Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters











Publication year range
1.
Mol Biol Rep ; 47(6): 4867-4873, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32410138

ABSTRACT

The multiple physiological effects of the indoleamine melatonin, are mediated primarily by its two G protein-coupled MT1 and MT2 receptors. Treatment with histone deacetylase (HDAC) inhibitors, including valproic acid (VPA) and trichostatin A, upregulates melatonin receptors in cultured cells and the rat brain. VPA increases histone H3 acetylation of the MT1 gene promoter in rat C6 glioma cells, indicating that this epigenetic mechanism is involved in upregulation of MT1 expression. Since HDAC inhibitors can alter DNA methylation, the possible involvement of this other epigenetic mechanism, in the regulation of MT1 expression, was examined. RT-qPCR and western blotting studies confirmed that treatment with the DNA demethylating agent, 5-azacytidine (AZA; 10 or 20 µM) for 24 or 48 h, suppressed DNA methyltransferase 1 mRNA and protein expression in C6 cells. Subsequent treatment with AZA (1-25 µM) for 24 h, revealed a significant concentration-dependent upregulation of MT1 mRNA expression. Moreover, a combination of 5 µM AZA plus 3 mM VPA caused a synergistic upregulation of the MT1 receptor, which exceeded the sum of the independent effects of these drugs. These results show that DNA methylation plays a role in the regulation of the MT1 receptor, consistent with the established effects of this major epigenetic mechanism on gene transcription. Combinatorial epigenetic regulation of melatonin receptor expression could provide novel strategies for enhancing the oncostatic, neuroprotective and other therapeutic benefits of this pleiotropic indoleamine and its receptor agonists.


Subject(s)
Azacitidine/pharmacology , Glioma/metabolism , Receptor, Melatonin, MT1/metabolism , Animals , Azacitidine/metabolism , Cell Line , Cell Line, Tumor , Epigenesis, Genetic/drug effects , Gene Expression/drug effects , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Glioma/genetics , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Melatonin/metabolism , Melatonin/pharmacology , RNA, Messenger/metabolism , Rats , Receptor, Melatonin, MT1/genetics , Receptors, Melatonin/metabolism , Transcriptional Activation/drug effects , Valproic Acid/pharmacology
2.
Br J Pharmacol ; 175(16): 3209-3219, 2018 08.
Article in English | MEDLINE | ID: mdl-28967098

ABSTRACT

Melatonin, the primary indoleamine hormone of the mammalian pineal gland, is known to have a plethora of neuroregulatory, neuroprotective and other properties. Melatonergic signalling is mediated by its two GPCRs, MT1 and MT2 , which are widely expressed in the mammalian CNS. Melatonin levels and receptor expression often show a decrease during normal ageing, and this reduction may be accelerated in some disease states. Depleted melatonergic signalling has been associated with neuropsychiatric dysfunction and impairments in cognition, memory, neurogenesis and neurorestorative processes. The anticonvulsant and mood stabilizer, valproic acid (VPA), up-regulates melatonin MT1 and/or MT2 receptor expression in cultured cells and in the rat brain. VPA is known to affect gene expression through several mechanisms, including the modulation of intracellular kinase pathways and transcription factors, as well as the inhibition of histone deacetylase (HDAC) activity. Interestingly, other HDAC inhibitors, such as trichostatin A, which are structurally distinct from VPA, can also up-regulate melatonin receptor expression, unlike a VPA analogue, valpromide, which lacks HDAC inhibitory activity. Moreover, VPA increases histone H3 acetylation along the length of the MT1 gene promoter in rat C6 cells. These findings indicate that an epigenetic mechanism, linked to histone hyperacetylation/chromatin remodelling and associated changes in gene transcription, is involved in the up-regulation of melatonin receptors by VPA. Epigenetic induction of MT1 and/or MT2 receptor expression, in areas where these receptors are lost because of ageing, injury or disease, may be a promising therapeutic avenue for the management of CNS dysfunction and other disorders. LINKED ARTICLES: This article is part of a themed section on Recent Developments in Research of Melatonin and its Potential Therapeutic Applications. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.16/issuetoc.


Subject(s)
Mental Disorders/genetics , Neurodegenerative Diseases/genetics , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT2/genetics , Animals , Epigenesis, Genetic , Gene Expression , Humans , Mental Disorders/metabolism , Neurodegenerative Diseases/metabolism , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/metabolism
3.
Eur Neuropsychopharmacol ; 27(8): 828-832, 2017 08.
Article in English | MEDLINE | ID: mdl-28648552

ABSTRACT

We have reported that the anticonvulsant/mood stabilizer and histone deacetylase (HDAC) inhibitor valproate (VPA) induces expression of melatonin receptors both in vitro and in vivo, but the mechanisms involved were not known. Here we show that pharmacological inhibition of CREB, PKC, PI3K, or GSK3ß signaling pathways, which are known targets for VPA, do not prevent its upregulation of melatonin MT1 receptors in rat C6 glioma cells. M344, an HDAC inhibitor unrelated to VPA, mimics the effects of VPA on MT1 expression, whereas valpromide, a VPA derivative lacking HDAC inhibitory activity, does not. Furthermore, VPA, at a concentration which upregulates the MT1 receptor, induces histone H3 hyperacetylation along the length of the MT1 receptor promoter. These results show that an epigenetic mechanism involving histone acetylation underlies induction of MT1 receptor expression by VPA. Given the neuropsychiatric effects of melatonin coupled with evidence that VPA upregulates melatonin receptors in the rat brain, these findings suggest that the melatonergic system contributes to the psychotropic effects of VPA.


Subject(s)
Anticonvulsants/pharmacology , Epigenesis, Genetic/drug effects , Receptor, Melatonin, MT1/metabolism , Valproic Acid/pharmacology , Animals , CREB-Binding Protein/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Glioma/pathology , Histones/metabolism , Hydroxamic Acids/pharmacology , RNA, Messenger/metabolism , Rats , Receptor, Melatonin, MT1/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Vorinostat
4.
Exp Toxicol Pathol ; 69(2): 109-114, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27986376

ABSTRACT

The pesticide rotenone has been shown to cause systemic inhibition of mitochondrial complex I activity, with consequent degeneration of dopamine neurons along the nigrostriatal pathway, as observed in Parkinson's disease (PD). Recently, intracranial infusion of rotenone was found to increase the protein levels of the Lewy body constituents, α-synuclein and small ubiquitin-related modifier-1(SUMO-1), in the lesioned hemisphere of the mouse brain. These findings are supportive of a mouse model of PD, but information about the dopamine-synthesizing enzyme, tyrosine hydroxylase (TH), an essential marker of dopaminergic status, was not reported. Clarification of this issue is important because an intracranial rotenone mouse model of Parkinson's disease has not been established. Towards this end, the present study examined the effects of intracranial rotenone treatment on TH and α-synuclein immunohistochemistry in addition to forelimb motor function. Mice were unilaterally infused with either vehicle or rotenone (2µg/site) in both the medial forebrain bundle and the substantia nigra. The forelimb asymmetry (cylinder) test indicated a significant decrease in use of the contralateral forelimb in lesioned animals as compared to the sham group. Densitometric analysis revealed a significant depletion of TH immunofluorescence within the ipsilateral striatum and substantia nigra of lesioned animals. Moreover, a significant bilateral increase in α-synuclein immunofluorescence was found in the substantia nigra of lesioned mice, as compared to control animals. These findings indicate that this intracranial rotenone mouse model will be useful for studies of neurodegenerative disorders such as PD.


Subject(s)
Insecticides/toxicity , Rotenone/toxicity , Substantia Nigra/drug effects , Substantia Nigra/pathology , alpha-Synuclein/biosynthesis , Animals , Disease Models, Animal , Immunohistochemistry , Injections, Intraventricular , Insecticides/administration & dosage , Male , Mice , Parkinson Disease/physiopathology , Rotenone/administration & dosage , Up-Regulation
5.
Brain Res ; 1652: 89-96, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27693415

ABSTRACT

The indoleamine hormone melatonin protects dopamine neurons in the rat nigrostriatal pathway following 6-hydroxydopamine lesioning, and an increase in striatal melatonin levels has been detected in this model of Parkinson's disease. Melatonin induces the expression of tyrosine hydroxylase, the rate-limiting enzyme for dopamine synthesis, in the ventral midbrain, where G protein-coupled melatonin receptors are present. Based on the interaction between the melatonergic and dopaminergic systems, we hypothesized that 6-hydroxydopamine-induced degeneration of dopamine neurons would affect the expression of melatonin receptors in the nigrostriatal pathway. Following unilateral injection of 6-hydroxydopamine into the rat striatum or medial forebrain bundle, there was a significant increase in apomorphine-induced contralateral rotations in lesioned animals as compared to sham controls. A loss of tyrosine hydroxylase immunoreactivity and/or immunofluorescence in the striatum and substantia nigra was seen in animals lesioned in either the striatum or medial forebrain bundle, indicating degeneration of dopamine neurons. There were no significant differences in melatonin MT1 receptor protein expression in the striatum or substantia nigra, between intrastriatally lesioned animals and sham controls. In contrast, lesions in the medial forebrain bundle caused a significant increase in MT1 receptor mRNA expression (p<0.03) on the lesioned side of the ventral midbrain, as compared with the contralateral side. Given the presence of MT1 receptors on neurons in the ventral midbrain, these results suggest that a compensatory increase in MT1 transcription occurs to maintain expression of this receptor and neuroprotective melatonergic signaling in the injured brain.


Subject(s)
Corpus Striatum/metabolism , Parkinsonian Disorders/metabolism , Receptor, Melatonin, MT1/metabolism , Substantia Nigra/metabolism , Animals , Apomorphine/pharmacology , Blotting, Western , Corpus Striatum/pathology , Dopamine Agonists/pharmacology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Fluorescent Antibody Technique , Male , Medial Forebrain Bundle , Motor Activity/drug effects , Motor Activity/physiology , Oxidopamine , Parkinsonian Disorders/pathology , Polymerase Chain Reaction , RNA, Messenger/metabolism , Random Allocation , Rats, Sprague-Dawley , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism
6.
Brain Res ; 1633: 115-125, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26740407

ABSTRACT

Parkinson's disease is a major neurodegenerative disorder which primarily involves the loss of dopaminergic neurons in the substantia nigra and related projections in the striatum. The pesticide/neurotoxin, rotenone, has been shown to cause systemic inhibition of mitochondrial complex I activity in nigral dopaminergic neurons, with consequent degeneration of the nigrostriatal pathway, as observed in Parkinson's disease. A novel intrastriatal rotenone model of Parkinson's disease was used to examine the neuroprotective effects of chronic low-dose treatment with the antioxidant indoleamine, melatonin, which can upregulate neurotrophic factors and other protective proteins in the brain. Sham or lesioned rats were treated with either vehicle (0.04% ethanol in drinking water) or melatonin at a dose of 4 µg/mL in drinking water. The right striatum was lesioned by stereotactic injection of rotenone at three sites (4 µg/site) along its rostrocaudal axis. Apomorphine administration to lesioned animals resulted in a significant (p<0.001) increase in ipsilateral rotations, which was suppressed by melatonin. Nine weeks post-surgery, animals were sacrificed by transcardial perfusion. Subsequent immunohistochemical examination revealed a decrease in tyrosine hydroxylase immunoreactivity within the striatum and substantia nigra of rotenone-lesioned animals. Melatonin treatment attenuated the decrease in tyrosine hydroxylase in the striatum and abolished it in the substantia nigra. Stereological cell counts indicated a significant (p<0.05) decrease in dopamine neurons in the substantia nigra of rotenone-lesioned animals, which was confirmed by Nissl staining. Importantly, chronic melatonin treatment blocked the loss of dopamine neurons in rotenone-lesioned animals. These findings strongly support the therapeutic potential of long-term and low-dose melatonin treatment in Parkinson's disease.


Subject(s)
Antioxidants/pharmacology , Corpus Striatum/drug effects , Melatonin/pharmacology , Parkinsonian Disorders/pathology , Substantia Nigra/drug effects , Animals , Disease Models, Animal , Immunohistochemistry , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Rotenone/toxicity , Uncoupling Agents/toxicity
7.
Mol Cell Endocrinol ; 402: 57-63, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25578604

ABSTRACT

We have shown that melatonin induces histone hyperacetylation in vitro and in vivo. To clarify the mechanisms involved, we have now investigated its effects on histone acetylation and signaling pathways in human SH-SY5Y neuroblastoma cells, which express melatonin MT1 receptors. Melatonin caused significant concentration-dependent increases in both histone H3 and H4 acetylation. Blockade of melatonin receptors with luzindole abolished the histone hyperacetylating effect of melatonin, whereas inhibition of MAPK-ERK by PD98059 attenuated but did not block this effect. Melatonin treatment for 24-h decreased the levels of phospho-ERK1/2, but significantly increased Akt phosphorylation and protein expression of the histone acetyltransferase, p300. These findings suggest that the epigenetic effects of melatonin in SH-SY5Y cells are mediated by G protein-coupled MT1 melatonin receptors. Furthermore, upregulation of the histone acetyltransferase/transcriptional co-activator p300, along with phosphorylation of Akt, which is essential for p300 activation, appear to be key mechanisms underlying the epigenetic effects of melatonin.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Histones/metabolism , Melatonin/physiology , Acetylation , Cell Line, Tumor , Chromatin Assembly and Disassembly , Humans , Neuroblastoma , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Melatonin, MT1/antagonists & inhibitors , Receptor, Melatonin, MT1/metabolism , Signal Transduction , Tryptamines/pharmacology , p300-CBP Transcription Factors/metabolism
8.
Neurosci Lett ; 576: 84-7, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24909617

ABSTRACT

We have reported that clinically relevant concentrations of valproic acid (VPA) upregulate the G protein-coupled melatonin MT1 receptor in rat C6 glioma cells, and both MT1 and MT2 receptors in the rat hippocampus. The melatonin MT2 receptor is relatively enriched in the hippocampus, where it is thought to be involved in modulating synaptic plasticity and cognitive function. Importantly, a significant decrease in MT2 expression has been observed in the hippocampus of Alzheimer's patients. Therefore, we examined whether the global upregulation of this receptor (and also the MT1) by VPA, observed in earlier RT-PCR and real time PCR studies, could be localized to more discrete hippocampal regions, which are involved in cognitive function. In situ hybridization of rat brain slices, following chronic VPA treatment (3mg/mL or 4mg/mL in drinking water), revealed a significant upregulation of the MT2 receptor mRNA in the CA1, CA2, CA3 and dentate gyrus (DG) regions of the rat hippocampus. In contrast, the MT1 receptor was not detected in the hippocampus by in situ hybridization. The significant induction of melatonin MT2 receptor expression by VPA in hippocampal regions involved in learning, memory and/or neural stem cell proliferation, suggests that a combinatorial therapeutic strategy involving VPA together with melatonin or other MT2 agonists, would be beneficial in neurodegenerative disorders such as Alzheimer's disease.


Subject(s)
Hippocampus/drug effects , Neuroprotective Agents/pharmacology , Receptor, Melatonin, MT2/metabolism , Valproic Acid/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , CA2 Region, Hippocampal/drug effects , CA2 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/metabolism , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Hippocampus/metabolism , Male , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/genetics , Up-Regulation
9.
Basic Clin Pharmacol Toxicol ; 115(2): 216-21, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24460582

ABSTRACT

Valproic acid (VPA), a short-chain fatty acid, is used clinically as an anticonvulsant and mood stabilizer. Valproic acid also inhibits histone deacetylase activity, which is associated with histone hyperacetylation and changes in gene expression. In this study, we examined the effects of VPA on the expression of selected neurotrophic and differentiation factors in C17.2 neural stem cells. Reverse transcription-polymerase chain reaction analysis revealed a significant increase in conserved dopamine neurotrophic factor (CDNF) and glial cell line-derived neurotrophic factor mRNA expression, after treatment with clinically relevant concentrations of VPA (0.5 or 1.0 mM) for 24 hr. Increases in the protein expression of CDNF and mesencephalic astrocyte-derived neurotrophic factor were also observed, after similar treatment with VPA. In addition, significant increases in the mRNA levels of the early dopaminergic neuron marker, nuclear receptor-related protein 1(Nurr1), and the transcriptional regulator, early growth response protein 1 (Egr1), were observed after treatment with VPA for 24 hr. C17.2 neural stem cells exhibited high basal mRNA expression of brain-derived neurotrophic factor and SRY (sex determining region Y)-box 2 (Sox2), which was not altered by VPA treatment. Western analysis revealed hyperacetylation of histone H3 proteins in C17.2 cells after VPA treatment for 24 hr or 48 hr, suggesting involvement of an epigenetic mechanism in the observed gene induction by this drug. These findings support a role for VPA in modulating neurotrophic and differentiation factor expression, in keeping with its reported neuroprotective and neurodevelopmental effects.


Subject(s)
Antimanic Agents/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Neural Stem Cells/drug effects , Valproic Acid/pharmacology , Animals , Antimanic Agents/administration & dosage , Cell Line , Dose-Response Relationship, Drug , Epigenesis, Genetic , Gene Expression Regulation/drug effects , Glial Cell Line-Derived Neurotrophic Factor/drug effects , Glial Cell Line-Derived Neurotrophic Factor/genetics , Histone Deacetylase Inhibitors/administration & dosage , Mice , Nerve Growth Factors/drug effects , Nerve Growth Factors/genetics , Neural Stem Cells/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Valproic Acid/administration & dosage
10.
Neurosci Lett ; 541: 49-53, 2013 Apr 29.
Article in English | MEDLINE | ID: mdl-23416321

ABSTRACT

We have reported that melatonin induces histone hyperacetylation in mouse neural stem cells, suggesting an epigenetic role for this pleiotropic hormone. To support such a role, it is necessary to demonstrate that melatonin produces similar effects in vivo. Histone acetylation, following chronic treatment with melatonin (4µg/ml in drinking water for 17 days), was examined by western blotting in selected rat brain regions. Melatonin induced significant increases in histone H3 and histone H4 acetylation in the hippocampus. Histone H4 was also hyperacetylated in the striatum, but there were no significant changes in histone H3 acetylation in this brain region. No significant changes in the acetylation of either histone H3 or H4 were observed in the midbrain and cerebellum. An examination of kinase activation, which may be related to these changes, revealed that melatonin treatment increased the levels of phospho-ERK (extracellular signal-regulated kinase) in the hippocampus and striatum, but phospho-Akt (protein kinase B) levels were unchanged. These findings suggest that chromatin remodeling and associated changes in the epigenetic regulation of gene expression underlie the multiple physiological effects of melatonin.


