Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Pineal Res ; 68(2): e12629, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31808568

ABSTRACT

The pineal gland is a neuroendocrine organ responsible for production of the nocturnal hormone melatonin. A specific set of homeobox gene-encoded transcription factors govern pineal development, and some are expressed in adulthood. The brain-specific homeobox gene (Bsx) falls into both categories. We here examined regulation and function of Bsx in the mature pineal gland of the rat. We report that Bsx is expressed from prenatal stages into adulthood, where Bsx transcripts are localized in the melatonin-synthesizing pinealocytes, as revealed by RNAscope in situ hybridization. Bsx transcripts were also detected in the adult human pineal gland. In the rat pineal gland, Bsx was found to exhibit a 10-fold circadian rhythm with a peak at night. By combining in vivo adrenergic stimulation and surgical denervation of the gland in the rat with in vitro stimulation and transcriptional inhibition in cultured pinealocytes, we show that rhythmic expression of Bsx is controlled at the transcriptional level by the sympathetic neural input to the gland acting via adrenergic stimulation with cyclic AMP as a second messenger. siRNA-mediated knockdown (>80% reduction) in pinealocyte cultures revealed Bsx to be a negative regulator of other pineal homeobox genes, including paired box 4 (Pax4), but no effect on genes encoding melatonin-synthesizing enzymes was detected. RNA sequencing analysis performed on siRNA-treated pinealocytes further revealed that downstream target genes of Bsx are mainly involved in developmental processes. Thus, rhythmic Bsx expression seems to govern other developmental regulators in the mature pineal gland.


Subject(s)
Circadian Rhythm/physiology , Gene Expression Regulation, Developmental/physiology , Melatonin/metabolism , Nerve Tissue Proteins/biosynthesis , Pineal Gland/metabolism , Transcription Factors/biosynthesis , Animals , Female , Male , Pineal Gland/cytology , Rats, Sprague-Dawley
2.
J Pineal Res ; 68(1): e12616, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31609018

ABSTRACT

Homeobox genes generally encode transcription factors involved in regulating developmental processes. In the pineal gland, a brain structure devoted to nocturnal melatonin synthesis, a number of homeobox genes are also expressed postnatally; among these is the LIM homeobox 4 gene (Lhx4). We here report that Lhx4 is specifically expressed in the postnatal pineal gland of rats and humans. Circadian analyses revealed a fourfold rhythm in Lhx4 expression in the rat pineal gland, with rhythmic expression detectable from postnatal day 10. Pineal Lhx4 expression was confirmed to be positively driven by adrenergic signaling, as evidenced by in vivo modulation of Lhx4 expression by pharmacological (isoprenaline injection) and surgical (superior cervical ganglionectomy) interventions. In cultured pinealocytes, Lhx4 expression was upregulated by cyclic AMP, a second messenger of norepinephrine. By use of RNAscope technology, Lhx4 transcripts were found to be exclusively localized in melatonin-synthesizing pinealocytes. This prompted us to investigate the possible role of Lhx4 in regulation of melatonin-producing enzymes. By use of siRNA technology, we knocked down Lhx4 by 95% in cultured pinealocytes; this caused a reduction in transcripts encoding the melatonin-producing enzyme arylalkylamine N-acetyl transferase (Aanat). Screening the transcriptome of siRNA-treated pinealocytes by RNAseq revealed a significant impact of Lhx4 on the phototransduction pathway and on transcripts involved in development of the nervous system and photoreceptors. These data suggest that rhythmic expression of Lhx4 in the pineal gland is controlled via an adrenergic-cyclic AMP mechanism and that Lhx4 acts to promote nocturnal melatonin synthesis.


Subject(s)
LIM-Homeodomain Proteins , Melatonin/metabolism , Pineal Gland , Transcription Factors , Transcriptome/genetics , Adult , Animals , Arylalkylamine N-Acetyltransferase/genetics , Arylalkylamine N-Acetyltransferase/metabolism , Circadian Rhythm/genetics , Cyclic AMP/metabolism , Female , Humans , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Male , Melatonin/genetics , Middle Aged , Norepinephrine/metabolism , Pineal Gland/chemistry , Pineal Gland/cytology , Pineal Gland/growth & development , Pineal Gland/metabolism , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Transcription Factors/genetics , Transcription Factors/metabolism , Young Adult
3.
J Pineal Res ; 66(4): e12567, 2019 May.
Article in English | MEDLINE | ID: mdl-30803008

ABSTRACT

Homeobox genes encode transcription factors that regulate developmental processes; however, in the pineal gland, a neuroendocrine organ responsible for nocturnal melatonin synthesis, expression of the homeobox genes Otx2 (orthodenticle homeobox 2) and Crx (cone-rod homeobox) persists postnatally. We here show that OTX2 and CRX are exclusively present in melatonin-producing pinealocytes of the rat pineal gland. To understand the roles of Otx2 and Crx in the mature pineal gland, we used siRNA technology in cultured rat pinealocytes with the nocturnal situation mimicked by adding norepinephrine to the culture media. siRNA-induced knockdown of Otx2 was found to reduce expression levels of the enzymes involved in melatonin synthesis at both transcript and protein levels. Similar results were obtained when knocking down Crx. Knocking down Otx2 and Crx simultaneously produced an even larger reduction in both transcript and protein levels of the melatonin-producing enzymes and also reduced the levels of melatonin released to the culture media. These results suggest that Otx2 and Crx, both alone and in combination, act to control pineal melatonin synthesis.


Subject(s)
Homeodomain Proteins/metabolism , Melatonin/metabolism , Otx Transcription Factors/metabolism , Pineal Gland/metabolism , Trans-Activators/metabolism , Animals , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/genetics , Immunohistochemistry , Male , Otx Transcription Factors/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics
4.
J Neurochem ; 143(1): 100-111, 2017 10.
Article in English | MEDLINE | ID: mdl-28675567

ABSTRACT

The retinal and anterior neural fold homeobox gene (Rax) controls development of the eye and the forebrain. Postnatal expression of Rax in the brain is restricted to the pineal gland, a forebrain structure devoted to melatonin synthesis. The role of Rax in pineal function is unknown. In order to investigate the role of Rax in pineal function while circumventing forebrain abnormalities of the global Rax knockout, we generated an eye and pineal-specific Rax conditional knockout mouse. Deletion of Rax in the pineal gland did not affect morphology of the gland, suggesting that Rax is not essential for pineal gland development. In contrast, deletion of Rax in the eye generated an anophthalmic phenotype. In addition to the loss of central visual pathways, the suprachiasmatic nucleus of the hypothalamus housing the circadian clock was absent, indicating that the retinohypothalamic tract is required for the nucleus to develop. Telemetric analyses confirmed the lack of a functional circadian clock. Arylalkylamine N-acetyltransferase (Aanat) transcripts, encoding the melatonin rhythm-generating enzyme, were undetectable in the pineal gland of the Rax conditional knockout under normal conditions, whereas the paired box 6 homeobox gene, known to regulate pineal development, was up-regulated. By injecting isoproterenol, which mimics a nocturnal situation in the pineal gland, we were able to induce pineal expression of Aanat in the Rax conditional knockout mouse, but Aanat transcript levels were significantly lower than those of Rax-proficient mice. Our data suggest that Rax controls pineal gene expression and via Aanat may modulate melatonin synthesis.


Subject(s)
Circadian Rhythm/physiology , Eye Proteins/physiology , Genes, Homeobox/physiology , Homeodomain Proteins/physiology , Pineal Gland/metabolism , Suprachiasmatic Nucleus/metabolism , Transcription Factors/physiology , Visual Pathways/metabolism , Animals , Arylalkylamine N-Acetyltransferase/biosynthesis , Arylalkylamine N-Acetyltransferase/genetics , Eye Proteins/genetics , Female , Gene Expression Profiling/methods , Homeodomain Proteins/genetics , Male , Mice , Mice, 129 Strain , Mice, Knockout , Neuroendocrine Cells/metabolism , Retina/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics
5.
J Biol Chem ; 287(44): 37420-33, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22955284

ABSTRACT

Nectins belong to a family of immunoglobulin (Ig)-like cell-adhesion molecules comprising four members, nectin-1 through nectin-4. Nectins are involved in formation of the mechanical adhesive puncta adherentia junctions of synapses. Nectins share the same overall structural topology with an extracellular region containing three Ig modules, a transmembrane region, and a cytoplasmic region. In nectin-1, the first and second Ig module in the extracellular region are necessary for the trans-interaction with nectin-3 and formation of cis-dimers, respectively. The function of the third Ig module of nectin-1 remains unknown. We here report the structure in solution of the third, membrane-proximal Ig module of mouse nectin-1 (nectin-1 Ig3) solved by means of nuclear magnetic resonance (NMR) spectroscopy. It belongs to the C1 set of the Ig superfamily. Nectin-1 Ig3 was produced as a recombinant protein and induced neurite outgrowth in primary cultures of hippocampal and cerebellar granule neurons, an effect abolished by treatment with the fibroblast growth factor receptor (FGFR) inhibitor SU5402, or by transfection with a dominant-negative FGFR1 construct. We showed by surface plasmon resonance (SPR) analysis that nectin-1 Ig3 directly interacted with various isoforms of FGFR. Nectin-1 Ig3 induced phosphorylation of FGFR1c in the same manner as the whole nectin-1 ectodomain, and promoted survival of cerebellar granule neurons induced to undergo apoptosis. Finally, we constructed a peptide, nectide, by employing in silico modeling of various FGFR ligand-binding sites. Nectide mimicked all the effects of nectin-1 Ig3. We suggest that FGFR is a downstream signaling partner of nectin-1.


Subject(s)
Cell Adhesion Molecules/physiology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Amino Acid Sequence , Animals , Apoptosis , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/metabolism , Cell Survival , Crystallography, X-Ray , Fibroblast Growth Factor 2/physiology , HEK293 Cells , Hippocampus/cytology , Humans , Mice , Molecular Sequence Data , Nectins , Neurites/metabolism , Neurites/physiology , Neurons/cytology , Neurons/physiology , Phosphorylation , Primary Cell Culture , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Processing, Post-Translational , Protein Structure, Secondary , Protein Structure, Tertiary , Rats , Rats, Wistar , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptors, Platelet-Derived Growth Factor/chemistry , Signal Transduction , Surface Plasmon Resonance
6.
Neurochem Res ; 38(6): 1100-12, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23076630

ABSTRACT

The pineal gland is a neuroendocrine gland responsible for nocturnal synthesis of melatonin. During early development of the rodent pineal gland from the roof of the diencephalon, homeobox genes of the orthodenticle homeobox (Otx)- and paired box (Pax)-families are expressed and are essential for normal pineal development consistent with the well-established role that homeobox genes play in developmental processes. However, the pineal gland appears to be unusual because strong homeobox gene expression persists in the pineal gland of the adult brain. Accordingly, in addition to developmental functions, homeobox genes appear to be key regulators in postnatal phenotype maintenance in this tissue. In this paper, we review ontogenetic and phylogenetic aspects of pineal development and recent progress in understanding the involvement of homebox genes in rodent pineal development and adult function. A working model is proposed for understanding the sequential action of homeobox genes in controlling development and mature circadian function of the mammalian pinealocyte based on knowledge from detailed developmental and daily gene expression analyses in rats, the pineal phenotypes of homebox gene-deficient mice and studies on development of the retinal photoreceptor; the pinealocyte and retinal photoreceptor share features not seen in other tissues and are likely to have evolved from the same ancestral photodetector cell.


Subject(s)
Genes, Homeobox/physiology , Pineal Gland/growth & development , Pineal Gland/physiology , Animals , Circadian Rhythm/physiology , Gene Expression Regulation, Developmental , Homeodomain Proteins/physiology , Melatonin/biosynthesis , Mice , Otx Transcription Factors/genetics , Phenotype , Photoreceptor Cells, Invertebrate/physiology , Photoreceptor Cells, Vertebrate/physiology , Phylogeny , Pineal Gland/embryology , Rats , Trans-Activators/physiology
7.
Neurochem Res ; 38(6): 1219-28, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23135794

ABSTRACT

Snap25 (synaptosomal-associated protein) is a 25 kDa protein, belonging to the SNARE-family (soluble N-ethylmaleimide-sensitive factor attachment protein receptors) of proteins, essential for synaptic and secretory vesicle exocytosis. Snap25 has by immunohistochemistry been demonstrated in the rat pineal gland but the biological importance of this is unknown. In this study, we demonstrate a high expression of mRNA encoding Snap25 in all parts of the rat pineal complex, the superficial-, and deep-pineal gland, as well as in the pineal stalk. Snap25 showed a low pineal expression during embryonic stages with a strong increase in expression levels just after birth. The expression showed no day/night variations. Neither removal of the sympathetic input to the pineal gland by superior cervical ganglionectomy nor bilateral decentralization of the superior cervical ganglia significantly affected the expression of Snap25 in the gland. The pineal expression levels of Snap25 were not changed following intraperitoneal injection of isoproterenol. The strong expression of Snap25 in the pineal gland suggests the presence of secretory granules and microvesicles in the rat pinealocyte supporting the concept of a vesicular release. At the transcriptional level, this Snap25-based release mechanism does not exhibit any diurnal rhythmicity and is regulated independently of the sympathetic nervous input to the gland.


Subject(s)
Pineal Gland/embryology , Pineal Gland/metabolism , Synaptosomal-Associated Protein 25/biosynthesis , Animals , Circadian Rhythm/physiology , Male , Mice , Pineal Gland/innervation , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Superior Cervical Ganglion/physiology
8.
J Neurochem ; 118(6): 999-1007, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21749377

ABSTRACT

Retina and anterior neural fold homeobox (Rax) gene encodes a transcription factor essential for vertebrate eye development. Recent microarray studies indicate that Rax is expressed in the adult rat pineal gland and retina. The present study reveals that Rax expression levels in the rat change significantly during retinal development with a peak occurring at embryonic day 18, whereas Rax expression in the pineal is relatively delayed and not detectable until embryonic day 20. In both tissues, Rax is expressed throughout postnatal development into adulthood. In the mature rat pineal gland, the abundance of Rax transcripts increases 2-fold during the light period with a peak occurring at dusk. These findings are consistent with the evidence that Rax is of functional importance in eye development and suggest a role of Rax in the developing pineal gland. In addition, it would appear possible that Rax contributes to phenotype maintenance in the mature retina and pineal gland and may facilitate 24-h changes in the pineal transcriptome.


Subject(s)
Homeodomain Proteins/biosynthesis , Pineal Gland/growth & development , Pineal Gland/metabolism , Retina/growth & development , Retina/metabolism , Aging/physiology , Animals , Blotting, Western , Circadian Rhythm/physiology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , In Situ Hybridization , Male , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley
9.
J Neurochem ; 119(2): 262-74, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21797868

ABSTRACT

Cone-rod homeobox (Crx) encodes Crx, a transcription factor expressed selectively in retinal photoreceptors and pinealocytes, the major cell type of the pineal gland. In this study, the influence of Crx on the mammalian pineal gland was studied by light and electron microscopy and by use of microarray and qRTPCR technology, thereby extending previous studies on selected genes (Furukawa et al. 1999). Deletion of Crx was not found to alter pineal morphology, but was found to broadly modulate the mouse pineal transcriptome, characterized by a>2-fold down-regulation of 543 genes and a>2-fold up-regulation of 745 genes (p<0.05). Of these, one of the most highly up-regulated (18-fold) was Hoxc4, a member of the Hox gene family, members of which are known to control gene expression cascades. During a 24-h period, a set of 51 genes exhibited differential day/night expression in pineal glands of wild-type animals; only eight of these were also day/night expressed in the Crx⁻/⁻ pineal gland. However, in the Crx⁻/⁻ pineal gland 41 genes exhibited differential night/day expression that was not seen in wild-type animals. These findings indicate that Crx broadly modulates the pineal transcriptome and also influences differential night/day gene expression in this tissue. Some effects of Crx deletion on the pineal transcriptome might be mediated by Hoxc4 up-regulation.


Subject(s)
Gene Expression Profiling , Homeodomain Proteins/genetics , Pineal Gland/physiology , Trans-Activators/genetics , Animals , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Cloning, Molecular , Computational Biology , Data Interpretation, Statistical , Down-Regulation , Gene Deletion , Mice , Mice, Knockout , Microarray Analysis , Microscopy, Electron, Transmission , Pineal Gland/anatomy & histology , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic/genetics
10.
Biomed Res Int ; 2014: 946075, 2014.
Article in English | MEDLINE | ID: mdl-24877149

ABSTRACT

Nocturnal synthesis of melatonin in the pineal gland is controlled by a circadian rhythm in arylalkylamine N-acetyltransferase (AANAT) enzyme activity. In the rodent, Aanat gene expression displays a marked circadian rhythm; release of norepinephrine in the gland at night causes a cAMP-based induction of Aanat transcription. However, additional transcriptional control mechanisms exist. Homeobox genes, which are generally known to encode transcription factors controlling developmental processes, are also expressed in the mature rodent pineal gland. Among these, the cone-rod homeobox (CRX) transcription factor is believed to control pineal-specific Aanat expression. Based on recent advances in our understanding of Crx in the rodent pineal gland, we here suggest that homeobox genes play a role in adult pineal physiology both by ensuring pineal-specific Aanat expression and by facilitating cAMP response element-based circadian melatonin production.


Subject(s)
Arylalkylamine N-Acetyltransferase/biosynthesis , Circadian Rhythm/physiology , Gene Expression Regulation, Enzymologic/physiology , Homeodomain Proteins/metabolism , Melatonin/biosynthesis , Pineal Gland/metabolism , Trans-Activators/metabolism , Animals , Arylalkylamine N-Acetyltransferase/genetics , Cyclic AMP/genetics , Cyclic AMP/metabolism , Homeodomain Proteins/genetics , Melatonin/genetics , Mice , Rats , Response Elements/physiology , Trans-Activators/genetics
11.
Endocrinology ; 155(8): 2966-75, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24877634

ABSTRACT

The cone-rod homeobox (Crx) gene encodes a transcription factor in the retina and pineal gland. Crx deficiency influences the pineal transcriptome, including a reduced expression of arylalkylamine N-acetyltransferase (Aanat), a key enzyme in nocturnal pineal melatonin production. However, previous functional studies on pineal Crx have been performed in melatonin-deficient mice. In this study, we have investigated the role of Crx in the melatonin-proficient rat pineal gland. The current study shows that pineal Crx transcript levels exhibit a circadian rhythm with a peak in the middle of the night, which is transferred into daily changes in CRX protein. The study further shows that the sympathetic innervation of the pineal gland controls the Crx rhythm. By use of adenovirus-mediated short hairpin RNA gene knockdown targeting Crx mRNA in primary rat pinealocyte cell culture, we here show that intact levels of Crx mRNA are required to obtain high levels of Aanat expression, whereas overexpression of Crx induces Aanat transcription in vitro. This regulatory function of Crx is further supported by circadian analysis of Aanat in the pineal gland of the Crx-knockout mouse. Our data indicate that the rhythmic nature of pineal CRX protein may directly modulate the daily profile of Aanat expression by inducing nighttime expression of this enzyme, thus facilitating nocturnal melatonin synthesis in addition to its role in ensuring a correct tissue distribution of Aanat expression.


