Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters










Publication year range
1.
Mol Neurodegener ; 10: 27, 2015 Jul 11.
Article in English | MEDLINE | ID: mdl-26159703

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) underlies dementia for millions of people worldwide, and its occurrence is set to double in the next 20 years. Currently, approved drugs for treating AD only marginally ameliorate cognitive deficits, and provide limited symptomatic relief, while newer substances under therapeutic development are potentially years away from benefiting patients. Melatonin (MEL) for insomnia has been proven safe with >15 years of over-the-counter access in the US. MEL exerts multiple complementary mechanisms of action against AD in animal models; thus it may be an excellent disease-modifying therapeutic. While presumed to provide neuroprotection via activation of known G-protein-coupled melatonin receptors (MTNRs), some data indicate MEL acts intracellularly to protect mitochondria and neurons by scavenging reactive oxygen species and reducing free radical formation. We examined whether genetic deletion of MTNRs abolishes MEL's neuroprotective actions in the AßPP(swe)/PSEN1dE9 mouse model of AD (2xAD). Beginning at 4 months of age, both AD and control mice either with or without both MTNRs were administered either MEL or vehicle in drinking water for 12 months. RESULTS: Behavioral and cognitive assessments of 15-month-old AD mice revealed receptor-dependent effects of MEL on spatial learning and memory (Barnes maze, Morris Water Maze), but receptor-independent neuroprotective actions of MEL on non-spatial cognitive performance (Novel Object Recognition Test). Similarly, amyloid plaque loads in hippocampus and frontal cortex, as well as plasma Aß1-42 levels, were significantly reduced by MEL in a receptor-independent manner, in contrast to MEL's efficacy in reducing cortical antioxidant gene expression (Catalase, SOD1, Glutathione Peroxidase-1, Nrf2) only when receptors were present. Increased cytochrome c oxidase activity was seen in 16 mo AD mice as compared to non-AD control mice. This increase was completely prevented by MEL treatment of 2xAD/MTNR+ mice, but only partially prevented in 2xAD/MTNR- mice, consistent with mixed receptor-dependent and independent effects of MEL on this measure of mitochondrial function. CONCLUSIONS: These findings demonstrate that prophylactic MEL significantly reduces AD neuropathology and associated cognitive deficits in a manner that is independent of antioxidant pathways. Future identification of direct molecular targets for MEL action in the brain should open new vistas for development of better AD therapeutics.


Subject(s)
Alzheimer Disease/drug therapy , Melatonin/therapeutic use , Neuroprotective Agents/therapeutic use , Receptors, Melatonin/physiology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Amyloid beta-Peptides/blood , Amyloid beta-Protein Precursor/genetics , Animals , Anxiety/genetics , Disease Models, Animal , Drug Evaluation, Preclinical , Exploratory Behavior , Frontal Lobe/metabolism , Frontal Lobe/pathology , Glutathione Peroxidase/biosynthesis , Glutathione Peroxidase/genetics , Hippocampus/metabolism , Hippocampus/pathology , Maze Learning , Memory Disorders/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/physiology , Mutation , NF-E2-Related Factor 2/biosynthesis , NF-E2-Related Factor 2/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Peptide Fragments/blood , Plaque, Amyloid/pathology , Presenilin-1/genetics , Receptors, Melatonin/deficiency , Receptors, Melatonin/genetics , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Glutathione Peroxidase GPX1
2.
Neuroscience ; 277: 506-21, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25046530

ABSTRACT

Melatonin, an indoleamine hormone secreted into circulation at night primarily by the brain's pineal gland, has been shown to have a wide variety of actions on the development and physiology of neurons in the CNS. Acting via two G-protein-coupled membrane receptors (MT1 and MT2), melatonin modulates neurogenesis, synaptic functions, neuronal cytoskeleton and gene expression. In the present studies, we sought to characterize the behavior and neuronal biology of transgenic mice lacking both of these melatonin receptors as a way to understand the hormone's receptor versus non-receptor-mediated actions in CNS-dependent activities, such as learning and memory, anxiety, general motor performance and circadian rhythmicity. Assessment of these behaviors was complemented by molecular analyses of gene expression in the brain. Our results demonstrate mild behavioral hyperactivity and a lengthened circadian period of free-running motor activity in melatonin receptor-deficient mice as compared to receptor-intact control mice beginning at an early age. Significant improvement in cognitive performance was found using the Barnes Maze and the Y-Maze. No behavioral changes in anxiety levels were found. Electrophysiological measures in hippocampal slices revealed a clear enhancement of long-term potentiation in mice lacking melatonin receptors with no significant differences in paired-pulse facilitation. Quantitative analysis of brain protein expression levels of phosphoCREB and phosphoERK1/2 and key markers of synaptic activity (synapsin, glutamate receptor 1, spinophilin, and glutamic acid decarboxylase 1) revealed significant differences between the double-knockout and wild-type animals, consistent with the behavioral findings. Thus, genetic deletion of melatonin receptors produces mice with enhanced cognitive and motor performance, supporting the view that these receptors play an important role in neurobehavioral development.


Subject(s)
Frontal Lobe/physiology , Hippocampus/physiology , Long-Term Potentiation/physiology , Maze Learning/physiology , Receptor, Melatonin, MT1/deficiency , Receptor, Melatonin, MT2/deficiency , Animals , Anxiety/physiopathology , CREB-Binding Protein/metabolism , Circadian Rhythm/physiology , Drinking/physiology , Locomotion/physiology , MAP Kinase Signaling System/physiology , Male , Melatonin/blood , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Random Allocation , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT2/genetics
3.
Neuroscience ; 261: 74-84, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24365461

ABSTRACT

Melatonin is a neurohormone associated with circadian rhythms. A diurnal rhythm in olfactory sensitivity has been previously reported and melatonin receptor mRNAs have been observed in the olfactory bulb, but the effects of melatonin in the olfactory bulb have not been explored. First, we corroborated data from a previous study that identified melatonin receptor messenger RNAs in the olfactory bulb. We then investigated whether melatonin treatment would affect cells in the olfactory bulbs of rats. Using a combination of polymerase chain reaction (PCR), quantitative PCR (qPCR), cell culture, and electrophysiology, we discovered that melatonin receptors and melatonin synthesis enzymes were present in the olfactory bulb and we observed changes in connexin43 protein, GluR1 mRNA, GluR2 mRNA, Per1 mRNA, Cry2 mRNA, and K(+) currents in response to 2-iodomelatonin. Via qPCR, we observed that messenger RNAs encoding melatonin receptors and melatonin biosynthesis enzymes fluctuated in the olfactory bulb across 24h. Together, these data show that melatonin receptors are present in the olfactory bulb and likely affect olfactory function. Additionally, these data suggest that melatonin may be locally synthesized in the olfactory bulb.


Subject(s)
Melatonin/metabolism , Olfactory Bulb/metabolism , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/metabolism , Animals , Astrocytes/metabolism , Cells, Cultured , Circadian Rhythm , Female , In Vitro Techniques , Male , Mice , Mice, Knockout , Neurons/metabolism , Olfactory Bulb/enzymology , Potassium/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT2/genetics
4.
Mol Cell Endocrinol ; 311(1-2): 120-5, 2009 Nov 13.
Article in English | MEDLINE | ID: mdl-19616057

ABSTRACT

We reported earlier that gonadotropin-releasing hormone (GnRH) activates period1 (mPer1) gene expression in immortalized gonadotropes through protein kinase C and p42/44 mitogen-activated protein kinase pathways. GnRH stimulation also leads to the upregulation of early growth response protein 1 (EGR-1), a critical transcription factor for GnRH-induced luteinizing hormone beta (LHbeta) synthesis. The parallels between the GnRH-LHbeta and the GnRH-mPer1 pathways led us to explore whether EGR-1 is involved in the regulation of mPer1 expression in gonadotropes. Of particular interest was the presence of an EGR-1 binding site in the proximal promoter of the mPer1 gene. Stimulation of LbetaT2 gonadotrope cells with a GnRH agonist caused the rapid induction of Egr-1 mRNA, which was rapidly followed by mPer1 expression. Chromatin immunoprecipitation revealed that the mPer1 promoter can bind EGR-1, while site-directed mutagenesis experiments confirmed the involvement of Egr-1 sequences in maintaining basal and allowing GnRH-stimulated mPer1 transcription. By means of RNA interference experiments, it could also be demonstrated that silencing of Egr-1 expression resulted in markedly lower mPer1 transcript levels. This silencing effect of the Egr-1 siRNA could be rescued by transfecting the cells with an EGR-1 overexpression vector. In summary, these results all point to a role for the EGR-1 protein in transactivating both the LHbeta as well as the mPer1 gene in pituitary gonadotrope cells.


Subject(s)
Early Growth Response Protein 1/metabolism , Gonadotrophs/drug effects , Gonadotrophs/metabolism , Gonadotropin-Releasing Hormone/pharmacology , Period Circadian Proteins/metabolism , Animals , Buserelin/pharmacology , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Chromatin Immunoprecipitation , Early Growth Response Protein 1/genetics , Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Gene Silencing/drug effects , Genes, Reporter , Gonadotrophs/cytology , Gonadotrophs/enzymology , Gonadotropin-Releasing Hormone/agonists , Luciferases/metabolism , Mice , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mutagenesis, Site-Directed , Period Circadian Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Time Factors
5.
Mol Cell Endocrinol ; 278(1-2): 36-43, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17928134

ABSTRACT

The pituitary gland's ability to respond to the hypothalamic hormone GnRH (gonadotropin-releasing hormone) depends directly on the gonadotrope-specific expression of the GnRH receptor (GnRHR), a G-protein coupled transmembrane protein coded by the GnRHR gene. In the present study, we have investigated the potential regulatory role of seven noncanonical E-box enhancer sequences within the 856bp proximal 5'-flanking region of the mGnRHR gene in regulating transcription. These sequences are known to mediate the action of clock gene proteins on the expression of a diverse array of genes both central and peripheral. In the present studies the expression of all of the cognate clock genes was identified in the alphaT3-1 gonadotrope cell line. Additionally, luteinizing hormone-immunoreactive cells in the adult rodent pituitary gland were also shown to co-express the PERIOD-1 protein. By means of chromatin immunoprecipitation of alphaT3-1 nuclear extracts we were able to capture promoter fragments of the GnRHR and Period-1 genes, indicating that E-boxes in these promoters bind the CLOCK protein. RNA interference experiments with alphaT3-1 cells in which Bmal1 expression was attenuated also confirmed the involvement of E-boxes in transcriptional regulation of the mGnRHR gene. Subsequent luciferase reporter assay experiments with GnRHR constructs possessing intact or mutated E-boxes confirmed the use of these sequences for the regulation of mGnRH-R/luc expression. Transient overexpression of the dominant negative E-box-binding factor CLOCK-Delta19, or the inhibitory clock protein mPER1, markedly reduced CLOCK/BMAL1-driven mGnRH-R/luc expression in a dose-dependent fashion. Our data implicate the clock genes as important factors controlling GnRHR expression in murine gonadotrope cells.


Subject(s)
E-Box Elements/physiology , Gene Expression Regulation , Gonadotrophs/metabolism , Receptors, LHRH/genetics , Trans-Activators/metabolism , ARNTL Transcription Factors , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , CLOCK Proteins , Cell Line , Chromatin Immunoprecipitation , E-Box Elements/genetics , Eye Proteins/genetics , Eye Proteins/metabolism , Genes, Reporter , Luciferases/genetics , Mice , Period Circadian Proteins , RNA Interference , Receptors, LHRH/antagonists & inhibitors , Sequence Deletion , Trans-Activators/genetics
6.
J Neuroendocrinol ; 18(4): 279-89, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16503923

ABSTRACT

Circadian rhythms of behaviour and gene expression are coupled to endogenous neuronal oscillators located in the hypothalamic suprachiasmatic nuclei (SCN), which are synchronised by the environmental light cycle. Besides light, other factors such as the pineal hormone melatonin, temperature and feeding have entraining properties. During senescence, the circadian system becomes weaker and susceptible to desynchronisation. It is unknown to what extent age-related changes are the result of the deterioration of the hypothalamic master clock. Supplementing ageing mice with melatonin as well as maintaining them on a hypocaloric diet extends the life span and delays age-related diseases. By means of DNA microarrays and the quantitative polymerase chain reaction, we have conducted an exploratory study to compare the effect of long-term melatonin substitution (MEL) and caloric restriction (CR) on circadian gene expression in hypothalamic samples, which contained the SCN as well as other important nuclei involved in nutrient balance, reproduction, and so on. Over 4% of the hypothalamic transcripts showed an overt circadian rhythm in expression, and many of these contain E boxes in their promoter regions, suggesting a direct regulation by circadian clock genes. MEL and CR significantly influenced some of these rhythmically expressed transcripts, but often in opposite ways. Importantly, our studies emphasise that the apparent direction of treatment effects (i.e. up-regulation versus down-regulation) depends on the time of day at which the samples are compared.


Subject(s)
Aging/physiology , Caloric Restriction , Circadian Rhythm/genetics , Hypothalamus/metabolism , Melatonin/physiology , Animals , Biological Clocks/genetics , Biological Clocks/physiology , E-Box Elements/physiology , Gene Expression Profiling , Gene Expression Regulation , Male , Mice , Mice, Inbred Strains , Oligonucleotide Array Sequence Analysis
7.
Neuroreport ; 14(4): 613-8, 2003 Mar 24.
Article in English | MEDLINE | ID: mdl-12657897

ABSTRACT

Hypothalamic GnRH (gonadotropin-releasing hormone) neurons play a critical role in the initiation and maintenance of reproduction competence. Using the mouse GnRH neuronal cell line, GT1-7, we have characterized the expression of the gene mPer1, a recognized key element of the mammalian circadian clockwork. Both mPer1 transcripts and the 136 kDa mPER1 gene product could be detected in these cells. Immunocytochemical analysis also confirmed expression of mPER1 both in vitro and in vivo in GnRH neurons. Activation of cyclic AMP signalling pathways in vitro elevated GnRH secretion as well as mPer1 expression and nuclear mPER1 immunoreactivity. As mPER1 is known to feedback on transcriptional activities in many cell models, the data presented here point to a role for mPER1 in the regulation of gene expression in GnRH neurons, and thus in the control of neuroendocrine activities.


Subject(s)
Gene Expression Regulation/physiology , Gonadotropin-Releasing Hormone/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , Animals , Cell Cycle Proteins , Cells, Cultured , Colforsin/pharmacology , Gonadotropin-Releasing Hormone/analysis , Immunoblotting/methods , Immunohistochemistry/methods , Mice , Neuroprotective Agents/pharmacology , Nuclear Proteins/genetics , Period Circadian Proteins , Preoptic Area/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Vasoactive Intestinal Peptide/pharmacology
8.
J Clin Endocrinol Metab ; 88(2): 908-13, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12574232

ABSTRACT

The circadian timing of spontaneous human deliveries results in births occurring statistically more often during the nocturnal phase of the 24-h cycle. The neuroendocrine mechanisms underlying this physiological phenomenon are not understood. In an effort to test the hypothesis that melatonin may serve as an endocrine signal for coordinating myometrial events in the human, we determined the mRNA expression of both MT1 and MT2 melatonin receptor isoforms in pregnant as well as nonpregnant myometrial biopsies by means of RT-PCR and in situ hybridization histochemistry. Additionally, we could demonstrate specific, high affinity iodomelatonin binding to myometrial tissues of both pregnant and nonpregnant women. Primary cultures of myocytes responded differentially from melatonin in terms of cAMP signaling depending on the reproductive state. These results imply that melatonin may have the potential to modulate myometrial function in the human, a finding that could open up new possibilities for the development of novel therapeutic agents.


Subject(s)
Melatonin/pharmacokinetics , Myometrium/physiology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Adult , Cells, Cultured , Female , Gene Expression/physiology , Humans , Iodine Radioisotopes , Middle Aged , Myometrium/cytology , Pregnancy , RNA, Messenger/analysis , Radioligand Assay , Receptors, Melatonin
9.
J Neuroendocrinol ; 14(1): 14-8, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11903808

ABSTRACT

The role of cGMP in the avian pineal is not well understood. Although the light-sensitive secretion of melatonin is a well-known output of the circadian oscillator, pharmacologically elevated cGMP levels do not result in altered melatonin secretory amplitude or phase. This suggests that pineal cGMP signalling does not couple the endogenous circadian oscillator to the expression of melatonin rhythms. Nonetheless, the free-running rhythm of cGMP signalling implies a link to the circadian oscillator in chick pinealocytes. As the circadian rhythm of cGMP levels in vitro is not altered by pharmacological inhibition of phosphodiesterase activity, we infer that the synthesis, rather than the degradation of cGMP, is under circadian control. In vitro experiments with the nitric oxide synthase (NOS) inhibitor NG-nitro-L-arginine as well as with an inhibitor of the NO-sensitive soluble guanylyl cyclase (sGC), showed that the NOS-sGC pathway does not play a major role in the circadian control of cGMP generation. In organ culture experiments, we demonstrated that C-type natriuretic peptide (CNP), but not atrial natriuretic peptide (ANP), elevated daytime levels of cGMP. As CNP acts on the membrane guanylyl cyclase isoform B (GC-B), which is expressed at very high levels in mammalian pineals, we investigated the effect of the membrane GC-specific inhibitor HS-142 on chick pineal cGMP levels. CNP-induced daytime cGMP levels were reduced by HS-142. More importantly, 'spontaneously' high nocturnal levels of cGMP in vitro were reduced to daytime levels by acute addition of HS-142. These data implicate endogenous nocturnal CNP release and subsequent activation of GC-B as the major input driving cGMP synthesis in the chick pineal organ.


Subject(s)
Chickens/physiology , Cyclic GMP/metabolism , Guanylate Cyclase/metabolism , Pineal Gland/enzymology , Pineal Gland/metabolism , Animals , Circadian Rhythm/physiology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Male , Membranes/enzymology , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/metabolism , Organ Culture Techniques , Signal Transduction/drug effects , Signal Transduction/physiology
10.
Brain Res Mol Brain Res ; 75(2): 321-9, 2000 Feb 22.
Article in English | MEDLINE | ID: mdl-10686354

ABSTRACT

The generation and function(s) of the signalling molecule cyclic GMP (cGMP) in brain are still poorly understood. One mechanism to raise intracellular cGMP levels is binding of C-type natriuretic peptide (CNP) to a membrane guanylyl cyclase (GC), termed GC-B. Here, we demonstrate an exceptionally strong expression of GC-B in the pineal gland. Crosslinking experiments performed with 125I-Tyr(0)-CNP and membranes from various rat tissues identified the receptor as a 130-kDa protein, expressed at highest levels in pineal membranes. Receptor autoradiography on brain sections confirmed a striking density of CNP binding sites in pineal tissue, whereas binding sites for the related atrial natriuretic peptide (ANP) predominate in other regions of the brain. Incubations of freshly dissected whole pineal glands in either the absence or presence of natriuretic peptides followed by immunohistochemical analyses of cGMP revealed strong accumulations of cGMP in response to CNP but not to ANP in the majority of pinealocytes. Stimulation of soluble GC (sGC) activity by use of sodium nitroprusside (SNP) resulted in a very similar pattern of cGMP immunostaining, indicating a co-expression at high levels of particulate and soluble forms of GC. These findings point to a major role of cGMP signalling in pinealocytes and suggest an important regulatory function for CNP.


Subject(s)
Guanylate Cyclase/biosynthesis , Natriuretic Peptide, C-Type/biosynthesis , Pineal Gland/metabolism , Receptors, Atrial Natriuretic Factor/biosynthesis , Animals , Cyclic GMP/metabolism , Guanylate Cyclase/metabolism , In Vitro Techniques , Male , Pineal Gland/enzymology , Rats , Rats, Wistar , Receptors, Enterotoxin , Receptors, Guanylate Cyclase-Coupled , Receptors, Peptide/metabolism
11.
J Pineal Res ; 27(3): 170-82, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10535767

ABSTRACT

Phosphorylation of cyclic AMP response element binding protein (CREB) at amino acid serine 133 appears as an important link between the norepinephrine (NE)-induced activation of second messenger systems and the stimulation of melatonin biosynthesis. Here we investigated in the rat pineal gland: 1) the type of protein kinase that mediates CREB phosphorylation: and 2) its impact on melatonin biosynthesis. Immunochemical or immunocytochemical demonstration of serine133-phosphorylated cyclic AMP regulated element binding protein (pCREB) and radioimmunological detection of melatonin revealed that only cyclic AMP-dependent protein kinase (PKA) inhibitors suppressed NE-induced CREB phosphorylation and stimulation of melatonin biosynthesis, whereas inhibitors of cyclic GMP-dependent protein kinase (PKG), mitogen-activated protein kinase kinase, protein kinase C, or calcium-calmodulin-dependent protein kinase (CaMK) were ineffective. Investigations with cyclic AMP-agonist pairs that selectively activate either PKA type I or II link NE-induced CREB phosphorylation and stimulation of melatonin biosynthesis to the activation of PKA type II. Our data suggest that PKA type II plays an important role in the transcriptional control of melatonin biosynthesis in the rat pineal organ.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Melatonin/biosynthesis , Pineal Gland/metabolism , Animals , Cell Nucleus/enzymology , Cyclic AMP Response Element-Binding Protein/analysis , Cyclic AMP-Dependent Protein Kinase Type II , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Immunohistochemistry , Male , Norepinephrine/pharmacology , Phosphorylation , Phosphoserine/analysis , Phosphoserine/metabolism , Pineal Gland/chemistry , Pineal Gland/drug effects , Protein Kinase Inhibitors , Rats , Rats, Wistar
12.
J Neurosci ; 19(9): 3326-36, 1999 May 01.
Article in English | MEDLINE | ID: mdl-10212292

ABSTRACT

Neurotransmitter-driven activation of transcription factors is important for control of neuronal and neuroendocrine functions. We show with an in vivo approach that the norepinephrine cAMP-dependent rhythmic hormone production in rat pineal gland is accompanied by a temporally regulated switch in the ratio of a transcriptional activator, phosphorylated cAMP-responsive element-binding protein (pCREB), and a transcriptional inhibitor, inducible cAMP early repressor (ICER). pCREB accumulates endogenously at the beginning of the dark period and declines during the second half of the night. Concomitant with this decline, the amount of ICER rises. The changing ratio between pCREB and ICER shapes the in vivo dynamics in mRNA and, thus, protein levels of arylalkylamine-N-acetyltransferase, the rate-limiting enzyme of melatonin synthesis. Consequently, a silenced ICER expression in pinealocytes leads to a disinhibited arylalkylamine-N-acetyltransferase transcription and a primarily enhanced melatonin synthesis.


Subject(s)
Arylamine N-Acetyltransferase/genetics , Brain/metabolism , Circadian Rhythm , Cyclic AMP Response Element-Binding Protein/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Melatonin/biosynthesis , Pineal Gland/metabolism , Animals , Arylamine N-Acetyltransferase/metabolism , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP Response Element Modulator , Cyclic AMP Response Element-Binding Protein/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation/drug effects , Male , Norepinephrine/pharmacology , Organ Culture Techniques , Phosphorylation , Pineal Gland/cytology , Pineal Gland/drug effects , RNA, Messenger/genetics , Rats , Rats, Wistar , Recombinant Proteins/biosynthesis , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transfection
13.
Brain Res ; 825(1-2): 95-103, 1999 Apr 17.
Article in English | MEDLINE | ID: mdl-10216177

ABSTRACT

Genetically mutant anophthalmic rats lacking a complete visual system due to the absence of eyeballs and optic nerves up to the optic chiasma were used as a model to study photo-regulated physiological activities. The photoreception in these mutant rats was determined by measuring the neuroendocrine response of the pineal gland-melatonin levels in the serum, and synaptic ribbon complexes (SRCs) in the pinealocytes. These parameters were studied in both normal and anophthalmic rats maintained under light-dark (LD 12:12), continuous dark (DD) and light (LL) conditions. Both normal and mutant anophthalmic animals showed nocturnal increases in serum melatonin levels and in the number and diameter of SRC and their vesicles in the pinealocytes in LD. The daily rhythms persisted even upon transfer to DD both in normal and mutant rats, whereas in LL, the nocturnal elevation of both the parameters disappeared. These observations suggested that congenitally blind rats can perceive light. The studies of these parameters in both normal and mutant rats in reversed-LD conditions confirmed that pineal rhythms can be entrained by light-dark cycles in congenitally anophthalmic mutant rats through a nonvisual system for light perception.


Subject(s)
Blindness/physiopathology , Circadian Rhythm/physiology , Melatonin/blood , Pineal Gland/physiology , Animals , Blindness/genetics , Darkness , Eye Enucleation , Lighting , Male , Motor Activity/physiology , Rats , Rats, Mutant Strains , Synapses/physiology
14.
J Histochem Cytochem ; 47(3): 411-20, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10026243

ABSTRACT

Knowledge about intracellular signal transduction cascades is largely based on investigations of cultured cells whose responses to different stimuli are typically quantified via RIA, ELISA, or immunoblots. These techniques, which require relatively large amounts of biological material, are performed with homogenized cells and therefore do not allow localization of the molecules under investigation. We describe a protocol for recording dose-response curves directly from immunocytochemical preparations using rat pinealocytes as a model system. The cells were exposed to beta-adrenergic stimuli inducing the phosphorylation of the transcription factor CREB (mediated by PKA), an increase in ICER protein levels, and synthesis and release of melatonin. Melatonin concentrations were determined by ELISA. cPKA, phosphorylated CREB, and ICER were demonstrated by immunocytochemistry and immunoblots. Dose-response curves were recorded by measuring the integrated density of the immunoreactive sites with an image analysis program. Dose-response curves from immunoblots and immunocytochemical preparations showed almost identical dynamics, validating the immunocytochemical approach, which minimizes the amount of biological material needed for such studies, allows combined quantification and localization of biomolecules, and may even be more sensitive than immunoblotting.


Subject(s)
Immunohistochemistry/methods , Pineal Gland/metabolism , Repressor Proteins , Signal Transduction , Animals , Cells, Cultured , Cyclic AMP/antagonists & inhibitors , Cyclic AMP Response Element Modulator , Cyclic AMP Response Element-Binding Protein/metabolism , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Image Processing, Computer-Assisted , Immunoblotting , Male , Melatonin/biosynthesis , Phosphorylation , Rats , Rats, Wistar , Reproducibility of Results
15.
Adv Exp Med Biol ; 460: 95-107, 1999.
Article in English | MEDLINE | ID: mdl-10810506

ABSTRACT

NPY is considered to play an important role in pineal function, because it is co-stored with the dominant pineal transmitter noradrenaline. However, little evidence from the literature suggests that NPY alone is a strong regulator of melatonin synthesis or secretion and it is therefore more likely that NPY modulates noradrenergic neurotransmission in the rat pineal gland. The purpose of the present studies was to determine the nature and origin of NPYergic inputs to, and the type of specific NPY receptor subtypes in, the rat pineal gland. Gel filtration and immunocytochemistry using region-specific antisera revealed that all proNPY present in intrapineal nerve fibres is cleaved to amidated NPY and a C-terminal flanking peptide of NPY (CPON). The vast majority of NPY content in the pineal gland was found to be of sympathetic origin. Receptor autoradiography showed that only a few NPY specific binding sites were present in the superficial pineal gland. A reverse transcriptase polymerase chain reaction detected sequences of only NPY receptor subtype Y1 and not other NPY receptor subtypes in pineal extracts. These results together with the available literature imply that NPY under certain conditions is co-released with noradrenaline and exerts its actions either presynaptically or on the pinealocyte through a Y1 receptor. The available data indicate that NPY has no effect alone, but acts in concert with noradrenaline. A presynaptic action regulating noradrenaline neurotransmission is also possible. NPY has been reported only to act on melatonin secretion in vitro, and it remains to be established what function NPY plays in the pineal gland in vivo. This paper discuss possible modulatory actions of NPY being a predominant sympathetic transmitter.


Subject(s)
Neuropeptide Y/analysis , Pineal Gland/chemistry , Pineal Gland/cytology , Receptors, Neuropeptide Y/analysis , Animals , Autoradiography , Chromatography, Gel , Immunohistochemistry , Male , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Pineal Gland/metabolism , Rats , Rats, Wistar , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/metabolism , Superior Cervical Ganglion/physiology
16.
Adv Exp Med Biol ; 460: 109-31, 1999.
Article in English | MEDLINE | ID: mdl-10810507

ABSTRACT

The rodent pineal organ transduces a photoneural input into a hormonal output. This photoneuroendocrine transduction leads to highly elevated levels of the hormone melatonin at night-time which serves as a message for darkness. The melatonin rhythm depends on transcriptional, translational and posttranslational regulation of the arylalkylamine-N-acetyltransferase, the key enzyme of melatonin biosynthesis. These regulatory mechanisms are fundamentally linked to two second messenger systems, namely the cAMP- and the Ca(2+)-signal transduction pathways. Our data gained by molecular biology, immunohistochemistry and single-cell imaging demonstrate a time- and substance-specific activation of these signaling pathways and provide a framework for the understanding of the complex signal transduction cascades in the rodent pineal gland which in concert not only regulate the basic profile but also fine-tune the circadian rhythm in melatonin synthesis.


Subject(s)
Cell Membrane/physiology , Cell Nucleus/physiology , Melatonin/biosynthesis , Pineal Gland/physiology , Repressor Proteins , Signal Transduction , Animals , Arylamine N-Acetyltransferase/genetics , Calcium/physiology , Cyclic AMP/physiology , Cyclic AMP Response Element Modulator , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA-Binding Proteins/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics , Male , Mice , Mice, Inbred C3H , Organ Culture Techniques , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-1/genetics , Second Messenger Systems , Transcription, Genetic
18.
Mol Cell Endocrinol ; 144(1-2): 131-8, 1998 Sep 25.
Article in English | MEDLINE | ID: mdl-9863633

ABSTRACT

By differential screening of a rat pineal cDNA library we identified earlier a novel transcript having a 57% nucleotide homology and a 45% amino acid identity with a plant fusca-gene (fus6) to which a corresponding human sequence (gps1) has recently been reported. Expression of this mammalian fusca homologue (mfh) was seen in a variety of mammalian tissues, including kidney, pineal and retina, but it was particularly strong in the testes. Northern blot analysis demonstrated that the rat testicular mfh message increases markedly from day 28 onwards. Additionally, by in situ hybridization, mfh was localized primarily to the seminiferous tubules with a stage-dependent distribution pattern, a result which was confirmed by immunohistochemistry with antibodies raised against a synthetic MFH oligopeptide. Western blotting also revealed strong signals of the expected molecular weight in testicular extracts from several species. In view of its homology to fus6, a plant gene known to be involved in repressing photomorphogenesis in darkness, the conservation of mfh in mammals suggests a potential function for MFH in signaling pathways involved in the regulation of mammalian differentiation and development.


