Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Neurobiol Dis ; 176: 105944, 2023 01.
Article in English | MEDLINE | ID: mdl-36493974

ABSTRACT

Many patients with autism spectrum disorders (ASD) show disturbances in their sleep/wake cycles, and they may be particularly vulnerable to the impact of circadian disruptors. We have previously shown that a 2-weeks exposure to dim light at night (DLaN) disrupts diurnal rhythms, increases repetitive behaviors and reduces social interactions in contactin-associated protein-like 2 knock out (Cntnap2 KO) mice. The deleterious effects of DLaN may be mediated by intrinsically photosensitive retinal ganglion cells (ipRGCs) expressing the photopigment melanopsin, which is maximally sensitive to blue light (480 nm). In this study, the usage of a light-emitting diode array enabled us to shift the spectral properties of the DLaN while keeping the intensity of the illumination at 10 lx. First, we confirmed that the short-wavelength enriched lighting produced strong acute suppression of locomotor activity (masking), robust light-induced phase shifts, and cFos expression in the suprachiasmatic nucleus in wild-type (WT) mice, while the long-wavelength enriched lighting evoked much weaker responses. Opn4DTA mice, lacking the melanopsin expressing ipRGCs, were resistant to DLaN effects. Importantly, shifting the DLaN stimulus to longer wavelengths mitigated the negative impact on the activity rhythms and 'autistic' behaviors (i.e. reciprocal social interactions, repetitive grooming) in the Cntnap2 KO as well as in WT mice. The short-, but not the long-wavelength enriched, DLaN triggered cFos expression in in the basolateral amygdala (BLA) as well as in the peri-habenula region raising that possibility that these cell populations may mediate the effects. Broadly, our findings are consistent with the recommendation that spectral properties of light at night should be considered to optimize health in neurotypical as well as vulnerable populations.


Subject(s)
Circadian Rhythm , Retinal Ganglion Cells , Mice , Animals , Circadian Rhythm/physiology , Retinal Ganglion Cells/metabolism , Suprachiasmatic Nucleus , Light , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism
2.
Front Nutr ; 9: 1034743, 2022.
Article in English | MEDLINE | ID: mdl-36407529

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 ß-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.

3.
Neurobiol Sleep Circadian Rhythms ; 11: 100070, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34307964

ABSTRACT

The dorsal striatum forms part of the basal ganglia circuit that is a major regulator of voluntary motor behavior. Dysfunction in this circuit is a critical factor in the pathology of neurological (Parkinson's and Huntington's disease) as well as psychiatric disorders. In this study, we employed in vivo real-time monitoring of multiple unit neural activity (MUA) in the dorsal striatum of freely moving mice. We demonstrate that the striatum exhibits robust diurnal and circadian rhythms in MUA that peak in the night. These rhythms are dependent upon the central circadian clock located in the suprachiasmatic nucleus (SCN) as lesions of this structure caused the loss of rhythmicity measured in the striatum. Nonetheless, chronic treatment of methamphetamine (METH) makes circadian rhythms appear in MUA recorded from the striatum of SCN-lesioned mice. These data demonstrate that the physiological properties of neurons in the dorsal striatum are regulated by the circadian system and that METH drives circadian rhythms in striatal physiology in the absence of the SCN. The finding of SCN-driven circadian rhythms in striatal physiology has important implications for an understanding of the temporal regulation of motor control as well as revealing how disease processes may disrupt this regulation.

5.
Neurobiol Dis ; 145: 105064, 2020 11.
Article in English | MEDLINE | ID: mdl-32889171

ABSTRACT

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.


Subject(s)
Autism Spectrum Disorder , Central Nervous System Depressants/pharmacology , Circadian Rhythm/drug effects , Lighting/adverse effects , Melatonin/pharmacology , Animals , Autism Spectrum Disorder/genetics , Behavior, Animal/drug effects , Disease Models, Animal , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics
6.
J Biol Rhythms ; 33(5): 535-554, 2018 10.
Article in English | MEDLINE | ID: mdl-30084274

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.


Subject(s)
Circadian Rhythm , Fasting , Huntington Disease/therapy , Animals , Cohort Studies , Disease Models, Animal , Heart Rate , Male , Mice , Mice, Transgenic , Motor Activity , Photoperiod , Quality of Life
7.
Diabetes ; 67(3): 486-495, 2018 03.
Article in English | MEDLINE | ID: mdl-29212780

ABSTRACT

Diabetes mellitus (DM) is associated with increased plasma levels of arginine-vasopressin (AVP), which may aggravate hyperglycemia and nephropathy. However, the mechanisms by which DM may cause the increased AVP levels are not known. Electrophysiological recordings in supraoptic nucleus (SON) slices from streptozotocin (STZ)-induced DM rats and vehicle-treated control rats revealed that γ-aminobutyric acid (GABA) functions generally as an excitatory neurotransmitter in the AVP neurons of STZ rats, whereas it usually evokes inhibitory responses in the cells of control animals. Furthermore, Western blotting analyses of Cl- transporters in the SON tissues indicated that Na+-K+-2Cl- cotransporter isotype 1 (a Cl- importer) was upregulated and K+-Cl- cotransporter isotype 2 (KCC2; a Cl- extruder) was downregulated in STZ rats. Treatment with CLP290 (a KCC2 activator) significantly lowered blood AVP and glucose levels in STZ rats. Last, investigation that used rats expressing an AVP-enhanced green fluorescent protein fusion gene showed that AVP synthesis in AVP neurons was much more intense in STZ rats than in control rats. We conclude that altered Cl- homeostasis that makes GABA excitatory and enhanced AVP synthesis are important changes in AVP neurons that would increase AVP secretion in DM. Our data suggest that Cl- transporters in AVP neurons are potential targets of antidiabetes treatments.


Subject(s)
Arginine Vasopressin/metabolism , Diabetes Mellitus, Experimental/metabolism , GABAergic Neurons/metabolism , Hypothalamus/metabolism , Neurosecretory Systems/metabolism , Supraoptic Nucleus/metabolism , Animals , Arginine Vasopressin/blood , Arginine Vasopressin/chemistry , Arginine Vasopressin/genetics , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Electrophysiological Phenomena/drug effects , GABAergic Neurons/drug effects , GABAergic Neurons/pathology , Hypoglycemic Agents/therapeutic use , Hypothalamus/drug effects , Hypothalamus/pathology , Hypothalamus/physiopathology , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Membrane Transport Modulators/therapeutic use , Microscopy, Fluorescence , Neurosecretory Systems/drug effects , Neurosecretory Systems/pathology , Neurosecretory Systems/physiopathology , Oxytocin/chemistry , Oxytocin/genetics , Oxytocin/metabolism , Prodrugs/therapeutic use , Rats, Sprague-Dawley , Rats, Transgenic , Rats, Wistar , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Streptozocin , Supraoptic Nucleus/drug effects , Supraoptic Nucleus/pathology , Supraoptic Nucleus/physiopathology , Symporters/agonists , Symporters/metabolism , Synaptic Transmission/drug effects , K Cl- Cotransporters
8.
ASN Neuro ; 7(6)2015.
Article in English | MEDLINE | ID: mdl-26553726

ABSTRACT

The neural activity patterns of suprachiasmatic nucleus (SCN) neurons are dynamically regulated throughout the circadian cycle with highest levels of spontaneous action potentials during the day. These rhythms in electrical activity are critical for the function of the circadian timing system and yet the mechanisms by which the molecular clockwork drives changes in the membrane are not well understood. In this study, we sought to examine how the clock gene Period1 (Per1) regulates the electrical activity in the mouse SCN by transiently and selectively decreasing levels of PER1 through use of an antisense oligodeoxynucleotide. We found that this treatment effectively reduced SCN neural activity. Direct current injection to restore the normal membrane potential partially, but not completely, returned firing rate to normal levels. The antisense treatment also reduced baseline [Ca(2+)]i levels as measured by Fura2 imaging technique. Whole cell patch clamp recording techniques were used to examine which specific potassium currents were altered by the treatment. These recordings revealed that the large conductance [Ca(2+)]i-activated potassium currents were reduced in antisense-treated neurons and that blocking this current mimicked the effects of the anti-sense on SCN firing rate. These results indicate that the circadian clock gene Per1 alters firing rate in SCN neurons and raise the possibility that the large conductance [Ca(2+)]i-activated channel is one of the targets.


