Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem Lett ; 92: 129408, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37429500

ABSTRACT

A series of thalidomide analogues, where the fused benzene ring in the phthalimide moiety was converted into two separated diphenyl rings in maleimide moiety and N-aminoglutarimide moiety was replaced by substituted phenyl moiety, were synthesized and evaluated for their NO inhibitory activities on BV2 cells stimulated with lipopolysaccharide (LPS). Among the synthesized compounds, the dimethylaminophenyl analogue 1s (IC50 = 7.1 µM) showed significantly higher inhibitory activity than the glutarimide analogue 1a (IC50 > 50 µM) and suppressed NO production dose-dependently without cytotoxicity. In addition, 1s inhibited the production of pro-inflammatory cytokines and the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) by blocking nuclear factor-kappa B (NF-κB) and p38 MAPK pathways. These results demonstrated that 1s showed good anti-inflammatory activity and could become a leading compound for the treatment of neuroinflammatory diseases.


Subject(s)
Lipopolysaccharides , Pyrroles , Lipopolysaccharides/pharmacology , Pyrroles/metabolism , Anti-Inflammatory Agents , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Microglia/metabolism , Cyclooxygenase 2/metabolism
2.
Antioxidants (Basel) ; 12(3)2023 Mar 05.
Article in English | MEDLINE | ID: mdl-36978896

ABSTRACT

Cognitive decline and memory impairment induced by oxidative brain damage are the critical pathological hallmarks of Alzheimer's disease (AD). Based on the potential neuroprotective effects of AD-1 small molecule, we here explored the possible underlying mechanisms of the protective effect of AD-1 small molecule against scopolamine-induced oxidative stress, neuroinflammation, and neuronal apoptosis. According to our findings, scopolamine administration resulted in increased AChE activity, MDA levels, and decreased antioxidant enzymes, as well as the downregulation of the antioxidant response proteins of Nrf2 and HO-1 expression; however, treatment with AD-1 small molecule mitigated the generation of oxidant factors while restoring the antioxidant enzymes status, in addition to improving antioxidant protein levels. Similarly, AD-1 small molecule significantly increased the protein expression of neuroprotective markers such as BDNF and CREB and promoted memory processes in scopolamine-induced mice. Western blot analysis showed that AD-1 small molecule reduced activated microglia and astrocytes via the attenuation of iba-1 and GFAP protein expression. We also found that scopolamine enhanced the phosphorylation of NF-κB/MAPK signaling and, conversely, that AD-1 small molecule significantly inhibited the phosphorylation of NF-κB/MAPK signaling in the brain regions of hippocampus and cortex. We further found that scopolamine promoted neuronal loss by inducing Bax and caspase-3 and reducing the levels of the antiapoptotic protein Bcl-2. In contrast, AD-1 small molecule significantly decreased the levels of apoptotic markers and increased neuronal survival. Furthermore, AD-1 small molecule ameliorated scopolamine-induced impairments in spatial learning behavior and memory formation. These findings revealed that AD-1 small molecule attenuated scopolamine-induced cognitive and memory dysfunction by ameliorating AChE activity, oxidative brain damage, neuroinflammation, and neuronal apoptosis.

3.
Bioorg Med Chem Lett ; 79: 129061, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36371018

ABSTRACT

A series of rimonabant analogues, where the N-aminopiperidine moiety was replaced by various amines and an additional carbonyl group, were synthesized and their inhibition of nitric oxide (NO) production was evaluated in lipopolysaccharide (LPS)-induced BV2 microglial cells. Among the synthesized compounds, the morpholine analogue 7y (IC50 = 4.71 ±â€¯0.11 µM) showed significantly higher inhibitory activity than rimonabant (IC50 = 16.17 ±â€¯0.56 µM), and suppressed NO production dose-dependently without cytotoxicity. In addition, 7y inhibited the expression of iNOS, COX-2 and pro-inflammatory cytokines and attenuated LPS-induced activation of nuclear factor-kappa B (NF-κB) and ERK MAPK phosphorylation in BV2 cells. These results demonstrated that 7y exerted anti-inflammatory effects by ERK pathway in BV2 cells, which can be used for the prevention and treatment of neuroinflammatory diseases.


Subject(s)
Anti-Inflammatory Agents , Lipopolysaccharides , Rimonabant , Anti-Inflammatory Agents/pharmacology , Cyclooxygenase 2/metabolism , Lipopolysaccharides/pharmacology , Microglia , NF-kappa B/metabolism , Nitric Oxide , Nitric Oxide Synthase Type II/metabolism , Rimonabant/analogs & derivatives , Rimonabant/chemistry , Rimonabant/pharmacology
4.
Biol Pharm Bull ; 44(9): 1230-1238, 2021.
Article in English | MEDLINE | ID: mdl-34471051

ABSTRACT

A series of salicylic acid analogues of celecoxib where the phenylsulfonamide moiety in the structure of celecoxib is replaced by salicylic acid moiety was synthesized and tested for in vitro cyclooxygenase (COX)-1 and COX-2 enzyme inhibition. Among the series, 5-substituted-2-hydroxy-benzoic acid analogues (7a-7h) generally showed better inhibitory activities on both enzymes than 4-substituted-2-hydroxy-benzoic acid analogues (12a-12h). In particular, the chloro analogue 7f which had the highest inhibitory effect (IC50 = 0.0057 µM) to COX-1 with excellent COX-1 selectivity (SI = 768) can be classified as a new potent and selective COX-1 inhibitor. The high inhibitory potency of 7f was rationalized through the docking simulation of this analogue in the active site of COX-1 enzyme.


Subject(s)
Celecoxib/analogs & derivatives , Cyclooxygenase 1/metabolism , Cyclooxygenase Inhibitors/pharmacology , Salicylates/pharmacology , Catalytic Domain/drug effects , Celecoxib/chemistry , Cyclooxygenase Inhibitors/chemical synthesis , Enzyme Assays , Molecular Docking Simulation , Molecular Structure , Salicylates/chemical synthesis , Structure-Activity Relationship
5.
Bioorg Med Chem Lett ; 36: 127780, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33422605

ABSTRACT

Based on our previous report that 3-morpholino-1-phenylpropan-1-one 2, one of the fluoxetine's simplified morpholino analogue, inhibited nitric oxide (NO) production, in this paper, various substituted benzene analogues with morpholine hydrochloride of 2 were synthesized and their inhibitory effects on NO production in lipopolysaccharide (LPS)-induced BV2 cells were tested. Among the synthesized compounds, 2-trifluoromethyl analogue 16n (IC50 = 8.6 µM) showed a significantly higher inhibitory activity than that of the parent compound 2a (IC50 > 50 µM) and suppressed NO production dose-dependently without cytotoxicity. Compound 16n also inhibited iNOS expression in LPS-induced BV2 cells at 2, 10 and 20 µM concentrations. These results suggest that compound 16n inhibited NO production by suppressing the expression of iNOS and can be used as a lead structure for developing new inhibitor of NO production.


Subject(s)
Chlorides/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Morpholines/pharmacology , Nitric Oxide/antagonists & inhibitors , Animals , Cell Line , Chlorides/chemical synthesis , Chlorides/chemistry , Dose-Response Relationship, Drug , Lipopolysaccharides/pharmacology , Mice , Molecular Structure , Morpholines/chemical synthesis , Morpholines/chemistry , Nitric Oxide/biosynthesis , Structure-Activity Relationship
6.
Int J Mol Sci ; 21(21)2020 Nov 02.
Article in English | MEDLINE | ID: mdl-33147699

ABSTRACT

Microglia-mediated neuroinflammation is one of the key mechanisms involved in acute brain injury and chronic neurodegeneration. This study investigated the inhibitory effects of 2-hydroxy-4-methylbenzoic anhydride (HMA), a novel synthetic derivative of HTB (3-hydroxy-4-trifluoromethylbenzoic acid) on neuroinflammation and underlying mechanisms in activated microglia in vitro and an in vivo mouse model of Parkinson's disease (PD). In vitro studies revealed that HMA significantly inhibited lipopolysaccharide (LPS)-stimulated excessive release of nitric oxide (NO) in a concentration dependent manner. In addition, HMA significantly suppressed both inducible NO synthase and cyclooxygenase-2 (COX-2) at the mRNA and protein levels in LPS-stimulated BV-2 microglia cells. Moreover, HMA significantly inhibited the proinflammatory cytokines such as interleukin (IL)-1beta, IL-6, and tumor necrosis factor-alpha in LPS-stimulated BV-2 microglial cells. Furthermore, mechanistic studies ensured that the potent anti-neuroinflammatory effects of HMA (0.1, 1.0, and 10 µM) were mediated by phosphorylation of nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha (IκBα) in LPS-stimulated BV-2 cells. In vivo evaluations revealed that intraperitoneal administration of potent neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP, 20 mg/kg, four times a 1 day) in mice resulted in activation of microglia in the brain in association with severe behavioral deficits as assessed using a pole test. However, prevention of microglial activation and attenuation of Parkinson's disease (PD)-like behavioral changes was obtained by oral administration of HMA (30 mg/kg) for 14 days. Considering the overall results, our study showed that HMA exhibited strong anti-neuroinflammatory effects at lower concentrations than its parent compound. Further work is warranted in other animal and genetic models of PD for evaluating the efficacy of HMA to develop a potential therapeutic agent in the treatment of microglia-mediated neuroinflammatory disorders, including PD.


Subject(s)
Benzoates/pharmacology , Cyclooxygenase 2/metabolism , Inflammation/drug therapy , Neurons/drug effects , Parkinson Disease/drug therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Administration, Oral , Animals , Cell Survival , Disease Models, Animal , Drug Design , In Vitro Techniques , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Models, Theoretical , Neuroglia/metabolism , Nitric Oxide/metabolism , Peptides/chemistry , Phosphorylation , Salicylates/chemistry , Signal Transduction
7.
Neuropharmacology ; 166: 107928, 2020 04.
Article in English | MEDLINE | ID: mdl-31887307

ABSTRACT

Parkinson's disease (PD) is characterized by the selective loss of nigrostriatal dopamine neurons associated with microglial activation. Inhibition of the inflammatory response elicited by activated microglia could be an effective strategy to alleviate the progression of PD. Here, we synthesized 2-(5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazol-3-yl)-N-(2-hydroxyethyl)-2-oxoacetamide (CDMPO) and studied its protective anti-inflammatory mechanisms following lipopolysaccharide (LPS)-induced neuroinflammation in vitro and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicity in vivo. CDMPO and its parent compound, rimonabant, significantly attenuated nitric oxide (NO) production in LPS-stimulated primary microglia and BV2 cells. Furthermore, CDMPO significantly inhibited the release of proinflammatory cytokines and prostaglandin E2 (PGE2) by activated BV2 cells, also suppressed expression of inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2). Mechanistically, CDMPO attenuated LPS-induced activation of nuclear factor-kappa B (NF-κB), inhibitor of kappa B alpha (IκBα), and p38 phosphorylation in BV2 cells. MPTP intoxication of mice results in glial activation, tyrosine hydroxylase (TH) depletion, and significant behavioral deficits. Prophylactic treatment with CDMPO decreased proinflammatory molecules via NF-κB and p38 mitogen-activated protein kinase signaling, resulting in protection of dopaminergic neurons and improved behavioral impairments. These results suggest that CDMPO is a promising neuroprotective agent for the prevention and treatment of microglia-mediated neuroinflammatory conditions and may be useful for behavioral improvement in PD phenotype.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Inflammation Mediators/antagonists & inhibitors , Locomotion/drug effects , Microglia/drug effects , Parkinsonian Disorders/drug therapy , Rimonabant/pharmacology , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Cannabinoid Receptor Antagonists/chemistry , Cannabinoid Receptor Antagonists/pharmacology , Cannabinoid Receptor Antagonists/therapeutic use , Cell Line , Dose-Response Relationship, Drug , Inflammation Mediators/metabolism , Locomotion/physiology , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/psychology , Rimonabant/analogs & derivatives , Rimonabant/therapeutic use
8.
Neurotherapeutics ; 16(2): 523-537, 2019 04.
Article in English | MEDLINE | ID: mdl-30680637

ABSTRACT

Ethyl pyruvate (EP) is a simple aliphatic ester of pyruvic acid and has been shown to have protective properties, which have been attributed to its anti-inflammatory, anti-oxidative, and anti-apoptotic functions. In an effort to develop better derivatives of EP, we previously synthesized DEOPA (N,N-diethyl-2-oxopropanamide, a novel isoster of EP) which has greater neuroprotective effects than EP, probably due to its anti-inflammatory and anti-excitotoxic effects. In the present study, we synthesized 3 DEOPA derivatives, in which its diethylamino group was substituted with diisopropylamino, dipropylamino, or diisobutylamino groups. Among them, DIPOPA (N,N-diisopropyl-2-oxopropanamide) containing diisopropylamino group had a greater neuroprotective effect than DEOPA or EP when administered intravenously to a rat middle cerebral artery occlusion (MCAO) model at 9 h after MCAO. Furthermore, DIPOPA had a wider therapeutic window than DEOPA and a marked reduction of infarct volume was accompanied by greater neurological and behavioral improvements. In particular, DIPOPA exerted robust anti-inflammatory effects, as evidenced by marked suppressions of microglia activation and neutrophil infiltration in the MCAO model, in microglial cells, and in neutrophil-endothelial cocultures at lower concentration, and did so more effectively than DEOPA. In particular, DIPOPA remarkably suppressed neutrophil infiltration into brain parenchyma, and this effect was attributed to the expressional inhibitions of cell adhesion molecules in neutrophils of brain parenchyma and in circulating neutrophils via NF-κB inhibition. Together, these results indicate the robust neuroprotective effects of DIPOPA are attributable to its anti-inflammatory effects and suggest that DIPOPA offers a potential therapeutic means of ameliorating cerebral ischemic injury and other inflammation-related pathologies.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Brain Ischemia/drug therapy , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/therapeutic use , Pyruvates/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Male , Neuroprotective Agents/pharmacology , Pyruvates/pharmacology , Rats , Rats, Sprague-Dawley
9.
FASEB J ; 33(3): 4314-4326, 2019 03.
Article in English | MEDLINE | ID: mdl-30566396

ABSTRACT

The overactivity of cannabinoid 1 receptor (CB1R) is associated with obesity and type 2 diabetes. First-generation CB1R antagonists, such as rimonabant, offered therapeutic advantages for the control of obesity and related metabolic abnormalities, but their therapeutic potential was limited by undesirable neuropsychiatric side effects. Here, we evaluated AJ5012 as a novel potent peripheral CB1R antagonist and, using this antagonist, investigated the role of peripheral CB1R on adipose tissue inflammation in obese mouse models. AJ5012 had a high degree of CB1R and cannabinoid 2 receptor selectivity but a low brain:plasma concentration ratio without eliciting centrally mediated neurobehavioral effects. In diet-induced obese (DIO) mice, AJ5012 did not reduce food intake but did induce a significant weight loss, likely owing to an increased energy expenditure. It was as effective as rimonabant for the improvement of hormonal or metabolic abnormalities, glycemic control, and insulin sensitivity. The treatment of DIO and leptin receptor-deficient mice with AJ5012 also exhibited effects comparable to rimonabant for the prevention of macrophage infiltration, activation of the nucleotide-binding domain and leucine-rich repeat protein 3 inflammasome, and production of proinflammatory cytokines, which resulted in the suppression of adipose tissue inflammation. In addition to macrophage, activation of CB1R in 3T3-L1 adipocytes induced the expression of proinflammatory genes, which was fully inhibited by AJ5012. Our findings identified AJ5012 as a novel peripheral CB1R antagonist and suggest that peripheral CB1R blockade might break the links between insulin resistance and adipose tissue inflammation.-Han, J. H., Shin, H., Park, J.-Y., Rho, J. G., Son, D. H., Kim, K. W., Seong, J. K., Yoon, S.-H., Kim, W. A novel peripheral cannabinoid 1 receptor antagonist, AJ5012, improves metabolic outcomes and suppresses adipose tissue inflammation in obese mice.


Subject(s)
Adipose Tissue/drug effects , Hypoglycemic Agents/pharmacology , Inflammation/drug therapy , Inflammation/metabolism , Obesity/drug therapy , Obesity/metabolism , Receptor, Cannabinoid, CB1/antagonists & inhibitors , 3T3 Cells , Adipose Tissue/metabolism , Animals , CHO Cells , Cricetulus , Cytokines/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Energy Metabolism/drug effects , Female , Humans , Inflammasomes/drug effects , Inflammasomes/metabolism , Insulin Resistance/physiology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , RAW 264.7 Cells , Receptor, Cannabinoid, CB2/metabolism , Rimonabant/metabolism , Weight Loss/drug effects
10.
Biol Pharm Bull ; 40(9): 1475-1482, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28592715

ABSTRACT

A series of methyl ester of clovamide analogues, where the hydroxyl group of catechol moiety in caffeic acid and L-3,4-dihydroxyphenylalanine (L-dopa) was replaced with various functional groups, were synthesized and their inhibitory effects on nitric oxide (NO) production and inducible NO synthase (iNOS) expression in lipopolysaccharide (LPS)-induced BV2 cells were tested. Among the synthesized compounds, 3,5-ditrifluoromethyl analogue 9l (IC50=2.8 µM) exhibited a potency about 26.3 times greater than that of the parent compound 9a (IC50=73.6 µM) and suppressed NO production dose-dependently without cytotoxicity. Compound 9l also inhibited iNOS expression in LPS-induced BV2 cells at 2.5, 5 and 10 µM concentrations. These results suggested that the dihydroxyl group of catechol moiety in caffeic acid unit is not essential for the suppression of NO production and that 9l has potential as a potent inhibitor of NO production.


Subject(s)
Microglia/metabolism , Nitric Oxide/biosynthesis , Tyrosine/analogs & derivatives , Animals , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Caffeic Acids , Cell Line , Levodopa , Lipopolysaccharides , Mice , Nitric Oxide Synthase Type II/metabolism , Plant Extracts , Structure-Activity Relationship , Tyrosine/chemical synthesis , Tyrosine/chemistry , Tyrosine/pharmacology
12.
Sci Rep ; 7: 42891, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28220827

ABSTRACT

Ethyl pyruvate (EP) is a simple aliphatic ester of pyruvic acid and has been shown to have robust neuroprotective effects via its anti-inflammatory, anti-oxidative, and anti-apoptotic functions. In an effort to develop novel EP derivatives with greater protective potencies than EP, we generated four EP isosteres, among them the neuroprotective potency of N,N-diethyl-2-oxopropanamide (DEOPA), in which the ethoxy group of EP was replaced with diethylamine, was far greater than that of EP. When DEOPA was administered intravenously (5 mg/kg) to rat middle cerebral artery occlusion (MCAO) model at 6 hrs post-surgery, it suppressed infarct formation, ameliorated neurological and sensory/motor deficits, and inhibited microglial activation and neutrophil infiltrations in the postischemic brain more effectively than EP. In particular, DEOPA markedly suppressed LPS-induced nitrite production and cytokine/chemokine inductions in microglia, neutrophils, and endothelial cells and these effects are attributable to inhibition of the activity of NF-κB by suppressing IκB-α degradation and p65 to DNA binding. In addition, DEOPA suppressed NMDA-induced neuronal cell death in primary cortical neuron cultures by NAD replenishment and suppression of NF-κB activity. Together, these results indicate DEOPA has multi-modal protective effects against ischemic brain damage targeting numerous cell types in the brain and also against other inflammation-related diseases.


Subject(s)
Amides/pharmacology , Anti-Inflammatory Agents/pharmacology , Brain/drug effects , Neuroprotective Agents/pharmacology , Amides/therapeutic use , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Brain/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/etiology , Cell Adhesion/drug effects , Cell Movement/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Infarction, Middle Cerebral Artery/complications , Male , Microglia/cytology , Microglia/drug effects , Microglia/metabolism , NF-KappaB Inhibitor alpha/antagonists & inhibitors , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/therapeutic use , Neutrophils/cytology , Neutrophils/metabolism , Nitric Oxide/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factor RelA/chemistry , Transcription Factor RelA/metabolism
13.
Neurosci Lett ; 643: 59-64, 2017 03 16.
Article in English | MEDLINE | ID: mdl-28189746

ABSTRACT

2-Hydroxy-4-trifluoromethylbenzoic acid (HTB) is a metabolite of triflusal (TF), and has been reported to exert anti-inflammatory effect. In this study, the authors investigated whether HTB has a neuroprotective effect against ischemic brain injuries. We showed that intravenous administration of HTB (5mg/kg) 30min before or 1, 3, or 6h after middle cerebral artery occlusion (MCAO) reduced brain infarct to 10.4±3.3%, 16.9±2.3%, 22.2±1.5% and 40.7±7.5%, respectively, of that of treatment-naive MCAO controls, and the therapeutic time window extended to 9h after MCAO (40.7±7.5%). Furthermore, HTB suppressed infarct formation, protected motor activities, and ameliorated neurological deficits more effectively than by TF or salicylic acid (SA). HTB markedly suppressed microglial activation and proinflammatory cytokines expressions in the postischemic brain and in BV2 cells and suppressed LPS-induced nitrite production by inhibiting IkB degradation. In addition, HTB suppressed NMDA-induced neuronal cell death more effectively than TF or SA in primary cortical neuron cultures. Together, these results indicate that HTB has multi-modal protective effects against ischemic brain damage that encompass anti-inflammatory, anti-excitotoxicity, and anti-Zn2+-toxicity effects.


Subject(s)
Brain Ischemia/drug therapy , Brain/drug effects , Salicylates/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Aspirin/pharmacology , Brain/metabolism , Brain Ischemia/metabolism , Cytokines/metabolism , Infarction, Middle Cerebral Artery/metabolism , Male , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Rats, Sprague-Dawley
14.
Sci Rep ; 6: 31843, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27545301

ABSTRACT

Postischemic brain damage in stroke is proceded with complicated pathological events, and so multimodal drug treatments may offer better therapeutic means for improving clinical outcomes. Here, we report robust neuroprotective effects of a novel compound, 2-((2-oxopropanoyl)oxy)-4-(trifluoromethyl)benzoic acid (OPTBA), a 2-hydroxy-4-trifluoromethyl benzoic acid (HTB, a metabolite of triflusal)-pyruvate ester. Intravenous administration of OPTBA (5 mg/kg) 3 or 6 h after middle cerebral artery occlusion (MCAO) in Sprague-Dawley rats reduced infarct volumes to 38.5 ± 11.4% and 46.5 ± 15.3%, respectively, of that of MCAO controls, and ameliorated motor impairment and neurological deficits. Importantly, neuroprotective effects of OPTBA were far greater than those afforded by combined treatment of HTB and pyruvate. Furthermore, OPTBA suppressed microglial activation and proinflammatory cytokine inductions more effectively than HTB/pyruvate co-treatment in the postischemic brain and LPS-treated cortical slice cultures and also attenuated NMDA-induced neuronal death in hippocampal slice cultures. LC-MS analysis demonstrated that OPTBA was hydrolyzed to HTB and pyruvate with a t1/2 of 38.6 min in blood and 7.2 and 2.4 h in cortex and striatum, respectively, and HTB was maintained for more than 24 h both in blood and brain tissue. Together these results indicate OPTBA acts directly and via its hydrolysis products, thus acting as a multimodal neuroprotectant in the postischemic brain.


Subject(s)
Benzoates/administration & dosage , Brain Ischemia/drug therapy , Neuroprotective Agents/administration & dosage , Stroke/prevention & control , Animals , Benzoates/chemical synthesis , Benzoates/chemistry , Blood-Brain Barrier/chemistry , Brain Ischemia/immunology , Cytokines/metabolism , Disease Models, Animal , Hydrolysis , Male , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Rats , Rats, Sprague-Dawley , Salicylates/chemistry , Stroke/immunology
15.
J Med Chem ; 59(10): 4676-87, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27007292

ABSTRACT

Axon regeneration after injury in the central nervous system is hampered in part because if an age-dependent decline in the intrinsic axon growth potential, and one of the strategies to stimulate axon growth in injured neurons involves pharmacological manipulation of implicated signaling pathways. Here we report phenotypic cell-based screen of chemical libraries and structure-activity-guided optimization that resulted in the identification of compound 7p which promotes neurite outgrowth of cultured primary neurons derived from the hippocampus, cerebral cortex, and retina. In an animal model of optic nerve injury, compound 7p was shown to induce growth of GAP-43 positive axons, indicating that the in vitro neurite outgrowth activity of compound 7p translates into stimulation of axon regeneration in vivo. Further optimization of compound 7p and elucidation of the mechanisms by which it elicits axon regeneration in vivo will provide a rational basis for future efforts to enhance treatment strategies.


Subject(s)
Acetamides/pharmacology , Axons/drug effects , Drug Discovery , Nerve Regeneration/drug effects , Sulfonamides/pharmacology , Acetamides/chemical synthesis , Acetamides/chemistry , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Mice , Molecular Structure , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry
16.
Neuropharmacology ; 105: 411-419, 2016 06.
Article in English | MEDLINE | ID: mdl-26850126

ABSTRACT

Ethyl pyruvate (EP) is a simple aliphatic ester of pyruvic acid and has been shown to have robust protective effect in various pathological conditions. A variety of mechanisms have been reported to underlie the protective effects of EP, which include anti-inflammatory, anti-oxidative, and anti-apoptotic functions. Recently, we reported that EP suppressed high mobility group box 1 (HMGB1) release from primary microglial cells via direct Ca(2+) chelation. In the present study, we investigated whether and how EP chelates Zn(2+) in neurons when it is present at toxic levels. In cortical neurons treated with 40 µM of Zn(2+) for 24 h, both EP and pyruvate significantly suppressed neuronal cell death, although the potency of pyruvate was greater than that of EP, and that NAD replenishment contributed to the neuroprotective effects of both pyruvate and EP. However, when cortical neurons were exposed to acute treatment of Zn(2+) (400 µM, 15 min), EP, but not pyruvate, significantly suppressed neuronal death, despite the fact that NAD replenishment by EP was weaker than that by pyruvate. Spectrophotometric studies revealed that EP directly chelates Zn(2+), and this was confirmed in physiological contexts, such as, NMDA-treated primary cortical cultures and OGD-subjected hippocampal slice cultures, in which EP suppressed intracellular Zn(2+) elevation and neuronal cell death. In addition, EP markedly reduced the expressions of PARP-1 and of the NADPH oxidase subunit in Zn(2+)-treated primary cortical neurons, well known Zn(2+)-induced downstream processes. Together, these results show EP suppresses Zn(2+) induced neurotoxicity via dual functions, chelating Zn(2+) and promoting NAD replenishment.


Subject(s)
Chelating Agents/pharmacology , NAD/metabolism , Neuroprotective Agents/pharmacology , Pyruvates/pharmacology , Zinc/toxicity , Animals , Cell Death/drug effects , Glucose/deficiency , Hypoxia/prevention & control , N-Methylaspartate/metabolism , Neurons/drug effects , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species , Zinc/metabolism
17.
Neuropharmacology ; 97: 46-57, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25983275

ABSTRACT

The selective loss of dopaminergic neurons in Parkinson's disease (PD) is associated with microglial activation. Therefore, the importance of early therapeutic intervention to inhibit microglial activation would be an effective strategy to alleviate the progression of PD. α-Asarone, an active compound found in Araceae and Annonaceae plant species has been used to improve various disease conditions including central nervous system disorders. In the present study the in vitro and in vivo therapeutic effects of α-asarone isolated from the rhizome of Acorus gramineus Solander was evaluated on microglia-mediated neuroinflammation and neuroprotection. Lipopolysaccharide (LPS)-stimulated BV-2 microglial cells were used to evaluate in vitro effects. 1-methyl-4 phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP)-induced mouse model of PD was developed to study the neuroprotective effects of α-asarone in vivo. The results indicated that α-asarone significantly attenuated the LPS-stimulated increase in neuroinflammatory responses and suppressed pro-inflammatory cytokine production in BV-2 cells. Mechanistic study revealed that α-asarone inhibited the LPS-stimulated activation via regulation of nuclear factor kappa-B by blocking degradation of inhibitor kappa B-alpha signaling in BV-2 microglial cells. In in vivo studies, MPTP intoxication to mice resulted in brain microglial activation and significant behavioral deficits. Prophylactic treatment with α-asarone suppressed microglial activation and attenuated PD-like behavioral impairments as assessed by the Y-maze and pole tests. Taken together, these data demonstrate that α-asarone is a promising neuroprotective agent that should be further evaluated and developed for future prevention and treatment of microglia-mediated neuroinflammatory conditions including PD.


Subject(s)
Anisoles/pharmacology , MPTP Poisoning/drug therapy , Microglia/drug effects , NF-kappa B/metabolism , Neuroprotective Agents/pharmacology , Allylbenzene Derivatives , Animals , Anisoles/chemistry , Anisoles/isolation & purification , Brain/drug effects , Brain/immunology , Brain/pathology , Cell Line , Cytokines/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Dopaminergic Neurons/physiology , Dose-Response Relationship, Drug , Lipopolysaccharides , MPTP Poisoning/immunology , MPTP Poisoning/pathology , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice, Inbred C57BL , Microglia/immunology , Microglia/pathology , Motor Activity/drug effects , Motor Activity/physiology , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/physiology , Neuroprotective Agents/chemistry , Neuroprotective Agents/isolation & purification
18.
Brain Res Bull ; 112: 25-34, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25596423

ABSTRACT

Neuroinflammation is one of the critical pathological mechanisms influencing various neurodegenerative disorders. Most of the neurodegenerative diseases involve over-activation of microglial cells contributing to the demise of neurons. The objective of the current study is to evaluate the anti-inflammatory effect of novel synthetic clovamide derivative on the suppression of microglial activation in an in vitro and in vivo model of neuroinflammation. We have used lipopolysaccharide (LPS) to induce an inflammatory response in murine BV-2 microglial cells. Molecular tools like immunocytochemistry and immunoblotting were used to study the activity of novel synthetic clovamide derivative to inhibit inflammation induced by LPS in microglial cells. In in vivo experiments, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) intoxicated mouse model of neuroinflammation was developed to investigate the anti-neuroinflammatory effects of DPTP [3-(3,4-Dihydroxy-phenyl)-2-[4-(3-trifluoromethylphenyl)-but-2-enoylamino]-propionic acid methyl ester]. DPTP was observed to reduce the proinflammatory response in BV-2 cells induced by LPS. Further investigation revealed that DPTP attenuated phosphorylation of c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK), which was accompanied by a decrease in nuclear translocation of nuclear factor-κB (NF-κB) in LPS-treated BV2 microglia. Moreover, prophylactic treatment with DPTP (20mg/kg) for 7 days suppressed MPTP induced glial activation and behavioral impairment. Overall, our findings suggested that, DPTP exerts anti-neuroinflammatory effects against activated microglia in an in vitro and in vivo model and hence might be a promising therapeutic agent for alleviating the evolvement of neurodegenerative diseases associated with microglial activation.


Subject(s)
Inflammation/drug therapy , Neuroprotective Agents/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Brain/drug effects , Brain/physiopathology , Cell Line , Cells, Cultured , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Inflammation/physiopathology , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/physiology , NF-kappa B/metabolism , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/physiology , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Phosphorylation/drug effects , Rats, Sprague-Dawley , Tyrosine/analogs & derivatives , Tyrosine/chemistry
19.
Molecules ; 19(12): 19361-75, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25429561

ABSTRACT

Microglia-induced neuroinflammation is an important pathological mechanism influencing various neurodegenerative disorders. Excess activation of microglia produces a myriad of proinflammatory mediators that decimate neurons. Hence, therapeutic strategies aimed to suppress the activation of microglia might lead to advancements in the treatment of neurodegenerative diseases. In this study, we synthesized a novel ethyl pyruvate derivative, named EOP (S-ethyl 2-oxopropanethioate) and studied its effects on lipopolysaccharide (LPS)-induced production of nitric oxide (NO) in rat primary microglia and mouse BV-2 microglia. EOP significantly decreased the production of NO, inducible nitric oxide synthase, cyclooxygenase and other proinflammatory cytokines, such as interleukin (IL)-6, IL-1ß and tumor necrosis factor-α, in LPS-stimulated BV-2 microglia. The phosphorylation levels of extracellular regulated kinase, p38 mitogen-activated protein kinase, and nuclear translocation of NF-κB were also inhibited by EOP in LPS-activated BV-2 microglial cells. Overall, our observations indicate that EOP might be a promising therapeutic agent to diminish the development of neurodegenerative diseases associated with microglia activation.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Microglia/enzymology , NF-kappa B/metabolism , Pyruvates/chemistry , Pyruvates/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Death/drug effects , Cell Line , Cell Survival/drug effects , Cyclooxygenase 2/metabolism , Cytokines/metabolism , I-kappa B Proteins/metabolism , Mice , Microglia/drug effects , NF-KappaB Inhibitor alpha , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/metabolism , Phosphorylation/drug effects , Protein Transport/drug effects , Proteolysis/drug effects , Pyruvates/chemical synthesis , Rats , Signal Transduction/drug effects
20.
Pharmacol Rep ; 66(3): 471-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24905526

ABSTRACT

Activated microglia cells are well recognized as mediators of neuroinflammation, as they release nitric oxide and pro-inflammatory cytokines in various neuroinflammatory diseases. Thus, suppressing microglial activation may alleviate neuroinflammatory and neurodegenerative processes. In the present study, we synthesized and investigated the anti-neuroinflammatory effect of a novel HTB (2-hydroxy-4-trifuoromethylbenzoic acid) derivative in lipopolysaccharide (LPS)-stimulated microglial cells. Among the synthesized derivatives, the BECT [But-2-enedioic acid bis-(2-carboxy-5-trifluoromethyl-phenyl) ester] significantly decreased production of nitric oxide and other pro-inflammatory cytokines including tumor necrosis factor-α, interleukin-1ß, and interleukin-6 in microglial cells. BECT also mitigated the expression of inducible nitric oxide synthase and cyclooxygenase-2 at both the mRNA and protein levels. Further mechanistic studies demonstrated that the HTB derivative inhibited phosphorylation of JNK and p38 mitogen-activated protein kinase and nuclear translocation of nuclear factor kappa-B in LPS-stimulated BV-2 microglial cells. Thus BECT, our novel synthesized compound have anti-inflammatory activity in microglial cells, and may have therapeutic potential for treating neuroinflammatory diseases.


Subject(s)
Inflammation/drug therapy , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/drug effects , NF-kappa B/metabolism , Salicylates/pharmacology , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Cell Line , Cyclooxygenase 2/metabolism , Inflammation/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Mice , Microglia/drug effects , Microglia/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Phosphorylation/drug effects , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...