Subject(s)
Arylalkylamine N-Acetyltransferase/metabolism , Circadian Rhythm , Homeodomain Proteins/metabolism , Pineal Gland/metabolism , Trans-Activators/metabolism , Animals , Cells, Cultured , Male , Melatonin/biosynthesis , Mice , Mice, Knockout , Otx Transcription Factors/metabolism , Pineal Gland/enzymology , Pineal Gland/innervation , Rats , Rats, Sprague-Dawley
12.
Brain Struct Funct ; 218(2): 551-62, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22527123

ABSTRACT

The circadian master clock of the mammalian brain resides in the suprachiasmatic nucleus (SCN) of the hypothalamus. At the molecular level, the clock of the SCN is driven by a transcriptional/posttranslational autoregulatory network with clock gene products as core elements. Recent investigations have shown the presence of peripheral clocks in extra-hypothalamic areas of the central nervous system. However, knowledge on the clock gene network in the cerebral cortex is limited. We here show that the mammalian clock genes Per1, Per2, Per3, Cry1, Cry2, Bmal1, Clock, Nr1d1 and Dbp are expressed in the rat neocortex. Among these, Per1, Per2, Per3, Cry1, Bmal1, Nr1d1 and Dbp were found to exhibit daily rhythms. The amplitude of circadian oscillation in neocortical clock gene expression was damped and the peak delayed as compared with the SCN. Lesions of the SCN revealed that rhythmic clock gene expression in the neocortex is dependent on the SCN. In situ hybridization and immunohistochemistry showed that products of the canonical clock gene Per2 are located in perikarya throughout all areas of the neocortex. These findings show that local circadian oscillators driven by the SCN reside within neurons of the neocortex.


Subject(s)
Circadian Rhythm Signaling Peptides and Proteins/metabolism , Circadian Rhythm , Neocortex/metabolism , Neurons/metabolism , Animals , Circadian Rhythm Signaling Peptides and Proteins/genetics , Gene Expression Regulation , Immunohistochemistry , In Situ Hybridization , Male , Neocortex/cytology , Neural Pathways/cytology , Neural Pathways/metabolism , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/metabolism , Time Factors
13.
Chronobiol Int ; 29(10): 1289-99, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23131067

ABSTRACT

The central circadian clock of the mammalian brain resides in the suprachiasmatic nucleus (SCN) of the hypothalamus. At the molecular level, the circadian clockwork of the SCN constitutes a self-sustained autoregulatory feedback mechanism reflected by the rhythmic expression of clock genes. However, recent studies have shown the presence of extrahypothalamic oscillators in other areas of the brain including the cerebellum. In the present study, the authors unravel the cerebellar molecular clock by analyzing clock gene expression in the cerebellum of the rat by use of radiochemical in situ hybridization and quantitative real-time polymerase chain reaction. The authors here show that all core clock genes, i.e., Per1, Per2, Per3, Cry1, Cry2, Clock, Arntl, and Nr1d1, as well as the clock-controlled gene Dbp, are expressed in the granular and Purkinje cell layers of the cerebellar cortex. Among these genes, Per1, Per2, Per3, Cry1, Arntl, Nr1d1, and Dbp were found to exhibit circadian rhythms in a sequential temporal manner similar to that of the SCN, but with several hours of delay. The results of lesion studies indicate that the molecular oscillatory profiles of Per1, Per2, and Cry1 in the cerebellum are controlled, though possibly indirectly, by the central clock of the SCN. These data support the presence of a circadian oscillator in the cortex of the rat cerebellum.


Subject(s)
Cerebellum/metabolism , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Circadian Rhythm , Suprachiasmatic Nucleus/metabolism , Animals , Cerebellum/pathology , Circadian Rhythm Signaling Peptides and Proteins/genetics , Cryptochromes/genetics , Male , RNA, Messenger/physiology , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction/methods , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL