3 and 6 months of age; however, deficits to BACHD female mouse activity levels, rhythm precision, and behavioral fragmentation are either delayed or less severe relative to males. These sex differences are associated with a smaller suprachiasmatic nucleus (SCN) in BACHD male mice at age of symptom onset (3 months), but are not associated with sex-specific differences in SCN daytime electrical activity deficits, or peptide expression (arginine vasopressin, vasoactive intestinal peptide) within the SCN. Notably, BACHD females exhibited delayed motor coordination deficits, as measured using rotarod and challenge beam. These findings suggest a sex specific factor plays a role both in non-motor and motor symptom progression for the BACHD mouse.


Assuntos
Ritmo Circadiano/genética , Modelos Animais de Doenças , Doença de Huntington/fisiopatologia , Camundongos Transgênicos/genética , Núcleo Supraquiasmático/fisiopatologia , Animais , Arginina Vasopressina/genética , Arginina Vasopressina/metabolismo , Cromossomos Artificiais Bacterianos/genética , Progressão da Doença , Feminino , Efeito Fundador , Expressão Gênica , Humanos , Doença de Huntington/genética , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Masculino , Camundongos , Atividade Motora , Teste de Desempenho do Rota-Rod , Fatores Sexuais , Núcleo Supraquiasmático/anormalidades , Núcleo Supraquiasmático/metabolismo , Fatores de Tempo , Peptídeo Intestinal Vasoativo/genética , Peptídeo Intestinal Vasoativo/metabolismo
9.
Elife ; 42015 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-26652002

RESUMO

Robust sleep/wake rhythms are important for health and cognitive function. Unfortunately, many people are living in an environment where their circadian system is challenged by inappropriate meal- or work-times. Here we scheduled food access to the sleep time and examined the impact on learning and memory in mice. Under these conditions, we demonstrate that the molecular clock in the master pacemaker, the suprachiasmatic nucleus (SCN), is unaltered while the molecular clock in the hippocampus is synchronized by the timing of food availability. This chronic circadian misalignment causes reduced hippocampal long term potentiation and total CREB expression. Importantly this mis-timed feeding resulted in dramatic deficits in hippocampal-dependent learning and memory. Our findings suggest that the timing of meals have far-reaching effects on hippocampal physiology and learned behaviour.


Assuntos
Ritmo Circadiano , Comportamento Alimentar , Memória , Animais , Métodos de Alimentação , Hipocampo/fisiologia , Camundongos , Núcleo Supraquiasmático/fisiologia
10.
Horm Behav ; 75: 55-63, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26226656

RESUMO

We measured diurnal rhythms of food intake, as well as body weight and composition, while varying three major classes of sex-biasing factors: activational and organizational effects of gonadal hormones, and sex chromosome complement (SCC). Four Core Genotypes (FCG) mice, comprising XX and XY gonadal males and XX and XY gonadal females, were either gonad-intact or gonadectomized (GDX) as adults (2.5months); food intake was measured second-by-second for 7days starting 5weeks later, and body weight and composition were measured for 22weeks thereafter. Gonadal males weighed more than females. GDX increased body weight/fat of gonadal females, but increased body fat and reduced body weight of males. After GDX, XX mice had greater body weight and more fat than XY mice. In gonad-intact mice, males had greater total food intake and more meals than females during the dark phase, but females had more food intake and meals and larger meals than males during the light phase. GDX reduced overall food intake irrespective of gonad type or SCC, and eliminated differences in feeding between groups with different gonads. Diurnal phase of feeding was influenced by all three sex-biasing variables. Gonad-intact females had earlier onset and acrophase (peak) of feeding relative to males. GDX caused a phase-advance of feeding, especially in XX mice, leading to an earlier onset of feeding in GDX XX vs. XY mice, but earlier acrophase in GDX males relative to females. Gonadal hormones and SCC interact in the control of diurnal rhythms of food intake.


Assuntos
Ritmo Circadiano , Ingestão de Alimentos/fisiologia , Hormônios Gonadais/sangue , Caracteres Sexuais , Cromossomos Sexuais/fisiologia , Animais , Composição Corporal/fisiologia , Peso Corporal , Ritmo Circadiano/genética , Ingestão de Alimentos/genética , Feminino , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fatores Sexuais
11.
Neuron ; 85(5): 895-8, 2015 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-25741718

RESUMO

Circadian behavior in mammals is coordinated by neurons within the suprachiasmatic nucleus (SCN). In this issue, Lee et al. (2015) and Mieda et al. (2015) applied state-of-the-art genetic tools to dissect the microcircuits within the SCN generating circadian rhythmic behavior.


Assuntos
Fatores de Transcrição ARNTL/deficiência , Arginina Vasopressina/metabolismo , Relógios Circadianos/fisiologia , Ritmo Circadiano/fisiologia , Interneurônios/fisiologia , Neurônios/fisiologia , Neuropeptídeos/biossíntese , Núcleo Supraquiasmático/fisiologia , Animais
12.
Neurobiol Dis ; 77: 155-64, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25779967

RESUMO

Disturbances in the sleep/wake cycle are prevalent in patients with Rett syndrome (RTT). We sought to determine whether the circadian system is disrupted in a RTT model, Mecp2(-/y) mice. We found that MeCP2 mutants showed decreased strength and precision of daily rhythms of activity coupled with extremely fragmented sleep. The central circadian clock (suprachiasmatic nucleus) exhibited significant reduction in the number of neurons expressing vasoactive intestinal peptide (VIP) as well as compromised spontaneous neural activity. The molecular clockwork was disrupted both centrally in the SCN and in peripheral organs, indicating a general disorganization of the circadian system. Disruption of the molecular clockwork was observed in fibroblasts of RTT patients. Finally, MeCP2 mutant mice were vulnerable to circadian disruption as chronic jet lag accelerated mortality. Our finds suggest an integral role of MeCP2 in the circadian timing system and provides a possible mechanistic explanation for the sleep/wake distrubances observed in RTT patients. The work raises the possibility that RTT patients may benefit from a temporally structured environment.


Assuntos
Transtornos Cronobiológicos/etiologia , Modelos Animais de Doenças , Síndrome de Rett/complicações , Potenciais de Ação/genética , Animais , Células Cultivadas , Transtornos Cronobiológicos/genética , Embrião de Mamíferos , Regulação da Expressão Gênica/genética , Técnicas In Vitro , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/genética , Mutação/genética , Técnicas de Patch-Clamp , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Síndrome de Rett/genética , Transtornos do Sono-Vigília/etiologia
13.
ASN Neuro ; 6(1): e00133, 2014 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-24328694

RESUMO

Sleep disorders are common in neurodegenerative diseases including Huntington's disease (HD) and develop early in the disease process. Mitochondrial alterations are believed to play a critical role in the pathophysiology of neurodegenerative diseases. In the present study, we evaluated the circadian system of mice after inhibiting mitochondrial complex II of the respiratory chain with the toxin 3-nitropropionic acid (3-NP). We found that a subset of mice treated with low doses of 3-NP exhibited severe circadian deficit in behavior. The temporal patterning of sleep behavior is also disrupted in some mice with evidence of difficulty in the initiation of sleep behavior. Using the open field test during the normal sleep phase, we found that the 3-NP-treated mice were hyperactive. The molecular clockwork responsible for the generation of circadian rhythms as measured by PER2::LUCIFERASE was disrupted in a subset of mice. Within the SCN, the 3-NP treatment resulted in a reduction in daytime firing rate in the subset of mice which had a behavioral deficit. Anatomically, we confirmed that all of the treated mice showed evidence for cell loss within the striatum but we did not see evidence for gross SCN pathology. Together, the data demonstrates that chronic treatment with low doses of the mitochondrial toxin 3-NP produced circadian deficits in a subset of treated mice. This work does raise the possibility that the neural damage produced by mitochondrial dysfunction can contribute to the sleep/circadian dysfunction seen so commonly in neurodegenerative diseases.


Assuntos
Relógios Circadianos/fisiologia , Mitocôndrias/efeitos dos fármacos , Sono/fisiologia , Animais , Relógios Circadianos/efeitos dos fármacos , Convulsivantes/toxicidade , Técnicas de Introdução de Genes , Doença de Huntington/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Nitrocompostos/toxicidade , Técnicas de Patch-Clamp , Proteínas Circadianas Period/metabolismo , Propionatos/toxicidade , Sono/efeitos dos fármacos , Núcleo Supraquiasmático/efeitos dos fármacos , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/patologia
14.
PLoS One ; 8(7): e69993, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23936129

RESUMO

Sleep and circadian disruptions are commonly reported by patients with neurodegenerative diseases, suggesting these may be an endophenotype of the disorders. Several mouse models of Huntington's disease (HD) that recapitulate the disease progression and motor dysfunction of HD also exhibit sleep and circadian rhythm disruption. Of these, the strongest effects are observed in the transgenic models with multiple copies of mutant huntingtin gene. For developing treatments of the human disease, knock-in (KI) models offer advantages of genetic precision of the insertion and control of mutation copy number. Therefore, we assayed locomotor activity and immobility-defined sleep in a new model of HD with an expansion of the KI repeats (Q175). We found evidence for gene dose- and age-dependent circadian disruption in the behavior of the Q175 line. We did not see evidence for loss of cells or disruption of the molecular oscillator in the master pacemaker, the suprachiasmatic nucleus (SCN). The combination of the precise genetic targeting in the Q175 model and the observed sleep and circadian disruptions make it tractable to study the interaction of the underlying pathology of HD and the mechanisms by which the disruptions occur.


Assuntos
Ritmo Circadiano/genética , Dosagem de Genes , Doença de Huntington/genética , Doença de Huntington/fisiopatologia , Sono/genética , Fatores Etários , Alelos , Animais , Modelos Animais de Doenças , Genótipo , Humanos , Proteína Huntingtina , Doença de Huntington/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Atividade Motora/genética , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo
15.
Endocrinology ; 154(4): 1501-12, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23439698

RESUMO

Compelling reasons to study the role of sex in the circadian system include the higher rates of sleep disorders in women than in men and evidence that sex steroids modulate circadian control of locomotor activity. To address the issue of sex differences in the circadian system, we examined daily and circadian rhythms in wheel-running activity, electrical activity within the suprachiasmatic nucleus, and PER2::LUC-driven bioluminescence of gonadally-intact adult male and female C57BL/6J mice. We observed greater precision of activity onset in 12-hour light, 12-hour dark cycle for male mice, longer activity duration in 24 hours of constant darkness for female mice, and phase-delayed PER2::LUC bioluminescence rhythm in female pituitary and liver. Next, in order to investigate whether sex differences in behavior are sex chromosome or gonadal sex dependent, we used the 4 core genotypes (FCG) mouse model, in which sex chromosome complement is independent of gonadal phenotype. Gonadal males had more androgen receptor expression in the suprachiasmatic nucleus and behaviorally reduced photic phase shift response compared with gonadal female FCG mice. Removal of circulating gonadal hormones in adults, to test activational vs organizational effects of sex revealed that XX animals have longer activity duration than XY animals regardless of gonadal phenotype. Additionally, we observed that the activational effects of gonadal hormones were more important for regulating activity levels in gonadal male mice than in gonadal female FCG mice. Taken together, sex differences in the circadian rhythms of activity, neuronal physiology, and gene expression were subtle but provide important clues for understanding the pathophysiology of the circadian system.


