Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Shock ; 56(1): 142-149, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33351449

ABSTRACT

ABSTRACT: Sepsis-associated encephalopathy (SAE) is a diffuse brain dysfunction associated with sepsis. The development of an effective strategy for early diagnosis and therapeutic intervention is essential for the prevention of poor prognosis of SAE. Translocator protein 18 kDa (TSPO) is a mitochondrial protein implicated in steroidogenesis and inflammatory responses. Despite accumulating evidence that implicates TSPO in the neuroinflammatory response of the central nervous system, the possible role of TSPO in SAE remains unclear. The aim of this study is to address a role of TSPO in neuroinflammation using mice 24 h after systemic injection of LPS, which consistently demonstrated microglial activation and behavioral inhibition. Quantitative polymerase chain reaction analysis revealed that hippocampal TSPO expression was induced following the systemic LPS injection, associated with an increase in pro-inflammatory cytokines such as tumor necrosis factor-α and interleukin-1ß. Interestingly, pretreatment with the TSPO antagonist, ONO-2952, or germ-line deletion of the TSPO gene exhibited an anti-inflammatory effect with significant suppression of LPS-induced production of those cytokines. These effects demonstrated by the ONO-2952 or TSPO knockout were associated with significant recovery from behavioral inhibition, as shown by improved locomotor activity in the open field analysis. Histological analysis revealed that ONO-2952 pretreatment suppressed the LPS-induced activation of TSPO-expressing microglia in the hippocampus of mice. Collectively, these results suggest that TSPO plays a critical role in the SAE mouse model. Based on this finding, monitoring TSPO activity, as well as the progress of endotoxemia and its sequelae in the animal model, would deepen our understanding of the underlying molecular mechanism of SAE.


Subject(s)
Endotoxemia/drug therapy , Endotoxemia/genetics , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/genetics , Receptors, GABA/genetics , Sepsis-Associated Encephalopathy/drug therapy , Sepsis-Associated Encephalopathy/genetics , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
2.
J Pharmacol Exp Ther ; 376(2): 250-260, 2021 02.
Article in English | MEDLINE | ID: mdl-33257316

ABSTRACT

ONO-4641 is a second-generation sphingosine 1-phosphate (S1P) receptor modulator that exhibits selectivity for S1P receptors 1 and 5. Treatment with ONO-4641 leads to a reduction in magnetic resonance imaging disease measures in patients with relapsing-remitting multiple sclerosis. The objective of this study was to explore the potential impact of ONO-4641 treatment based on its immunomodulatory effects. Severe aplastic anemia is a bone marrow (BM) failure disease typically caused by aberrant immune destruction of blood progenitors. Although the T helper type 1-mediated pathology is well described for aplastic anemia, the molecular mechanisms driving disease progression remain undefined. We evaluated the efficacy of ONO-4641 in a mouse model of aplastic anemia. ONO-4641 reduced the severity of BM failure in a dose-dependent manner, resulting in higher blood and BM cell counts. By evaluating the mode of action, we found that ONO-4641 inhibited the infiltration of donor-derived T lymphocytes to the BM. ONO-4641 also induced the accumulation of hematopoietic stem cells in the BM of model mice. These observations indicate, for the first time, that S1P receptor modulators demonstrate efficacy in the mouse model of aplastic anemia and suggest that treatment with ONO-4641 might delay the progression of aplastic anemia. SIGNIFICANCE STATEMENT: ONO-4641 is a second-generation sphingosine 1-phosphate (S1P) receptor modulator selective for S1P receptors 1 and 5. In this study, we demonstrated that ONO-4641 regulates the trafficking of T lymphocytes along with hematopoietic stem and progenitor cells, leading to alleviation of pancytopenia and destruction of bone marrow in a bone marrow failure-induced mouse model mimicking human aplastic anemia.


Subject(s)
Anemia, Aplastic/drug therapy , Azetidines/pharmacology , Hematopoietic Stem Cells/drug effects , Naphthalenes/pharmacology , Sphingosine 1 Phosphate Receptor Modulators/pharmacology , T-Lymphocytes/drug effects , Anemia, Aplastic/immunology , Animals , Azetidines/therapeutic use , Cell Movement , Cells, Cultured , Hematopoietic Stem Cells/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Naphthalenes/therapeutic use , Sphingosine 1 Phosphate Receptor Modulators/therapeutic use , Sphingosine-1-Phosphate Receptors/metabolism , T-Lymphocytes/physiology
3.
Neuropharmacology ; 162: 107835, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31682855

ABSTRACT

In preclinical models, it has been reported that social defeat stress activates microglial cells in the CNS. Translocator protein 18 kDa (TSPO) is a mitochondrial protein expressed on microglia in the CNS that has been proposed to be a useful biomarker for brain injury and inflammation. We hypothesized that a TSPO antagonist, ONO-2952, would inhibit the neuroinflammation induced by microglial hyperactivation and associated depressive-like behaviors. An in vitro analysis showed that ONO-2952 suppressed the release of pro-inflammatory cytokines and mitochondrial reactive oxygen species in cultured microglia stimulated by lipopolysaccharide. In mice submitted to chronic social defeat stress, microglia predominantly expressed TSPO in limbic areas implicated in depressive-like behaviors, including the amygdala, ventral hippocampus and nucleus accumbens, in which an increase in the production of pro-inflammatory cytokines in vivo were associated. Treating animals with ONO-2952 during chronic social defeat stress ameliorated impairments in social avoidance and anxiety-like behaviors and suppressed pro-inflammatory cytokine production, suggesting that ONO-2952 exerted an anti-stress effect in this animal model of depression. Thus, targeting TSPO as a candidate for the development of antidepressants that reduce susceptibility to chronic stress could pave the way toward therapeutic interventions for relapse prophylaxis in depression.


Subject(s)
Antidepressive Agents/pharmacology , Brain/drug effects , Cyclopropanes/pharmacology , GABA-A Receptor Antagonists/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Microglia/drug effects , Receptors, GABA/drug effects , Social Defeat , Stress, Psychological/metabolism , Amygdala/drug effects , Amygdala/metabolism , Animals , Anxiety/metabolism , Anxiety/psychology , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Brain/metabolism , Cytokines/drug effects , Cytokines/metabolism , Disease Models, Animal , Elevated Plus Maze Test , Hindlimb Suspension , Hippocampus/drug effects , Hippocampus/metabolism , In Vitro Techniques , Lipopolysaccharides/toxicity , Mice , Microglia/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Open Field Test , Reactive Oxygen Species/metabolism , Receptors, GABA/metabolism , Social Behavior , Stress, Psychological/psychology
4.
J Pharmacol Exp Ther ; 366(1): 58-65, 2018 07.
Article in English | MEDLINE | ID: mdl-29674331

ABSTRACT

GABAA receptors containing α5 subunits (GABAAα5) are highly expressed in the hippocampus and negatively involved in memory processing, as shown by the fact that GABAAα5-deficient mice show higher hippocampus-dependent performance than wild-type mice. Accordingly, small-molecule GABAAα5 negative allosteric modulators (NAMs) are known to enhance spatial learning and memory in rodents. Here we introduce a new, orally available GABAAα5 NAM that improves hippocampal functions. ONO-8590580 [1-(cyclopropylmethyl)-5-fluoro-4-methyl-N-[5-(1-methyl-1H-imidazol-4-yl)-2-pyridinyl]-1H-benzimidazol-6-amine] binds to the benzodiazepine binding sites on recombinant human α5-containing GABAA receptors with a Ki of 7.9 nM, and showed functionally selective GABAAα5 NAM activity for GABA-induced Cl- channel activity with a maximum 44.4% inhibition and an EC50 of 1.1 nM. In rat hippocampal slices, tetanus-induced long-term potentiation of CA1 synapse response was significantly augmented in the presence of 300 nM ONO-8590580. Orally administered ONO-8590580 (1-20 mg/kg) dose-dependently occupied hippocampal GABAAα5 in a range of 40%-90% at 1 hour after intake. In the rat passive avoidance test, ONO-8590580 (3-20 mg/kg, by mouth) significantly prevented (+)-MK-801 hydrogen maleate (MK-801)-induced memory deficit. In addition, ONO-8590580 (20 mg/kg, p.o.) was also effective in improving the cognitive deficit induced by scopolamine and MK-801 in the rat eight-arm radial maze test with equal or greater activity than 0.5 mg/kg donepezil. No anxiogenic-like or proconvulsant effect was associated with ONO-8590580 at 20 mg/kg p.o. in the elevated plus maze test or pentylenetetrazole-induced seizure test, respectively. In sum, ONO-8590580 is a novel GABAAα5 NAM that enhances hippocampal memory function without an anxiogenic or proconvulsant risk.


Subject(s)
Cognition/drug effects , Imidazoles/pharmacology , Long-Term Potentiation/drug effects , Pyridines/pharmacology , Receptors, GABA-A/metabolism , Allosteric Regulation/drug effects , Animals , Avoidance Learning/drug effects , HEK293 Cells , Hippocampus/drug effects , Hippocampus/physiology , Humans , Imidazoles/therapeutic use , Male , Maze Learning/drug effects , Pyridines/therapeutic use , Rats , Rats, Sprague-Dawley
6.
J Pharmacol Exp Ther ; 360(3): 457-465, 2017 03.
Article in English | MEDLINE | ID: mdl-27986860

ABSTRACT

We have previously shown that ONO-2952, a novel 18-kDa translocator protein (TSPO) antagonist, inhibits stress-induced accumulation of neurosteroids and noradrenaline release in the rat brain and alleviates the subsequent symptomatic responses with a brain TSPO occupancy of 50% or more. In this study, we measured ONO-2952 brain TSPO occupancy in conscious rhesus monkeys using positron emission tomography (PET) with 11C-PBR28 as ligand for translational research to clinical application. PET scans were performed after single and repeated oral administration of ONO-2952 at several dose levels for each animal, with sequential arterial blood sampling. In vitro binding studies showed that ONO-2952 potently binds to brain TSPO in monkeys with an affinity equivalent to that in rats. ONO-2952, given orally before PET scans, dose dependently decreased 11C-PBR28 uptake without marked brain region specificity. Results of the quantitative analysis using arterial input function revealed that TSPO occupancy after ONO-2952 single and repeated oral administration tended to increase in parallel with its plasma concentration, reaching the highest level of 100%. These findings indicate that ONO-2952 has sufficient brain distribution in primates and that ONO-2952 TSPO occupancy in humans can also be determined using PET.


Subject(s)
Brain , Consciousness/drug effects , Cyclopropanes/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Positron-Emission Tomography/methods , Acetamides/pharmacology , Administration, Oral , Animals , Brain/diagnostic imaging , Brain/drug effects , Brain/metabolism , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Central Nervous System Agents/pharmacology , Consciousness/physiology , Dose-Response Relationship, Drug , Haplorhini , Pyridines/pharmacology , Radiopharmaceuticals/pharmacology , Rats , Receptors, GABA-A/metabolism , Tissue Distribution
7.
Neuropharmacology ; 99: 51-66, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26189762

ABSTRACT

Accumulating evidence has shown the pathophysiological significance of the translocator protein 18 kDa (TSPO) in the central nervous system. In this study, we evaluated the beneficial effects of ONO-2952, a novel TSPO antagonist in rat stress models. ONO-2952 potently bound both rat and human TSPO (Ki=0.330-9.30 nmol/L) with high selectivity over other receptors, transporters, ion channels and enzymes. ONO-2952 inhibited both neurosteroid accumulation and noradrenaline release in the brain of rats exposed to acute stress. The inhibitory effect of ONO-2952 on stress-induced noradrenaline release was attenuated by co-treatment with the TSPO agonist CB34 in a dose-dependent manner. ONO-2952, at 0.3 mg/kg or higher, dose-dependently suppressed restraint stress-induced defecation in rats with brain TSPO occupancy of more than 50%. In addition, ONO-2952, at 1 mg/kg or higher, suppressed conditioned fear stress-induced freezing behavior in rats with an efficacy equivalent to that of diazepam, given orally at 3 mg/kg. Results of the passive avoidance learning test revealed that ONO-2952, unlike diazepam, did not affect learning and memory even at doses 10 times higher than its effective doses in the stress models. The present findings indicate that ONO-2952 is a promising candidate for the treatment of stress-related disorders.


Subject(s)
Cyclopropanes/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Psychotropic Drugs/pharmacology , Stress, Psychological/drug therapy , Amygdala/drug effects , Amygdala/metabolism , Animals , Avoidance Learning/drug effects , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Line, Tumor , Cyclopropanes/chemistry , Cyclopropanes/pharmacokinetics , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Freezing Reaction, Cataleptic/drug effects , Heterocyclic Compounds, 4 or More Rings/chemistry , Heterocyclic Compounds, 4 or More Rings/pharmacokinetics , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Male , Psychotropic Drugs/chemistry , Psychotropic Drugs/pharmacokinetics , Rats, Sprague-Dawley , Rats, Wistar , Receptors, GABA/metabolism , Receptors, GABA-A/metabolism , Restraint, Physical , Stress, Psychological/metabolism
8.
Bioorg Med Chem ; 19(20): 5955-66, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21930387

ABSTRACT

To identify structurally novel CRF1 receptor antagonists, a series of bicyclic core antagonists, pyrazolo[1,5-a]pyrimidines, triazolo[1,5-a]pyrimidines, imidazo[1,2-a]pyrimidines and pyrazolo[1,5-a][1,3,5]triazines were designed, synthesized and evaluated as CRF1 receptor antagonists. Compounds 2-27 showed binding affinity (IC(50)=4.2-418 nM) and antagonist activity (EC(50)=4.0-889 nM). Compound 5 was found to show oral efficacy in an Elevated Plus Maze test in rats. Further chemical modification of them led us to discovery of the tricyclic core antagonists pyrazolo[1,5-a]pyrrolo[3,2-e]pyrimidines. The discovery process of these compounds is presented, as is the study of the structure-activity relationship.


Subject(s)
Pyrimidines/chemistry , Pyrimidines/pharmacology , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/chemistry , Animals , Humans , Kinetics , Male , Protein Binding , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
9.
Bioorg Med Chem ; 19(18): 5432-45, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21865047

ABSTRACT

To identify an orally active corticotropin-releasing factor 1 receptor antagonist, a series of 6,7-dihydro-5H-cyclopenta[d]pyrazolo[1,5-a]pyrimidines and their derivatives were designed, synthesized and evaluated. An in vitro study followed by in vivo and pharmacokinetic studies of these heterotricyclic compounds led us to the discovery of an orally active CRF1 receptor antagonist. The results of a structure-activity relationship study are presented.


Subject(s)
Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Animals , CHO Cells , Cricetinae , Dose-Response Relationship, Drug , Drug Design , Male , Maze Learning/drug effects , Molecular Structure , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship , Swine
10.
Brain Res ; 1089(1): 79-91, 2006 May 17.
Article in English | MEDLINE | ID: mdl-16643860

ABSTRACT

Although astrocytes express gamma-aminobutyric acid subtype-A (GABAA) receptors in the mature brain, GABAA receptor expression in a cultivation state remains controversial. In this study, we investigated the alteration of astrocytic GABAA receptor expression in in vitro and in vivo studies to elucidate the relevance of astrocytic activation to reductions of astrocytic GABAA receptors. The GABA-evoked Cl- current (GABAA response) in cultured astrocytes was determined by recording in the whole-cell mode using a conventional patch-clamp technique under voltage-clamp conditions. The respective amplitudes of GABAA responses on days in vitro 1, 3-5, 7-10, and 12-15 were 1019+/-97, 512+/-76, 84+/-21, and 22+/-9 pA, respectively, suggesting that the GABAA response subsequently diminished with in vitro aging. In immunohistochemical and biochemical analyses, the expression of GABAA receptor beta-subunit decreased, whereas expressions of glial fibrillary acidic protein (GFAP) and S100B, hallmarks of astrocytic activation, increased dramatically in the cultured astrocytes with in vitro aging. With the use of [3H]SR95531, a GABAA-specific ligand, at 24 h after transient focal ischemia, binding was significantly reduced in the astrocytic fractions without affecting the synaptosomal fractions, and decreases in the mRNA expression level of GABAA receptor beta-subunits were concurrently observed. Interestingly, the loss of GABAA response in cultured astrocytes was mitigated by co-culturing with neurons or treatments with monoclonal S100B antibodies. These results indicate that astrocytic GABAA receptors are reduced with in vitro aging and cerebral ischemia, presumably through the overproduction of S100B in activated astrocytes.


Subject(s)
Aging/metabolism , Astrocytes/metabolism , Brain Ischemia/metabolism , Cerebral Cortex/metabolism , Gliosis/metabolism , Receptors, GABA-A/metabolism , Animals , Animals, Newborn , Biomarkers/metabolism , Brain Ischemia/physiopathology , Cell Communication/physiology , Cell Membrane/physiology , Cells, Cultured , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Coculture Techniques , Down-Regulation/physiology , Female , Glial Fibrillary Acidic Protein/metabolism , Gliosis/physiopathology , Male , Membrane Potentials/physiology , Nerve Growth Factors/metabolism , Patch-Clamp Techniques , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Rats, Wistar , Receptors, GABA-A/genetics , S100 Calcium Binding Protein beta Subunit , S100 Proteins/metabolism , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...