Subject(s)
Circadian Clocks/physiology , Neurons/physiology , Period Circadian Proteins/metabolism , Suprachiasmatic Nucleus/physiology , Animals , Calcium/metabolism , Calcium Signaling/physiology , Cations, Divalent/metabolism , Circadian Clocks/drug effects , Electric Stimulation , Gene Knock-In Techniques , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Period Circadian Proteins/genetics , Suprachiasmatic Nucleus/drug effects , Tissue Culture Techniques
9.
eNeuro ; 2(4)2015.
Article in English | MEDLINE | ID: mdl-26464996

ABSTRACT

Circadian timing systems, like most physiological processes, cannot escape the effects of aging. With age, humans experience decreased duration and quality of sleep. Aged mice exhibit decreased amplitude and increased fragmentation of the activity rhythm, and lengthened circadian free-running period in both light-dark (LD) and constant dark (DD) conditions. Several studies have shown that aging impacts neural activity rhythms in the central circadian clock in the suprachiasmatic nucleus (SCN). However, evidence for age-related disruption of circadian oscillations of clock genes in the SCN has been equivocal. We hypothesized that daily exposure to LD cycles masks the full impact of aging on molecular rhythms in the SCN. We performed ex vivo bioluminescent imaging of cultured SCN slices of young and aged PER2::luciferase knock-in (PER2::LUC) mice housed under LD or prolonged DD conditions. Under LD conditions, the amplitude of PER2::LUC rhythms differed only slightly between SCN explants from young and aged animals; under DD conditions, the PER2::LUC rhythms of aged animals showed markedly lower amplitudes and longer circadian periods than those of young animals. Recordings of PER2::LUC rhythms in individual SCN cells using an electron multiplying charge-coupled device camera revealed that aged SCN cells showed longer circadian periods and that the rhythms of individual cells rapidly became desynchronized. These data suggest that aging degrades the SCN circadian ensemble, but that recurrent LD cycles mask these effects. We propose that these changes reflect a decline in pacemaker robustness that could increase vulnerability to environmental challenges, and partly explain age-related sleep and circadian disturbances.

10.
Cell Rep ; 12(9): 1407-13, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26299967

ABSTRACT

Female reproductive function changes during aging with the estrous cycle becoming more irregular during the transition to menopause. We found that intermittent shifts of the light-dark cycle disrupted regularity of estrous cycles in middle-aged female mice, whose estrous cycles were regular under unperturbed 24-hr light-dark cycles. Although female mice deficient in Cry1 or Cry2, the core components of the molecular circadian clock, exhibited regular estrous cycles during youth, they showed accelerated senescence characterized by irregular and unstable estrous cycles and resultant infertility in middle age. Notably, tuning the period length of the environmental light-dark cycles closely to the endogenous one inherent in the Cry-deficient females restored the regularity of the estrous cycles and, consequently, improved fertility in middle age. These results suggest that reproductive potential can be strongly influenced by age-related changes in the circadian system and normal reproductive functioning can be rescued by the manipulation of environmental timing signals.


Subject(s)
Aging/physiology , Circadian Rhythm , Estrous Cycle/physiology , Fertility/physiology , Photoperiod , Animals , Cryptochromes/genetics , Estrous Cycle/genetics , Female , Fertility/genetics , Mice , Mice, Inbred C57BL
11.
J Neurophysiol ; 110(5): 1097-106, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23741043

ABSTRACT

The neuropeptide vasoactive intestinal peptide (VIP) is expressed at high levels in the neurons of the suprachiasmatic nucleus (SCN). While VIP is known to be important to the input and output pathways from the SCN, the physiological effects of VIP on electrical activity of SCN neurons are not well known. Here the impact of VIP on firing rate of SCN neurons was investigated in mouse slice cultures recorded during the night. The application of VIP produced an increase in electrical activity in SCN slices that lasted several hours after treatment. This is a novel mechanism by which this peptide can produce long-term changes in central nervous system physiology. The increase in action potential frequency was blocked by a VIP receptor antagonist and lost in a VIP receptor knockout mouse. In addition, inhibitors of both the Epac family of cAMP binding proteins and cAMP-dependent protein kinase (PKA) blocked the induction by VIP. The persistent increase in spike rate following VIP application was not seen in SCN neurons from mice deficient in Kv3 channel proteins and was dependent on the clock protein PER1. These findings suggest that VIP regulates the long-term firing rate of SCN neurons through a VIPR2-mediated increase in the cAMP pathway and implicate the fast delayed rectifier (FDR) potassium currents as one of the targets of this regulation.


Subject(s)
Neurons/physiology , Suprachiasmatic Nucleus/physiology , Vasoactive Intestinal Peptide/pharmacology , Animals , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/physiology , Guanine Nucleotide Exchange Factors/physiology , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Period Circadian Proteins/metabolism , Shaw Potassium Channels/genetics , Shaw Potassium Channels/physiology , Signal Transduction , Suprachiasmatic Nucleus/drug effects
12.
Diabetes ; 62(10): 3469-78, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23775768

ABSTRACT

There is a correlation between circadian disruption, type 2 diabetes mellitus (T2DM), and islet failure. However, the mechanisms underlying this association are largely unknown. Pancreatic islets express self-sustained circadian clocks essential for proper ß-cell function and survival. We hypothesized that exposure to environmental conditions associated with disruption of circadian rhythms and susceptibility to T2DM in humans disrupts islet clock and ß-cell function. To address this hypothesis, we validated the use of Per-1:LUC transgenic rats for continuous longitudinal assessment of islet circadian clock function ex vivo. Using this methodology, we subsequently examined effects of the continuous exposure to light at night (LL) on islet circadian clock and insulin secretion in vitro in rat islets. Our data show that changes in the light-dark cycle in vivo entrain the phase of islet clock transcriptional oscillations, whereas prolonged exposure (10 weeks) to LL disrupts islet circadian clock function through impairment in the amplitude, phase, and interislet synchrony of clock transcriptional oscillations. We also report that exposure to LL leads to diminished glucose-stimulated insulin secretion due to a decrease in insulin secretory pulse mass. Our studies identify potential mechanisms by which disturbances in circadian rhythms common to modern life can predispose to islet failure in T2DM.


Subject(s)
CLOCK Proteins/metabolism , Circadian Clocks , Circadian Rhythm , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Light , Period Circadian Proteins/metabolism , Animals , Blood Glucose/metabolism , Body Weight , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Disease Susceptibility , Immunohistochemistry , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Motor Activity , Rats , Rats, Transgenic
13.
J Neurosci ; 32(46): 16193-202, 2012 Nov 14.
Article in English | MEDLINE | ID: mdl-23152603

ABSTRACT

Aging produces a decline in the amplitude and precision of 24 h behavioral, endocrine, and metabolic rhythms, which are regulated in mammals by a central circadian pacemaker within the suprachiasmatic nucleus (SCN) and local oscillators in peripheral tissues. Disruption of the circadian system, as experienced during transmeridian travel, can lead to adverse health consequences, particularly in the elderly. To test the hypothesis that age-related changes in the response to simulated jet lag will reflect altered circadian function, we examined re-entrainment of central and peripheral oscillators from young and old PER2::luciferase mice. As in previous studies, locomotor activity rhythms in older mice required more days to re-entrain following a shift than younger mice. At the tissue level, effects of age on baseline entrainment were evident, with older mice displaying earlier phases for the majority of peripheral oscillators studied and later phases for cells within most SCN subregions. Following a 6 h advance of the light:dark cycle, old mice displayed slower rates of re-entrainment for peripheral tissues but a larger, more rapid SCN response compared to younger mice. Thus, aging alters the circadian timing system in a manner that differentially affects the re-entrainment responses of central and peripheral circadian clocks. This pattern of results suggests that a major consequence of aging is a decrease in pacemaker amplitude, which would slow re-entrainment of peripheral oscillators and reduce SCN resistance to external perturbation.


Subject(s)
Aging/physiology , Central Nervous System/physiology , Circadian Rhythm/physiology , Peripheral Nervous System/physiology , Animals , Behavior, Animal/physiology , Biological Clocks/physiology , Brain/physiology , Data Interpretation, Statistical , Image Processing, Computer-Assisted , Jet Lag Syndrome/physiopathology , Luciferases/genetics , Luciferases/physiology , Luminescence , Male , Mice , Mice, Neurologic Mutants , Motor Activity/physiology , Period Circadian Proteins/genetics , Period Circadian Proteins/physiology , Suprachiasmatic Nucleus/physiology , Tissue Culture Techniques
14.
J Biol Rhythms ; 26(5): 423-33, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21921296

ABSTRACT

Type 2 diabetes mellitus (T2DM) is complex metabolic disease that arises as a consequence of interactions between genetic predisposition and environmental triggers. One recently described environmental trigger associated with development of T2DM is disturbance of circadian rhythms due to shift work, sleep loss, or nocturnal lifestyle. However, the underlying mechanisms behind this association are largely unknown. To address this, the authors examined the metabolic and physiological consequences of experimentally controlled circadian rhythm disruption in wild-type (WT) Sprague Dawley and diabetes-prone human islet amyloid polypeptide transgenic (HIP) rats: a validated model of T2DM. WT and HIP rats at 3 months of age were exposed to 10 weeks of either a normal light regimen (LD: 12:12-h light/dark) or experimental disruption in the light-dark cycle produced by either (1) 6-h advance of the light cycle every 3 days or (2) constant light protocol. Subsequently, blood glucose control, beta-cell function, beta-cell mass, turnover, and insulin sensitivity were examined. In WT rats, 10 weeks of experimental disruption of circadian rhythms failed to significantly alter fasting blood glucose levels, glucose-stimulated insulin secretion, beta-cell mass/turnover, or insulin sensitivity. In contrast, experimental disruption of circadian rhythms in diabetes-prone HIP rats led to accelerated development of diabetes. The mechanism subserving early-onset diabetes was due to accelerated loss of beta-cell function and loss of beta-cell mass attributed to increases in beta-cell apoptosis. Disruption of circadian rhythms may increase the risk of T2DM by accelerating the loss of beta-cell function and mass characteristic in T2DM.


Subject(s)
Circadian Rhythm/physiology , Diabetes Mellitus, Type 2/physiopathology , Insulin-Secreting Cells/physiology , Animals , Blood Glucose/metabolism , Humans , Insulin/metabolism , Insulin Resistance , Insulin Secretion , Insulin-Secreting Cells/cytology , Islet Amyloid Polypeptide/physiology , Male , Motor Activity , Photoperiod , Rats , Rats, Sprague-Dawley , Rats, Transgenic
15.
J Neurosci ; 31(28): 10201-5, 2011 Jul 13.
Article in English | MEDLINE | ID: mdl-21752996

ABSTRACT

Disruptions in sleep/wake cycles, including decreased amplitude of rhythmic behaviors and fragmentation of the sleep episodes, are commonly associated with aging in humans and other mammals. While there are undoubtedly many factors contributing to these changes, a body of literature is emerging, suggesting that an age-related decline in the central circadian clock in the suprachiasmatic nucleus (SCN) may be a key element responsible. To explore age-related changes in the SCN, we have performed in vivo multiunit neural activity (MUA) recordings from the SCN of freely moving young (3-5 months) and middle-aged (13-18 months) mice. Importantly, the amplitude of day-night difference in MUA was significantly reduced in the older mice. We also found that the neural activity rhythms are clearly degraded in the subparaventricular zone, one of the main neural outputs of the SCN. Surprisingly, parallel studies indicate that the molecular clockwork in the SCN as measured by PER2 exhibited only minor deficits at this same age. Thus, the circadian output measured at the level of neural activity rhythms in the SCN is degraded by aging, and this decline occurs before the disruption of key components of the molecular clockwork.


Subject(s)
Aging/physiology , Biological Clocks/physiology , Circadian Rhythm/physiology , Animals , Electrodes, Implanted , Electrophysiology , Male , Mice , Motor Activity/physiology , Period Circadian Proteins/metabolism , Suprachiasmatic Nucleus/physiology
16.
J Biol Rhythms ; 26(3): 200-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21628547

ABSTRACT

The neuropeptide vasoactive intestinal polypeptide (VIP) has emerged as a key candidate molecule mediating the synchronization of rhythms in clock gene expression within the suprachiasmatic nucleus (SCN). In addition, neurons expressing VIP are anatomically well positioned to mediate communication between the SCN and peripheral oscillators. In this study, we examined the temporal expression profile of 3 key circadian genes: Per1, Per2 , and Bmal1 in the SCN, the adrenal glands and the liver of mice deficient for the Vip gene (VIP KO), and their wild-type counterparts. We performed these measurements in mice held in a light/dark cycle as well as in constant darkness and found that rhythms in gene expression were greatly attenuated in the VIP-deficient SCN. In the periphery, the impact of the loss of VIP varied with the tissue and gene measured. In the adrenals, rhythms in Per1 were lost in VIP-deficient mice, while in the liver, the most dramatic impact was on the phase of the diurnal expression rhythms. Finally, we examined the effects of the loss of VIP on ex vivo explants of the same central and peripheral oscillators using the PER2::LUC reporter system. The VIP-deficient mice exhibited low amplitude rhythms in the SCN as well as altered phase relationships between the SCN and the peripheral oscillators. Together, these data suggest that VIP is critical for robust rhythms in clock gene expression in the SCN and some peripheral organs and that the absence of this peptide alters both the amplitude of circadian rhythms as well as the phase relationships between the rhythms in the SCN and periphery.


Subject(s)
ARNTL Transcription Factors/metabolism , Circadian Rhythm , Gene Expression Regulation , Period Circadian Proteins/metabolism , Suprachiasmatic Nucleus/metabolism , Vasoactive Intestinal Peptide/physiology , Adrenal Glands/metabolism , Animals , Genes, Reporter , In Situ Hybridization , Liver/metabolism , Luciferases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Photoperiod
17.
J Biol Rhythms ; 25(2): 92-102, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20348460

ABSTRACT

African sleeping sickness is characterized by alterations in rhythmic functions. It is not known if the disease affects the expression of clock genes, which are the molecular basis for rhythm generation. We used a chronic rat model of experimental sleeping sickness, caused by the extracellular parasite Trypanosoma brucei brucei (Tb brucei), to study the effects on clock gene expression. In tissue explants of pituitary glands from Period1-luciferase (Per1-luc) transgenic rats infected with Tb brucei, the period of Per1-luc expression was significantly shorter. In explants containing the suprachiasmatic nuclei (SCN), the Per1-luc rhythms were flat in 21% of the tissues. We also examined the relative expression of Per1, Clock, and Bmal1 mRNA in the SCN, pineal gland, and spleen from control and infected rats using qPCR. Both Clock and Bmal1 mRNA expression was reduced in the pineal gland and spleen following Tb brucei infection. Infected rats were periodic both in core body temperature and in locomotor activity; however, early after infection, we observed a significant decline in the amplitude of the locomotor activity rhythm. In addition, both activity and body temperature rhythms exhibited decreased regularity and "robustness." In conclusion, although experimental trypanosome infection has previously been shown to cause functional disturbances in SCN neurons, only 21% of the SCN explants had disturbed Per1-luc rhythms. However, our data show that the infection overall alters molecular clock function in peripheral clocks including the pituitary gland, pineal gland, and spleen.


Subject(s)
Gene Expression Regulation , Inflammation , Period Circadian Proteins/physiology , Trypanosoma brucei brucei/metabolism , Trypanosomiasis, African/parasitology , Animals , Animals, Genetically Modified , Biological Clocks , Body Temperature , Male , Neurons/metabolism , Period Circadian Proteins/genetics , Pineal Gland/metabolism , Pituitary Gland/metabolism , Rats , Rats, Wistar , Spleen/metabolism , Suprachiasmatic Nucleus/metabolism
18.
Steroids ; 75(3): 203-12, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20096720

ABSTRACT

Circadian rhythms in physiology and behavior are known to be influenced by the estrous cycle in female rodents. The clock genes responsible for the generation of circadian oscillations are widely expressed both within the central nervous system and peripheral tissues, including those that comprise the reproductive system. To address whether the estrous cycle affects rhythms of clock gene expression in peripheral tissues, we first examined rhythms of clock gene expression (Per1, Per2, Bmal1) in reproductive (uterus, ovary) and non-reproductive (liver) tissues of cycling rats using quantitative real-time PCR (in vivo) and luminescent recording methods to measure circadian rhythms of PER2 expression in tissue explant cultures from cycling PER2::LUCIFERASE (PER2::LUC) knockin mice (ex vivo). We found significant estrous variations of clock gene expression in all three tissues in vivo, and in the uterus ex vivo. We also found that exogenous application of estrogen and progesterone altered rhythms of PER2::LUC expression in the uterus. In addition, we measured the effects of ovarian steroids on clock gene expression in a human breast cancer cell line (MCF-7 cells) as a model for endocrine cells that contain both the steroid hormone receptors and clock genes. We found that progesterone, but not estrogen, acutely up-regulated Per1, Per2, and Bmal1 expression in MCF-7 cells. Together, our findings demonstrate that the timing of the circadian clock in reproductive tissues is influenced by the estrous cycle and suggest that fluctuating steroid hormone levels may be responsible, in part, through direct effects on the timing of clock gene expression.


Subject(s)
Biological Clocks/genetics , Circadian Rhythm/physiology , Estrogens/pharmacology , Estrous Cycle/physiology , Gene Expression Regulation/drug effects , Ovary/metabolism , Progesterone/pharmacology , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , Cell Line, Tumor , Estrogens/metabolism , Female , Humans , Mice , Mice, Transgenic , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Progesterone/metabolism , Rats , Rats, Wistar
19.
J Biol Rhythms ; 24(1): 55-63, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19150929

ABSTRACT

The mammalian circadian system is orchestrated by a master pacemaker in the brain, but many peripheral tissues also contain independent or quasi-independent circadian oscillators. The adaptive significance of clocks in these structures must lie, in large part, in the phase relationships between the constituent oscillators and their micro- and macroenvironments. To examine the relationship between postnatal development, which is dependent on endogenous programs and maternal/environmental influences, and the phase of circadian oscillators, the authors assessed the circadian phase of pineal, liver, lung, adrenal, and thyroid tissues cultured from Period 1-luciferase (Per1-luc ) rat pups of various postnatal ages. The liver, thyroid, and pineal were rhythmic at birth, but the phases of their Per1-luc expression rhythms shifted remarkably during development. To determine if the timing of the phase shift in each tissue could be the result of changing environmental conditions, the behavior of pups and their mothers was monitored. The circadian phase of the liver shifted from the day to night around postnatal day (P) 22 as the pups nursed less during the light and instead ate solid food during the dark. Furthermore, the phase of Per1-luc expression in liver cultures from nursing neonates could be shifted experimentally from the day to the night by allowing pups access to the dam only during the dark. Peak Per1-luc expression also shifted from midday to early night in thyroid cultures at about P20, concurrent with the shift in eating times. The phase of Per1-luc expression in the pineal gland shifted from day to night coincident with its sympathetic innervation at around P5. Per1-luc expression was rhythmic in adrenal cultures and peaked around the time of lights-off throughout development; however, the amplitude of the rhythm increased at P25. Lung cultures were completely arrhythmic until P12 when the pups began to leave the nest. Taken together, the data suggest that the molecular machinery that generates circadian oscillations matures at different rates in different tissues and that the phase of at least some peripheral organs is malleable and may shift as the organ's function changes during development.


Subject(s)
Brain/metabolism , Circadian Rhythm , Intracellular Signaling Peptides and Proteins/physiology , Animals , Animals, Newborn , Biological Clocks , Female , Homozygote , Intracellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Male , Models, Biological , Oscillometry , Period Circadian Proteins , Rats , Suprachiasmatic Nucleus/metabolism , Time Factors
20.
Am J Physiol Endocrinol Metab ; 295(5): E1025-31, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18728223

ABSTRACT

Fluctuations in circulating estrogen and progesterone levels associated with the estrous cycle alter circadian rhythms of physiology and behavior in female rodents. Endogenously applied estrogen shortens the period of the locomotor activity rhythm in rodents. We recently found that estrogen implants affect Period (Per) gene expression in the suprachiasmatic nucleus (SCN; central clock) and uterus of rats in vivo. To explore whether estrogen directly influences the circadian clock in the SCN and/or tissues of the reproductive system, we examined the effects of 17beta-estradiol (E(2)) on PER2::LUCIFERASE (PER2::LUC) expression in tissue explant cultures from ovariectomized PER2::LUC knockin mice. E(2) applied to explanted cultures shortened the period of rhythmic PER2::LUC expression in the uterus but did not change the period of PER2::LUC expression in the SCN. Raloxifene, a selective estrogen receptor modulator and known E(2) antagonist in uterine tissues, attenuated the effect of E(2) on the period of the PER2::LUC rhythm in the uterus. These data indicate that estrogen directly affects the timing of the molecular clock in the uterus via an estrogen receptor-mediated response.


Subject(s)
Cell Cycle Proteins/metabolism , Circadian Rhythm/physiology , Estrogens/pharmacology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Uterus/drug effects , Animals , Cell Cycle Proteins/genetics , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Female , Gene Expression Regulation/drug effects , In Vitro Techniques , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Ovariectomy , Period Circadian Proteins , Progesterone/pharmacology , Raloxifene Hydrochloride/pharmacology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Suprachiasmatic Nucleus/drug effects , Suprachiasmatic Nucleus/metabolism , Transcription Factors/genetics , Uterus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...