Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 939
Filter
Add more filters

Complementary Medicines
Publication year range
1.
J Med Food ; 25(7): 695-709, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35834631

ABSTRACT

Spirulina platensis is a cyanobacterium with high protein content and presenting neuroprotective effects. Now, we studied a protein-enriched fraction (SPF), on behavior, neurochemical and immunohistochemical (IHC) assays in hemiparkinsonian rats, distributed into the groups: SO (sham-operated), 6-hydroxydopamine (6-OHDA), and 6-OHDA (treated with SPF, 5 and 10 mg/kg, p.o., 15 days). Afterward, animals were subjected to behavioral tests and euthanized, and brain areas used for neurochemical and IHC assays. SPF partly reversed the changes in the apomorphine-induced rotations, open field and forced swim tests, and also the decrease in striatal dopamine and 3,4-dihydroxyphenylacetic acid contents seen in hemiparkinsonian rats. Furthermore, SPF reduced brain oxidative stress and increased striatal expressions of tyrosine hydroxylase and dopamine transporter and significantly reduced hippocampal inducible nitric oxide synthase, cyclooxygenase-2 and glial fibrillary acidic protein expressions. The data suggest that the protein fraction from S. platensis, through its brain anti-inflammatory and antioxidative actions, exerts neuroprotective effects that could benefit patients affected by neurodegenerative diseases, like Parkinson's disease.


Subject(s)
Neuroprotective Agents , Parkinson Disease , Spirulina , Tissue Extracts , Animals , Brain/drug effects , Corpus Striatum/drug effects , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Neuroprotection , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidopamine , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Rats , Spirulina/metabolism , Tissue Extracts/metabolism , Tissue Extracts/pharmacology , Tissue Extracts/therapeutic use
2.
Mol Neurobiol ; 59(1): 590-602, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34741234

ABSTRACT

DNA oxidative damage can cause telomere attrition or dysfunction that triggers cell senescence and apoptosis. The hypothesis of this study is that folic acid decreases apoptosis in neural stem cells (NSCs) by preventing oxidative stress-induced telomere attrition. Primary cultures of NSCs were incubated for 9 days with various concentrations of folic acid (0-40 µM) and then incubated for 24 h with a combination of folic acid and an oxidant (100-µM hydrogen peroxide, H2O2), antioxidant (10-mM N-acetyl-L-cysteine, NAC), or vehicle. Intracellular folate concentration, apoptosis rate, cell proliferative capacity, telomere length, telomeric DNA oxidative damage, telomerase activity, intracellular reactive oxygen species (ROS) levels, cellular oxidative damage, and intracellular antioxidant enzyme activities were determined. The results showed that folic acid deficiency in NSCs decreased intracellular folate concentration, cell proliferation, telomere length, and telomerase activity but increased apoptosis, telomeric DNA oxidative damage, and intracellular ROS levels. In contrast, folic acid supplementation dose-dependently increased intracellular folate concentration, cell proliferative capacity, telomere length, and telomerase activity but decreased apoptosis, telomeric DNA oxidative damage, and intracellular ROS levels. Exposure to H2O2 aggravated telomere attrition and oxidative damage, whereas NAC alleviated the latter. High doses of folic acid prevented telomere attrition and telomeric DNA oxidative damage by H2O2. In conclusion, inhibition of telomeric DNA oxidative damage and telomere attrition in NSCs may be potential mechanisms of inhibiting NSC apoptosis by folic acid.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Folic Acid/pharmacology , Neural Stem Cells/drug effects , Oxidative Stress/drug effects , Telomere/drug effects , Animals , Cell Proliferation/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hydrogen Peroxide/pharmacology , Neural Stem Cells/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Telomere/metabolism
3.
Neurosci Lett ; 765: 136251, 2021 11 20.
Article in English | MEDLINE | ID: mdl-34536508

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease caused by the death of dopaminergic neurons in the substantia nigra pars compacta (SNpc), characterized by motor dysfunction. While PD symptoms are well treated with L-DOPA, continuous use can cause L-DOPA-induced dyskinesia (LID). We have previously demonstrated that sub-anesthetic ketamine attenuated LID development in rodents, measured by abnormal involuntary movements (AIMs), and reduced the density of maladaptive striatal dendritic mushroom spines. Microglia may play a role by phagocytosing maladaptive neuronal spines. In this exploratory study, we hypothesized that ketamine would prevent AIMs and change microglia ramified morphology - an indicator of a microglia response. Unilaterally 6-hydroxydopamine (6-OHDA)-lesioned rats were primed with daily injections of L-DOPA for 14 days, treated on days 0 and 7 for 10-hours with sub-anesthetic ketamine (i.p.), and we replicated that this attenuated LID development. We further extended our prior work by showing that while ketamine treatment did lead to an increase of striatal interleukin-6 in dyskinetic rats, indicating a modulation of an inflammatory response, it did not change microglia number or morphology in the dyskinetic striatum. Yet an increase of CD68 in the SNpc of 6-OHDA-lesioned hemispheres post-ketamine indicates increased microglia phagocytosis suggestive of a lingering microglial response to 6-OHDA injury in the SNpc pointing to possible anti-inflammatory action in the PD model in addition to anti-dyskinetic action. In conclusion, we provide further support for sub-anesthetic ketamine treatment of LID. The mechanisms of action for ketamine, specifically related to inflammation and microglia phagocytic functions, are emerging, and require further examination.


Subject(s)
Dyskinesia, Drug-Induced/prevention & control , Excitatory Amino Acid Antagonists/administration & dosage , Ketamine/administration & dosage , Levodopa/administration & dosage , Parkinson Disease/drug therapy , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Corpus Striatum/drug effects , Corpus Striatum/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/pathology , Humans , Levodopa/adverse effects , Male , Microglia/drug effects , Microglia/pathology , Phagocytosis/drug effects , Rats , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Substantia Nigra/drug effects , Substantia Nigra/pathology
4.
Biosci Rep ; 41(8)2021 08 27.
Article in English | MEDLINE | ID: mdl-34355745

ABSTRACT

Traditional Chinese medicine detoxification prescription Chaihu-jia-Longgu-Muli decoction (CLMD) relieves depressive symptoms in patients withdrawing from methamphetamine. In the present study, we assessed the effects of CLMD on methamphetamine withdrawal in rats. A methamphetamine-intoxicated rat model was established. Rats were randomly divided into the control, model, high-dosage, medium-dosage, and low-dosage groups, receiving high, medium, and low doses of CLMD, respectively. Weekly body weight measurements revealed that rats treated with methamphetamine had the lowest body weight. The conditioned place preference (CPP) experiment revealed that methamphetamine-intoxicated rats stayed significantly longer in the drug-paired chamber than the control rats. However, after administering high-dosage CLMD, the amount of time the rats spent in the drug-paired chamber was significantly less than that of the model rats. Our open-field test revealed that the model group had lower crossing and rearing scores than the control group. Additionally, rats that received CLMD treatment exhibited higher crossing and rearing scores than the model rats. Striatal dopamine (DA), 5-hydroxytryptamine (5-HT), and endorphins (ß-EP) and serum interleukin (IL)-1α and IL-2 concentrations were estimated. Rats in the model group had lower striatal DA, 5-HT, and ß-EP and higher serum IL-1α and IL-2 concentrations than those in the control group. High-dosage CLMD administration significantly changed the concentrations of these molecules, such that they approached normal concentrations. In general, CLMD could prevent the development of methamphetamine-induced withdrawal symptoms in rats by increasing the DA, 5-HT, and ß-EP and lowering the IL-1α and IL-2 concentrations.


Subject(s)
Behavior, Animal/drug effects , Central Nervous System Stimulants , Conditioning, Psychological/drug effects , Corpus Striatum/drug effects , Drugs, Chinese Herbal/pharmacology , Methamphetamine , Substance Withdrawal Syndrome/drug therapy , Animals , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine/metabolism , Interleukin-1alpha/blood , Interleukin-2/blood , Male , Open Field Test/drug effects , Rats, Sprague-Dawley , Serotonin/metabolism , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/psychology , beta-Endorphin/metabolism
5.
Pharmacol Res Perspect ; 9(5): e00855, 2021 10.
Article in English | MEDLINE | ID: mdl-34423920

ABSTRACT

Several therapeutic options are currently available to treat excessive daytime sleepiness (EDS) in patients suffering from narcolepsy or obstructive sleep apnea. However, there are no comparisons between the various wake-promoting agents in terms of mechanism of action, efficacy, or safety. The goal of this study was to compare amphetamine, modafinil, solriamfetol, and pitolisant at their known primary pharmacological targets, histamine H3 receptors (H3R), dopamine, norepinephrine, and serotonin transporters, and in various in vivo preclinical models in relation to neurochemistry, locomotion, behavioral sensitization, and food intake. Results confirmed that the primary pharmacological effect of amphetamine, modafinil, and solriamfetol was to increase central dopamine neurotransmission, in part by inhibiting its transporter. Furthermore, solriamfetol increased levels of extracellular dopamine in the nucleus accumbens, and decreased the 3,4-dihydroxyphenyl acetic acid (DOPAC)/DA ratio in the striatum, as reported for modafinil and amphetamine. All these compounds produced hyperlocomotion, behavioral sensitization, and hypophagia, which are common features of psychostimulants and of compounds with abuse potential. In contrast, pitolisant, a selective and potent H3R antagonist/inverse agonist that promotes wakefulness, had no effect on striatal dopamine, locomotion, or food intake. In addition, pitolisant, devoid of behavioral sensitization by itself, attenuated the hyperlocomotion induced by either modafinil or solriamfetol. Therefore, pitolisant presents biochemical, neurochemical, and behavioral profiles different from those of amphetamine and other psychostimulants such as modafinil or solriamfetol. In conclusion, pitolisant is a differentiated therapeutic option, when compared with psychostimulants, for the treatment of EDS, as this agent does not show any amphetamine-like properties within in vivo preclinical models.