Assuntos
Relógios Circadianos/fisiologia , Ritmo Circadiano/fisiologia , Hormônios Gonadais/fisiologia , Atividade Motora/fisiologia , Núcleo Supraquiasmático/fisiologia , Potenciais de Ação/genética , Potenciais de Ação/fisiologia , Glândulas Suprarrenais/metabolismo , Animais , Relógios Circadianos/genética , Ritmo Circadiano/genética , Feminino , Genótipo , Hormônios Gonadais/genética , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/genética , Miocárdio/metabolismo , Técnicas de Patch-Clamp , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Fenótipo , Hipófise/metabolismo , Receptores Androgênicos/metabolismo , Cromossomos Sexuais , Fatores Sexuais , Núcleo Supraquiasmático/metabolismo
16.
Exp Neurol ; 243: 57-66, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23353924

RESUMO

Sleep disorders are nearly ubiquitous among patients with Parkinson's disease (PD), and they manifest early in the disease process. While there are a number of possible mechanisms underlying these sleep disturbances, a primary dysfunction of the circadian system should be considered as a contributing factor. Our laboratory's behavioral phenotyping of a well-validated transgenic mouse model of PD reveals that the electrical activity of neurons within the master pacemaker of the circadian system, the suprachiasmatic nuclei (SCN), is already disrupted at the onset of motor symptoms, although the core features of the intrinsic molecular oscillations in the SCN remain functional. Our observations suggest that the fundamental circadian deficit in these mice lies in the signaling output from the SCN, which may be caused by known mechanisms in PD etiology: oxidative stress and mitochondrial disruption. Disruption of the circadian system is expected to have pervasive effects throughout the body and may itself lead to neurological and cardiovascular disorders. In fact, there is much overlap in the non-motor symptoms experienced by PD patients and in the consequences of circadian disruption. This raises the possibility that the sleep and circadian dysfunction experienced by PD patients may not merely be a subsidiary of the motor symptoms, but an integral part of the disease. Furthermore, we speculate that circadian dysfunction can even accelerate the pathology underlying PD. If these hypotheses are correct, more aggressive treatment of the circadian misalignment and sleep disruptions in PD patients early in the pathogenesis of the disease may be powerful positive modulators of disease progression and patient quality of life.


Assuntos
Ritmo Circadiano/fisiologia , Modelos Animais de Doenças , Doença de Parkinson/fisiopatologia , Animais , Humanos , Camundongos , Camundongos Transgênicos , Estresse Oxidativo/fisiologia , Doença de Parkinson/epidemiologia , Doença de Parkinson/genética , Transtornos do Sono-Vigília/epidemiologia , Transtornos do Sono-Vigília/genética , Transtornos do Sono-Vigília/fisiopatologia , Núcleo Supraquiasmático/fisiologia
17.
J Physiol ; 590(23): 6213-26, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22988135

RESUMO

The circadian system co-ordinates the temporal patterning of behaviour and many underlying biological processes. In some cases, the regulated outputs of the circadian system, such as activity, may be able to feed back to alter core clock processes. In our studies, we used four wheel-access conditions (no access; free access; early night; and late night) to manipulate the duration and timing of activity while under the influence of a light-dark cycle. In wild-type mice, scheduled wheel access was able to increase ambulatory activity, inducing a level of exercise driven at various phases of the light-dark cycle. Scheduled exercise also manipulated the magnitude and phasing of the circadian-regulated outputs of heart rate and body temperature. At a molecular level, the phasing and amplitude of PER2::LUCIFERASE (PER2::LUC) expression rhythms in the SCN and peripheral tissues of Per2::Luc knockin mice were altered by scheduled exercise. We then tested whether scheduled wheel access could improve deficits observed in vasointestinal polypeptide-deficient mice under the influence of a light-dark cycle. We found that scheduled wheel access during the late night improved many of the behavioural, physiological and molecular deficits previously described in vasointestinal polypeptide-deficient mice. Our results raise the possibility that scheduled exercise could be used as a tool to modulate daily rhythms and, when applied, may counteract some of the negative impacts of ageing and disease on the circadian system.


Assuntos
Ritmo Circadiano/fisiologia , Condicionamento Físico Animal/fisiologia , Peptídeo Intestinal Vasoativo/fisiologia , Animais , Comportamento Animal/fisiologia , Temperatura Corporal/fisiologia , Expressão Gênica , Frequência Cardíaca/fisiologia , Camundongos , Camundongos Transgênicos , Proteínas Circadianas Period/genética
18.
Minerva Pneumol ; 51(3): 93-106, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23687390

RESUMO

Sleep disorders are common in patients with neurogenerative diseases and manifest early in the disease process. Among a number of possible mechanisms underlying the sleep disturbances, there is evidence that dysfunction in the circadian system is a contributing factor. Focusing on a mouse model of Huntington's disease has enabled us to determine that at the onset of symptoms, spontaneous electrical activity of neurons within the central clock is disrupted even though the molecular clockwork is still functional. These findings suggest that the fundamental deficit contributing to disordered sleep is reduced SCN output. The mechanism underlying this deficit is not yet known, but mitochondrial dysfunction and oxidative stress are likely involved. Disruption of circadian output from the SCN would be expected to have wide ranging impact on the body including SCN regulated brain regions and the heart. In fact, there is a great deal of overlap in the non-motor symptoms experienced by HD patients and the consequences of circadian disruption. This raises the possibility that the disordered sleep and circadian function experienced by HD patients may be an integral part of the disease. Furthermore, we speculate that circadian dysfunction may accelerate the pathology underlying HD. If these hypotheses are correct, we should focus on treating circadian misalignment and sleep disruptions early in disease progression.

19.
PLoS Curr ; 3: RRN1266, 2011 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-22069044

RESUMO

Huntington's disease is a progressive, neurodegenerative disorder that presents with a triad of clinical symptoms, which include movement abnormalities, emotional disturbance and cognitive impairment. Recent studies reported dysfunction of the autonomic nervous system in Huntington's disease patients, which may contribute to the increased incidence of cardiovascular events in this patient population that often leads to death. We measured the baroreceptor reflex, a process dependent on proper autonomic function, in the BACHD mouse model of Huntington's disease. We found a blunted response of the baroreceptor reflex as well as significantly higher daytime blood pressure in BACHD mice compared to WT controls, which are both indications of autonomic dysfunction. BACHD mice had increased heart weight to tibia length ratios at 7 and 12 mo of age suggesting hypertrophic changes of the heart, which we speculate is a response to the increased blood pressure and aberrant baroreceptor reflex. Despite these structural changes, the hearts of BACHD mice continue to function normally as assessed by echocardiographic analysis. Studies of autonomic and cardiovascular function in BACHD mice may help elucidate the pathophysiology of Huntington's disease and aid in the development of clinical strategies to offset the incidence of fatal cardiovascular events in the Huntington's disease patient population.

20.
Exp Neurol ; 232(1): 66-75, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21864527

RESUMO

Many Parkinson's disease (PD) patients exhibit sleep disorders as part of their symptoms with evidence suggesting that REM sleep disorders may be intimately associated with this disease. Possible dysfunction in the circadian system in PD has received less attention, yet problems in circadian timing are common in neurodegenerative diseases. In the present study, we examined the expression of daily and circadian rhythms in the alpha-synuclein overexpressing (ASO) transgenic line. We found selective deficits in the expression of circadian rhythms of locomotor activity, including lower night-time activity and greater fragmentation in the wheel-running activity in this PD model. These alterations were prominent in young adult (3-4 mo) ASO mice and worsened progressively with age, consistent with prior reports of age-related loss of motor skills. The temporal distribution of sleep was also altered in the ASO mice compared to littermate controls. In the ASO mice, the peak/trough expression of the clock gene PERIOD2 was normal in the master pacemaker of the circadian system: the suprachiasmatic nucleus (SCN); however, the daytime firing rate of SCN neurons was reduced in the mutant mice. Together, this data raises the possibility that a weakening of circadian output is a core feature of PD. The reduction in magnitude of circadian output would be expected to have functional consequences throughout the body.


Assuntos
Ritmo Circadiano , Doença de Parkinson/fisiopatologia , Núcleo Supraquiasmático/fisiopatologia , Envelhecimento/metabolismo , Animais , Contagem de Células , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Transgênicos , Atividade Motora , Doença de Parkinson/metabolismo , Proteínas Circadianas Period/metabolismo , Núcleo Supraquiasmático/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...