Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Front Neurosci ; 13: 1281, 2019.
Article in English | MEDLINE | ID: mdl-31866806

ABSTRACT

Results from a variety of sources indicate a role for pituitary adenylate cyclase-activating polypeptide (PACAP) in light/glutamate-induced phase resetting of the circadian clock mediated by the retinohypothalamic tract (RHT). Attempts to block or remove PACAP's contribution to clock-resetting have generated phenotypes that differ in their responses to light or glutamate. For example, previous studies of circadian behaviors found that period-maintenance and early-night phase delays are intact in PACAP-null mice, yet there is a consistent deficit in behavioral phase-resetting to light stimulation in the late night. Here we report rodent stimulus-response characteristics of PACAP release from the RHT, and map these to responses of the suprachiasmatic nucleus (SCN) in intact and PACAP-deficient mouse hypothalamus with regard to phase-resetting. SCN of PACAP-null mice exhibit normal circadian rhythms in neuronal activity, but are "blind" to glutamate stimulating phase-advance responses in late night, although not in early night, consistent with previously reported selective lack of late-night light behavioral responsiveness of these mice. Induction of CREB phosphorylation, a hallmark of the light/glutamate response of the SCN, also is absent in SCN-containing ex vivo slices from PACAP-deficient mouse hypothalamus. PACAP replacement to the SCN of PACAP-null mice restored wild-type phase-shifting of firing-rate patterns in response to glutamate applied to the SCN in late night. Likewise, ex vivo SCN of wild-type mice post-orbital enucleation are unresponsive to glutamate unless PACAP also is restored. Furthermore, we demonstrate that the period of efficacy of PACAP at SCN nerve terminals corresponds to waxing of PACAP mRNA expression in ipRGCs during the night, and waning during the day. These results validate the use of PACAP-deficient mice in defining the role and specificity of PACAP as a co-transmitter with glutamate in ipRGC-RHT projections to SCN in phase advancing the SCN circadian rhythm in late night.

2.
PLoS One ; 9(8): e103103, 2014.
Article in English | MEDLINE | ID: mdl-25084275

ABSTRACT

Circadian rhythms of behavior and physiology are driven by the biological clock that operates endogenously but can also be entrained to the light-dark cycle of the environment. In mammals, the master circadian pacemaker is located in the suprachiasmatic nucleus (SCN), which is composed of individual cellular oscillators that are driven by a set of core clock genes interacting in transcriptional/translational feedback loops. Light signals can trigger molecular events in the SCN that ultimately impact on the phase of expression of core clock genes to reset the master pacemaker. While transcriptional regulation has received much attention in the field of circadian biology in the past, other mechanisms including targeted protein degradation likely contribute to the clock timing and entrainment process. In the present study, proteome-wide screens of the murine SCN led to the identification of ubiquitin protein ligase E3 component N-recognin 4 (UBR4), a novel E3 ubiquitin ligase component of the N-end rule pathway, as a time-of-day-dependent and light-inducible protein. The spatial and temporal expression pattern of UBR4 in the SCN was subsequently characterized by immunofluorescence microscopy. UBR4 is expressed across the entire rostrocaudal extent of the SCN in a time-of-day-dependent fashion. UBR4 is localized exclusively to arginine vasopressin (AVP)-expressing neurons of the SCN shell. Upon photic stimulation in the early subjective night, the number of UBR4-expressing cells within the SCN increases. This study is the first to identify a novel E3 ubiquitin ligase component, UBR4, in the murine SCN and to implicate the N-end rule degradation pathway as a potential player in regulating core clock mechanisms and photic entrainment.


Subject(s)
Circadian Clocks/genetics , Gene Expression Regulation , Light , Microtubule-Associated Proteins/genetics , Suprachiasmatic Nucleus/metabolism , Ubiquitin-Protein Ligases/genetics , Animals , Calmodulin-Binding Proteins , Cell Line , Circadian Rhythm/genetics , Male , Mice , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , Peptides/genetics , Peptides/metabolism , Photoperiod , Protein Binding , Protein Transport , Proteome , Proteomics/methods , Reproducibility of Results , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
3.
Essays Biochem ; 49(1): 103-17, 2011 Jun 30.
Article in English | MEDLINE | ID: mdl-21819387

ABSTRACT

In vitro assays have localized circadian pacemakers to individual cells, revealed genetic determinants of rhythm generation, identified molecular players in cell-cell synchronization and determined physiological events regulated by circadian clocks. Although they allow strict control of experimental conditions and reduce the number of variables compared with in vivo studies, they also lack many of the conditions in which cellular circadian oscillators normally function. The present review highlights methods to study circadian timing in cultured mammalian cells and how they have shaped the hypothesis that all cells are capable of circadian rhythmicity.


