Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Neuropharmacology ; 126: 12-24, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28807675

ABSTRACT

We have previously demonstrated the unexpected neuroprotection of the anti-cancer agent SU4312 in cellular models associated with Parkinson's disease (PD). However, the precise mechanisms underlying its neuroprotection are still unknown, and the effects of SU4312 on rodent models of PD have not been characterized. In the current study, we found that the protection of SU4312 against 1-methyl-4-phenylpyridinium ion (MPP+)-induced neurotoxicity in PC12 cells was achieved through the activation of transcription factor myocyte enhancer factor 2D (MEF2D), as evidenced by the fact that SU4312 stimulated myocyte enhancer factor 2 (MEF2) transcriptional activity and prevented the inhibition of MEF2D protein expression caused by MPP+, and that short hairpin RNA (ShRNA)-mediated knockdown of MEF2D significantly abolished the neuroprotection of SU4312. Additionally, Western blotting analysis revealed that SU4312 potentiated pro-survival PI3-K/Akt pathway to down-regulate MEF2D inhibitor glycogen synthase kinase-3beta (GSK3ß). Furthermore, using the in vivo PD model of C57BL/6 mice insulted with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), we found that intragastrical administration of SU4312 (0.2 and 1 mg/kg) greatly ameliorated Parkinsonian motor defects, and restored protein levels of MEF2D, phosphorylated-Ser473-Akt and phosphorylated-Ser9-GSK3ß. Meanwhile, SU4312 effectively reversed the decrease in protein expression of tyrosine hydroxylase in substantia nigra pars compacta dopaminergic neurons, inhibited oxidative stress, maintained mitochondrial biogenesis and partially prevented the depletion of dopamine and its metabolites. Very encouragingly, SU4312 was able to selectively inhibit monoamine oxidase-B (MAO-B) activity both in vitro and in vivo, with an IC50 value of 0.2 µM. These findings suggest that SU4312 provides therapeutic benefits in cellular and animal models of PD, possibly through multiple mechanisms including enhancement of MEF2D through the activation of PI3-K/Akt pathway, maintenance of mitochondrial biogenesis and inhibition of MAO-B activity. SU4312 thus may be an effective drug candidate for the prevention or even modification of the pathological processes of PD.


Subject(s)
Antiparkinson Agents/administration & dosage , Indoles/administration & dosage , MPTP Poisoning/metabolism , Monoamine Oxidase/metabolism , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cortical Bone/drug effects , Cortical Bone/metabolism , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , In Vitro Techniques , MEF2 Transcription Factors/metabolism , MPTP Poisoning/drug therapy , Male , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/physiology , Monoamine Oxidase Inhibitors/administration & dosage , PC12 Cells , Rats , Signal Transduction/drug effects
2.
Sci Rep ; 5: 10256, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26194093

ABSTRACT

ß-amyloid (Aß) oligomers have been closely implicated in the pathogenesis of Alzheimer's disease (AD). We found, for the first time, that bis(heptyl)-cognitin, a novel dimeric acetylcholinesterase (AChE) inhibitor derived from tacrine, prevented Aß oligomers-induced inhibition of long-term potentiation (LTP) at concentrations that did not interfere with normal LTP. Bis(heptyl)-cognitin also prevented Aß oligomers-induced synaptotoxicity in primary hippocampal neurons. In contrast, tacrine and donepezil, typical AChE inhibitors, could not prevent synaptic impairments in these models, indicating that the modification of Aß oligomers toxicity by bis(heptyl)-cognitin might be attributed to a mechanism other than AChE inhibition. Studies by using dot blotting, immunoblotting, circular dichroism spectroscopy, and transmission electron microscopy have shown that bis(heptyl)-cognitin altered Aß assembly via directly inhibiting Aß oligomers formation and reducing the amount of preformed Aß oligomers. Molecular docking analysis further suggested that bis(heptyl)-cognitin presumably interacted with the hydrophobic pockets of Aß, which confers stabilizing powers and assembly alteration effects on Aß. Most importantly, bis(heptyl)-cognitin significantly reduced cognitive impairments induced by intra-hippocampal infusion of Aß oligomers in mice. These results clearly demonstrated how dimeric agents prevent Aß oligomers-induced synaptic and memory impairments, and offered a strong support for the beneficial therapeutic effects of bis(heptyl)-cognitin in the treatment of AD.


Subject(s)
Amyloid beta-Peptides/toxicity , Memory Disorders/chemically induced , Memory Disorders/drug therapy , Neuroprotective Agents/therapeutic use , Synapses/pathology , Tacrine/analogs & derivatives , Tacrine/therapeutic use , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/ultrastructure , Animals , Cells, Cultured , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/therapeutic use , Electric Stimulation , Hippocampus/pathology , Hydrophobic and Hydrophilic Interactions , Long-Term Potentiation/drug effects , Male , Mice, Inbred ICR , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Protein Structure, Secondary , Rats, Sprague-Dawley , Rats, Wistar , Synapses/drug effects , Tacrine/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/metabolism
3.
Br J Pharmacol ; 168(5): 1201-14, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23062100

ABSTRACT

BACKGROUND AND PURPOSE: SU4312, a potent and selective inhibitor of VEGF receptor-2 (VEGFR-2), has been designed to treat cancer. Recent studies have suggested that SU4312 can also be useful in treating neurodegenerative disorders. In this study, we assessed neuroprotection by SU4312 against 1-methyl-4-phenylpyridinium ion (MPP(+) )-induced neurotoxicity and further explored the underlying mechanisms. EXPERIMENTAL APPROACH: MPP(+) -treated neurons and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated zebrafish were used to study neuroprotection by SU4312. NOS activity was assayed in vitro to examine direct interactions between SU4312 and NOS isoforms. KEY RESULTS: SU4312 unexpectedly prevented MPP(+) -induced neuronal apoptosis in vitro and decreased MPTP-induced loss of dopaminergic neurons, reduced expression of mRNA for tyrosine hydroxylase and impaired swimming behaviour in zebrafish. In contrast, PTK787/ZK222584, a well-studied VEGFR-2 inhibitor, failed to prevent neurotoxicity, suggesting that the neuroprotective actions of SU4312 were independent of its anti-angiogenic action. Furthermore, SU4312 exhibited non-competitive inhibition of purified neuronal NOS (nNOS) with an IC(50) value of 19.0 µM but showed little or no effects on inducible and endothelial NOS. Molecular docking simulations suggested an interaction between SU4312 and the haem group within the active centre of nNOS. CONCLUSIONS AND IMPLICATION: SU4312 exhibited neuroprotection against MPP(+) at least partly via selective and direct inhibition of nNOS. Because SU4312 could reach the brain in rats, our study also offered a support for further development of SU4312 to treat neurodegenerative disorders, particularly those associated with NO-mediated neurotoxicity.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Antineoplastic Agents/therapeutic use , Indoles/therapeutic use , Neuroprotective Agents/therapeutic use , Nitric Oxide Synthase Type I/antagonists & inhibitors , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Antineoplastic Agents/pharmacology , Behavior, Animal/drug effects , Brain/metabolism , Cell Death/drug effects , Cell Line, Tumor , Embryo, Nonmammalian/anatomy & histology , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/physiology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Indoles/pharmacology , Molecular Docking Simulation , Motor Activity/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Nitric Oxide/metabolism , PC12 Cells , Rats , Rats, Sprague-Dawley , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...