Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
1.
Mol Psychiatry ; 23(8): 1717-1730, 2018 08.
Article in English | MEDLINE | ID: mdl-28924188

ABSTRACT

Dopamine in prefrontal cortices is implicated in cognitive and emotional functions, and the dysfunction of prefrontal dopamine has been associated with cognitive and emotional deficits in mental illnesses. These findings have led to clinical trials of dopamine-targeting drugs and brain imaging of dopamine receptors in patients with mental illnesses. Rodent studies have suggested that dopaminergic pathway projecting to the medial prefrontal cortex (mPFC) suppresses stress susceptibility. Although various types of mPFC neurons express several dopamine receptor subtypes, previous studies neither isolated a role of dopamine receptor subtype nor identified the site of its action in mPFC. Using social defeat stress (SDS) in mice, here we identified a role of dopamine D1 receptor subtype in mPFC excitatory neurons in suppressing stress susceptibility. Repeated social defeat stress (R-SDS) reduces the expression of D1 receptor subtype in mPFC of mice susceptible to R-SDS. Knockdown of D1 receptor subtype in whole neuronal populations or excitatory neurons in mPFC facilitates the induction of social avoidance by SDS. Single social defeat stress (S-SDS) induces D1 receptor-mediated extracellular signal-regulated kinase phosphorylation and c-Fos expression in mPFC neurons. Whereas R-SDS reduces dendritic lengths of mPFC layer II/III pyramidal neurons, S-SDS increases arborization and spines of apical dendrites of these neurons in a D1 receptor-dependent manner. Collectively, our findings show that D1 receptor subtype and related signaling in mPFC excitatory neurons mediate acute stress-induced dendritic growth of these neurons and contribute to suppression of stress susceptibility. Therefore, we propose that D1 receptor-mediated dendritic growth in mPFC excitatory neurons suppresses stress susceptibility.


Subject(s)
Dendrites/metabolism , Prefrontal Cortex/metabolism , Receptors, Dopamine D1/metabolism , Resilience, Psychological , Stress, Psychological/metabolism , Animals , Avoidance Learning/physiology , Cell Enlargement , Dendrites/pathology , Disease Models, Animal , Disease Susceptibility/metabolism , Dominance-Subordination , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Knockdown Techniques , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Prefrontal Cortex/pathology , Proto-Oncogene Proteins c-fos/metabolism , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Receptors, Dopamine D1/genetics , Stress, Psychological/pathology
2.
Br J Pharmacol ; 164(5): 1534-46, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21518336

ABSTRACT

BACKGROUND AND PURPOSE: The aim of this study was to evaluate the anti-convulsant effects of magnolol (6, 6', 7, 12-tetramethoxy-2, 2'-dimethyl-1-ß-berbaman, C18H18O2) and the mechanisms involved. EXPERIMENTAL APPROACH: Mice were treated with magnolol (20, 40 and 80 mg·kg(-1)) 30 min before injection with pentylenetetrazol (PTZ, 60 mg·kg(-1), i.p.). The anti-seizure effects of magnolol were analysed using seizure models of behaviour, EEG and in vitro electrophysiology and c-Fos expression in the hippocampus and cortex. KEY RESULTS: Magnolol at doses of 40 and 80 mg·kg(-1) significantly delayed the onset of myoclonic jerks and generalized clonic seizures, and decreased the seizure stage and mortality compared with those of the vehicle-treated animals. EEG recordings showed that magnolol (40 and 80 mg·kg(-1)) prolonged the latency of seizure onset and decreased the number of seizure spikes. The anti-epileptic effect of magnolol was reversed by the GABA(A)/benzodiazepine receptor antagonist flumazenil. Pretreatment with flumazenil decreased the effects of magnolol on prolongation of seizure latency and decline of seizure stage. In a Mg(2+)-free model of epileptiform activity, using multi-electrode array recordings in mouse hippocampal slices, magnolol decreased spontaneous epileptiform discharges. Magnolol also significantly decreased seizure-induced Fos immunoreactivity in the piriform cortex, dentate gyrus and hippocampal area CA1. These effects were attenuated by pretreatment with flumazenil. CONCLUSIONS AND IMPLICATIONS: These findings indicate that the inhibitory effects of magnolol on epileptiform activity were mediated by the GABA(A) /benzodiazepine receptor complex.


Subject(s)
Anticonvulsants/therapeutic use , Biphenyl Compounds/therapeutic use , Lignans/therapeutic use , Magnolia/chemistry , Receptors, GABA-A/metabolism , Seizures/drug therapy , Animals , Anticonvulsants/administration & dosage , Anticonvulsants/isolation & purification , Behavior, Animal/drug effects , Biphenyl Compounds/administration & dosage , Biphenyl Compounds/isolation & purification , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Dose-Response Relationship, Drug , Electroencephalography , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Lignans/administration & dosage , Lignans/isolation & purification , Male , Mice , Mice, Inbred Strains , Molecular Structure , Plant Bark/chemistry , Seizures/metabolism
3.
Neuroscience ; 163(1): 296-307, 2009 Sep 29.
Article in English | MEDLINE | ID: mdl-19531375

ABSTRACT

Cerebral ischemia/reperfusion injury is characterized by the development of inflammatory response, in which vascular macrophages and endogenous microglia are involved. Recent studies showed marked induction of hematopoietic prostaglandin D synthase (HPGDS) after ischemic/reperfusion injury and its localization in microglia, but the molecular mechanism(s) of HPGDS actions in cerebral ischemia is not clear. To clarify the role of HPGDS in cerebral ischemia, C57BL/6 mice and bone marrow chimera mice with cerebral ischemia/reperfusion injury were treated with (4-benzhydryloxy-(1) {3-(1H-tetrazol-5-yl)-propyl}piperidine (HQL-79), a specific inhibitor of HPGDS. The bone marrow chimera mice exhibit expression of enhanced green fluorescent protein (EGFP) in bone marrow/blood-derived monocytes/macrophages. Mice were subjected to ischemia/reperfusion and either treated with HQL-79 (n=44) or vehicle (n=44). Brain sections prepared at 72 h and 7 days after reperfusion were analyzed for neuronal nuclei (NeuN), HPGDS, ionized calcium-binding adapter molecule 1 (Iba1), inducible NO synthase (iNOS), nitrotyrosine, nuclear factor kappa B (NF-kB) and cyclooxygenase-2 (COX-2). The mortality rate (80%) and infarct size were larger in HQL-79- than vehicle-treated mice (58.7+/-8.5 versus 45.2+/-4.9 mm(3); mean+/-SEM, P<0.0001) at 7 days after reperfusion. HQL-79 reduced NeuN expression in the transition area and Iba1 expression (P<0.0001) in the ischemic peri- and penumbra area, but increased COX-2 (P<0.05) and NF-kB expression (P<0.05) in ischemic penumbra and increased formation of nitrotyrosine (P<0.0001) and iNOS (P<0.0001) in the ischemic core area at 72 h and 7 days after reperfusion. In EGFP chimera mice, HQL-79 increased the migration of Iba1/EGFP-positive bone marrow-derived monocytes/macrophages, and simultaneously upregulated iNOS expression in the ischemic core area (P<0.0001), but increased intrinsic microglia/macrophages in ischemic peri-area and penumbra (P<0.0001) at 72 h and 7 days after reperfusion, suggesting involvement of monocytes/macrophages in HQL-79-induced expansion of ischemic injury. Our results demonstrated that the neuroprotective effects of HPGDS in our model are mediated by suppression of activation and infiltration of inflammatory cells.


Subject(s)
Brain/drug effects , Encephalitis/drug therapy , Hypoxia-Ischemia, Brain/drug therapy , Intramolecular Oxidoreductases/metabolism , Ischemic Attack, Transient/drug therapy , Isomerases/metabolism , Lipocalins/metabolism , Reperfusion Injury/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Biomarkers/metabolism , Bone Marrow Transplantation/methods , Brain/enzymology , Brain/physiopathology , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/physiology , Disease Models, Animal , Encephalitis/physiopathology , Encephalitis/prevention & control , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Green Fluorescent Proteins/genetics , Hypoxia-Ischemia, Brain/physiopathology , Hypoxia-Ischemia, Brain/prevention & control , Intramolecular Oxidoreductases/antagonists & inhibitors , Ischemic Attack, Transient/physiopathology , Ischemic Attack, Transient/prevention & control , Isomerases/antagonists & inhibitors , Lipocalins/antagonists & inhibitors , Macrophages/drug effects , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/physiology , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Nitric Oxide Synthase Type II/metabolism , Piperidines/pharmacology , Piperidines/therapeutic use , Reperfusion Injury/physiopathology , Reperfusion Injury/prevention & control , Transplantation Chimera
4.
Neuroscience ; 160(1): 248-54, 2009 Apr 21.
Article in English | MEDLINE | ID: mdl-19254753

ABSTRACT

Prostaglandin D(2) (PGD(2)) is the most abundant prostaglandin produced in the brain. It is a metabolite of arachidonic acid and synthesized by prostaglandin D(2) synthases (PGDS) via the cyclooxygenase pathway. Two distinct types of PGDS have been identified: hematopoietic prostaglandin D synthase (H-PGDS) and lipocalin-type prostaglandin D synthase (L-PGDS). Because relatively little is known about the role of L-PGDS in the CNS, here we examined the outcomes in L-PGDS knockout and wild-type (WT) mice after two different cerebral ischemia models, transient middle cerebral artery (MCA) occlusion (tMCAO) and permanent distal middle cerebral artery occlusion (pMCAO). In the tMCAO model, the MCA was occluded with a monofilament for 90 min and then reperfused for 4 days. In the pMCAO model, the distal part of the MCA was permanently occluded and the mice were sacrificed after 7 days. Percent corrected infarct volume and neurological score were determined after 4 and 7 days, respectively. L-PGDS knockout mice had significantly greater infarct volume and brain edema than did WT mice after tMCAO (P<0.01). Similarly, L-PGDS knockout mice showed greater infarct volume and neurological deficits as compared to their WT counterparts after pMCAO (P<0.01). Using the two models enabled us to study the role of L-PGDS in both early (tMCAO) and delayed (pMCAO) ischemic processes. Our findings suggest that L-PGDS is beneficial for protecting the brain against transient and permanent cerebral ischemia. These results provide a better understanding of the role played by the enzymes that control eicosanoid synthesis and how they can be utilized as potential targets to prevent damage following either acute or potentially chronic neurological disorders.


Subject(s)
Infarction, Middle Cerebral Artery/enzymology , Intramolecular Oxidoreductases/metabolism , Lipocalins/metabolism , Animals , Blood Gas Analysis , Blood Pressure/physiology , Body Temperature/physiology , Brain/blood supply , Brain/enzymology , Brain/pathology , Brain Chemistry/physiology , Brain Edema/pathology , Cerebrovascular Circulation/physiology , Disease Models, Animal , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/psychology , Intramolecular Oxidoreductases/genetics , Lipocalins/genetics , Male , Mice , Mice, Knockout , Severity of Illness Index , Water/chemistry
5.
Neuropharmacology ; 53(4): 534-41, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17681557

ABSTRACT

Epilepsy is characterized by neuronal hyperexcitability and hypersynchronization. Disruption of electroencephalographically (EEG) synchronized epileptiform discharges may be a possible therapy for epilepsy. In the present study, to clarify the role of EEG desynchronization on epilepsy, we investigated the effect of modafinil, a potent wake-promoting substance with EEG desynchronization activity, on epilepsy in mice and clarified the receptors involved in the suppression of seizure caused by maximal electroshock (MES) and pentylenetetrazol (PTZ) kindling models. Modafinil given at 22.5, 45, and 90 mg/kg, i.p. significantly decreased the incidence of tonic hindleg extension in MES seizure models, and protected against PTZ-induced convulsive behaviors in a dose-dependent manner. In addition, modafinil at 180 mg/kg exerted an antiepileptic effect in the MES model; however, at the same dosage it increased the seizure stage in the PTZ-kindling model. The antiepileptic effect in both MES and PTZ models was antagonized by the adrenergic alpha(1) receptor antagonist terazosin, but not by the adrenergic alpha(2) receptor antagonist yohimbine or by dopaminergic receptor antagonists, SCH-23390 (for D(1) receptors) and haloperidol (for D(2) ones). Pyrilamine, a histaminergic H(1) receptor antagonist, counteracted the antiepileptic action of modafinil in the PTZ induced-kindling model, but not in the MES seizure model. Taken together, the present findings indicate that modafinil exerted its antiepileptic effect via adrenergic alpha(1) and histaminergic H(1) receptors, and might be of potential use in the treatment of epilepsy.


Subject(s)
Benzhydryl Compounds/pharmacology , Epilepsy/prevention & control , Neuroprotective Agents/pharmacology , Animals , Disease Models, Animal , Electroencephalography , Electroshock , Male , Mice , Mice, Inbred Strains , Modafinil , Pentylenetetrazole/pharmacology , Seizures/etiology , Seizures/prevention & control
6.
Prog Neurobiol ; 83(5): 332-47, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17532111

ABSTRACT

Adenosine A2A receptors localized in the dorsal striatum are considered as a new target for the development of antiparkinsonian drugs. Co-administration of A2A receptor antagonists has shown a significant improvement of the effects of l-DOPA. The present review emphasizes the possible application of A2A receptor antagonists in pathological conditions other than parkinsonism, including drug addiction, sleep disorders and pain. In addition to the dorsal striatum, the ventral striatum (nucleus accumbens) contains a high density of A2A receptors, which presynaptically and postsynaptically regulate glutamatergic transmission in the cortical glutamatergic projections to the nucleus accumbens. It is currently believed that molecular adaptations of the cortico-accumbens glutamatergic synapses are involved in compulsive drug seeking and relapse. Here we review recent experimental evidence suggesting that A2A antagonists could become new therapeutic agents for drug addiction. Morphological and functional studies have identified lower levels of A2A receptors in brain areas other than the striatum, such as the ventrolateral preoptic area of the hypothalamus, where adenosine plays an important role in sleep regulation. Although initially believed to be mostly dependent on A1 receptors, here we review recent studies that demonstrate that the somnogenic effects of adenosine are largely mediated by hypothalamic A2A receptors. A2A)receptor antagonists could therefore be considered as a possible treatment for narcolepsy and other sleep-related disorders. Finally, nociception is another adenosine-regulated neural function previously thought to mostly involve A1 receptors. Although there is some conflicting literature on the effects of agonists and antagonists, which may partly be due to the lack of selectivity of available drugs, the studies in A2A receptor knockout mice suggest that A2A receptor antagonists might have some therapeutic potential in pain states, in particular where high intensity stimuli are prevalent.


Subject(s)
Basal Ganglia/metabolism , Hypothalamus/metabolism , Pain/metabolism , Receptor, Adenosine A2A/metabolism , Sleep Wake Disorders/metabolism , Substance-Related Disorders/metabolism , Adenosine/metabolism , Adenosine A1 Receptor Antagonists , Adenosine A2 Receptor Antagonists , Animals , Basal Ganglia/drug effects , Basal Ganglia/physiopathology , Humans , Hypothalamus/drug effects , Hypothalamus/physiopathology , Neural Pathways/drug effects , Neural Pathways/metabolism , Neural Pathways/physiopathology , Pain/drug therapy , Pain/physiopathology , Receptor, Adenosine A1/metabolism , Sleep Wake Disorders/drug therapy , Sleep Wake Disorders/physiopathology , Substance-Related Disorders/drug therapy , Substance-Related Disorders/physiopathology
7.
Neuroscience ; 145(2): 520-9, 2007 Mar 16.
Article in English | MEDLINE | ID: mdl-17241746

ABSTRACT

Hematopoietic prostaglandin D synthase is a key enzyme in synthesis of prostaglandin D. Hematopoietic prostaglandin D synthase is expressed in microglia of the developing mouse brain. This study determined the serial changes and cellular localization of hematopoietic prostaglandin D synthase, and its role in cerebral ischemia/reperfusion injury using C57BL/6 mice (n=84) and bone marrow chimera mice (n=16). The latter mice were selected based on their expression of enhanced green fluorescent protein in bone marrow/blood-derived monocytes/macrophages. The middle cerebral artery was occluded for 60 min, followed by reperfusion. Hematopoietic prostaglandin D synthase expression was examined by immunohistochemistry and Western blotting. Hematopoietic prostaglandin D synthase-positive cells were mainly expressed in the peri-ischemic area at 12 h (P<0.05) and 24 h (P<0.001) after reperfusion, while they were mostly found in the transition area at 48-72 h postreperfusion (P<0.001). There was a significant increase in staining intensity as well as number of hematopoietic prostaglandin D synthase-positive cells in the ischemic core at 5-7 (P<0.001) days postreperfusion. Hematopoietic prostaglandin D synthase-positive cells also co-expressed ionized calcium-binding adapter molecule 1, a marker of microglia/macrophages, and cyclooxygenase-2, but not markers of neurons, oligodendrocytes and astrocytes. Until 72 h postreperfusion, many enhanced green fluorescent protein-positive cells were negative for hematopoietic prostaglandin D synthase, but the number of hematopoietic prostaglandin D synthase-enhanced green fluorescent protein coexpressing cells increased significantly at 5-7 days after reperfusion. Our results indicate that hematopoietic prostaglandin D synthase is mainly produced by endogenous microglia until 72 h after reperfusion, but at 7 days after reperfusion, it is also produced by migrating bone marrow/blood-derived macrophages in the ischemic brain tissue. We speculate that hematopoietic prostaglandin D synthase in the brain has different functions during early and late phases of ischemia.


Subject(s)
Brain Ischemia/enzymology , Brain/enzymology , Intramolecular Oxidoreductases/metabolism , Macrophages/enzymology , Microglia/enzymology , Reperfusion Injury/enzymology , Animals , Brain/blood supply , Brain/physiopathology , Brain Ischemia/physiopathology , Calcium-Binding Proteins/metabolism , Cell Count , Cell Movement/physiology , Cell Proliferation , Cyclooxygenase 2/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hematopoiesis/physiology , Lipocalins , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins , Prostaglandin D2/biosynthesis , Reperfusion Injury/physiopathology , Transplantation Chimera , Up-Regulation
8.
Clin Exp Allergy ; 36(8): 1028-38, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16911359

ABSTRACT

BACKGROUND: Prostaglandin (PG)D(2) and E(2), two major cyclooxygenase (COX) products, are generated by PGD(2) synthase (PGDS) and PGE(2) synthase (PGES), respectively, and appear to mediate airway inflammation. OBJECTIVE: We sought to determine the role of PGDS and PGES in the pathophysiology of chronic rhinosinusitis (CRS). METHODS: The study examined the expression of PGDS and PGES in nasal polyps of 22 CRS patients. As controls, uncinate process mucosae were obtained from 12 CRS patients not having nasal polyps and five subjects without sinusitis. Immunohistochemistry and quantitative real-time PCR were used to evaluate the expression. RESULTS: Both PGDS and PGES were detected in nasal polyps by immunohistochemistry. Significantly greater levels of PGDS mRNA and lesser levels of PGES mRNA were observed in the nasal polyps as compared with uncinate process mucosae, and an inverse correlation between PGDS and PGES expression was observed. Levels of PGDS mRNA in nasal polyps were positively correlated with degree of infiltration by EG2+ eosinophils, whereas the levels of PGES were inversely correlated. Significantly increased levels of PGDS and conversely decreased levels of PGES were observed in asthmatics as compared with non-asthmatics. In addition, PGDS and PGES levels were positively and inversely correlated with the radiological severity of sinusitis, respectively. CONCLUSIONS: These results suggest that PGDS and PGES display an opposite and important role in the pathophysiology of CRS such as polyp formation, and more specifically, a biased expression of these synthases might contribute to the development of CRS by affecting eosinophilic inflammation.


Subject(s)
Intramolecular Oxidoreductases/physiology , Isoenzymes/physiology , Rhinitis/enzymology , Sinusitis/enzymology , Adolescent , Adult , Chronic Disease , Cyclooxygenase 1/analysis , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/analysis , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Eosinophilia/enzymology , Female , Humans , Immunohistochemistry/methods , Intramolecular Oxidoreductases/analysis , Intramolecular Oxidoreductases/genetics , Isoenzymes/analysis , Isoenzymes/genetics , Lipocalins , Male , Nasal Mucosa/enzymology , Nasal Polyps/enzymology , Nasal Polyps/immunology , Prostaglandin-E Synthases , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Rhinitis/immunology , Sinusitis/immunology , Statistics, Nonparametric
9.
Neuropathol Appl Neurobiol ; 32(1): 64-73, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16409554

ABSTRACT

Lipocalin-type prostaglandin D synthase (L-PGDS), which is mainly synthesized in leptomeningeal cells and oligodendrocytes (OLs) in rodents and humans, is secreted into the human cerebrospinal fluid (CSF) as beta-trace. L-PGDS protects OLs and neurones against apoptosis in twitcher mice, a murine model of Krabbe's disease, and is the second only to a stress protein, alphaB-crystallin, as the most abundant gene product upregulated in the demyelinating focus of multiple sclerosis (MS). Here we report that although the CSF level of L-PGDS is not increased in MS patients, L-PGDS is increased in the white matter of MS patients, especially in the shadow plaque as compared with the normal white matter. L-PGDS immunoreactivity was intensely expressed in OLs within the shadow plaques and in hypertrophied astrocytes within the chronic plaques of MS patients. Both L-PGDS-positive OLs and astrocytes expressed a stress protein, alphaB-crystallin. These results suggest that the upregulation of L-PGDS occurs in OLs and astrocytes as a stress reaction.


Subject(s)
Astrocytes/metabolism , Intermediate Filament Proteins/biosynthesis , Intramolecular Oxidoreductases/biosynthesis , Multiple Sclerosis/metabolism , Nerve Tissue Proteins/biosynthesis , Oligodendroglia/metabolism , Protein Kinases/biosynthesis , Adult , Aged , Aged, 80 and over , Chronic Disease , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Lipocalins , Male , Middle Aged , Multiple Sclerosis/pathology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation , alpha-Crystallin B Chain
10.
Neuroscience ; 134(4): 1377-90, 2005.
Article in English | MEDLINE | ID: mdl-16039802

ABSTRACT

Recent research has shown that neurons in the ventrolateral preoptic nucleus are crucial for sleep by inhibiting wake-promoting systems, but the process that triggers their activation at sleep onset remains to be established. Since evidence indicates that sleep induced by adenosine, an endogenous sleep-promoting substance, requires activation of brain A(2A) receptors, we examined the hypothesis that adenosine could activate ventrolateral preoptic nucleus sleep neurons via A(2A) adenosine receptors in rat brain slices. Following on from our initial in vitro identification of these neurons as uniformly inhibited by noradrenaline and acetylcholine arousal transmitters, we established that the ventrolateral preoptic nucleus comprises two intermingled subtypes of sleep neurons, differing in their firing responses to serotonin, inducing either an inhibition (Type-1 cells) or an excitation (Type-2 cells). Since both cell types contained galanin and expressed glutamic acid decarboxylase-65/67 mRNAs, they potentially correspond to the sleep promoting neurons inhibiting arousal systems. Our pharmacological investigations using A(1) and A(2A) adenosine receptors agonists and antagonists further revealed that only Type-2 neurons were excited by adenosine via a postsynaptic activation of A(2A) adenosine receptors. Hence, the present study is the first demonstration of a direct activation of the sleep neurons by adenosine. Our results further support the cellular and functional heterogeneity of the sleep neurons, which could enable their differential contribution to the regulation of sleep. Adenosine and serotonin progressively accumulate during arousal. We propose that Type-2 neurons, which respond to these homeostatic signals by increasing their firing are involved in sleep induction. In contrast, Type-1 neurons would likely play a role in the consolidation of sleep.


Subject(s)
Adenosine/metabolism , Neurons/cytology , Preoptic Area/cytology , Receptor, Adenosine A2A/metabolism , Sleep/physiology , Adenosine A2 Receptor Agonists , Adenosine A2 Receptor Antagonists , Animals , Neurons/metabolism , Organ Culture Techniques , Patch-Clamp Techniques , Preoptic Area/physiology , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Serotonin/metabolism
11.
Neuroscience ; 130(4): 1029-40, 2005.
Article in English | MEDLINE | ID: mdl-15652998

ABSTRACT

CNS activity is generally coupled to the vigilance state, being primarily active during wakefulness and primarily inactive during deep sleep. During periods of high neuronal activity, a significant volume of oxygen is used to maintain neuronal membrane potentials, which subsequently produces cytotoxic reactive oxygen species (ROS). Glutathione, a major endogenous antioxidant, is an important factor protecting against ROS-mediated neuronal degeneration. Glutathione has also been proposed to be a sleep-promoting substance, yet the relationship between sleep and cerebral oxidation remains unclear. Here we report that i.c.v. infusion of the organic peroxide t-butyl-hydroperoxide at a concentration below that triggering neurodegeneration (0.1 micromol/100 microl/10 h) promotes sleep in rats. Also, microinjection (2 nmol, 2 microl) or microdialysis (100 microM, 20 min) of t-butyl-hydroperoxide into the preoptic/anterior hypothalamus (POAH) induces the release of the sleep-inducing neuromodulators, nitric oxide and adenosine, without causing neurodegeneration. Nitric oxide and adenosine release was inhibited by co-dialysis of the N-methyl-D-aspartate receptor antagonist, d(-)-2-amino-5-phosphonopentanoic acid (D-AP5; 1 mM), suggesting that glutamate-induced neuronal excitation mediates the peroxide-induced release of nitric oxide and adenosine. Indeed, Ca2+ release from mitochondria and delayed-onset Ca2+ influx via N-methyl-D-aspartate receptors was visualized during peroxide exposure using Ca2+ indicator proteins (YC-2.1 and mitochondrial-targeted Pericam) expressed in organotypic cultures of the POAH. In the in vitro models, t-butyl-hydroperoxide (50 microM) causes dendritic swelling followed by the intracellular Ca2+ mobilization, and D-AP5 (100 microM) or glutathione (500 microM) inhibited t-butyl-hydroperoxide-induced intracellular Ca2+ mobilization and protected POAH neurons from oxidative stress. These data suggest that low-level subcortical oxidation under the control of an antioxidant system may trigger sleep via the Ca(2+)-dependent release of sleep-inducing neuromodulators in the POAH, and thus we propose that a moderate increase of ROS during wakefulness in the neuronal circuits regulating sleep may be an initial trigger in sleep induction.


Subject(s)
Brain/metabolism , Energy Metabolism/physiology , Neurons/metabolism , Oxygen Consumption/physiology , Reactive Oxygen Species/metabolism , Sleep/physiology , Adenosine/metabolism , Animals , Anterior Hypothalamic Nucleus/drug effects , Anterior Hypothalamic Nucleus/metabolism , Brain/drug effects , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/metabolism , Glutathione/metabolism , Glutathione/pharmacology , Male , Microtubule-Associated Proteins/drug effects , Microtubule-Associated Proteins/metabolism , Neurons/drug effects , Nitric Oxide/metabolism , Organ Culture Techniques , Oxidative Stress/drug effects , Oxidative Stress/physiology , Preoptic Area/drug effects , Preoptic Area/metabolism , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Sleep/drug effects , tert-Butylhydroperoxide/pharmacology
12.
Arch Ital Biol ; 142(4): 533-9, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15493554

ABSTRACT

(1) Prostaglandin D2 is essential for the maintenance of the sleep state. (2) The adenosine and A2A receptor system is a link between the humoral and neural mechanisms of sleep-wake regulation. (3) Prostaglandin D2 plays a crucial role in the homeostatic regulation of NREM sleep. Finally, it may not be too far-fetched to say that prostaglandin D2 was most likely the endogenous sleep substance described by Piéron and Ishimori about 100 years ago, and possibly the sleep-inducing factor reported by Professor Jouvet and coworkers some twenty years ago.


Subject(s)
Brain/physiology , Intramolecular Oxidoreductases/genetics , Prostaglandin D2/biosynthesis , Receptors, Immunologic/genetics , Receptors, Prostaglandin/genetics , Sleep/physiology , Animals , Brain/anatomy & histology , Brain/enzymology , Cerebrospinal Fluid/metabolism , Homeostasis/genetics , Humans , Lipocalins , Mice , Prostaglandin D2/genetics , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Sleep/genetics
13.
Article in English | MEDLINE | ID: mdl-15301794

ABSTRACT

We investigated the tissue distribution and cellular localization of microsomal PGE synthase-1 (mPGES-1) and cyclooxygenase (COX)-1 and -2 in male monkey reproductive organs. Western blotting revealed that monkey mPGES-1 was expressed most intensely in the seminal vesicles, moderately in the testis, and weakly in the epididymis and vas deferens. The tissue distribution profile was quite different from those profiles for rats, rabbits, and pigs, e.g., rat mPGES-1 was the most abundant in the vas deferens, and the rabbit and pig enzymes, in the testis. Immunohistochemical staining with mouse monoclonal anti-human mPGES-1 antibody revealed that monkey mPGES-1 was localized in spermatogonia, Sertoli cells, and primary spermatocytes of testis and in epithelial cells of the epididymis, vas deferens, and seminal vesicles. In monkeys, COX-1 was localized in epithelial cells of the epididymis and vas deferens, whereas COX-2 was dominantly found in epithelial cells of the seminal vesicles.


Subject(s)
Intramolecular Oxidoreductases/metabolism , Isoenzymes/metabolism , Macaca fascicularis , Microsomes/enzymology , Prostaglandin-Endoperoxide Synthases/metabolism , Testis/enzymology , Animals , Antibodies, Monoclonal/immunology , Cyclooxygenase 1 , Cyclooxygenase 2 , Humans , Immunohistochemistry , Intramolecular Oxidoreductases/immunology , Male , Membrane Proteins , Prostaglandin-E Synthases , Rabbits , Rats , Species Specificity , Swine , Testis/cytology
14.
Neuroscience ; 107(4): 653-63, 2001.
Article in English | MEDLINE | ID: mdl-11720788

ABSTRACT

Considerable evidence indicates that adenosine may be an endogenous somnogen, yet the mechanism through which it promotes sleep is unknown. Adenosine may act via A1 receptors to promote sleep, but an A2a receptor antagonist can block the sleep induced by prostaglandin D(2). We previously reported that prostaglandin D(2) activates sleep-promoting neurons of the ventrolateral preoptic area, and we hypothesized that an A2a receptor agonist also should activate these neurons. Rats were instrumented for sleep recordings, and an injection cannula was placed in the subarachnoid space just anterior to the ventrolateral preoptic area. After an 8-10-day recovery period, the A2a receptor agonist CGS21680 (20 pmol/min) or saline was infused through the injection cannula, and the animals were killed 2 h later. The brains were stained using Fos immunohistochemistry, and the pattern of Fos expression was studied in the entire brain. CGS21680 increased non-rapid eye movement sleep and markedly increased the expression of Fos in the ventrolateral preoptic area and basal leptomeninges, but it reduced Fos expression in wake-active brain regions such as the tuberomammillary nucleus. CGS21680 also induced Fos in the shell and core of the nucleus accumbens and in the lateral subdivision of the central nucleus of the amygdala. To determine whether these effects may have been mediated through A1 receptors, an additional group of rats received subarachnoid infusion of the A1 receptor agonist N(6)-cyclopentyladenosine (2 pmol/min). In contrast to CGS21680, infusion of N(6)-cyclopentyladenosine into the subarachnoid space produced only a small decrease in rapid eye movement sleep, and the pattern of Fos expression induced by N(6)-cyclopentyladenosine was notable only for decreased Fos in regions near the infusion site. These findings suggest that an adenosine A2a receptor agonist may activate cells of the leptomeninges or nucleus accumbens that increase the activity of ventrolateral preoptic area neurons. These ventrolateral preoptic area neurons may then coordinate the inhibition of multiple wake-promoting regions, resulting in sleep.


Subject(s)
Adenosine/analogs & derivatives , Neurons/metabolism , Preoptic Area/cytology , Proto-Oncogene Proteins c-fos/biosynthesis , Purinergic P1 Receptor Agonists , Sleep/drug effects , Adenosine/pharmacology , Animals , Antihypertensive Agents/pharmacology , Brain Chemistry/drug effects , Male , Neurons/chemistry , Phenethylamines/pharmacology , Preoptic Area/physiology , Proto-Oncogene Proteins c-fos/analysis , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A2A , Specific Pathogen-Free Organisms , Subarachnoid Space , Wakefulness/drug effects
15.
Electrophoresis ; 22(16): 3433-7, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11669522

ABSTRACT

Charge microheterogeneity of the beta-trace protein (beta-TP = lipocalin-type prostaglandin D synthase) in the cerebrospinal fluid (CSF) of patients with various neurological disorders was analyzed by capillary isoelectric focusing (CIEF). Under the conditions employed, beta-TP in the low-molecular-weight protein fraction of CSF was separated into at least four isoforms with different p/ values. An isoform with the pl value of 4.6-4.8 was usually the most abundant. The total beta-TP level in the CSF was determined by enzyme-linked immunosorbent assay (ELISA) to be elevated in patients recovering from organic damage to the CNS and those with pathological brain atrophy. Changes in the total beta-TP level in the CSF were occasionally accompanied by those in its charge microheterogeneity, as revealed by CIEF. Such quantitative and qualitative changes in beta-TP in human CSF indicated changes in its pathophysiological roles in association with various neurological disorders.


Subject(s)
Electrophoresis, Capillary/methods , Intramolecular Oxidoreductases/cerebrospinal fluid , Adult , Aged , Diagnostic Techniques, Neurological , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Isoelectric Focusing/methods , Isoenzymes/cerebrospinal fluid , Lipocalins , Male , Middle Aged , Nervous System Diseases/cerebrospinal fluid , Nervous System Diseases/diagnosis
16.
Proc Natl Acad Sci U S A ; 98(20): 11674-9, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11562489

ABSTRACT

Infusion of prostaglandin (PG) D(2) into the lateral ventricle of the brain induced an increase in the amount of non-rapid eye movement sleep in wild-type (WT) mice but not in mice deficient in the PGD receptor (DP). Immunofluorescence staining of WT mouse brain revealed that DP immunoreactivity was dominantly localized in the leptomeninges (LM) of the basal forebrain but that PGD synthase immunoreactivity was widely distributed in the LM of the entire brain. Electron microscopic observation indicated that DP-immunoreactive particles were predominantly located on the plasma membranes of arachnoid trabecular cells of the LM. The region with the highest DP immunoreactivity was clearly defined as bilateral wings in the LM of the basal forebrain located lateral to the optic chiasm in the proximity of the ventrolateral preoptic area, one of the putative sleep centers, and the tuberomammillary nucleus, one of the putative wake centers. The LM of this region contained DP mRNA 70-fold higher than that in the cortex as judged from the results of quantitative reverse transcription-PCR. PGD(2) infusion into the subarachnoid space of this region increased the extracellular adenosine level more than 2-fold in WT mice but not in the DP-deficient mice. These results indicate that DPs in the arachnoid trabecular cells of the basal forebrain mediate an increase in the extracellular adenosine level and sleep induction by PGD(2).


Subject(s)
Receptors, Calcitriol/genetics , Sleep/physiology , Adenosine/metabolism , Amino Acid Sequence , Anesthesia, General , Animals , Arachnoid/physiology , Base Sequence , DNA Primers , Electroencephalography , Electromyography , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Intramolecular Oxidoreductases/analysis , Kinetics , Lipocalins , Medulla Oblongata/physiology , Mice , Mice, Knockout , Molecular Sequence Data , Neocortex/physiology , Pentobarbital/pharmacology , Perfusion , Polymerase Chain Reaction , Prostaglandin D2/pharmacology , RNA/genetics , RNA/isolation & purification , RNA, Messenger/analysis , RNA, Messenger/genetics , Receptors, Calcitriol/analysis , Receptors, Calcitriol/chemistry , Sleep Stages/physiology , Sleep, REM/physiology , Subarachnoid Space/drug effects , Subarachnoid Space/metabolism
17.
Proc Natl Acad Sci U S A ; 98(17): 9965-70, 2001 Aug 14.
Article in English | MEDLINE | ID: mdl-11493714

ABSTRACT

Orexin neurons are exclusively localized in the lateral hypothalamic area and project their fibers to the entire central nervous system, including the histaminergic tuberomammillary nucleus (TMN). Dysfunction of the orexin system results in the sleep disorder narcolepsy, but the role of orexin in physiological sleep-wake regulation and the mechanisms involved remain to be elucidated. Here we provide several lines of evidence that orexin A induces wakefulness by means of the TMN and histamine H(1) receptor (H1R). Perfusion of orexin A (5 and 25 pmol/min) for 1 hr into the TMN of rats through a microdialysis probe promptly increased wakefulness for 2 hr after starting the perfusion by 2.5- and 4-fold, respectively, concomitant with a reduction in rapid eye movement (REM) and non-REM sleep. Microdialysis studies showed that application of orexin A to the TMN increased histamine release from both the medial preoptic area and the frontal cortex by approximately 2-fold over the baseline for 80 to 160 min in a dose-dependent manner. Furthermore, infusion of orexin A (1.5 pmol/min) for 6 hr into the lateral ventricle of mice produced a significant increase in wakefulness during the 8 hr after starting infusion to the same level as the wakefulness observed during the active period in wild-type mice, but not at all in H1R gene knockout mice. These findings strongly indicate that the arousal effect of orexin A depends on the activation of histaminergic neurotransmission mediated by H1R.


Subject(s)
Arousal/drug effects , Carrier Proteins/pharmacology , Histamine/physiology , Hypothalamic Area, Lateral/drug effects , Hypothalamus/physiology , Intracellular Signaling Peptides and Proteins , Nerve Tissue Proteins/drug effects , Neuropeptides/pharmacology , Receptors, Histamine H1/drug effects , Sleep/drug effects , Wakefulness/drug effects , Animals , Electroencephalography , Electromyography , Frontal Lobe/physiology , Hypothalamic Area, Lateral/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microdialysis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Orexin Receptors , Orexins , Preoptic Area/physiology , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled , Receptors, Histamine H1/deficiency , Receptors, Histamine H1/genetics , Receptors, Histamine H1/physiology , Receptors, Neuropeptide
18.
J Biol Chem ; 276(37): 34918-27, 2001 Sep 14.
Article in English | MEDLINE | ID: mdl-11418589

ABSTRACT

We have recently shown that two distinct prostaglandin (PG) E(2) synthases show preferential functional coupling with upstream cyclooxygenase (COX)-1 and COX-2 in PGE(2) biosynthesis. To investigate whether other lineage-specific PG synthases also show preferential coupling with either COX isozyme, we introduced these enzymes alone or in combination into 293 cells to reconstitute their functional interrelationship. As did the membrane-bound PGE(2) synthase, the perinuclear enzymes thromboxane synthase and PGI(2) synthase generated their respective products via COX-2 in preference to COX-1 in both the -induced immediate and interleukin-1-induced delayed responses. Hematopoietic PGD(2) synthase preferentially used COX-1 and COX-2 in the -induced immediate and interleukin-1-induced delayed PGD(2)-biosynthetic responses, respectively. This enzyme underwent stimulus-dependent translocation from the cytosol to perinuclear compartments, where COX-1 or COX-2 exists. COX selectivity of these lineage-specific PG synthases was also significantly affected by the concentrations of arachidonate, which was added exogenously to the cells or supplied endogenously by the action of cytosolic or secretory phospholipase A(2). Collectively, the efficiency of coupling between COXs and specific PG synthases may be crucially influenced by their spatial and temporal compartmentalization and by the amount of arachidonate supplied by PLA(2)s at a moment when PG production takes place.


Subject(s)
Isoenzymes/physiology , Phospholipases A/physiology , Prostaglandin-Endoperoxide Synthases/physiology , Prostaglandins/biosynthesis , Arachidonic Acid/metabolism , Calcimycin/pharmacology , Cell Line , Cyclooxygenase 1 , Cyclooxygenase 2 , Humans , Isoenzymes/analysis , Membrane Proteins , Phospholipases A/analysis , Phospholipases A2 , Prostaglandin-Endoperoxide Synthases/analysis , Transfection
19.
J Immunol ; 166(9): 5782-91, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11313422

ABSTRACT

Xeroderma pigmentosum group A (XPA) gene-deficient mice easily developed skin cancers by the application of topical chemical carcinogens as well as by UV irradiation. As certain chemical carcinogens have been shown to be immunosuppressive, we examined the inflammatory and immunosuppressive effects of dimethylbenz(a)anthracene (DMBA) on XPA mice. Compared with wild-type mice, XPA mice showed greater ear swelling and reduction of epidermal Langerhans cells after DMBA application. Topical application of DMBA impaired the induction of contact hypersensitivity, initiated either locally or at distant sites. These DMBA-induced local and systemic immunosuppressions were more greatly enhanced in XPA mice than in wild-type mice. DMBA application induced pronounced production of PGE(2), IL-10, and TNF-alpha in the skin of XPA mice. Treatment with indomethacin, a potent inhibitor of PG biosynthesis, inhibited DMBA-induced inflammation and local immunosuppression. In XPA mice, increased serum IL-10 was detected after DMBA treatment. Excess production of PGE(2), TNF-alpha, and IL-10 after DMBA application may be involved in the enhanced local and systemic immunosuppression in DMBA-treated XPA mice. Susceptibility to DMBA-induced skin tumors in XPA mice may be due to easy impairment of the immune system by DMBA in addition to a defect in the repair of DMBA-DNA adduct. Enhanced immunosuppression by chemical carcinogens as well as the mutagenicity of these mutagens might be associated with the high incidence of internal malignancies seen in XP patients. Moreover, these results supported the hypothesis that persistent DNA damage is a trigger for the production of immunoregulatory cytokines.


Subject(s)
9,10-Dimethyl-1,2-benzanthracene/toxicity , Carcinogens/toxicity , DNA-Binding Proteins/genetics , Dinoprostone/biosynthesis , Immunosuppressive Agents/toxicity , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/immunology , 9,10-Dimethyl-1,2-benzanthracene/antagonists & inhibitors , Adjuvants, Immunologic/antagonists & inhibitors , Adjuvants, Immunologic/toxicity , Administration, Cutaneous , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Apyrase/biosynthesis , Carcinogens/antagonists & inhibitors , Dermatitis, Contact/genetics , Dermatitis, Contact/immunology , Dermatitis, Contact/prevention & control , Dinitrofluorobenzene/administration & dosage , Disease Models, Animal , Ear/pathology , Edema/chemically induced , Edema/genetics , Edema/immunology , Edema/prevention & control , Female , Immunosuppressive Agents/antagonists & inhibitors , Indomethacin/administration & dosage , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Interleukin-10/biosynthesis , Interleukin-10/blood , Interleukin-10/metabolism , Langerhans Cells/drug effects , Langerhans Cells/enzymology , Langerhans Cells/pathology , Mice , Mice, Hairless , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Skin/immunology , Skin/metabolism , Skin/pathology , Tumor Necrosis Factor-alpha/biosynthesis , Up-Regulation/genetics , Up-Regulation/immunology , Xeroderma Pigmentosum/metabolism , Xeroderma Pigmentosum/pathology , Xeroderma Pigmentosum Group A Protein
20.
Nephron ; 87(4): 321-7, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11287775

ABSTRACT

OBJECTIVE: Circulating levels of lipocalin-type prostaglandin D synthase (L-PGDS)/beta-trace reportedly increase in renal failure as well as in cardiovascular injuries. We investigated the alterations of L-PGDS in urine and plasma in the early stage of type-2 diabetic patients. METHOD: Thirty-six type-2 diabetic patients and 29 normal subjects were studied. Overnight spot urine and plasma samples were obtained in the morning. L-PGDS was measured by ELISA method using anti-L-PGDS antibody. Variables indicating renal function were determined. RESULTS: Plasma L-PGDS concentration was slightly higher in the patients with diabetes mellitus than in the control subjects, whereas the urinary L-PGDS excretion almost doubled in the diabetic patients as compared with that in the control subjects. Plasma L-PGDS was determined by plasma creatinine (Cr) concentration while urinary L-PGDS excretion was correlated solely with urinary protein excretion. There was no relationship between plasma L-PGDS concentration and urinary L-PGDS excretion. The averaged plasma concentration of L-PGDS in the diabetics with a normal Cr level in plasma, corresponding to that in the controls, was determined by the plasma Cr concentration. On the other hand, the urinary L-PGDS excretion was determined by the amount of proteinuria and greater in the diabetics with a normal Cr level in plasma than in the controls even when the patients exhibited urinary protein excretion equal to that in the control subjects. CONCLUSIONS: Urinary L-PGDS excretion increased in the early stage of kidney injury in patients with type-2 diabetes mellitus. The urinary excretion was correlated independently with urinary protein excretion even when there was no difference in urinary protein or albumin excretions, thereby suggesting that urinary L-PGDS excretion is possibly a more sensitive indicator of renal injuries than proteinuria. Urinary L-PGDS may thus predict the progression of renal injuries in diabetic patients.


Subject(s)
Diabetes Mellitus, Type 2/urine , Diabetic Nephropathies/urine , Intramolecular Oxidoreductases/urine , Analysis of Variance , Biomarkers/urine , Blood Glucose/metabolism , Cholesterol/blood , Creatinine/blood , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/physiopathology , Diabetic Nephropathies/diagnosis , Diabetic Nephropathies/physiopathology , Enzyme-Linked Immunosorbent Assay , Glycated Hemoglobin/analysis , Humans , Intramolecular Oxidoreductases/blood , Kidney Function Tests , Lipocalins , Middle Aged , Reference Values , Regression Analysis , Sensitivity and Specificity , Triglycerides/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...