Subject(s)
Brain/drug effects , Histones/metabolism , Melatonin/pharmacology , Acetylation , Animals , Brain/metabolism , Epigenesis, Genetic , Male , Melatonin/physiology , Rats , Rats, Sprague-Dawley
12.
Biomed Pharmacother ; 66(3): 232-6, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22436652

ABSTRACT

Histone deacetylase (HDAC) inhibitors represent a novel class of drugs that selectively induce cell cycle arrest and apoptosis in transformed cells. This study examined, for the first time, the effects of the relatively new HDAC inhibitor, M344 [4-dimethylamino-N-(6-hydroxycarbamoylhexyl)-benzamide], on the proliferation of MCF-7 breast cancer cells. MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assays revealed significant concentration- and time-dependent decreases in MCF-7 cell proliferation following treatment with M344 (1-100µM). In contrast to the significant induction of p21(waf1/cip1) mRNA expression following treatment with M344 (10µM) for 1 or 3 days, there was a significant decrease in p53 mRNA expression, although p53 protein levels were unchanged. Similar treatment with M344 also induced expression of the pro-apoptotic genes, Puma and Bax, together with the morphological features of apoptosis, in MCF-7 cells. The results of this study reinforce previous findings indicating that HDAC inhibitors are an important group of oncostatic drugs, and show that M344 is a potent suppressor of breast cancer cell proliferation.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Female , Humans , Proto-Oncogene Proteins/genetics , Tumor Suppressor Protein p53/genetics , Vorinostat , bcl-2-Associated X Protein/genetics
13.
Int J Neuropsychopharmacol ; 15(9): 1343-50, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22243807

ABSTRACT

We have reported that clinically relevant concentrations of valproic acid (VPA) up-regulate the G-protein-coupled melatonin MT1 receptor in rat C6 glioma cells. To determine whether this effect occurs in vivo, the effects of chronic VPA treatment on the expression of both melatonin receptor subtypes, MT1 and MT2, were examined in the rat brain. Reverse transcription-polymerase chain reaction (RT-PCR) and real-time PCR analyses revealed significant increases in MT1 and MT2 mRNA expression in the hippocampus, following VPA (4 mg/ml drinking water) treatment for 17 d. Increases in the mRNA and protein expression of the novel neurotrophic factors, conserved dopamine neurotrophic factor and mesencephalic astrocyte-derived neurotrophic factor, were detected in the hippocampus and/or striatum. In addition, significant changes in persephin, glial cell line-derived neurotrophic factor and brain-derived neurotrophic factor mRNA expression were observed. The robust multi-fold induction of MT1 and MT2 receptors in the hippocampus suggests a role for the melatonergic system in the psychotropic effects of VPA.


Subject(s)
Anticonvulsants/pharmacology , Nerve Growth Factors/biosynthesis , Receptor, Melatonin, MT1/drug effects , Receptor, Melatonin, MT2/drug effects , Valproic Acid/pharmacology , Animals , Blotting, Western , Body Weight/drug effects , Drinking/drug effects , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Neostriatum/drug effects , Neostriatum/metabolism , Nerve Tissue Proteins/metabolism , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Up-Regulation/drug effects
14.
Eur J Pharmacol ; 589(1-3): 45-8, 2008 Jul 28.
Article in English | MEDLINE | ID: mdl-18550052

ABSTRACT

C6 glioma cells were treated with clinically relevant concentrations of valproic acid (0.5 or 1.0 mM) for 1-7 days and RT-PCR used to examine expression of the melatonin MT(1) receptor and selected epigenetic modulators. Valproic acid caused significant time-dependent changes in the mRNA expression of the melatonin MT(1) receptor, histone deacetylase (HDAC) 1, 2 and 3, and methyl CpG binding protein 2 (MeCP2). A structurally distinct HDAC inhibitor, trichostatin A, also caused a significant concentration-dependent induction of melatonin MT(1) receptor mRNA expression, suggesting involvement of an epigenetic mechanism. The ability of clinical concentrations of valproic acid to significantly alter melatonin MT(1) receptor expression, suggests a role for this receptor in the diverse neuropharmacological and oncostatic effects of this agent.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glioma/genetics , Histone Deacetylases/genetics , Methyl-CpG-Binding Protein 2/genetics , Receptor, Melatonin, MT1/genetics , Valproic Acid/pharmacology , Animals , Brain Neoplasms/enzymology , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/drug effects , Glioma/enzymology , Histone Deacetylase 1 , Histone Deacetylase 2 , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Methyl-CpG-Binding Protein 2/metabolism , RNA, Messenger/metabolism , Rats , Receptor, Melatonin, MT1/metabolism , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
15.
J Pineal Res ; 45(3): 277-84, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18373554

ABSTRACT

We have reported the induction of glial cell line-derived neurotrophic factor, a potent survival factor for dopaminergic neurons, in the C17.2 neural stem cell line following in vitro treatment with melatonin. Furthermore, we have detected the melatonin MT(1) receptor in these cells. Given these findings and recent evidence that melatonin may play a role in cellular differentiation, we examined whether this indoleamine induces morphological and transcriptional changes suggestive of a neuronal phenotype in C17.2 cells. Moreover, in order to extend preliminary evidence of a potential role for melatonin in epigenetic modulation, its effects on the mRNA expression of several histone deacetylase (HDAC) isoforms and on histone acetylation were examined. Physiological concentrations of melatonin (nanomolar range) increased neurite-like extensions and induced mRNA expression of the neural stem cell marker, nestin, the early neuronal marker beta-III-tubulin and the orphan nuclear receptor nurr1 in C17.2 cells. The indoleamine also significantly increased mRNA expression for various HDAC isoforms, including HDAC3, HDAC5, and HDAC7. Importantly, treatment with melatonin for 24 hr caused a significant increase in histone H3 acetylation, which is associated with chromatin remodeling and gene transcription. Since the melatonin MT(2) receptor was not detected in C17.2 cells, it is likely that the MT(1) receptor is involved in mediating these physiological effects of melatonin. These findings suggest novel roles for melatonin in stem cell differentiation and epigenetic modulation of gene transcription.


Subject(s)
Epigenesis, Genetic , Histone Deacetylases/genetics , Histones/metabolism , Melatonin/metabolism , Neurons/metabolism , Stem Cells/metabolism , Acetylation , Animals , Cell Differentiation , Cell Line , Chromatin Assembly and Disassembly , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Histone Deacetylases/metabolism , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Neurites/metabolism , Neurons/cytology , Neurons/ultrastructure , Nuclear Receptor Subfamily 4, Group A, Member 2 , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/genetics , Receptor, Melatonin, MT2/metabolism , Stem Cells/ultrastructure , Transcription Factors/metabolism , Transcription, Genetic , Tubulin/genetics , Tubulin/metabolism
16.
J Pineal Res ; 43(3): 245-54, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17803521

ABSTRACT

Melatonin has multiple roles including neuroprotection. Melatonin signaling involves diverse targets including two G-protein-coupled receptors, MT(1) and MT(2), which have both been localized to the nigrostriatal pathway. Previous studies in our laboratory demonstrated preservation of tyrosine hydroxylase immunoreactivity, following chronic treatment with a physiological dose of melatonin, in the 6-hydroxydopamine rat model of Parkinson's disease. Additionally, we reported the presence of the melatonin MT(1) receptor subtype in cultured C17.2 neural stem cells (NSCs). In the present study, we examined the effects of C17.2 NSC transplantation on dopaminergic denervation following 6-hydroxydopamine lesioning in the rat striatum. Moreover, based on our detection of the MT(1) in these cells, we examined the effects of combined C17.2 NSC transplantation and melatonin treatment, following striatal lesioning. Behavioral studies indicated a marked inhibition of apomorphine-induced rotations in lesioned animals that received C17.2 NSC transplantation, melatonin, or the combined regimen. In addition, these treatments resulted in a significant protection of tyrosine hydroxylase immunoreactivity in the striatum and substantia nigra of lesioned animals, when compared with untreated controls. Lesioned animals treated with C17.2 NSCs, melatonin or a combination of both agents exhibited no significant differences in the number of tyrosine hydroxylase-positive cells in the substantia nigra or ventral tegmental area ipsilateral or contralateral to the lesioned striatum. These findings suggest that stem cell therapy and concomitant use of neuroprotective agents such as melatonin could be a viable approach in Parkinson's disease.


Subject(s)
Melatonin/therapeutic use , Neurons , Oxidopamine/metabolism , Parkinson Disease/metabolism , Parkinson Disease/pathology , Stem Cell Transplantation , Animals , Behavior, Animal/drug effects , Cell Line , Disease Models, Animal , Male , Parkinson Disease/drug therapy , Parkinson Disease/surgery , Rats , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/metabolism
17.
Neurosci Lett ; 419(3): 202-6, 2007 Jun 04.
Article in English | MEDLINE | ID: mdl-17482356

ABSTRACT

We have previously reported in vivo preservation of tyrosine hydroxylase (TH), the rate-limiting enzyme in dopamine synthesis, following treatment with physiological doses of melatonin, in a 6-hydroxydopamine model of Parkinson's disease. Based on these findings, we postulated that melatonin would similarly modulate the expression of TH in vitro. Therefore, using human SH-SY5Y neuroblastoma cells which can differentiate into dopaminergic neurons following treatment with retinoic acid, we first examined whether these cells express melatonin receptors. Subsequently, the physiological dose-dependent effects of melatonin on TH expression were examined in both undifferentiated and differentiated cells. The novel detection of the G protein-coupled melatonin MT(1) receptor in SH-SY5Y cells by RT-PCR was confirmed by sequencing and Western blotting. In addition, following treatment of SH-SY5Y cells with melatonin (0.1-100 nM) for 24h, Western analysis revealed a significant increase in TH protein levels. A biphasic response, with significant increases in TH protein at 0.5 and 1 nM melatonin and a reversal at higher doses was seen in undifferentiated cells; whereas in differentiated cells, melatonin was effective at doses of 1 and 100 nM. These findings suggest a physiological role for melatonin in modulating TH expression, possibly via the MT(1) receptor.


Subject(s)
Melatonin/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Tyrosine 3-Monooxygenase/biosynthesis , Blotting, Western , Cell Differentiation , Cell Line, Tumor , Dose-Response Relationship, Drug , Gene Expression , Humans , Neuroblastoma/metabolism , Neurons/cytology , RNA, Messenger/analysis , Receptor, Melatonin, MT1/drug effects , Receptor, Melatonin, MT1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tyrosine 3-Monooxygenase/drug effects
18.
Eur J Pharmacol ; 560(1): 17-22, 2007 Mar 29.
Article in English | MEDLINE | ID: mdl-17303109

ABSTRACT

We have reported that valproic acid upregulates melatonin MT1 receptor expression in rat C6 glioma cells. In addition to its anticonvulsant and mood stabilizing properties, valproic acid can also inhibit the growth of cancer cells. Since the melatonin MT1 receptor has been implicated in the oncostatic action of melatonin on human MCF-7 breast cancer cells, the effect of valproic acid on its expression was examined in this cell line. Treatment of MCF-7 cells with valproic acid (0.5 or 1 mM) for 24 or 72 h caused a significant increase in melatonin MT1 receptor mRNA or protein expression, as shown by reverse transcription-polymerase chain reaction (RT-PCR) analysis and western blotting, respectively. MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assays revealed a concentration-dependent inhibition of MCF-7 cell proliferation by valproic acid (0.5, 1.0 or 5 mM), but melatonin (1 or 10 nM) was ineffective alone or in combination with valproic acid, in the first (MCF-7A) subline examined. However, in subsequent experiments using a different (MCF-7B) subline, which expressed higher levels of MT1 receptor mRNA and showed modest sensitivity to melatonin, a combination of this hormone with valproic acid produced a significant synergistic inhibition of cell proliferation. These findings indicate that clinically relevant concentrations of valproic acid upregulate melatonin MT1 receptor expression in human breast cancer cells. Moreover, the enhanced antiproliferative effect observed with a combination of valproic acid and melatonin suggests that a similar therapeutic approach may be beneficial in breast cancer.


Subject(s)
Anticonvulsants/pharmacology , Breast Neoplasms/metabolism , Melatonin/pharmacology , Receptor, Melatonin, MT1/metabolism , Valproic Acid/pharmacology , Animals , Anticonvulsants/administration & dosage , Apoptosis/drug effects , Blotting, Western , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Therapy, Combination , Gene Expression , Humans , Melatonin/administration & dosage , Rats , Receptor, Melatonin, MT1/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/drug effects , Valproic Acid/administration & dosage
19.
Brain Res ; 1068(1): 230-6, 2006 Jan 12.
Article in English | MEDLINE | ID: mdl-16375867

ABSTRACT

There is considerable evidence that pharmacological doses of the pineal hormone, melatonin, are neuroprotective in diverse models of neurodegeneration including Parkinson's disease. However, there is limited information about the effects of physiological doses of this hormone in similar models. In this study, rats were chronically treated with melatonin via drinking water following partial 6-hydroxydopamine lesioning in the striatum. The two doses of melatonin (0.4 microg/ml and 4.0 microg/ml) were within the reported physiological concentrations present in the serum and cerebrospinal fluid respectively. At 2 weeks after surgery, the higher dose of melatonin significantly attenuated rotational behavior in hemi-parkinsonian rats compared to similarly lesioned animals receiving either vehicle (P < 0.001) or the lower dose of melatonin (P < 0.01). Animals were perfused or sacrificed 10 weeks after commencing melatonin treatment for immunohistochemical or mRNA studies. Animals treated with 4.0 microg/ml melatonin exhibited normal tyrosine hydroxylase (TH) immunoreactivity in the lesioned striatum, whereas little or no TH immunofluorescence was visible in similarly lesioned animals receiving vehicle. In contrast, semiquantitative RT-PCR analysis revealed no group differences in TH mRNA, suggesting spontaneous recovery of this transcript as observed previously in partially lesioned animals. There were no significant differences in striatal GDNF mRNA levels between sham and lesioned animals. However, there was a significant (P < 0.01) increase in GDNF mRNA expression in the intact contralateral striata of lesioned animals treated with vehicle. Interestingly, melatonin treatment attenuated this novel compensatory contralateral increase in striatal GDNF expression, presumably due to its neuroprotective effect. These findings support a physiological role for melatonin in protecting against parkinsonian neurodegeneration in the nigrostriatal system.


Subject(s)
Melatonin/pharmacology , Neuroprotective Agents , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/prevention & control , Animals , Apomorphine/antagonists & inhibitors , Apomorphine/pharmacology , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/genetics , Densitometry , Dopamine Agonists/pharmacology , Functional Laterality/physiology , Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor/genetics , Immunohistochemistry , Male , Neostriatum/drug effects , Neostriatum/enzymology , Oxidopamine , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Receptor, Melatonin, MT1/drug effects , Receptors, Melatonin/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Stereotyped Behavior/drug effects , Sympatholytics , Tyrosine 3-Monooxygenase/metabolism
20.
J Neurochem ; 95(5): 1227-36, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16313512

ABSTRACT

Valproic acid (VPA) is a potent anti-epileptic and effective mood stabilizer. It is known that VPA enhances central GABAergic activity and activates the mitogen-activated protein kinase-extracellular signal-regulated kinase (MAPK-ERK) pathway. It can also inhibit various isoforms of the enzyme, histone deacetylase (HDAC), which is associated with modulation of gene transcription. Recent in vivo studies indicate a neuroprotective role for VPA, which has been found to up-regulate the expression of brain-derived neurotrophic factor (BDNF) in the rat brain. Given the interaction between the pineal hormone, melatonin, and GABAergic systems in the central nervous system, the effects of VPA on the expression of the mammalian melatonin receptor subtypes, MT1 and MT2, were examined in rat C6 glioma cells. The effects of VPA on the expression of glial cell line-derived neurotrophic factor (GDNF) and BDNF were also examined. RT-PCR studies revealed a significant induction of melatonin MT1 receptor mRNA in C6 cells following treatment with 3 or 5 mm VPA for 24 h or 5 mm VPA for 48 h. Western analysis and immunocytochemical detection confirmed that the VPA-induced increase in MT1 mRNA results in up-regulation of MT1 protein expression. Blockade of the MAPK-ERK pathway by PD98059 enhanced the effect of VPA on MT1 expression, suggesting a negative role for this pathway in MT1 receptor regulation. In addition, significant increases in BDNF, GDNF and HDAC mRNA expression were observed after treatment with VPA for 24 or 48 h. Taken together, the present findings suggest that the neuroprotective properties of VPA involve modulation of neurotrophic factors and receptors for melatonin, which is also thought to play a role in neuroprotection. Moreover, the foregoing suggests that combinations of VPA and melatonin could provide novel therapeutic strategies in neurological and psychiatric disorders.


Subject(s)
Enzyme Inhibitors/pharmacology , Nerve Growth Factors/metabolism , Receptors, Melatonin/metabolism , Up-Regulation/drug effects , Valproic Acid/pharmacology , Animals , Blotting, Northern/methods , Blotting, Western/methods , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Interactions , Glioma , Histone Deacetylase 1 , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Immunohistochemistry/methods , Mice , Nerve Growth Factors/genetics , RNA, Messenger/metabolism , Receptors, Melatonin/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods
SELECTION OF CITATIONS
SEARCH DETAIL