Subject(s)
GTP-Binding Proteins , Gene Expression Regulation, Developmental , Plant Proteins/genetics , Proteins , Repressor Proteins , Testis/metabolism , Animals , Blotting, Northern , Brain/metabolism , COP9 Signalosome Complex , Female , Gene Library , Humans , Immunohistochemistry , In Situ Hybridization , Intracellular Signaling Peptides and Proteins , Male , Molecular Weight , Organ Specificity , Ovary/metabolism , Plant Proteins/metabolism , RNA, Messenger/metabolism , Rats , Seminiferous Tubules/cytology , Seminiferous Tubules/growth & development , Seminiferous Tubules/metabolism , Sequence Homology, Nucleic Acid , Spermatozoa/growth & development , Spermatozoa/metabolism , Testis/growth & development
19.
J Neuroendocrinol ; 9(3): 177-82, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9089468

ABSTRACT

Former studies have indicated an influence of natriuretic peptides on LHRH secretion. In this report we demonstrate local synthesis of CNP in immortalized LHRH neurons (GT1-7 cells). Using reverse transcription-polymerase chain reaction and RNase protection assays a transcript for the CNP precursor was identified in these cells. Immunocytochemical data revealed the presence of the peptide CNP in GT1 cells, using a specific polyclonal antiserum against CNP. Electron microscopic immunohistochemical investigations also showed the strongest CNP-immunoreactivity in some small vesicles, providing initial evidence for the potential secretion of this peptide by immortalized LHRH neurons. Subsequent experiments demonstrated also that CNP elevates LHRH production in static cultures of GT1 cells. These data show for the first time the co-production of the functionally relevant natriuretic peptide, CNP, by immortalized LHRH neurons. Together with the recent demonstration of CNP receptor expression by these cells, we suggest that CNP may represent a novel autocrine regulator of LHRH neuronal activity. It remains to be elucidated, however, to what extent CNP expression in immortalized LHRH neurons reflects a co-localization in situ of CNP and LHRH peptides.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Protein Biosynthesis , Animals , Blotting, Southern , Cell Line, Transformed , Hypothalamus/ultrastructure , Immunohistochemistry , Mice , Microscopy, Immunoelectron , Natriuretic Peptide, C-Type , Polymerase Chain Reaction , RNA-Directed DNA Polymerase
20.
J Neurochem ; 68(2): 770-7, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9003068

ABSTRACT

We investigated the expression of regulatory (R) and catalytic (C) subunits of cyclic AMP-dependent protein kinase (cAK; ATP:protein phosphotransferase; EC 2.7.1.37) in the bovine pineal gland. In total RNA extracts of bovine pineal glands moderate levels of RI alpha/RII beta and high levels of C alpha and C beta mRNA were found. We were able to detect a strong signal for RII and C subunit at the protein level, whereas RI was apparently absent. Probing sections of the intact bovine pineal gland with RI and RII antibodies stained only RII in pinealocytes. Pairs of cyclic AMP analogues complementing each other in activation of type II cAK, but not cAKI-directed analogue pairs, showed synergistic stimulation of melatonin synthesis. Moreover, melatonin synthesis stimulated by the physiological activator norepinephrine in pineal cell cultures was inhibited by cAK antagonists. Taken together these results show the presence of RII regulatory and both C alpha and C beta catalytic subunits and thus cAKII holoenzyme in the bovine pineal gland. The almost complete inhibition of norepinephrine-mediated melatonin synthesis by the cAK antagonists emphasizes the dominant role of cyclic AMP as the second messenger and cAK as the transducer in bovine pineal signal transduction.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Isoenzymes/metabolism , Melatonin/biosynthesis , Pineal Gland/enzymology , Animals , Blotting, Northern , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cattle , Cells, Cultured/drug effects , Cells, Cultured/enzymology , Cyclic AMP/agonists , Cyclic AMP/antagonists & inhibitors , Gene Expression Regulation, Enzymologic/physiology , Immunohistochemistry , Melatonin/genetics , Pineal Gland/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...