Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Biol Rhythms ; 36(4): 410-418, 2021 08.
Article in English | MEDLINE | ID: mdl-33969745

ABSTRACT

Heavy water lengthens the periods of circadian rhythms in various plant and animal species. Many studies have reported that drinking heavy water lengthens the periods of circadian activity rhythms of rodents by slowing the clock mechanism in the suprachiasmatic nucleus (SCN), the mammalian circadian center. The SCN clock is stable and robust against disturbance, due to its intercellular network. It is unclear whether this robustness provides resistance to the effects of heavy water. Here, we report that heavy water lengthened the rhythm period of clock gene expression of the SCN and peripheral tissues in vitro using a PERIOD2::LUCIFERASE bioluminescence reporter. Our results show that the period-elongation rate of the SCN is similar to those of other tissues. Therefore, the intercellular network of the SCN is not resistant to the period-elongation effect of heavy water.


Subject(s)
Circadian Clocks , Animals , Circadian Rhythm , Deuterium Oxide , Mice , Mice, Transgenic , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Suprachiasmatic Nucleus/metabolism
2.
Connect Tissue Res ; 62(5): 519-530, 2021 09.
Article in English | MEDLINE | ID: mdl-32619127

ABSTRACT

Purpose: Collagen IV is a component of the basement membrane (BM) that provides mechanical support for muscle fibers. In addition, transcription factor 4 (TCF4) is highly expressed in muscle connective tissue fibroblasts and regulates muscle regeneration. However, the expression of collagen IV and TCF4 (+) cells in response to exercise-induced muscle injury is not well known. Here, we investigated the expression and localization of collagen IV and TCF4 (+) cells during the recovery process after muscle injury induced by different exercise loads.Materials and Methods: Muscle injury was observed in the soleus muscle of young Wistar rats after 12 or 18 sets-downhill running (DR) on a treadmill. After running, the rats were permitted to recover for a period of 0.5 days, 2 days, or 7 days.Results: Ectopic localization of collagen IV in injured muscle fibers was observed after DR, and the number increased at 0.5 days after 18 sets DR and at 2 days after 12 or 18 sets DR as compared to the number observed at baseline. BM disruption was observed after DR. TCF4 (+) cells appeared in the inside and around injured muscle fibers at 0.5 day of recovery. After 18 sets DR, TCF4 (+) cells were more abundant for a longer period than that observed after 12 sets DR.Conclusions: DR induces BM disruption accompanied by muscle fiber damage. It is possible that BM destruction may be accompanied by muscle damage and that TCF4 (+) cells contribute to muscle fiber and BM recovery.


Subject(s)
Basement Membrane , Muscle, Skeletal , Animals , Collagen , Muscle Fibers, Skeletal , Rats , Rats, Wistar
3.
Brain Res ; 1714: 73-80, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30771316

ABSTRACT

The suprachiasmatic nucleus (SCN) is the center of the mammalian circadian system. Environmental photic signals shifts the phase of the circadian rhythm in the SCN except during the dead zone, when the photic signal is gated somewhere on the way from the retina to the neurons in the SCN. Here we examined the phase of the dead zone after an abrupt delay of the LD cycles for several days by observing the mc-Fos induction in the SCN by light pulses. After an abrupt shift of the LD cycles, the dead zone showed a slow phase shift, about two hours per day, which was well corresponded with the slow phase shift of the rest-activity cycles. In our previous study we demonstrated that, after an abrupt shift of the LD cycles, the SCN showed transient endogenous desynchronization between shell and core regions that showed a slow and a rapid shift of the circadian rhythms, respectively. Therefore, the present findings on the phase shift of the dead zone after the LD cycles shift suggest that the phase of the dead zone is under the control of the timing signals from the shell region of the SCN.


Subject(s)
Circadian Rhythm/physiology , Suprachiasmatic Nucleus/metabolism , Animals , Biological Clocks/physiology , Light , Male , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Neurons/metabolism , Nuclear Proteins/metabolism , Period Circadian Proteins/metabolism , Photic Stimulation/methods , Photoperiod , Proto-Oncogene Proteins c-fos/analysis , Retina/metabolism , Suprachiasmatic Nucleus/physiology
4.
Sci Rep ; 8(1): 854, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29339832

ABSTRACT

In mammals, the principal circadian oscillator exists in the hypothalamic suprachiasmatic nucleus (SCN). In the SCN, CLOCK works as an essential component of molecular circadian oscillation, and ClockΔ19 mutant mice show unique characteristics of circadian rhythms such as extended free running periods, amplitude attenuation, and high-magnitude phase-resetting responses. Here we investigated what modifications occur in the spatiotemporal organization of clock gene expression in the SCN of ClockΔ19 mutants. The cultured SCN, sampled from neonatal homozygous ClockΔ19 mice on an ICR strain comprising PERIOD2::LUCIFERASE, demonstrated that the Clock gene mutation not only extends the circadian period, but also affects the spatial phase and period distribution of circadian oscillations in the SCN. In addition, disruption of the synchronization among neurons markedly attenuated the amplitude of the circadian rhythm of individual oscillating neurons in the mutant SCN. Further, with numerical simulations based on the present studies, the findings suggested that, in the SCN of the ClockΔ19 mutant mice, stable oscillation was preserved by the interaction among oscillating neurons, and that the orderly phase and period distribution that makes a phase wave are dependent on the functionality of CLOCK.


Subject(s)
CLOCK Proteins/metabolism , Neurons/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Behavior, Animal , CLOCK Proteins/genetics , Circadian Rhythm/physiology , Cyclic AMP/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Models, Theoretical , Mutagenesis , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Signal Transduction/drug effects , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/drug effects , Tetrodotoxin/pharmacology
5.
Exp Gerontol ; 98: 153-161, 2017 11.
Article in English | MEDLINE | ID: mdl-28803135

ABSTRACT

Aging is known to lead to the impaired recovery of muscle after disuse as well as the increased susceptibility of the muscle to damage. Here, we show that, in the older rats, reloading after disuse atrophy, causes the damage of the muscle fibers and the basement membrane (BM) that structurally support the muscle fibers. Male Wistar rats of 3-(young) and 20-(older) months of age were subjected to hindlimb-unloading for 2weeks followed by reloading for a week. In the older rats, the soleus muscles showed necrosis and central nuclei fiber indicating the regeneration of muscle fibers. Furthermore, ectopic immunoreactivity of collagen IV, a major component of the BM, remained mostly associated with the necrotic appearance, suggesting that the older rats were impaired with the ability of repairing the damaged BM. Further, after unloading and reloading, the older rats did not show a significant alteration, although the young rats showed clear response of Col4a1 and Col4a2 genes, both coding for collagen IV. In addition, during the recovery phase, the young rats showed increase in the amount of Hsp47 and Sparc mRNA, which are protein folding-related factor genes, while the older rats did not show any significant variation. Taken together, our findings suggest that the atrophic muscle fibers of the older rats induced by unloading were vulnerable to the weight loading, and that attenuated reactivity of the BM-synthesizing fibroblast to gravity contributes to the fragility of muscle fibers in the older animals.


Subject(s)
Aging/metabolism , Basement Membrane/metabolism , Muscle Contraction , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Age Factors , Aging/genetics , Aging/pathology , Animals , Basement Membrane/ultrastructure , Collagen Type IV/genetics , Collagen Type IV/metabolism , Disease Models, Animal , Gene Expression Regulation , HSP47 Heat-Shock Proteins/genetics , HSP47 Heat-Shock Proteins/metabolism , Hindlimb Suspension , Male , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/ultrastructure , Muscle Strength , Muscle, Skeletal/physiopathology , Muscle, Skeletal/ultrastructure , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , Necrosis , Osteonectin/genetics , Osteonectin/metabolism , Rats, Wistar , Recovery of Function , Time Factors
6.
Acta Histochem Cytochem ; 50(2): 95-104, 2017 Apr 27.
Article in English | MEDLINE | ID: mdl-28522884

ABSTRACT

Both prokineticin receptor 2 (pkr2) and prokineticin 2 (pk2) gene-deficient mice have hypoplasia of the main olfactory bulb (MOB). This hypoplasia has been attributed to disruption of the glomerulus that is caused by loss of afferent projection from olfactory sensory neurons (OSN), and to the impaired migration of granule cells, a type of interneuron. In the present study, we examined whether migration of the second type of interneuron, periglomerular cells (PGC), is dependent on the pkr2 expression by observing the localization of distinct subpopulations of PGC: calretinin (CR)-, calbindin (CB)- and tyrosine hydroxylase (TH)-expressing neurons. In the Pkr2-/- mice, the construction of the layered structure of the MOB was partially preserved, with the exception of the internal plexiform layer (IPL) and the glomerular layer (GL). In the outermost layer of the MOB, abundant CR- and CB-immunopositive neurons were observed in the hypoplastic olfactory bulb. In addition, although markedly decreased, TH-immunopositive neurons were also observed in the outermost cell-dense region in the Pkr2-/-. The findings suggest that the migration of PGC to the MOB, as well as the migration from the core to the surface region of the MOB, is not driven by the PK2-PKR2 system.

7.
Eur J Neurosci ; 38(6): 2832-41, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23869693

ABSTRACT

The suprachiasmatic nucleus (SCN) is the mammalian circadian rhythm center. Individual oscillating neurons have different endogenous circadian periods, but they are usually synchronized by an intercellular coupling mechanism. The differences in the period of each oscillating neuron have been extensively studied; however, the clustering of oscillators with similar periods has not been reported. In the present study, we artificially disrupted the intercellular coupling among oscillating neurons in the SCN and observed regional differences in the periods of the oscillating small-latticed regions of the SCN using a transgenic rat carrying a luciferase reporter gene driven by regulatory elements from a per2 clock gene (Per2::dluc rat). The analysis divided the SCN into two regions--aregion with periods shorter than 24 h (short-period region, SPR) and another with periods longer than 24 h (long-period region, LPR). The SPR was located in the smaller medial region of the dorsal SCN, whereas the LPR occupied the remaining larger region. We also found that slices containing the medial region of the SCN generated shorter circadian periods than slices that contained the lateral region of the SCN. Interestingly, the SPR corresponded well with the region where the SCN phase wave is generated. We numerically simulated the relationship between the SPR and a large LPR. A mathematical model of the SCN based on our findings faithfully reproduced the kinetics of the oscillators in the SCN in synchronized conditions, assuming the existence of clustered short-period oscillators.


Subject(s)
Circadian Clocks , Neurons/metabolism , Period Circadian Proteins/metabolism , Suprachiasmatic Nucleus/physiology , Animals , Circadian Clocks/drug effects , Colforsin/pharmacology , Male , Period Circadian Proteins/genetics , Rats , Rats, Transgenic , Rats, Wistar , Suprachiasmatic Nucleus/drug effects
8.
Endocrinology ; 153(5): 2277-86, 2012 May.
Article in English | MEDLINE | ID: mdl-22434077

ABSTRACT

The suprachiasmatic nucleus is the master circadian clock and resets the peripheral clocks via various pathways. Glucocorticoids and daily feeding are major time cues for entraining most peripheral clocks. However, recent studies have suggested that the dominant timing factor differs among organs and tissues. In our current study, we reveal differences in the entrainment properties of the peripheral clocks in the liver, kidney, and lung through restricted feeding (RF) and antiphasic corticosterone (CORT) injections in adrenalectomized rats. The peripheral clocks in the kidney and lung were found to be entrained by a daily stimulus from CORT administration, irrespective of the meal time. In contrast, the liver clock was observed to be entrained by an RF regimen, even if daily CORT injections were given at antiphase. These results indicate that glucocorticoids are a strong zeitgeber that overcomes other entrainment factors regulating the peripheral oscillators in the kidney and lung and that RF is a dominant mediator of the entrainment ability of the circadian clock in the liver.


Subject(s)
Circadian Clocks/drug effects , Corticosterone/pharmacology , Feeding Behavior/physiology , Glucocorticoids/pharmacology , Suprachiasmatic Nucleus/drug effects , Adrenalectomy , Animals , Circadian Clocks/physiology , Circadian Rhythm/drug effects , Circadian Rhythm/physiology , Gene Expression/drug effects , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Lung/drug effects , Lung/metabolism , Male , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Rats , Rats, Wistar , Suprachiasmatic Nucleus/physiology
9.
Eur J Neurosci ; 26(10): 2731-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17973924

ABSTRACT

The mammalian hypothalamic suprachiasmatic nucleus (SCN) is the master oscillator that regulates the circadian rhythms of the peripheral oscillators. Previous studies have demonstrated that the transplantation of embryonic SCN tissues into SCN-lesioned arrhythmic mice restores the behavioral circadian rhythms of these animals. In our present study, we examined the clock gene expression profiles in a transplanted SCN and peripheral tissues, and also analysed the circadian rhythm of the locomotor activity in SCN-grafted mice. These experiments were undertaken to elucidate whether the transplanted SCN generates a dynamic circadian oscillation and maintains the phase relationships that can be detected in intact mice. The grafted SCN indeed showed dynamic circadian expression rhythms of clock genes such as mPeriod1 (mPer1) and mPeriod2 (mPer2). Furthermore, the phase differences between the expression rhythms of these genes in the grafted SCN and the locomotor activity rhythms of the transplanted animals were found to be very similar to those in intact animals. Moreover, in the liver, kidney and skeletal muscles of the transplanted animals, the phase angles between the circadian rhythm of the grafted SCN and that of the peripheral tissues were maintained as in intact animals. However, in the SCN-grafted animals, the amplitudes of the mPer1 and mPer2 rhythms were attenuated in the peripheral tissues. Our current findings therefore indicate that a transplanted SCN has the capacity to generate a dynamic intrinsic circadian oscillation, and can also lock the normal phase angles among the SCN, locomotor activity and peripheral oscillators in a similar manner as in intact control animals.


Subject(s)
Brain Tissue Transplantation , Circadian Rhythm/physiology , Gene Expression/physiology , Nuclear Proteins/metabolism , Suprachiasmatic Nucleus/metabolism , Suprachiasmatic Nucleus/transplantation , Animals , Behavior, Animal , Embryo, Mammalian , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Nuclear Proteins/genetics , Suprachiasmatic Nucleus/injuries , Suprachiasmatic Nucleus/surgery , Time Factors
10.
Nat Cell Biol ; 9(11): 1327-34, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17952058

ABSTRACT

Singularity behaviour in circadian clocks--the loss of robust circadian rhythms following exposure to a stimulus such as a pulse of bright light--is one of the fundamental but mysterious properties of clocks. To quantitatively perturb and accurately measure the dynamics of cellular clocks, we synthetically produced photo-responsiveness within mammalian cells by exogenously introducing the photoreceptor melanopsin and continuously monitoring the effect of photo-perturbation on the state of cellular clocks. Here we report that a critical light pulse drives cellular clocks into singularity behaviour. Our theoretical analysis consistently predicts and subsequent single-cell level observation directly proves that desynchronization of individual cellular clocks underlies singularity behaviour. Our theoretical framework also explains why singularity behaviours have been experimentally observed in various organisms, and it suggests that desynchronization is a plausible mechanism for the observable singularity of circadian clocks. Importantly, these in vitro and in silico findings are further supported by in vivo observations that desynchronization underlies the multicell-level amplitude decrease in the rat suprachiasmatic nucleus induced by critical light pulses.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Light , Rod Opsins/physiology , Animals , Biological Clocks/drug effects , Biological Clocks/radiation effects , Cell Line, Tumor , Cells, Cultured , Circadian Rhythm/drug effects , Circadian Rhythm/radiation effects , Humans , In Situ Hybridization, Fluorescence , Male , Mice , NIH 3T3 Cells , Rats , Rats, Wistar , Rod Opsins/pharmacology
11.
Brain Res ; 1098(1): 9-18, 2006 Jul 07.
Article in English | MEDLINE | ID: mdl-16780815

ABSTRACT

Most biological phenomena, including behavior and metabolic pathways, are governed by an internal clock system that is circadian (i.e., with a period of approximately 24 h) and is reset by light exposure from outside. In order to understand the molecular basis of the resetting mechanism of the clock, we attempted to isolate light-inducible transcripts in the suprachiasmatic nucleus, where the master clock resides, using a new gene expression profiling procedure. We identified 87 such transcripts, successfully cloned 60 of them and confirmed their light inducibility. Six of the 60 were already known to be light inducible and 17 are protein-coding transcripts registered in the public database that were not known to be light inducible. Induction is subjective night specific in most of the transcripts. Interestingly, 6 of the transcripts exhibit rhythmic expression in a circadian manner in the suprachiasmatic nucleus.


Subject(s)
Circadian Rhythm/genetics , Suprachiasmatic Nucleus/physiology , Animals , Cloning, Molecular , Gene Expression Regulation , In Situ Hybridization , Light , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , RNA/biosynthesis , RNA/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Suprachiasmatic Nucleus/radiation effects
12.
Proc Natl Acad Sci U S A ; 103(10): 3716-21, 2006 Mar 07.
Article in English | MEDLINE | ID: mdl-16537451

ABSTRACT

Three mammalian Period (Per) genes, termed Per1, Per2, and Per3, have been identified as structural homologues of the Drosophila circadian clock gene, period (per). The three Per genes are rhythmically expressed in the suprachiasmatic nucleus (SCN), the central circadian pacemaker in mammals. The phases of peak mRNA levels for the three Per genes in the SCN are slightly different. Light sequentially induces the transcripts of Per1 and Per2 but not of Per3 in mice. These data and others suggest that each Per gene has a different but partially redundant function in mammals. To elucidate the function of Per1 in the circadian system in vivo, we generated two transgenic rat lines in which the mouse Per1 (mPer1) transcript was constitutively expressed under the control of either the human elongation factor-1alpha (EF-1alpha) or the rat neuron-specific enolase (NSE) promoter. The transgenic rats exhibited an approximately 0.6-1.0-h longer circadian period than their wild-type siblings in both activity and body temperature rhythms. Entrainment in response to light cycles was dramatically impaired in the transgenic rats. Molecular analysis revealed that the amplitudes of oscillation in the rat Per1 (rPer1) and rat Per2 (rPer2) mRNAs were significantly attenuated in the SCN and eyes of the transgenic rats. These results indicate that either the level of Per1, which is raised by overexpression, or its rhythmic expression, which is damped or abolished in over expressing animals, is critical for normal entrainment of behavior and molecular oscillation of other clock genes.


Subject(s)
Circadian Rhythm/genetics , Nuclear Proteins/genetics , Animals , Animals, Genetically Modified , Base Sequence , Behavior, Animal/physiology , Cell Cycle Proteins , Circadian Rhythm/physiology , Eye/metabolism , Female , Gene Expression , Male , Mice , Nuclear Proteins/physiology , Period Circadian Proteins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Suprachiasmatic Nucleus/metabolism , Transcription Factors/genetics , Transcription Factors/physiology
13.
J Biol Chem ; 279(11): 10237-42, 2004 Mar 12.
Article in English | MEDLINE | ID: mdl-14701801

ABSTRACT

The regulatory factor X (RFX) family of transcription factors is characterized by a unique and highly conserved 76-amino acid residue DNA-binding domain. Mammals have five RFX genes, but the physiological functions of their products are unknown, with the exception of RFX5. Here a mouse RFX4 transcript was identified that encodes a peptide of 735 amino acids, including the DNA-binding domain. Its expression was localized in the suprachiasmatic nucleus, the central pacemaker site of the circadian clock. Also, light exposure was found to induce its gene expression in a subjective night-specific manner. Polyclonal antibodies were prepared, and an 80-kDa band was detected in the suprachiasmatic nucleus by Western hybridization. A histochemical study showed a localization of the products in the nucleus. This is the first report on mouse RFX4, which contains the RFX DNA-binding motif. Our investigation may provide clues to the physiological function of RFX4.


Subject(s)
Hypothalamus/metabolism , Transcription Factors/biosynthesis , Transcription Factors/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , Brain/pathology , COS Cells , Circadian Rhythm , DNA/metabolism , DNA, Complementary/metabolism , DNA-Binding Proteins/chemistry , Immunohistochemistry , In Situ Hybridization , Light , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Peptides/chemistry , Protein Binding , Protein Structure, Tertiary , RNA/chemistry , RNA, Messenger/metabolism , Regulatory Factor X Transcription Factors , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
14.
Curr Biol ; 13(8): 664-8, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12699623

ABSTRACT

The mammalian master clock driving circadian rhythmicity in physiology and behavior resides within the suprachiasmatic nuclei (SCN) of the hypothalamus. SCN neurons contain a molecular oscillator composed of a set of clock genes that acts in intertwined negative and positive feedback loops [1]. In addition, all peripheral tissues analyzed thus far have been shown to contain circadian oscillators [2]. This raises the question of whether the central circadian pacemaker in the SCN is sufficient to evoke behavioral rhythms or whether peripheral circadian clockworks are also required. Mice with a mutated CLOCK protein (a transcriptional activator of E box-containing clock and clock output genes) or lacking both CRYPTOCHROMES, mCRY1 and mCRY2 proteins (inhibitors of E box-mediated transcription), lack circadian rhythmicity in behavior [3,4]. Here, we show that transplantation of mouse fetal SCN tissue into the hypothalamus restores free-running circadian behavioral rhythmicity in Clock mutant or mCry1/mCry2 double knockout mice. The periodicity of the emerged rhythms is determined by the genetic constitution (i.e., wild-type or mCry2 knockout) of the grafted SCN. Since transplanted mCry1/mCry2-deficient mice do not have functional circadian oscillators [5] other than those present in the grafted hypothalamus region, these findings suggest that the SCN can generate circadian behavioral rhythms in the absence of distant peripheral oscillators in the brain or elsewhere.


Subject(s)
Circadian Rhythm/genetics , Circadian Rhythm/physiology , Drosophila Proteins , Eye Proteins , Photoreceptor Cells, Invertebrate , Suprachiasmatic Nucleus/physiology , Suprachiasmatic Nucleus/transplantation , Animals , Biological Clocks/physiology , CLOCK Proteins , Cryptochromes , Flavoproteins/genetics , Hypothalamus/anatomy & histology , Immunohistochemistry , Locomotion/physiology , Mice , Mice, Knockout , Receptors, G-Protein-Coupled , Trans-Activators/genetics
15.
Brain Res Mol Brain Res ; 110(1): 1-6, 2003 Jan 31.
Article in English | MEDLINE | ID: mdl-12573527

ABSTRACT

We identified the Dexamethasone-induced RAS protein 1 (Dexras1) gene as a cycling gene in the suprachiasmatic nucleus (SCN). Investigation of the whole brain using in situ hybridization demonstrated the localization of the expression of the gene in the SCN, thalamus, piriform cortex and hippocampus. However, rhythmic expression of the gene was observed only in the SCN. The rhythmic change in gene expression during 1 day was approximately five-fold, and the maximum expression was observed during subjective night. Real-time PCR using the SCN, paraventricular nucleus and cortex confirmed these results. Next, we analyzed the expression of the Dexras1 gene in the SCN of cryptochrome (Cry) 1 and 2 double knockout mice. We found that the rhythmic expression disappeared. The results indicate that Dexras1 rhythmicity and levels are dependent upon CRYs. This is the first time that the G protein, which may be involved in the input pathway, has been isolated as a cycling gene in the SCN.


Subject(s)
Circadian Rhythm/genetics , Drosophila Proteins , Eye Proteins , GTP-Binding Proteins , Monomeric GTP-Binding Proteins/genetics , Photoreceptor Cells, Invertebrate , Suprachiasmatic Nucleus/physiology , ras Proteins , Animals , Cryptochromes , Dexamethasone , Flavoproteins/genetics , Gene Expression Regulation , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monomeric GTP-Binding Proteins/metabolism , Polymerase Chain Reaction/methods , RNA, Messenger/analysis , Receptors, G-Protein-Coupled
SELECTION OF CITATIONS
SEARCH DETAIL
...