Subject(s)
Astrocytes/physiology , Circadian Rhythm/physiology , Molecular Imaging/methods , Neurons/physiology , Suprachiasmatic Nucleus/physiology , Action Potentials/physiology , Animals , Cell Line, Tumor , Cells, Cultured , Circadian Clocks/physiology , Electrophysiological Phenomena , Humans , Neurons/metabolism
4.
J Neurosci ; 31(23): 8342-50, 2011 Jun 08.
Article in English | MEDLINE | ID: mdl-21653839

ABSTRACT

Circadian clocks sustain daily oscillations in gene expression, physiology, and behavior, relying on transcription-translation feedback loops of clock genes for rhythm generation. Cultured astrocytes display daily oscillations of extracellular ATP, suggesting that ATP release is a circadian output. We hypothesized that the circadian clock modulates ATP release via mechanisms that regulate acute ATP release from glia. To test the molecular basis for circadian ATP release, we developed methods to measure in real-time ATP release and Bmal1::dLuc circadian reporter expression in cortical astrocyte cultures from mice of different genotypes. Daily rhythms of gene expression required functional Clock and Bmal1, both Per1 and Per2, and both Cry1 and Cry2 genes. Similarly, high-level, circadian ATP release also required a functional clock mechanism. Whereas blocking IP(3) signaling significantly disrupted ATP rhythms with no effect on Bmal1::dLuc cycling, blocking vesicular release did not alter circadian ATP release or gene expression. We conclude that astrocytes depend on circadian clock genes and IP(3) signaling to express daily rhythms in ATP release.


Subject(s)
Adenosine Triphosphate/metabolism , Astrocytes/metabolism , Cerebral Cortex/metabolism , Circadian Rhythm/physiology , Analysis of Variance , Animals , Astrocytes/cytology , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cryptochromes/genetics , Cryptochromes/metabolism , Immunohistochemistry , Mice , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
J Biol Rhythms ; 26(6): 561-4, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22215614

ABSTRACT

Recent demonstration of the histone acetyltransferase activity of the Clock gene greatly expanded the regulatory role of circadian clocks in gene transcription. Clock and its partner Bmal1 are responsible for the generation of circadian oscillations that are synchronized (entrained) to the external light cycle. Entraining light often produces long-lasting changes in the endogenous period called aftereffects. Aftereffects are light-dependent alterations in the speed of free-running rhythms that persist for several weeks upon termination of light exposure. How light causes such long-lasting changes is unknown. However, the persistent nature of circadian aftereffects in conjunction with the long-term effects of epigenetic modifications on development and various aspects of brain physiology prompted us to hypothesize that the histone acetyltransferase CLOCK was required for circadian aftereffects. The authors exposed Clock knockout mice to 25-hour light cycles and report that these mice retain the ability to display circadian aftereffects, indicating that Clock is dispensable for this form of circadian plasticity.


Subject(s)
Acetyltransferases/metabolism , Biological Clocks/physiology , CLOCK Proteins/metabolism , Circadian Rhythm/physiology , Acetyltransferases/genetics , Animals , Behavior, Animal/physiology , CLOCK Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/metabolism
6.
PLoS One ; 4(10): e7476, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19829696

ABSTRACT

BACKGROUND: Circadian clocks control daily rhythms including sleep-wake, hormone secretion, and metabolism. These clocks are based on intracellular transcription-translation feedback loops that sustain daily oscillations of gene expression in many cell types. Mammalian astrocytes display circadian rhythms in the expression of the clock genes Period1 (Per1) and Period2 (Per2). However, a functional role for circadian oscillations in astrocytes is unknown. Because uptake of extrasynaptic glutamate depends on the presence of Per2 in astrocytes, we asked whether glutamate uptake by glia is circadian. METHODOLOGY/PRINCIPAL FINDINGS: We measured glutamate uptake, transcript and protein levels of the astrocyte-specific glutamate transporter, Glast, and the expression of Per1 and Per2 from cultured cortical astrocytes and from explants of somatosensory cortex. We found that glutamate uptake and Glast mRNA and protein expression were significantly reduced in Clock/Clock, Per2- or NPAS2-deficient glia. Uptake was augmented when the medium was supplemented with dibutyryl-cAMP or B27. Critically, glutamate uptake was not circadian in cortical astrocytes cultured from rats or mice or in cortical slices from mice. CONCLUSION/SIGNIFICANCE: We conclude that glutamate uptake levels are modulated by CLOCK, PER2, NPAS2, and the composition of the culture medium, and that uptake does not show circadian variations.


Subject(s)
Astrocytes/cytology , Gene Expression Regulation , Glutamic Acid/metabolism , Animals , Animals, Genetically Modified , Cerebral Cortex/metabolism , Circadian Rhythm , Culture Media/metabolism , Dose-Response Relationship, Drug , Glutamic Acid/chemistry , Mice , Mice, Inbred C57BL , Neuroglia/metabolism , Rats , Rats, Wistar , Transcription Factors/metabolism
7.
J Biol Rhythms ; 24(2): 126-34, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19382381

ABSTRACT

Circadian rhythms in physiology and behavior are temporally synchronized to the day/night cycle through the action of light on the circadian clock. In mammals, transduction of the photic signal reaching the circadian oscillator in the suprachiasmatic nucleus (SCN) occurs through the release of glutamate and pituitary adenylate cyclase-activating peptide (PACAP). The authors' study aimed at clarifying the role played by PACAP in photic resetting and entrainment. They investigated the circadian response to light of PACAPnullmice lacking the 5th exon of the PACAP coding sequence. Specifically, they examined free-running rhythms, entrainment to 12-h light:12-h dark (LD)cycles, the phase-response curve (PRC) to single light pulses, entrainment to a23-h T-cycle, re-entrainment to 6-h phase shifts in LD cycles, and light-induced c-Fos expression. PACAP-null and wild-type mice show similar free-running periods and similar entrainment to 12:12 LD cycles. However, the PRC of PACAP-null mice lacks a phase-advance portion. Surprisingly, despite the absence of phase advance to single light pulses, PACAP-null mice are able to entrain to a 23-h T-cycle, but with a significantly longer phase angle of entrainment than wild types. In addition, PACAP-null mice re-entrain more slowly to a 6-h phase advance of the LD cycle. Nevertheless, induction of c-Fos by light in late night is normal. In all experiments, PACAP-null mice show specific behavioral impairments in response to phase-advancing photic stimuli. These results suggest that PACAP is required for the normal integration of the phase advancing light signal by the SCN.


Subject(s)
Circadian Rhythm/physiology , Photoperiod , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Retina/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Behavior, Animal/physiology , Biological Clocks/physiology , Light , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics
8.
J Mol Neurosci ; 22(3): 223-30, 2004.
Article in English | MEDLINE | ID: mdl-14997016

ABSTRACT

The protein product of the immediate-early gene c-fos is expressed rhythmically in the shell region of the suprachiasmatic nucleus (SCN), the mammalian circadian clock. Recently, we found that exposure to an entraining light pulse caused a suppression of Fos expression in the SCN shell in rats. To study the hypothesis that suppression of Fos in the shell is a correlate of photic entrainment, we used rats that were treated with the retinal neurotoxin monosodium glutamate (MSG) during the neonatal period. In spite of retinal degeneration, MSG-treated rats entrained normally and displayed light-induced suppression of Fos within the SCN shell. These results support the view that light-induced suppression of Fos within the SCN shell is a cellular correlate of photic entrainment.


Subject(s)
Circadian Rhythm/radiation effects , Light , Proto-Oncogene Proteins c-fos/metabolism , Retina/physiology , Suprachiasmatic Nucleus/metabolism , Visual Pathways/physiology , Animals , Animals, Newborn , Circadian Rhythm/physiology , Denervation , Down-Regulation/physiology , Down-Regulation/radiation effects , Female , Male , Neurotoxins , Photic Stimulation/methods , Photoperiod , Rats , Rats, Wistar , Receptor, Nerve Growth Factor , Receptors, Nerve Growth Factor/metabolism , Retina/drug effects , Retinal Degeneration/chemically induced , Retinal Degeneration/physiopathology , Sodium Glutamate , Suprachiasmatic Nucleus/cytology , Up-Regulation/physiology , Up-Regulation/radiation effects
9.
J Mol Neurosci ; 21(2): 133-47, 2003.
Article in English | MEDLINE | ID: mdl-14593213

ABSTRACT

The circadian clock cells of the mammalian suprachiasmatic nucleus (SCN) generate oscillations in physiology and behavior that are synchronized (entrained) by the external light/dark (LD) cycle. The mechanisms that mediate the effect of light on the core molecular mechanism of the clock are not well understood, but evidence suggests that the Period2 gene, which encodes a key clock regulator (PER2), might be involved. We assessed the expression of PER2 immunoreactivity in the retinorecipient core and shell compartments of the SCN of rats entrained to cycles of discrete light pulses presented at the early subjective day (dawn) or night (dusk), or housed in constant light. We found that in animals entrained to a 0.5 h:23.5-h LD cycle (light falls near dawn), PER2 expression is rhythmic both in the shell and in the core regions of the SCN and indistinguishable from that seen in the SCN of control rats housed in complete darkness. Similarly, the pattern of PER2 expression in the SCN of rats entrained to a 0.5-h:25.5-h LD cycle (light falls near dusk) resembled that in dark-housed controls. We also found that presentation of a discrete light pulse in the early subjective night did not induce PER2 protein expression in the SCN, even 6 h after photic stimulation. Finally, in constant light-housed, behaviorally arrhythmic rats, PER2 expression in the SCN was low and nonrhythmic. These results show that rhythmic PER2 expression occurs both in the shell and core regions of the rat SCN. Furthermore, they show that the expression of PER2 in the SCN is not regulated by entraining light. Finally, constant light-induced behavioral arrhythmicity is associated with a disruption of rhythmic PER2 expression in the whole SCN. Together, the results are consistent with a proposed role for PER2 in the core mechanism of the circadian clock but argue against an important role for PER2 in the mechanism mediating photic entrainment.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Nuclear Proteins/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Cell Cycle Proteins , Dark Adaptation/physiology , Down-Regulation/physiology , Gene Expression/physiology , Male , Neurons/cytology , Neurons/metabolism , Period Circadian Proteins , Photic Stimulation , Rats , Rats, Wistar , Suprachiasmatic Nucleus/cytology , Transcription Factors , Up-Regulation/physiology
10.
J Mol Neurosci ; 21(1): 73-89, 2003.
Article in English | MEDLINE | ID: mdl-14500998

ABSTRACT

In mammals, circadian rhythms are generated by a light-entrainable oscillator located in the hypothalamic suprachiasmatic nucleus (SCN). Light signals reach the SCN via a dedicated retinal pathway, the retinohypothalamic tract (RHT). One question that continues to elude scientists is whether the circadian system has its own dedicated photoreceptor or photoreceptors. It is well established that conventional photoreceptors, rods and cones, are not required for circadian photoreception, suggesting that the inner retinal layer might contribute to circadian photoreception. Melanopsin, a novel photo pigment expressed in retinal ganglion cells (RGCs), has been proposed recently as a candidate circadian photoreceptor. Melanopsin-containing RGCs are intrinsically photosensitive, form part of the RHT, and contain neurotransmitters known to play a critical role in the circadian response to light. Furthermore, melanopsin-containing RGCs do not depend on inputs from rods and cones to transmit light signals to the SCN. However, based on a review of the available information about melanopsin and on new data from our laboratory, we propose that melanopsin, in itself, is not necessary for circadian photoreception. In fact, it appears that of the known photoreceptor systems, none, in and of itself, is necessary for circadian photoreception. Instead, it appears that within the photoreceptive systems there is some degree of redundancy, each contributing in some way to photic entrainment.


Subject(s)
Circadian Rhythm/physiology , Retina/metabolism , Rod Opsins/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Animals, Newborn , Antibodies, Monoclonal/pharmacology , Circadian Rhythm/drug effects , Denervation , Eye Enucleation , Immunohistochemistry , Immunotoxins/pharmacology , Light Signal Transduction/drug effects , Light Signal Transduction/physiology , Male , N-Glycosyl Hydrolases , Neurotoxins/pharmacology , Photic Stimulation/methods , Rats , Rats, Wistar , Receptor, Nerve Growth Factor/drug effects , Receptor, Nerve Growth Factor/metabolism , Retina/cytology , Retina/drug effects , Retinal Ganglion Cells/drug effects , Ribosome Inactivating Proteins, Type 1 , Saporins , Sensory Deprivation , Sodium Glutamate , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/drug effects , Visual Pathways/cytology , Visual Pathways/drug effects , Visual Pathways/metabolism
12.
Exp Neurol ; 176(2): 377-89, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12359180

ABSTRACT

Photic entrainment of circadian rhythms in mammals is mediated through a direct retinal projection to the core region of the suprachiasmatic nucleus (SCN), the circadian clock. A proportion of this projection contains the low-affinity p75 neurotrophic receptor (p75NTR). Neonatal monosodium glutamate (MSG) treatment, which dramatically reduces p75NTR immunoreactivity in the SCN has no impact on photic entrainment. In order to clarify the contribution of p75NTR fibers in photic entrainment, targeted lesions of the p75NTR-immunoreactive SCN plexus were performed using intracerebroventricular (ICV) or intrahypothalamic injections of the immunotoxin 192 IgG-saporin (SAP) in rats. SAP treatment effectively abolished p75NTR immunoreactivity within the SCN core. ICV SAP treatment produced three different behavioral activity patterns: Animals became arrhythmic, displayed a shorter free-running period, or remained rhythmic following the lesion. Arrhythmic animals had large hypothalamic lesion which encompassed the entire SCN. In rhythmic rats, ICV-SAP significantly reduced immunostaining for calbindin-D28k (CaBP) in the SCN, and rats with shortened free-running periods had the lowest number of CaBP immunoreactive cells. ICV SAP also attenuated light-induced Fos expression in the SCN core. Despite lack of p75NTR and reduced CaBP and Fos expression in the SCN, SAP-treated rhythmic rats displayed normal photic entrainment. Intrahypothalamic SAP treatment reduced CaBP expression in the SCN but had no effect on light-induced Fos expression, free-running rhythms, or photic entrainment. The data show that p75NTR-immunoreactive elements in the SCN are not required for photic entrainment.


Subject(s)
Antibodies, Monoclonal/pharmacology , Circadian Rhythm/drug effects , Immunotoxins/pharmacology , Oncogene Proteins v-fos/metabolism , Receptors, Nerve Growth Factor/biosynthesis , S100 Calcium Binding Protein G/biosynthesis , Suprachiasmatic Nucleus/metabolism , Activity Cycles/drug effects , Activity Cycles/radiation effects , Animals , Calbindin 1 , Calbindins , Circadian Rhythm/radiation effects , Drug Administration Routes , Immunohistochemistry , Light , Male , Motor Activity/drug effects , Motor Activity/radiation effects , N-Glycosyl Hydrolases , Photoperiod , Rats , Rats, Wistar , Receptor, Nerve Growth Factor , Ribosome Inactivating Proteins, Type 1 , Saporins , Suprachiasmatic Nucleus/anatomy & histology , Suprachiasmatic Nucleus/drug effects
13.
Horm Behav ; 41(1): 59-69, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11863384

ABSTRACT

In lactating rats, food restriction potentiates the already high levels of hypothalamic neuropeptide Y (NPY). To investigate the role that high levels of NPY might play in the prolongation of lactational infertility that typically accompanies a food restricted lactation we investigated the effects of chronic central infusions of NPY in ad libitum-fed lactating females. First, we compared the effects of intracerebroventricular (icv) infusion of NPY from Days 12-19 postpartum at a dose of 14.4 microg/day with a similar treatment in nonlactating females. In subsequent experiments we examined the effects of NPY infusions into the lateral ventricle at doses of 6 or 20 mug/day or unilaterally into the medial preoptic area at a dose of 1 microg/day from either Days 12-19 or 7-21 postpartum. Effects on food intake; female body weight; and, where appropriate, litter weight and length of lactational diestrus were compared between NPY and vehicle-treated females. As expected NPY infusion produced a robust increase in body weight and food intake in nonlactating females that was accompanied by a suppression of cyclicity. By contrast NPY treatment in lactating rats resulted in a marked decrease in litter growth and an earlier termination of lactational diestrus.


Subject(s)
Animals, Suckling/growth & development , Infertility, Female/physiopathology , Lactation/drug effects , Neuropeptide Y/pharmacology , Animals , Eating/drug effects , Female , Injections, Intraventricular , Preoptic Area/drug effects , Prolactin/metabolism , Rats , Rats, Wistar , Weight Gain/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...