Subject(s)
Amphetamine/pharmacology , Carbamates/pharmacology , Corpus Striatum/drug effects , Disorders of Excessive Somnolence/drug therapy , Feeding Behavior/drug effects , Locomotion/drug effects , Modafinil/pharmacology , Phenylalanine/analogs & derivatives , Piperidines/pharmacology , Wakefulness-Promoting Agents/pharmacology , 3,4-Dihydroxyphenylacetic Acid/metabolism , Adrenergic Uptake Inhibitors/pharmacology , Animals , Corpus Striatum/metabolism , Disorders of Excessive Somnolence/etiology , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/drug effects , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine Uptake Inhibitors/pharmacology , Drug Evaluation, Preclinical , Drug Inverse Agonism , Histamine Antagonists/pharmacology , Mice , Narcolepsy/drug therapy , Neostriatum/drug effects , Neostriatum/metabolism , Norepinephrine Plasma Membrane Transport Proteins/drug effects , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Phenylalanine/pharmacology , Receptors, Histamine H3 , Sleep Apnea, Obstructive/complications
6.
Nutrients ; 13(5)2021 May 10.
Article in English | MEDLINE | ID: mdl-34068460

ABSTRACT

Parkinson's disease (PD) is a debilitating neurodegenerative disease, which progresses over time, causing pathological depigmentation of the substantia nigra (SN) in the midbrain due to loss of dopaminergic neurons. Emerging studies revealed the promising effects of some nutrient compounds in reducing the risk of PD. One such nutrient compound that possess neuroprotective effects and prevents neurodegeneration is tocotrienol (T3), a vitamin E family member. In the present study, a single dose intracisternal injection of 250 µg 6-hydroxydopamine (6-OHDA) was used to induce parkinsonism in male Sprague Dawley (SD) rats. Forty-eight hours post injection, the SD rats were orally supplemented with alpha (α)- and gamma (γ)-T3 for 28 days. The neuroprotective effects of α- and γ-T3 were evaluated using behavioural studies and immunohistochemistry (IHC). The findings from this study revealed that supplementation of α- and γ-T3 was able to ameliorate the motor deficits induced by 6-OHDA and improve the neuronal functions by reducing inflammation, reversing the neuronal degradation, and preventing further reduction of dopaminergic neurons in the SN and striatum (STR) fibre density.


Subject(s)
Oxidopamine/toxicity , Parkinson Disease/drug therapy , Tocotrienols/pharmacology , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Immunohistochemistry , Male , Neuroprotective Agents/pharmacology , Parkinson Disease/etiology , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/etiology , Rats , Rats, Sprague-Dawley , Substantia Nigra/drug effects , Substantia Nigra/metabolism
7.
Arch Biochem Biophys ; 709: 108970, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34181873

ABSTRACT

Glutaric acidemia type 1 (GA1) is caused by glutaryl-CoA dehydrogenase deficiency that leads to a blockage in the metabolic route of the amino acids lysine and tryptophan and subsequent accumulation of glutaric acid (GA), 3-hydroxyglutaric acids and glutarylcarnitine (C5DC). Patients predominantly manifest neurological symptoms, associated with acute striatal degeneration, as well as progressive cortical and striatum injury whose pathogenesis is not yet fully established. Current treatment includes protein/lysine restriction and l-carnitine supplementation of (L-car). The aim of this work was to evaluate behavior parameters and pro-inflammatory factors (cytokines IL-1ß, TNF-α and cathepsin-D levels), as well as the anti-inflammatory cytokine IL10 in striatum of knockout mice (Gcdh-/-) and wild type (WT) mice submitted to a normal or a high Lys diet. The potential protective effects of L-car treatment on these parameters were also evaluated. Gcdh-/- mice showed behavioral changes, including lower motor activity (decreased number of crossings) and exploratory activity (reduced number of rearings). Also, Gcdh-/- mice had significantly higher concentrations of glutarylcarnitine (C5DC) in blood and cathepsin-D (CATD), interleukin IL-1ß and tumor factor necrosis alpha (TNF-α) in striatum than WT mice. Noteworthy, L-car treatment prevented most behavioral alterations, normalized CATD levels and attenuated IL-1ß levels in striatum of Gcdh-/- mice. Finally, IL-1ß was positively correlated with CATD and C5DC levels and L-car was negatively correlated with CATD. Our results demonstrate behavioral changes and a pro-inflammatory status in striatum of the animal model of GA1 and, most importantly, L-car showed important protective effects on these alterations.


Subject(s)
Amino Acid Metabolism, Inborn Errors/drug therapy , Brain Diseases, Metabolic/drug therapy , Carnitine/therapeutic use , Glutaryl-CoA Dehydrogenase/deficiency , Inflammation/drug therapy , Neuroprotective Agents/therapeutic use , Amino Acid Metabolism, Inborn Errors/genetics , Animals , Brain Diseases, Metabolic/genetics , Carnitine/analogs & derivatives , Carnitine/metabolism , Cathepsin D/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Glutaryl-CoA Dehydrogenase/genetics , Grooming/drug effects , Inflammation/genetics , Interleukin-1beta/metabolism , Locomotion/drug effects , Lysine/pharmacology , Mice, Knockout , Open Field Test/drug effects , Transforming Growth Factor beta/metabolism
8.
Article in English | MEDLINE | ID: mdl-34048863

ABSTRACT

Tardive dyskinesia (TD) is a movement disorder that appears after chronic use of drugs that block dopaminergic receptors such as antipsychotics. Besides the motor symptoms, patients with TD also present cognitive deficits. Neuroinflammatory mechanisms could be involved in the development of these symptoms. A previous study showed that cannabidiol (CBD), the major non-psychotomimetic compound of Cannabis sativa plant, prevents orofacial dyskinesia induced by typical antipsychotics by activating peroxisome proliferator-activated receptors gamma (PPARγ). Here, we investigated if CBD would also reverse haloperidol-induced orofacial dyskinesia and associated cognitive deficits. We also verified if these effects depend on PPARγ receptor activation. Daily treatment with haloperidol (3 mg/kg, 21 days) increased the frequency of vacuous chewing movements (VCM) and decreased the discrimination index in the novel object recognition test in male Swiss mice. CBD (60 mg/kg/daily) administered in the last 7 days of haloperidol treatment attenuated both behavioral effects. Furthermore, haloperidol increased IL-1ß and TNF-α levels in the striatum and hippocampus while CBD reverted these effects. The striatal and hippocampal levels of proinflammatory cytokines correlated with VCM frequency and discrimination index, respectively. Pretreatment with the PPARγ antagonist GW9662 (2 mg/kg/daily) blocked the behavioral effects of CBD. In conclusion, these results indicated that CBD could attenuate haloperidol-induced orofacial dyskinesia and improve non-motor symptoms associated with TD by activating PPARγ receptors.


Subject(s)
Antipsychotic Agents/adverse effects , Cannabidiol/pharmacology , Cognitive Dysfunction , Dyskinesias/drug therapy , PPAR gamma/therapeutic use , Tardive Dyskinesia/chemically induced , Animals , Anti-Dyskinesia Agents/adverse effects , Anti-Dyskinesia Agents/pharmacology , Behavior, Animal/drug effects , Cannabidiol/administration & dosage , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/prevention & control , Corpus Striatum/drug effects , Haloperidol/adverse effects , Haloperidol/pharmacology , Male , Mastication/drug effects , Mice , Neostriatum/drug effects , Rats , Rats, Wistar
9.
Neuroreport ; 32(10): 869-874, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34029288

ABSTRACT

OBJECTIVE: Recent studies have suggested that ninjin'yoeito (NYT), a traditional Japanese Kampo medicine, improves diminished motivation in humans and animals, rendering it a novel therapeutic option for impaired motivation. To better characterize the effect of NYT on motivation, we examined its effect on motivated behaviors in mice. METHODS: Mouse models of neurodegeneration-related apathy, in which striatal dopamine receptor type 2-expressing medium spiny neurons (D2-MSNs) were progressively damaged by diphtheria toxin expression, were chosen. RESULTS: The decrease in effort-based operant responding for rewards (sucrose pellets), indicative of the mouse's motivated behavior, in the affected mice was not suppressed by chronic treatment with NYT suspended in drinking water at 1% (w/v). Mice were then subjected to a sucrose preference test, wherein they freely chose to ingest tap water and a sucrose solution without being required to exert effort. The affected mice showed a decline in preference for sucrose over tap water, relative to nonaffected controls, indicating anhedonia-like traits. In contrast to the diminished operant behavior, the anhedonic behavior in the affected mice was prevented by the NYT administration. Furthermore, NYT did not affect the size of Drd2 mRNA disappearance in the striatum of affected mice, suggesting that the NYT effect was unrelated to DTA-mediated neurodegeneration. CONCLUSION: These results demonstrate that the beneficial effect of NYT on motivation is mediated, at least in part, through the potentiation of hedonic capacity by certain neuromodulatory pathways.


Subject(s)
Anhedonia/drug effects , Corpus Striatum/drug effects , Drugs, Chinese Herbal/pharmacology , Medicine, Kampo/methods , Motivation/drug effects , Receptors, Dopamine D2/biosynthesis , Anhedonia/physiology , Animals , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Corpus Striatum/metabolism , Gene Expression , Japan , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Motivation/physiology , Neurons/drug effects , Neurons/metabolism , Receptors, Dopamine D2/genetics
10.
J Ethnopharmacol ; 275: 114096, 2021 Jul 15.
Article in English | MEDLINE | ID: mdl-33823166

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The increasing use of "kidney"-nourishing Traditional Chinese Medicine (TCM) like Er-xian decoction (EXD) for management of menopausal symptoms and osteoporosis has aroused concerns about their safety, and whether they interact with prescription drugs as both of them act via estrogen receptors (ERs) and regulate serum estradiol. AIM OF THE STUDY: The present study aimed to evaluate whether EXD selectively exerted estrogenic activities and interacted with Selective Estrogen Receptor Modulators (SERMs). MATERIALS AND METHODS: In vivo, mature ovariectomized (OVX) rats were administrated with EXD or combined treatment of EXD and SERMs for 12 weeks. The tissue-selective effect of EXD and its interaction of SERMs were studied in four estrogen sensitive tissues, bone, brain, breast and uterus. In vitro, the interaction of extracts of EXD-treated serum and SERMs in four ER-positive cell lines. RESULTS: In OVX rats, EXD selectively alleviated estrogen deficiency-induced changes in the bone and brain without inducing any estrogenic effects in the breast or uterus. Two-way ANOVA indicated the presence of interactions between EXD and SERMs in OVX rats but EXD did not significantly alter the tissue responses to SERMs in the bone, breast or brain. Indeed, the combined use of EXD and SERMs appeared to suppress the estrogenic effect of raloxifene and tamoxifen in the uterus. Extract of EXD-treated serum directly stimulated cell proliferation or differentiation in human osteosarcoma MG-63, neuroblastoma SHSY5Y, breast cancer MCF-7, and endometrial Ishikawa cells. Two-way ANOVA revealed that EXD-treated serum interacted with SERMs at various concentrations and altered the effects of tamoxifen in MG-63 and MCF-7 cells. CONCLUSIONS: EXD exerted estrogenic effects in a tissue-selective manner and interacted with SERMs. Combined treatment of EXD and SERMs did not hamper the beneficial effects of SERMs on the bone or brain but appeared to moderate the estrogenic effect of SERMs in the uterus.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Estrogens/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Alkaline Phosphatase/metabolism , Animals , Body Weight/drug effects , Bone and Bones/drug effects , Bone and Bones/metabolism , Breast/drug effects , Breast/metabolism , Breast/pathology , Cell Line, Tumor , Central Nervous System/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/therapeutic use , Estradiol/pharmacology , Estradiol/therapeutic use , Estrogens/chemistry , Estrogens/therapeutic use , Female , Herb-Drug Interactions/physiology , Hormones/blood , Humans , Mammary Glands, Human/drug effects , Medicine, Chinese Traditional , Models, Biological , Ovariectomy/adverse effects , Raloxifene Hydrochloride/pharmacology , Raloxifene Hydrochloride/therapeutic use , Rats, Sprague-Dawley , Receptors, Estrogen/metabolism , Selective Estrogen Receptor Modulators/therapeutic use , Tamoxifen/pharmacology , Tamoxifen/therapeutic use , Uterus/drug effects , Uterus/metabolism , Uterus/pathology , Water
11.
Phytomedicine ; 83: 153474, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33548867

ABSTRACT

BACKGROUND: Limonene, a common terpene found in citrus fruits, is assumed to reduce stress and mood disorders. Dopamine and γ-aminobutyric acid (GABA) have been reported to play an important role in modulating anxiety in different parts of the brain. HYPOTHESIS/PURPOSE: Herein, we report the anxiolytic activity of limonene. In addition, we identified a possible mechanism underlying the effect of limonene on DAergic and GABAergic neurotransmission. STUDY DESIGN: In this study, mice were injected with saline in the control group and limonene in the test group before behavioral analysis. We performed immunoblotting and high-performance liquid chromatography (HPLC) analysis after the behavioral study. RESULTS: The limonene treated group showed increased locomotor activity and open-arm preference in the elevated plus maze experiment. Limonene treatment increased the expression of both tyrosine hydroxylase and GAD-67 proteins and significantly upregulated dopamine levels in the striatum. Furthermore, tissue dopamine levels were increased in the striatum of mice following limonene treatment, and depolarization-induced GABA release was enhanced by limonene pre-treatment in PC-12 cells. Interestingly, limonene-induced anxiolytic activity and GABA release augmentation were blocked by an adenosine A2A receptor (A2AR) antagonist. CONCLUSION: Our results suggest that limonene inhibits anxiety-related behavior through A2A receptor-mediated regulation of DAergic and GABAergic neuronal activity.


Subject(s)
Anti-Anxiety Agents/pharmacology , Corpus Striatum/drug effects , Limonene/pharmacology , Receptor, Adenosine A2A/metabolism , Animals , Anxiety/drug therapy , Anxiety/metabolism , Behavior, Animal/drug effects , Corpus Striatum/metabolism , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Locomotion/drug effects , Male , Mice, Inbred C57BL , Rats , Synaptic Transmission/drug effects , Tyrosine 3-Monooxygenase/metabolism , gamma-Aminobutyric Acid/metabolism
12.
Nutr Neurosci ; 24(1): 1-12, 2021 Jan.
Article in English | MEDLINE | ID: mdl-30822260

ABSTRACT

Background: Essential fatty acids (EFAs) and non-essential fatty acids (nEFAs) exert experimental and clinical neuroprotection in neurodegenerative diseases. The main EFAs, eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), nEFAs, and oleic acid (OA) contained in olive and fish oils are inserted into the cell membranes, but the exact mechanism through which they exert neuroprotection is still unknown. Objectives and Methods: In this study, we assessed the fatty acids content and membrane fluidity in striatal rat synaptosomes after fatty acid-rich diets (olive- or a fish-oil diet, 15% w/w). Then, we evaluated the effect of enriching striatum synaptosomes with fatty acids on the oxidative damage produced by the prooxidants ferrous sulfate (FeSO4) or quinolinic acid (QUIN). Results and Discussion: Lipid profile analysis in striatal synaptosomes showed that EPA content increased in the fish oil group in comparison with control and olive groups. Furthermore, we found that synaptosomes enriched with fatty acids and incubated with QUIN or FeSO4 showed a significant oxidative damage reduction. Results suggest that EFAs, particularly EPA, improve membrane fluidity and confer antioxidant effect.


Subject(s)
Cell Membrane/metabolism , Corpus Striatum/metabolism , Fatty Acids/metabolism , Oxidative Stress , Synaptosomes/metabolism , Animals , Cell Membrane/ultrastructure , Corpus Striatum/drug effects , Corpus Striatum/ultrastructure , Fatty Acids/administration & dosage , Fish Oils/administration & dosage , Male , Plant Oils/administration & dosage , Rats, Wistar , Reactive Oxygen Species/metabolism , Synaptosomes/ultrastructure
13.
Neuroimage ; 225: 117456, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33069863

ABSTRACT

Hallucinogenic agents have been proposed as potent antidepressants; this includes the serotonin (5-HT) receptor 2A agonist psilocybin. In human subjects, psilocybin alters functional connectivity (FC) within the default-mode network (DMN), a constellation of inter-connected regions that displays altered FC in depressive disorders. In this study, we investigated the effects of psilocybin on FC across the entire brain with a view to investigate underlying mechanisms. Psilocybin effects were investigated in lightly-anaesthetized mice using resting-state fMRI. Dual-regression analysis identified reduced FC within the ventral striatum in psilocybin- relative to vehicle-treated mice. Refinement of the analysis using spatial references derived from both gene expression maps and viral tracer projection fields revealed two distinct effects of psilocybin: it increased FC between 5-HT-associated networks and cortical areas, including elements of the murine DMN, thalamus, and midbrain; it decreased FC within dopamine (DA)-associated striatal networks. These results suggest that interactions between 5-HT- and DA-regulated neural networks contribute to the neural and therefore psychological effects of psilocybin. Furthermore, they highlight how information on molecular expression patterns and structural connectivity can assist in the interpretation of pharmaco-fMRI findings.


Subject(s)
Brain/drug effects , Default Mode Network/drug effects , Psilocybin/pharmacology , Serotonin 5-HT2 Receptor Agonists/pharmacology , Animals , Brain/diagnostic imaging , Brain/metabolism , Corpus Striatum/diagnostic imaging , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Default Mode Network/diagnostic imaging , Default Mode Network/metabolism , Dopamine/metabolism , Functional Neuroimaging , Magnetic Resonance Imaging , Mesencephalon/diagnostic imaging , Mesencephalon/drug effects , Mesencephalon/metabolism , Mice , Neural Pathways/diagnostic imaging , Neural Pathways/drug effects , Neural Pathways/metabolism , Rest , Serotonin/metabolism , Thalamus/diagnostic imaging , Thalamus/drug effects , Thalamus/metabolism
14.
Pharmacol Biochem Behav ; 200: 173087, 2021 01.
Article in English | MEDLINE | ID: mdl-33309825

ABSTRACT

Methamphetamine (Meth) seeking progressively increases after cessation from drug self-administration (incubation of Meth craving). We have previously shown that both dorsomedial and dorsolateral striatum (DMS and DLS) play critical roles in this incubation in male rats. Moreover, our recent anatomical tracing study examined afferent projections into DMS and demonstrated a novel role of projections from anterior intralaminar nucleus of thalamus (AIT) to DMS in incubation of Meth craving in male rats. Here we investigated projection-specific activation of afferent glutamate projections into DLS associated with incubated Meth seeking in female rats. We trained female rats to self-administer Meth (6-h/d for 10 d). On abstinence day 12, we injected cholera toxin subunit B (CTb, a retrograde tracer) unilaterally into DLS. On abstinence day 26, we tested rats for relapse to Meth seeking and measured Fos (a neuronal activity marker), and double-labeling of CTb and Fos in anterior cingulate cortex, anterior insula cortex, orbitofrontal cortex, basolateral amygdala, AIT, and parafascicular nuclei of thalamus. We observed neuronal activation in both cortical and thalamic regions associated with incubated Meth seeking. At the circuit level, AIT➔DLS projections were strongly activated, followed by other corticostriatal projections. Overall our results suggest that AIT to DLS may play a role in Meth seeking after prolonged abstinence in female rats.


Subject(s)
Central Nervous System Stimulants/pharmacology , Corpus Striatum/metabolism , Drug-Seeking Behavior/drug effects , Intralaminar Thalamic Nuclei/metabolism , Methamphetamine/pharmacology , Amphetamine-Related Disorders/metabolism , Animals , Cerebral Cortex/drug effects , Corpus Striatum/drug effects , Craving/drug effects , Female , Intralaminar Thalamic Nuclei/drug effects , Neurons/drug effects , Prefrontal Cortex/drug effects , Rats , Rats, Sprague-Dawley , Self Administration/methods , Thalamus/drug effects , Thalamus/metabolism
15.
Molecules ; 25(24)2020 Dec 14.
Article in English | MEDLINE | ID: mdl-33327458

ABSTRACT

(1) Background: Sesame has been popular as a healthy food since ancient times, and effects of the aroma component of roasted sesame are also expected. However, little research has been reported on its scent; (2) Methods: Jcl:ICR male mice were housed under water immersion stress for 24 h. Then, the scent of saline or sesame oil was inhaled to stress groups for 90 min. We investigated the effects of sesame oil aroma on the behavior and brains of mice; (3) Results: In an elevated plus maze test, the rate of entering to open arm and the staying time were decreased by the stress. These decrements were significantly enhanced by sesame oil aroma. Stress had a tendency to increase the serum corticosterone concentration, which was slightly decreased by the aroma. Expression of Kruppel-like factor-4 (Klf-4) and Dual-specificity phosphatase-1 (Dusp-1) in the striatum were increased by water immersion stress, and the level of Klf-4 and Dusp-1 in the striatum and hippocampus were significantly attenuated by sesame oil aroma (4) Conclusions: The present results strongly suggest that the odor component of sesame oil may have stress suppressing effects. Moreover, Klf-4 and Dusp-1 may be sensitive stress-responsive biomarkers.


Subject(s)
Anti-Anxiety Agents/pharmacology , Corpus Striatum/drug effects , Hippocampus/drug effects , Odorants/analysis , Sesame Oil/pharmacology , Stress, Psychological/drug therapy , Administration, Inhalation , Animals , Anti-Anxiety Agents/chemistry , Biomarkers/metabolism , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Corticosterone/blood , Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 1/metabolism , Gene Expression/drug effects , Hippocampus/metabolism , Hippocampus/physiopathology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred ICR , Sesame Oil/chemistry , Sesamum/chemistry , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Stress, Psychological/psychology , Swimming/psychology
16.
Brain Res ; 1749: 147145, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33035499

ABSTRACT

Degeneration of striatal neurons and cortical atrophy are pathological characteristics of glutaric acidemia type I (GA-I), a disease characterized by accumulation of glutaric acid (GA). The mechanisms that lead to neuronal loss and cognitive impairment are still unclear. The purpose of this study was to verify if acute exposure to GA during the neonatal period is sufficient to trigger apoptotic processes and lead to learning delay in early and late period. Besides, whether N-acetylcysteine (NAC) would protect against impairment induced by GA. Pups mice received a dose of GA (2.5 µmol/ g) or saline, 12 hs after birth, and were treated with NAC (250 mg/kg) or saline, up to 21th day of life. Although GA exhibited deficits in the procedural and working memories in 21 and 40-day-old mice, NAC protected against cognitive impairment. In striatum and cortex, NAC prevented glial cells activation (GFAP and Iba-1), decreased NGF, Bcl-2 and NeuN, the increase of lipid peroxidation and PARP induced by GA in both ages. NAC protected against increased p75NTR induced by GA, but not in cortex of 21-day-old mice. Thus, we showed that the integrity of striatal and cortical pathways has an important role for learning and suggested that sustained glial reactivity in neonatal period can be an initial trigger for delay of cognitive development. Furthermore, NAC protected against cognitive impairment induced by GA. This work shows that early identification of the alterations induced by GA is important to avoid future clinical complications and suggest that NAC could be an adjuvant treatment for this acidemia.


Subject(s)
Acetylcysteine/pharmacology , Cerebral Cortex/drug effects , Corpus Striatum/drug effects , Glutarates/pharmacology , Maze Learning/drug effects , Receptors, Nerve Growth Factor/metabolism , Animals , Apoptosis/drug effects , Cerebral Cortex/metabolism , Cognition/drug effects , Corpus Striatum/metabolism , Lipid Peroxidation/drug effects , Male , Mice , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects
17.
Neurosci Lett ; 738: 135344, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32889006

ABSTRACT

Prolonged exposure to manganese (Mn) may lead to toxic effects on the central nervous system (CNS). The mechanisms underlying neuronal death from exposure to Mn are not well understood but undoubtedly involve inflammatory processes. The aim of this study was to explore the effects of long-lasting intranasal Mn exposure in rats focusing on inflammatory processes and catecholamine (dopamine, norepinephrine) levels in the striatum and hippocampus. It was found that intranasal administration by instillation of MnCl2 solution once a day for 90 days leads to impaired movement and gait. We also observed that Mn concentration increased in the hippocampus (by 30 %) and in the striatum (by 220 %), dopamine (24 %) and DOPAC (35 %) were reduced in the striatum, and dopamine (190 %) and DOPAC (220 %) levels increased with simultaneously norepinephrine reduction (30 %) in the hippocampus. Observation of cytokine mRNA revealed increased expression of both assayed cytokines (IL-1ß and TNF-α) in the hippocampus. There was a 3-fold increase in the expression of IBA-1 mRNA, 2-fold increase in NFκB mRNA, and dramatic reduction in IkB mRNA in the striatum. Taken together, intranasal exposure to a high dose of MnCl2 induces neuroinflammation and neurotransmission disturbance, but the effects are specific for each studied brain region.


Subject(s)
Corpus Striatum/drug effects , Dopamine/metabolism , Hippocampus/drug effects , Inflammation/metabolism , Manganese/administration & dosage , Administration, Intranasal , Animals , Corpus Striatum/metabolism , Hippocampus/metabolism , Interleukin-1beta/metabolism , Male , Motor Activity/drug effects , Norepinephrine/metabolism , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/metabolism
18.
Med Sci Monit ; 26: e924658, 2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32738135

ABSTRACT

BACKGROUND Anxiety is one of the common comorbidities of Tourette syndrome (TS). The serotonin (5-HT) system is involved in both TS and anxiety. Jian-pi-zhi-dong decoction (JPZDD) is widely used. However, the mechanism remains unknown. In this study, a rat model of TS and comorbid anxiety was used to evaluate the effect of JPZDD on 5-HT and its receptor. MATERIAL AND METHODS 48 rats were divided into 4 groups randomly (n=12). The model was established by empty water bottle stimulation plus iminodipropionitrile injection for 3 weeks. Then the control and model groups were gavaged with saline, while the treatment groups were gavaged with fluoxetine hydrochloride (Flx) or JPZDD. Body weights were measured, and behavioral tests were evaluated with stereotypy and elevated plus maze. The morphologic characters were observed by hematoxylin and eosin staining. The content of 5-HT was detected by enzyme-linked immunosorbent assay and high-performance liquid chromatography. The expression of 5-HT2C receptor was detected by western blot and quantitative polymerase chain reaction. RESULTS The stereotypy score was lower and the time spent in the open arm was longer in the JPZDD group compared with the model group. After the treatment of Flx or JPZDD, the structure of neurons became gradually normal and the cells were arranged neatly. The contents of 5-HT in the treatment groups were higher compared with the model group in the striatum. The expression of 5-HT2C mRNA in the striatum of JPZDD and Flx groups decreased compared with the model group, and the JPZDD group was lower than the Flx group. CONCLUSIONS JPZDD alleviated both tic and anxiety symptoms and the mechanism may be via reducing the expression of 5-HT2C mRNA in the striatum, increasing the concentration of 5-HT, and enhancing the activity of the 5-HT system, which in turn exerts neuro-inhibition.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Anxiety/drug therapy , Drugs, Chinese Herbal/pharmacology , Receptor, Serotonin, 5-HT2C/genetics , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Tourette Syndrome/drug therapy , Animals , Anxiety/chemically induced , Anxiety/genetics , Anxiety/physiopathology , Behavior, Animal/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Disease Models, Animal , Fluoxetine/pharmacology , Gene Expression , Humans , Male , Maze Learning/drug effects , Nitriles/administration & dosage , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT2C/metabolism , Serotonin/metabolism , Tourette Syndrome/chemically induced , Tourette Syndrome/genetics , Tourette Syndrome/physiopathology , Treatment Outcome
19.
Biosci Trends ; 14(4): 271-278, 2020 Sep 21.
Article in English | MEDLINE | ID: mdl-32741856

ABSTRACT

Tourette's syndrome (TS) is an inherited neurologic disorder characterized by involuntary stereotyped motor and vocal tics. Its pathogenesis is still unclear and its treatment remains limited. Recent research has suggested the involvement of immune mechanisms in the pathophysiology of TS. Microglia are the brain's resident innate immune cells. They can mediate neuroinflammation and regulate brain development and homeostasis. A traditional Chinese medicine (TCM), Ningdong granule (NDG), has been found to be efficacious in the treatment of TS while causing few adverse reactions. In the current study, a rat model of 3,3'-iminodipropionitrile (IDPN)-induced TS was used to explore the regulating effects and mechanisms of NDG on microglia-mediated neuroinflammation. IDNP led to robust pathological changes and neurobehavioral complications, with activation of microglia in the striatum of rats with TS. After activation by IDNP, microglia strongly responded to this specific injury, and TNF-α, IL-6, and MCP-1 were released in the striatum and/or serum of rats with TS. Interestingly, NDG inhibited the activation of microglia and decreased the abnormal expression of TNF-α, IL-6, and MCP-1 in the striatum and/or serum of rats with TS, thus controlling tics. However, there were no significant changes in the striatum and/or serum of rats with TS after treatment with haloperidol. The anti-TS action of haloperidol might occur not through microglial activation and neuroinflammation but through the DAT system, thus controlling tics. In conclusion, microglia might play key roles in mediating neuroinflammatory responses in TS, triggering the release of TNF-α, IL-6, and MCP-1.NDG inhibited tics in rats with TS, and this mechanism may be associated with a reduction in the increased number of activated microglia and a decrease in the expression of pro-inflammatory cytokines and chemokines in the striatum and/or serum.


Subject(s)
Corpus Striatum/drug effects , Drugs, Chinese Herbal/pharmacology , Microglia/drug effects , Tourette Syndrome/drug therapy , Animals , Chemokine CCL2/blood , Chemokine CCL2/metabolism , Corpus Striatum/cytology , Corpus Striatum/immunology , Corpus Striatum/pathology , Disease Models, Animal , Drugs, Chinese Herbal/therapeutic use , Haloperidol/pharmacology , Haloperidol/therapeutic use , Humans , Interleukin-6/blood , Interleukin-6/metabolism , Male , Microglia/immunology , Microglia/metabolism , Nitriles/toxicity , Rats , Rats, Wistar , Tourette Syndrome/chemically induced , Tourette Syndrome/immunology , Tourette Syndrome/pathology , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism
20.
Psychopharmacology (Berl) ; 237(11): 3225-3236, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32651640

ABSTRACT

The asymptomatic and clinical stages of Parkinson's disease (PD) are associated with comorbid non-motor symptoms including gastrointestinal (GI) dysfunction. Although the neuroprotective and gastroprotective roles of kolaviron (KV) have been reported independently, whether KV-mediated GI-protective capacity could be beneficial in PD is unknown. We therefore investigated the modulatory effects of KV on the loss of dopaminergic neurons, locomotor abnormalities, and ileal oxidative damage when rats are lesioned in the nigrostriatal pathway. KV treatment markedly suppressed the behavioral deficit and apomorphine-induced rotations associated with rotenone lesioning. KV attenuated the loss of nigrostriatal dopaminergic neurons and perturbations in the striatal glucose-regulated protein (GRP78) and X-box binding protein 1 (XBP1) levels. Ileal epithelial injury following stereotaxic rotenone infusion was associated with oxidative stress and marked inhibition of acetylcholine esterase activity and reduced expression of occludin in the crypt and villi. While KV treatment attenuated the redox imbalance in the gut and enhanced occludin immunoreactivity, acetylcholinesterase activity was not affected. Our data demonstrate ileal oxidative damage as a characteristic non-motor gut dysfunction in PD while showing the potential dual efficacy of KV in the attenuation of both neural defects and gut abnormalities associated with PD.


Subject(s)
Corpus Striatum/drug effects , Flavonoids/administration & dosage , Gastrointestinal Microbiome/drug effects , Parkinsonian Disorders/prevention & control , Rotenone/toxicity , Substantia Nigra/drug effects , Animals , Corpus Striatum/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Flavonoids/isolation & purification , Gastrointestinal Microbiome/physiology , Male , Nerve Degeneration/chemically induced , Nerve Degeneration/drug therapy , Nerve Degeneration/prevention & control , Neuroprotective Agents/administration & dosage , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/metabolism , Plant Extracts/administration & dosage , Plant Extracts/isolation & purification , Rats , Rotenone/administration & dosage , Stereotaxic Techniques , Substantia Nigra/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL