Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Phytother Res ; 38(2): 1089-1103, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38168755

ABSTRACT

Autism spectrum disorder (ASD) is a multifaceted neuropsychiatric condition for which effective drug therapy for core clinical symptoms remains elusive. Lotusine, known for its neuroprotective properties in the treatment of neurological disorders, holds potential in addressing ASD. Nevertheless, its specific efficacy in ASD remains uncertain. This study aims to investigate the therapeutic potential of lotusine in ASD and elucidate the underlying molecular mechanisms. We induced an ASD mouse model through intracerebroventricular-propionic acid (ICV-PPA) injection for 7 days, followed by lotusine administration for 5 days. The efficacy of lotusine was evaluated through a battery of behavioral tests, including the three-chamber social test. The underlying mechanisms of lotusine action in ameliorating ASD-like behavior were investigated in the medial prefrontal cortex (mPFC) using whole-cell patch-clamp recordings, western blotting, immunofluorescence staining, molecular docking, and cellular thermal shift assay. The efficacy and mechanisms of lotusine were further validated in vitro. Lotusine effectively alleviated social deficits induced by ICV-PPA injection in mice by counteracting the reduction in miniature excitatory postsynaptic current frequency within the mPFC. Moreover, lotusine enhanced neuronal activity and ameliorated α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor dysfunction in ICV-PPA infusion mice by upregulating c-fos, p-GluA1 Ser 845, and p-GluA1 Ser 831 protein levels within the mPFC. Our findings also suggest that lotusine may exert its effects through modulation of the D1 dopamine receptor (DRD1). Furthermore, the rescuing effects of lotusine were nullified by a DRD1 antagonist in PC12 cells. In summary, our results revealed that lotusine ameliorates ASD-like behavior through targeted modulation of DRD1, ultimately enhancing excitatory synaptic transmission. These findings highlight the potential of lotusine as a nutritional supplement in the treatment of ASD.


Subject(s)
Autism Spectrum Disorder , Dopamine , Isoquinolines , Propionates , Rats , Mice , Animals , Dopamine/metabolism , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/metabolism , Molecular Docking Simulation , Receptors, Dopamine D1/metabolism , Prefrontal Cortex/metabolism , Disease Models, Animal
2.
Phytother Res ; 35(7): 3936-3944, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33856723

ABSTRACT

Anxiety disorders are a common frequently psychiatric symptom in patients that lead to disruption of daily life. Scutellarin (Scu) is the main component of Erigeron breviscapus, which has been used as a neuroprotective agent against glutamate-induced excitotoxicity. However, the potential effect of Scu on the stress-related neuropsychological disorders has not been clarified. In this study, Anxiety-like behavior was induced by acute restraint stress in mice. Scu were injected intraperitoneally (twice daily, 3 days). Results showed that Scu exhibited good protective activity on mice by decreasing transmitter release levels. Restraint stress caused significant anxiety like behavior in mice. Treatment of Scu could significantly improve the moving time of open arms in Elevated Plus Maze and central time on open field test. Scu treatment suppressed action potential firing frequency, restored excessive presynaptic quantal release, and down-regulated glutamatergic receptor expression levels in the prefrontal cortex (PFC) of stressed mice. GABAA Rα1 and GABAA γ2 expression in the brain PFC tissues of mice were nearly abrogated by Scu treatment. In stress-induced anxiety mice, stress can increase the frequency of mini excitatory postsynaptic currents (mEPSC), which can be reversed by Scu treatment. Therefore, Scu has a potent anxiolytic activity and may be valuable for the treatment of stress-induced anxiety disorders.


Subject(s)
Anxiety , Apigenin , Glucuronates , Neurotransmitter Agents/physiology , Animals , Anxiety/drug therapy , Apigenin/pharmacology , Glucuronates/pharmacology , Mice
3.
Neuromolecular Med ; 19(1): 57-68, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27544207

ABSTRACT

Activation of translocator protein (18 kDa) (TSPO) plays an important role to mediate rapid anxiolytic efficacy in stress response and stress-related disorders by the production of neurosteroids. However, little is known about the ligand of TSPO on the anxiety-like and depressive behaviors and the underlying mechanisms in chronic unpredictable mild stress (UCMS) mice. In the present study, a novel ligand of TSPO, ZBD-2 [N-benzyl-N-ethyl-2-(7,8-dihydro-7-benzyl-8-oxo-2-phenyl-9H-purin-9-yl) acetamide] synthesized by our laboratory, was used to evaluate the anxiolytic and antidepressant efficacy and to elucidate the underlying mechanisms. ZBD-2 (3 mg/kg) significantly attenuated anxiety-like and depressive behaviors in the UCMS mice, which was blocked by TSPO antagonist PK11195 (3 mg/kg). Treatment of ZBD-2 reversed the decrease in biogenic amines (norepinephrine, dopamine, and serotonin) in the brain region of hippocampus in the UCMS mice. The decreases in TSPO, GluN2B-containing N-methyl-D-aspartate (NMDA) receptors, GluA1, p-GluA1-Ser831, p-GluA1-Ser845, PSD-95, and GABAA-a2 were integrated with the increases of CaMKII and iNOS levels in the hippocampus of the UCMS mice. ZBD-2 significantly reversed the changes of above proteins. However, ZBD-2 or PK11195 treatment did not affect the levels of GluN2A-containing NMDA receptors and the total levels of GAD67. Our study provides strong evidences that ZBD-2 has a therapeutic effect on chronic stress-related disorders such as depression and anxiety through regulating the biogenic amine levels and the synaptic proteins in the hippocampus.


Subject(s)
Acetamides/therapeutic use , Anti-Anxiety Agents/therapeutic use , Antidepressive Agents/therapeutic use , Purinones/therapeutic use , Receptors, GABA/drug effects , Acetamides/pharmacology , Animals , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Anxiety/drug therapy , Anxiety/etiology , Depression/drug therapy , Depression/etiology , Disease Models, Animal , Drug Evaluation, Preclinical , Glutamate Decarboxylase/analysis , Hippocampus/chemistry , Hippocampus/drug effects , Isoquinolines/pharmacology , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/biosynthesis , Neurotransmitter Agents/analysis , Purinones/pharmacology , Receptors, N-Methyl-D-Aspartate/analysis , Stress, Psychological/drug therapy , Stress, Psychological/psychology
4.
Mol Neurobiol ; 53(2): 932-943, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25561437

ABSTRACT

Neuronal apoptosis and oxidative stress are involved in most of the neurodegenerative diseases, promoting neuron survival is critical for therapy. Silibinin (SLB), which is derived from the seeds of Silybinisus laborinum L., has been widely used as an antioxidant. Here we tested the neuroprotective effects of SLB and the involved molecular mechanisms. We demonstrated that SLB promoted neuron viability upon hydrogen peroxide (H2O2) challenge and reduced hypoxia/ischemia injury in the middle cerebral artery occlusion (MCAO) mouse model. SLB reversed the decreased level of procaspase-3 and balanced Bcl-2 and Bax expression upon H2O2 insult to inhibit cell apoptosis. Furthermore, SLB suppressed the activation of autophagy by decreasing microtubule-associated protein 1 light chain 3 (LC3-II) and Beclin-1 levels under oxidative stress accordingly. SLB phosphorylated protein kinase B (Akt-1) at Ser473 in a time- and dose-dependent manner. The inhibitor for phosphoinositide-3-kinase (PI3K) wortmannin abrogated SLB-induced phosphorylation of Akt-1 and mTOR, decreased the suppression of autophagy, and therefore abolished SLB-mediated neuroprotection. All the data suggested that SLB protected neurons by inhibiting both the mitochondrial and autophagic cell death pathways. This study opens new avenues for the use of SLB in treatment of central nervous system (CNS) diseases in which oxidative stress plays a major role in disease pathogenesis. Given that it occurs naturally with low toxicity and pleiotropic effects that benefit the nervous system, SLB acts potentially as a novel therapy for ischemic injury.


Subject(s)
Autophagy/drug effects , Brain Ischemia/drug therapy , Cerebral Cortex/pathology , Neurons/pathology , Oxidative Stress/drug effects , Reperfusion Injury/drug therapy , Silymarin/therapeutic use , Animals , Apoptosis/drug effects , Brain Ischemia/complications , Brain Ischemia/pathology , Cell Survival/drug effects , Cells, Cultured , Hydrogen Peroxide/toxicity , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Male , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Reperfusion Injury/complications , Reperfusion Injury/pathology , Silybin , Silymarin/chemistry , Silymarin/pharmacology
5.
Mol Brain ; 8(1): 60, 2015 Oct 09.
Article in English | MEDLINE | ID: mdl-26452469

ABSTRACT

Our previous studies have demonstrated the critical roles of calcium-stimulated adenylyl cyclase 1 (AC1) in the central nervous system in chronic pain. In the present study, we examined the analgesic effects of NB001, a selective inhibitor of AC1, on animal models of ankle joint arthritis and knee joint arthritis induced by complete Freund's adjuvant injection. NB001 treatment had no effect on joint edema, stiffness, and joint destruction. Furthermore, the treatment failed to attenuate the disease progression of arthritis. However, NB001 treatment (3 mg/kg) significantly weakened joint pain-related behavior in the mouse models of ankle joint arthritis and knee joint arthritis. Results indicated that NB001 exhibited an analgesic effect on the animal models of arthritis but was not caused by anti-inflammatory activities.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Analgesics/therapeutic use , Arthralgia/drug therapy , Adenosine Triphosphate/pharmacology , Adenosine Triphosphate/therapeutic use , Analgesics/pharmacology , Animals , Arthralgia/complications , Arthralgia/pathology , Arthralgia/physiopathology , Behavior, Animal , Disease Models, Animal , Freund's Adjuvant , Hyperalgesia/complications , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Inflammation/complications , Inflammation/drug therapy , Inflammation/pathology , Knee Joint/drug effects , Knee Joint/pathology , Male , Mice, Inbred C57BL , Pain/complications , Pain/drug therapy , Pain/pathology , Sensory Thresholds
6.
Phytomedicine ; 20(3-4): 343-50, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23219339

ABSTRACT

Oxymatrine (OMT) is a major bioactive component derived from Sophora flavescens Ait (kushen), which is widely used in Chinese medicine. Recent studies have shown that it has neuroprotective effects; however, its underlying mechanisms remain unclear. We focus on the mechanisms of pharmacologic action in OMT by detecting its pharmacological properties against focal cerebral ischemia in vivo and NMDA-induced neurotoxicity in vitro. OMT prevented cerebral ischemic injury in mice induced via a 2 h middle cerebral artery occlusion and a 24 h reperfusion, in vivo. In vitro cultured neurons challenged with N-methyl-D-aspartate (NMDA, 200 µM) for 30 min showed significant decrease in the viability of neurons; however, OMT was able to protect neurons against induced neurotoxicity via NMDA exposure. Western blot analysis revealed that OMT decreased the expression of Bax and repaired the balance of pro- and anti-apoptotic proteins. Furthermore, OMT significantly reversed the up-regulation of NR2B and inhibited the calcium overload in the cultured neurons after challenging the NMDA. OMT showed partial protection in the cortical neurons via down-regulation of NR2B containing NMDA receptors and up-regulation of Bcl-2 family. Our results provide new insights into the development of natural therapeutic anti-oxidants against ischemia.


Subject(s)
Alkaloids/therapeutic use , Brain Ischemia/prevention & control , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Quinolizines/therapeutic use , Receptors, N-Methyl-D-Aspartate/metabolism , Reperfusion Injury/prevention & control , Alkaloids/pharmacology , Animals , Calcium/metabolism , Cell Survival/drug effects , Cells, Cultured , Down-Regulation/drug effects , Drug Evaluation, Preclinical , Drugs, Chinese Herbal/therapeutic use , Male , Mice , Neurons/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Phytotherapy , Proto-Oncogene Proteins c-bcl-2/metabolism , Quinolizines/pharmacology , Sophora , bcl-2-Associated X Protein/metabolism
7.
Pharmacol Rep ; 62(5): 949-55, 2010.
Article in English | MEDLINE | ID: mdl-21098879

ABSTRACT

Hyperoside (Hyp) is a flavonoid compound isolated from a folk remedy, Rhododendron ponticum L. leaves. It has been shown to have neuroprotective effects both in vivo and in vitro. However, little is known about the effects of Hyp on the neuronal apoptosis induced by glutamate. The present study showed that Hyp significantly attenuated, in a concentration-dependent manner, the apoptosis induced by the exposure of cultured neurons to NMDA. Western blot analysis revealed that Hyp antagonized the expression of excess NR2B-containing NMDA receptors; however, it had no effect on the expression of NR2A-containing NMDA receptors. Our results demonstrate that the neuroprotective effect of Hyp owes, at least partially, to its differential modulation of NR2A- and NR2B-containing NMDA receptors.


Subject(s)
Apoptosis/drug effects , Neuroprotective Agents/pharmacology , Quercetin/analogs & derivatives , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Cells, Cultured , Glutamic Acid/metabolism , Neurons/cytology , Neurons/drug effects , Plant Leaves/chemistry , Prefrontal Cortex/cytology , Quercetin/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Rhododendron
8.
Mol Pain ; 5: 71, 2009 Dec 12.
Article in English | MEDLINE | ID: mdl-20003379

ABSTRACT

The midbrain periaqueductal grey (PAG) is a structure known for its roles in pain transmission and modulation. Noxious stimuli potentiate the glutamate synaptic transmission and enhance glutamate NMDA receptor expression in the PAG. However, little is known about roles of NMDA receptor subunits in the PAG in processing the persistent inflammatory pain. The present study was undertaken to investigate NR2A- and NR2B-containing NMDA receptors in the PAG and their modulation to the peripheral painful inflammation. Noxious stimuli induced by hind-paw injection of complete Freund's adjuvant (CFA) caused up-regulation of NR2B-containing NMDA receptors in the PAG, while NR2A-containing NMDA receptors were not altered. Whole-cell patch-clamp recordings revealed that NMDA receptor mediated mEPSCs were increased significantly in the PAG synapse during the chronic phases of inflammatory pain in mice. PAG local infusion of Ro 25-6981, an NR2B antagonist, notably prolonged the paw withdrawal latency to thermal radian heat stimuli bilaterally in rats. Hyperoside (Hyp), one of the flavonoids compound isolated from Rhododendron ponticum L., significantly reversed up-regulation of NR2B-containing NMDA receptors in the PAG and exhibited analgesic activities against persistent inflammatory stimuli in mice. Our findings provide strong evidence that up-regulation of NR2B-containing NMDA receptors in the PAG involves in the modulation to the peripheral persistent inflammatory pain.


Subject(s)
Glutamic Acid/metabolism , Inflammation/metabolism , Pain, Intractable/metabolism , Periaqueductal Gray/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Afferent Pathways/metabolism , Afferent Pathways/physiopathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Chronic Disease , Disease Models, Animal , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/physiology , Freund's Adjuvant , Inflammation/physiopathology , Male , Mice , Mice, Inbred C57BL , Nociceptors/metabolism , Organ Culture Techniques , Pain Measurement/methods , Pain, Intractable/physiopathology , Patch-Clamp Techniques , Periaqueductal Gray/physiopathology , Phenols/pharmacology , Piperidines/pharmacology , Quercetin/analogs & derivatives , Quercetin/pharmacology , Rats , Rats, Sprague-Dawley , Synaptic Transmission/physiology , Up-Regulation/drug effects , Up-Regulation/physiology
9.
Neuropharmacology ; 54(8): 1175-81, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18410946

ABSTRACT

Gentiopicroside is one of the secoiridoid compound isolated from Gentiana lutea. It exhibits analgesic activities in the mice. The anterior cingulate cortex (ACC) is a forebrain structure known for its roles in pain transmission and modulation. Painful stimuli potentiate the prefrontal synaptic transmission and induce glutamate NMDA NR2B receptor expression in the ACC. But little is known about Gentiopicroside on the persistent inflammatory pain and chronic pain-induced synaptic transmission changes in the ACC. The present study was undertaken to investigate its analgesic activities and central synaptic modulation to the peripheral painful inflammation. Gentiopicroside produced significant analgesic effects against persistent inflammatory pain stimuli in mice. Systemic administration of Gentiopicroside significantly reversed NR2B over-expression during the chronic phases of persistent inflammation caused by hind-paw administration of complete Freunds adjuvant (CFA) in mice. Whole-cell patch clamp recordings revealed that Gentiopicroside significantly reduced NR2B receptors mediated postsynaptic currents in the ACC. Our findings provide strong evidence that analgesic effects of Gentiopicroside involve down-regulation of NR2B receptors in the ACC to persistent inflammatory pain.


Subject(s)
Analgesics, Non-Narcotic , Glucosides/pharmacology , Glucosides/therapeutic use , Inflammation/complications , Inflammation/drug therapy , Iridoids/pharmacology , Iridoids/therapeutic use , Pain/drug therapy , Pain/etiology , Receptors, N-Methyl-D-Aspartate/biosynthesis , Animals , Blotting, Western , Chronic Disease , Cyclic AMP/metabolism , Down-Regulation/drug effects , Excitatory Postsynaptic Potentials/drug effects , Freund's Adjuvant , Glutamic Acid/physiology , Inflammation/chemically induced , Iridoid Glucosides , Mice , Mice, Inbred C57BL , Pain/psychology , Pain Measurement/drug effects , Patch-Clamp Techniques , Receptors, GABA-A/drug effects , Synaptic Transmission/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL