Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
Add more filters











Publication year range
1.
Biomolecules ; 14(8)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39199395

ABSTRACT

One aspect of reproducibility in preclinical research that is frequently overlooked is the physical condition in which physiological, pharmacological, or behavioural recordings are conducted. In this study, the physical conditions of mice were altered through the attachments of wireless electrophysiological recording devices (Neural Activity Tracker-1, NAT-1). NAT-1 devices are miniaturised multichannel devices with onboard memory for direct high-resolution recording of brain activity for >48 h. Such devices may limit the mobility of animals and affect their behavioural performance due to the added weight (total weight of approximately 3.4 g). The mice were additionally treated with saline (control), N-methyl-D-aspartate (NMDA) receptor antagonist MK801 (0.85 mg/kg), or the muscarinic acetylcholine receptor blocker scopolamine (0.65 mg/kg) to allow exploration of the effect of NAT-1 attachments in pharmacologically treated mice. We found only minimal differences in behavioural outcomes with NAT-1 attachments in standard parameters of locomotor activity widely reported for the open field test between the drug treatments. Hypoactivity was globally observed as a consistent outcome in the MK801-treated mice and hyperactivity in scopolamine groups regardless of NAT-1 attachments. These data collectively confirm the reproducibility for combined behavioural, pharmacological, and physiological endpoints even in the presence of lightweight wireless data loggers. The NAT-1 therefore constitutes a pertinent tool for investigating brain activity in, e.g., drug discovery and models of neuropsychiatric and/or neurodegenerative diseases with minimal effects on pharmacological and behavioural outcomes.


Subject(s)
Dizocilpine Maleate , Electroencephalography , Exploratory Behavior , Scopolamine , Animals , Scopolamine/pharmacology , Dizocilpine Maleate/pharmacology , Mice , Male , Exploratory Behavior/drug effects , Behavior, Animal/drug effects , Open Field Test/drug effects
2.
Behav Pharmacol ; 35(6): 338-350, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39051900

ABSTRACT

Increasing evidence indicates that neuroinflammation, oxidative stress, and neurotrophic factors play a key role in the pathophysiology of major depressive disorder (MDD). In addition, the attenuation of inflammatory response has been considered a putative mechanism for MDD treatment. PT-31 is an imidazolidine derivative and a putative α2-adrenoceptor agonist that has previously demonstrated antinociceptive activity. The present study aimed to investigate the effect of PT-31 on depressive-like behavior and lipopolysaccharide-induced neurochemical changes. To this end, mice received intraperitoneally saline or lipopolysaccharide (600 µg/kg), and 5 h postinjection animals were orally treated with saline, PT-31 (3, 10, and 30 mg/kg), or fluoxetine (30 mg/kg). Mice were subjected to the open field test (OFT) 6 and 24 h after lipopolysaccharide administration and to the tail suspension test (TST) 24 h postlipopolysaccharide. Subsequently, animals were euthanized, and brains were dissected for neurochemical analyses. The administration of lipopolysaccharide-induced sickness- and depressive-like behaviors, besides promoting an increase in myeloperoxidase activity and a reduction in brain-derived neurotrophic factor (BDNF) levels. Noteworthy, PT-31 3 mg/kg attenuated lipopolysaccharide-induced decreased locomotor activity 6 h after lipopolysaccharide in the OFT. All tested doses of PT-31 significantly reduced the immobility time of animals in the TST and attenuated lipopolysaccharide-induced increased myeloperoxidase activity in the cortex of mice. Our results demonstrate that PT-31 ameliorates behavioral changes promoted by lipopolysaccharide in OFT and TST, which is possibly mediated by attenuation of the inflammatory response.


Subject(s)
Adrenergic alpha-2 Receptor Agonists , Antidepressive Agents , Behavior, Animal , Brain-Derived Neurotrophic Factor , Depression , Lipopolysaccharides , Animals , Lipopolysaccharides/pharmacology , Mice , Male , Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Depression/drug therapy , Depression/metabolism , Adrenergic alpha-2 Receptor Agonists/pharmacology , Behavior, Animal/drug effects , Disease Models, Animal , Fluoxetine/pharmacology , Dose-Response Relationship, Drug , Open Field Test/drug effects , Brain/metabolism , Brain/drug effects , Hindlimb Suspension , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/metabolism
3.
Cardiovasc Toxicol ; 24(8): 766-775, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38850470

ABSTRACT

Cognitive impairment is a commonly observed complication following myocardial infarction; however, the underlying mechanisms are still not well understood. The most recent research suggests that extracellular signal-regulated kinase (ERK) plays a critical role in the development and occurrence of cognitive dysfunction-related diseases. This study aims to explore whether the ERK inhibitor U0126 targets the ERK/Signal Transducer and Activator of Transcription 1 (STAT1) pathway to ameliorate cognitive impairment after myocardial infarction. To establish a mouse model of myocardial infarction, we utilized various techniques including Echocardiography, Hematoxylin-eosin (HE) staining, Elisa, Open field test, Elevated plus maze test, and Western blot analysis to assess mouse cardiac function, cognitive function, and signal transduction pathways. For further investigation into the mechanisms of cognitive function and signal transduction, we administered the ERK inhibitor U0126 via intraperitoneal injection. Reduced total distance and activity range were observed in mice subjected to myocardial infarction during the open field test, along with decreased exploration of the open arms in the elevated plus maze test. However, U0126 treatment exhibited a significant improvement in cognitive decline, indicating a protective effect through the inhibition of the ERK/STAT1 signaling pathway. Hence, this study highlights the involvement of the ERK/STAT1 pathway in regulating cognitive dysfunction following myocardial infarction and establishes U0126 as a promising therapeutic target.


Subject(s)
Behavior, Animal , Butadienes , Cognition , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases , Mice, Inbred C57BL , Myocardial Infarction , Nitriles , STAT1 Transcription Factor , Animals , Myocardial Infarction/enzymology , Myocardial Infarction/physiopathology , Myocardial Infarction/pathology , Myocardial Infarction/drug therapy , Cognition/drug effects , Nitriles/pharmacology , Male , Butadienes/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Behavior, Animal/drug effects , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/enzymology , Cognitive Dysfunction/prevention & control , Signal Transduction/drug effects , Elevated Plus Maze Test , Open Field Test/drug effects , Mice
4.
Behav Pharmacol ; 35(5): 303-314, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38869060

ABSTRACT

Riparin A is a synthetic form of natural riparins. Acute scale studies that take into consideration the structure-activity relationship have shown preliminary evidence of antidepressant and anxiolytic effects of riparin A, similar to that already known for other riparins. However, for better pharmacological characterization of this new compound, further studies are required. The aim of this work was to evaluate the effect of chronic treatment with riparin A (10 mg/kg; intraperitoneally) on depressive-like behavior in the forced swimming test and tail suspension test, as well as the reduction of anhedonia in the sucrose preference test, and on anxiety-like behavior in the open field and elevated plus maze apparatus, triggered in rats previously subjected to unpredictable chronic mild stress by 4 weeks. In addition, a pentobarbital-induced sleep time test was also used. Riparin A reduced the duration of immobility in both the forced swimming test and tail suspension test, as well as attenuated the anhedonia in the sucrose preference test. Furthermore, riparin A appears to produce anxiolytic effects in rats exposed to an open field and elevated plus maze, while increasing the alertness/vigilance in rats submitted to pentobarbital-induced sleep time test, without altering their locomotor integrity. Our results suggest that chronic riparin A appears to be a potential pharmacological target for new studies on the control of depression- and anxiety-like behaviors in stressed rats.


Subject(s)
Antidepressive Agents , Anxiety , Depression , Disease Models, Animal , Rats, Wistar , Animals , Antidepressive Agents/pharmacology , Male , Depression/drug therapy , Rats , Anxiety/drug therapy , Anti-Anxiety Agents/pharmacology , Behavior, Animal/drug effects , Swimming/psychology , Anhedonia/drug effects , Stress, Psychological/drug therapy , Hindlimb Suspension , Maze Learning/drug effects , Mice , Open Field Test/drug effects
5.
Pharmacol Biochem Behav ; 240: 173790, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38761992

ABSTRACT

Ketamine is a rapid-acting antidepressant associated with various cognitive side effects. To mitigate these side effects while enhancing efficacy, it can be co-administered with other antidepressants. In our study, we adopted a similar strategy by combining ketamine with environmental enrichment, a potent sensory-motor paradigm, in adult male Wistar rats. We divided the animals into four groups based on a combination of housing conditions and ketamine versus vehicle injections. The groups included those housed in standard cages or an enriched environment for 50 days, which encompassed a 13-day-long behavioral testing period. Each group received either two doses of ketamine (20 mg/kg, IP) or saline as a vehicle. We tested the animals in the novel object recognition test (NORT), forced swim test (FST), open field test (OFT), elevated plus maze (EPM), and Morris water maze (MWM), which was followed by ex vivo c-Fos immunohistochemistry. We observed that combining environmental enrichment with ketamine led to a synergistic antidepressant effect. Environmental enrichment also ameliorated the spatial memory deficits caused by ketamine in the MWM. There was enhanced neuronal activity in the habenula of the enrichment only group following the probe trial of the MWM. In contrast, no differential activity was observed in enriched animals that received ketamine injections. The present study showed how environmental enrichment can enhance the antidepressant properties of ketamine while reducing some of its side effects, highlighting the potential of combining pharmacological and sensory-motor manipulations in the treatment of mood disorders.


Subject(s)
Antidepressive Agents , Ketamine , Memory Disorders , Rats, Wistar , Spatial Memory , Animals , Ketamine/pharmacology , Ketamine/administration & dosage , Male , Rats , Antidepressive Agents/pharmacology , Antidepressive Agents/administration & dosage , Memory Disorders/drug therapy , Memory Disorders/chemically induced , Spatial Memory/drug effects , Environment , Open Field Test/drug effects , Maze Learning/drug effects , Behavior, Animal/drug effects
6.
Article in English | MEDLINE | ID: mdl-38642730

ABSTRACT

Continuous antipsychotic treatment is often recommended to prevent relapse in schizophrenia. However, the efficacy of antipsychotic treatment appears to diminish in patients with relapsed schizophrenia and the underlying mechanisms are still unknown. Moreover, though the findings are inconclusive, several recent studies suggest that intermittent versus continuous treatment may not significantly differ in recurrence risk and therapeutic efficacy but potentially reduce the drug dose and side effects. Notably, disturbances in fatty acid (FA) metabolism are linked to the onset/relapse of schizophrenia, and patients with multi-episode schizophrenia have been reported to have reduced FA biosynthesis. We thus utilized an MK-801-induced animal model of schizophrenia to evaluate whether two treatment strategies of clozapine would affect drug response and FA metabolism differently in the brain. Schizophrenia-related behaviors were assessed through open field test (OFT) and prepulse inhibition (PPI) test, and FA profiles of prefrontal cortex (PFC) and hippocampus were analyzed by gas chromatography-mass spectrometry. Additionally, we measured gene expression levels of enzymes involved in FA synthesis. Both intermittent and continuous clozapine treatment reversed hypermotion and deficits in PPI in mice. Continuous treatment decreased total polyunsaturated fatty acids (PUFAs), saturated fatty acids (SFAs) and FAs in the PFC, whereas the intermittent administration increased n-6 PUFAs, SFAs and FAs compared to continuous administration. Meanwhile, continuous treatment reduced the expression of Fads1 and Elovl2, while intermittent treatment significantly upregulated them. This study discloses the novel findings that there was no significant difference in clozapine efficacy between continuous and intermittent administration, but intermittent treatment showed certain protective effects on phospholipid metabolism in the PFC.


Subject(s)
Antipsychotic Agents , Clozapine , Disease Models, Animal , Dizocilpine Maleate , Fatty Acids , Schizophrenia , Animals , Clozapine/pharmacology , Clozapine/administration & dosage , Schizophrenia/drug therapy , Schizophrenia/metabolism , Dizocilpine Maleate/pharmacology , Antipsychotic Agents/pharmacology , Antipsychotic Agents/administration & dosage , Fatty Acids/metabolism , Male , Mice , Brain/metabolism , Brain/drug effects , Prepulse Inhibition/drug effects , Mice, Inbred C57BL , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Open Field Test/drug effects
7.
Naunyn Schmiedebergs Arch Pharmacol ; 397(8): 6153-6163, 2024 08.
Article in English | MEDLINE | ID: mdl-38430231

ABSTRACT

Depression is a debilitating mood disorder affecting millions worldwide and continues to pose a significant global health burden. Due to the multifaceted nature of depression, the current treatment regimens are not up to mark in terms of their multitargeting potential and least side effect profile. Molecules within the isoflavone class demonstrate promising potential in alleviating depression and associated conditions, offering a multifaceted approach to manage mental health concerns. Therefore, the current study was designed to explore the potential of glycitein, an isoflavone in managing reserpine-induced depression and associated comorbidities in mice. Reserpine (0.5 mg/kg; i.p.) administration for the first 3 days induced depression and associated comorbidities as evidenced by increased immobility time in forced swim test (FST) and tail suspension test (TST), along with reduced locomotor activity in the open field test (OFT) and increased latency to reach the platform in the Morris water maze (MWM) test. Reserpine treatment also upregulated and downregulated the brain thiobarbituric acid reactive substance (TBARS) and glutathione (GSH) levels, respectively. Furthermore, reserpine administration also uplifted the level of TNF-α in the serum samples. Glycitein (3 mg/kg and 6 mg/kg; p.o.) treatment for 5 days prevented the depressive effect of reserpine. It also improved the spatial memory at both dose levels. Moreover, in biochemical analysis, glycitein also reduced the brain TBARS and serum tumor necrosis factor-alpha (TNF-α) levels. Whereas, no significant effect was seen on the brain GSH level. Glycitein (6 mg/kg) was found to be more effective than the 3 mg/kg dose of glycitein. Overall results delineate that glycitein has the potential to manage depression and impaired memory by inhibiting lipid peroxidation and inflammatory stress.


Subject(s)
Depression , Lipid Peroxidation , Reserpine , Tumor Necrosis Factor-alpha , Animals , Depression/drug therapy , Depression/metabolism , Depression/prevention & control , Depression/chemically induced , Mice , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/blood , Male , Lipid Peroxidation/drug effects , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Isoflavones/pharmacology , Isoflavones/therapeutic use , Behavior, Animal/drug effects , Disease Models, Animal , Brain/metabolism , Brain/drug effects , Glutathione/metabolism , Open Field Test/drug effects , Comorbidity
8.
Int J Mol Sci ; 23(2)2022 Jan 15.
Article in English | MEDLINE | ID: mdl-35055113

ABSTRACT

Methionine (Met) is considered the most toxic amino acid in mammals. Here, we investigated biochemical and behavioral impacts of ad libitum one-week feeding of high-Met diets on mice. Adult male mice were fed the standard rodent diet that contained 0.44% Met (1×) or a diet containing 16 graded Met doses (1.2×-13×). High-Met diets for one-week induced a dose-dependent decrease in body weight and an increase in serum Met levels with a 2.55 mM peak (versus basal 53 µM) on the 12×Met diet. Total homocysteine (Hcy) levels were also upregulated while concentrations of other amino acids were almost maintained in serum. Similarly, levels of Met and Hcy (but not the other amino acids) were highly elevated in the cerebrospinal fluids of mice on the 10×Met diet; the Met levels were much higher than Hcy and the others. In a series of behavioral tests, mice on the 10×Met diet displayed increased anxiety and decreased traveled distances in an open-field test, increased activity to escape from water soaking and tail hanging, and normal learning/memory activity in a Y-maze test, which were reflections of negative/positive symptoms and normal cognitive function, respectively. These results indicate that high-Met ad libitum feeding even for a week can induce bipolar disorder-like disease models in mice.


Subject(s)
Bipolar Disorder/psychology , Homocysteine/blood , Methionine/adverse effects , Amino Acids/blood , Amino Acids/cerebrospinal fluid , Animals , Bipolar Disorder/blood , Bipolar Disorder/cerebrospinal fluid , Bipolar Disorder/chemically induced , Disease Models, Animal , Drug Administration Schedule , Homocysteine/cerebrospinal fluid , Male , Methionine/blood , Methionine/cerebrospinal fluid , Mice , Open Field Test/drug effects , Up-Regulation
9.
Behav Brain Res ; 417: 113615, 2022 01 24.
Article in English | MEDLINE | ID: mdl-34606775

ABSTRACT

The increasing use of aluminum nanoparticles (nano-Al) leads to increased human exposure and might affect human health. Considering the suggested connection between aluminum exposure and Alzheimer's disease (AD) pathogenesis, there is a concern about the effect of nano-Al on cognitive function and brain health. This study was aimed to assess the effect of a 5-day oral gavage of aluminum oxide nanoparticle (nano-Al) on memory and the phosphorylation levels of hippocampal p38, JNK (c-Jun N-terminal kinase), ERK (extracellular signal-regulated kinase) as well as cleaved caspase-3 in mice. Adult male NMRI mice were treated with nano-Al in doses 5 and 10 mg/kg/oral gavage for 5 days. The test session of novel object recognition (NOR) task was performed on day 5. Following the NOR test, the hippocampi were isolated for western blot analysis to determine the total and phosphorylated levels of p38, JNK, ERK as well as cleaved caspase-3 proteins. The results showed that nano-Al oral gavage in doses of 5 and 10 mg/kg impairs NOR memory in mice. Moreover, the memory impairing effect of nano-Al coincided with a dose dependent increase in phosphorylated p38 and cleaved caspase-3 in the hippocampus. It also increased the ratio of phosphorylated to total content of ERK in the hippocampus while JNK signaling was not affected by nano-Al. This study showed that nano-Al in doses as low as 5 and 10 mg/ kg ingested for 5 days impairs NOR memory and activates p38, ERK and cleaved caspase-3 in the hippocampus.


Subject(s)
Aluminum/administration & dosage , Caspase 3/metabolism , Dose-Response Relationship, Drug , Hippocampus/metabolism , MAP Kinase Signaling System/drug effects , Nanoparticles , Open Field Test/drug effects , Alzheimer Disease/metabolism , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Memory/drug effects , Mice , Visual Perception
10.
Nitric Oxide ; 119: 29-40, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34896554

ABSTRACT

Nitric oxide is a small gaseous molecule that plays important roles in the majority of biological functions. Impairments of NO-related pathways contribute to the majority of neurological disorders, such as Alzheimer's disease (AD), and mental disorders, such as schizophrenia. Cognitive decline is one of the most serious impairments accompanying both AD and schizophrenia. In the present study, the activities of NO donors, slow (spermine NONOate) or fast (DETANONOate) releasers, and selective inhibitor of neuronal nitric oxide synthase N(ω)-propyl-l-arginine (NPLA) were investigated in pharmacological models of schizophrenia and AD. Cognitive impairments were induced by administration of MK-801 or scopolamine and were measured in novel object recognition (NOR) and Y-maze tests. The compounds were investigated at doses of 0.05-0.5 mg/kg. The dose-dependent effectiveness of all the compounds was observed in the NOR test, while only the highest doses of spermine NONOate and NPLA were active in the Y-maze test. DETANONOate was not active in the Y-maze test. The impact of the investigated compounds on motor coordination was tested at doses of 0.5 and 1 mg/kg. Only NPLA at a dose of 1 mg/kg slightly disturbed motor coordination in animals.


Subject(s)
Alzheimer Disease/drug therapy , Cognitive Dysfunction/drug therapy , Nitric Oxide Donors/therapeutic use , Nitric Oxide/metabolism , Nootropic Agents/therapeutic use , Schizophrenia/drug therapy , Alzheimer Disease/chemically induced , Animals , Arginine/analogs & derivatives , Arginine/therapeutic use , Cognitive Dysfunction/chemically induced , Dizocilpine Maleate , Enzyme Inhibitors/therapeutic use , Male , Mice , Nitric Oxide/antagonists & inhibitors , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitroso Compounds/therapeutic use , Open Field Test/drug effects , Rotarod Performance Test , Schizophrenia/chemically induced , Scopolamine , Spermine/analogs & derivatives , Spermine/therapeutic use
11.
Chem Biol Interact ; 351: 109767, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34863679

ABSTRACT

The present study investigated the role of neuroinflammation and brain oxidative stress induced by neonatal treatment with lipopolysaccharides (LPS) on the development of autism spectrum disorder (ASD)-like behaviors and disruptive hippocampal neurogenesis in rats by exploring the chemopreventive effects of alpha-glycosyl isoquercitrin (AGIQ) as an antioxidant. AGIQ was dietary administered to dams at 0.25% or 0.5% (w/w) from gestational day 18 until postnatal day (PND) 21 on weaning and then to pups until the adult stage on PND 77. The pups were intraperitoneally injected with LPS (1 mg/kg body weight) on PND 3. At PND 6, LPS alone increased Iba1+ and CD68+ cell numbers without changing the CD163+ cell number and strongly upregulated pro-inflammatory cytokine gene expression (Il1a, Il1b, Il6, Nfkb1, and Tnf) in the hippocampus, and increased brain malondialdehyde levels. At PND 10, pups decreased ultrasonic vocalization (USV), suggesting the induction of pro-inflammatory responses and oxidative stress to trigger communicative deficits. By contrast, LPS alone upregulated Nfe2l2 expression at PND 6, increased Iba1+, CD68+, and CD163+ cell numbers, and upregulated Tgfb1 at PND 21, suggesting anti-inflammatory responses until the weaning period. However, LPS alone disrupted hippocampal neurogenesis at weaning and suppressed social interaction parameters and rate of freezing time at fear acquisition and extinction during the adolescent stage. On PND 77, neuroinflammatory responses had mostly disappeared; however, disruptive neurogenesis and fear memory deficits were sustained. AGIQ ameliorated most changes on acute pro-inflammatory responses and oxidative stress at PND 6, and the effects on USVs at PND 10 and neurogenesis and behavioral parameters throughout the adult stage. These results suggested that neonatal LPS treatment induced acute but transient neuroinflammation, triggering the progressive disruption of hippocampal neurogenesis leading to abnormal behaviors in later life. AGIQ treatment was effective for ameliorating LPS-induced progressive changes by critically suppressing initial pro-inflammatory responses and oxidative stress.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Autistic Disorder/drug therapy , Glycosides/therapeutic use , Neuroprotective Agents/therapeutic use , Quercetin/analogs & derivatives , Animals , Animals, Newborn , Autistic Disorder/chemically induced , Autistic Disorder/pathology , Dentate Gyrus/drug effects , Dentate Gyrus/pathology , Female , Gene Expression/drug effects , Lipopolysaccharides , Male , Neurogenesis/drug effects , Neuroinflammatory Diseases/chemically induced , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/pathology , Open Field Test/drug effects , Oxidative Stress/drug effects , Pregnancy , Quercetin/therapeutic use , Rats, Sprague-Dawley , Social Interaction/drug effects
12.
Sci Rep ; 11(1): 22852, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34819526

ABSTRACT

Depression and anxiety, two of the most common mental health disorders, share common symptoms and treatments. Most pharmacological agents available to treat these disorders target monoamine systems. Currently, finding the most effective treatment for an individual is a process of trial and error. To better understand how disease etiology may predict treatment response, we studied mice exposed developmentally to the selective serotonin reuptake inhibitor (SSRI) fluoxetine (FLX). These mice show the murine equivalent of anxiety- and depression-like symptoms in adulthood and here we report that these mice are also behaviorally resistant to the antidepressant-like effects of adult SSRI administration. We investigated whether tianeptine (TIA), which exerts its therapeutic effects through agonism of the mu-opioid receptor instead of targeting monoaminergic systems, would be more effective in this model. We found that C57BL/6J pups exposed to FLX from postnatal day 2 to 11 (PNFLX, the mouse equivalent in terms of brain development to the human third trimester) showed increased avoidant behaviors as adults that failed to improve, or were even exacerbated, by chronic SSRI treatment. By contrast, avoidant behaviors in these same mice were drastically improved following chronic treatment with TIA. Overall, this demonstrates that TIA may be a promising alternative treatment for patients that fail to respond to typical antidepressants, especially in patients whose serotonergic system has been altered by in utero exposure to SSRIs.


Subject(s)
Antidepressive Agents, Second-Generation/toxicity , Antidepressive Agents, Tricyclic/pharmacology , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Brain/drug effects , Fluoxetine/toxicity , Selective Serotonin Reuptake Inhibitors/toxicity , Thiazepines/pharmacology , Animals , Animals, Newborn , Brain/growth & development , Feeding Behavior/drug effects , Mice, 129 Strain , Mice, Inbred C57BL , Open Field Test/drug effects
13.
Lipids Health Dis ; 20(1): 164, 2021 Nov 17.
Article in English | MEDLINE | ID: mdl-34789244

ABSTRACT

BACKGROUND: Quercetin (QUE) is a flavonol reported with anti-inflammatory and antioxidant activities, and previous results from the group of this study have demonstrated its neuroprotective effect against lipopolysaccharide-induced neuropsychiatric injuries. However, little is known about its potential effect on neuropsychiatric injuries induced or accompanied by metabolic dysfunction of glucose and lipids. METHODS: A nonalcoholic fatty liver disease (NAFLD) rat model was induced via a high-fat diet (HFD), and glucolipid parameters and liver function were measured. Behavioral performance was observed via the open field test (OFT) and the Morris water maze (MWM). The plasma levels of triggering receptor expressed on myeloid cells-1 (TREM1) and TREM2 were measured via enzyme-linked immunosorbent assay (ELISA). The protein expression levels of Synapsin-1 (Syn-1), Synaptatogmin-1 (Syt-1), TREM1 and TREM2 in the hippocampus were detected using western blotting. Morphological changes in the liver and hippocampus were detected by HE and Oil red or silver staining. RESULTS: Compared with the control rats, HFD-induced NAFLD model rats presented significant metabolic dysfunction, hepatocyte steatosis, and impaired learning and memory ability, as indicated by the increased plasma concentrations of total cholesterol (TC) and triglyceride (TG), the impaired glucose tolerance, the accumulated fat droplets and balloon-like changes in the liver, and the increased escaping latency but decreased duration in the target quadrant in the Morris water maze. All these changes were reversed in QUE-treated rats. Moreover, apart from improving the morphological injuries in the hippocampus, treatment with QUE could increase the decreased plasma concentration and hippocampal protein expression of TREM1 in NAFLD rats and increase the decreased expression of Syn-1 and Syt-1 in the hippocampus. CONCLUSIONS: These results suggested the therapeutic potential of QUE against NAFLD-associated impairment of learning and memory, and the mechanism might involve regulating the metabolic dysfunction of glucose and lipids and balancing the protein expression of synaptic plasticity markers and TREM1/2 in the hippocampus.


Subject(s)
Memory Disorders/drug therapy , Metabolic Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Non-alcoholic Fatty Liver Disease/drug therapy , Quercetin/therapeutic use , Animals , Blotting, Western , Disease Models, Animal , Glucose Tolerance Test , Liver/pathology , Male , Membrane Glycoproteins/blood , Memory Disorders/etiology , Metabolic Diseases/etiology , Morris Water Maze Test/drug effects , Non-alcoholic Fatty Liver Disease/complications , Open Field Test/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Immunologic/blood , Triggering Receptor Expressed on Myeloid Cells-1/blood
14.
Pharmacol Biochem Behav ; 211: 173300, 2021 12.
Article in English | MEDLINE | ID: mdl-34798097

ABSTRACT

The spontaneous object recognition (SOR) task is one of the most widely used behavioral protocols to assess visual memory in animals. However, only recently was it shown that nonhuman primates also perform well on this task. Here we further characterized this new monkey recognition memory test by assessing the performance of adult marmosets after an acute systemic administration of two putative amnesic agents: the competitive muscarinic acetylcholine receptor antagonist scopolamine (SCP; 0.05 mg/kg) and the noncompetitive N-methyl-d-aspartate glutamate receptor antagonist MK-801 (0.015 mg/kg). We also determined whether the acetylcholinesterase inhibitor donepezil (DNP; 0.50 mg/kg), a clinically-used cognitive enhancer, reverses memory deficits caused by either drug. The subjects had an initial 10 min sample trial where two identical neutral objects could be explored. After a 6 h retention interval, recognition was based on an exploratory preference for a new rather than familiar object during a 10 min test trial. Both SCP and MK-801 impaired the marmosets' performance on the SOR task, as both objects were explored equivalently. Co-administration of 0.50 mg/kg of DNP reversed the SCP- but not the MK-801-induced memory deficit. These results indicate that cholinergic and glutamatergic pathways mediate object recognition memory in the monkey SOR task.


Subject(s)
Dizocilpine Maleate/pharmacology , Open Field Test/drug effects , Recognition, Psychology/drug effects , Scopolamine/pharmacology , Acetylcholinesterase/metabolism , Animals , Callithrix/metabolism , Donepezil/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Haplorhini/metabolism , Male , Memory/drug effects , Memory Disorders/drug therapy , Memory Disorders/metabolism , Muscarinic Antagonists/pharmacology , Nootropic Agents/pharmacology , Receptors, Muscarinic/metabolism
15.
Biomed Pharmacother ; 144: 112303, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34673424

ABSTRACT

BACKGROUND: Depressive disorders induced by acute myocardial infarction (AMI) play a pivotal role in the deterioration of cardiac function, and Shuangxinfang (Psycho-cardiology Formula, PCF) was reported to alleviate heart function damage and improve depression-like behavior, but the complex mechanism in such process has not been clarified. METHODS: AMI models were established and PCF was administered in rats. Subjects were then assessed in open field test (OFT) and forced swimming test (FST) recapitulating symptoms of depressive disorder. Afterward, pharmacoproteomic profiling of the hippocampus and peri-infarct border zone (BZ) was performed using a label-free liquid chromatography-tandem mass spectrometry (LC-MS/MS) technique, to identify contributing proteins and pathways responsible for myocardial ischemia and behavioral allostasis. Bioinformatics analysis was processed for further investigation, while western blotting was employed for testing dominating proteins to validate proteomic results. RESULTS: Rats in the AMI group showed depression-like behavior in OFT and FST, which was improved by PCF. There were 131 differentially expressed proteins (DEPs) in BZ and 64 proteins in the hippocampus being detected and quantified shared by the sham group, the AMI group, and the PCF group. Subsequently, pertinent pathways and molecular functions were further identified. Altered molecules were discovered to be enriched in the apoptotic process, innate immune response, and NF-κB transcription factor activity in BZ, as well as chemical synaptic transmission, axon, collagen binding, cell adhesion, response to carbohydrate, laminin binding, and cellular response to nitric oxide in the hippocampus. Groups of signal transducers were also able to select multiple pathways, including innate immunity and arginine biosynthesis in the heart, also integrin signaling in the brain. DEPs were intersected from the myocardium and hippocampus to screen out the protein S100A9, which was up-regulated in the AMI group compared with the sham, and showed a down-regulation trend after treatment with PCF. CONCLUSION: Taken together, we present a comprehensive proteomics analysis of rat models with depression post-AMI. Reviewing the literatures concerned, it's hypothesized that macrophage/microglia inflammation mediated by S100A9 might be the pivotal pathogenic process of psycho-cardiology disease, as well as potential mechanisms for the treatment of PCF.


Subject(s)
Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Calgranulin B/metabolism , Cardiovascular Agents/pharmacology , Depression/drug therapy , Drugs, Chinese Herbal/pharmacology , Hippocampus/drug effects , Myocardial Infarction/drug therapy , Myocardium/metabolism , Proteomics , Animals , Chromatography, High Pressure Liquid , Chromatography, Reverse-Phase , Depression/metabolism , Depression/psychology , Disease Models, Animal , Drugs, Chinese Herbal/therapeutic use , Hippocampus/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Microglia/drug effects , Microglia/metabolism , Motor Activity/drug effects , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Open Field Test/drug effects , Protein Interaction Maps , Proteome , Rats, Sprague-Dawley , Tandem Mass Spectrometry , Ventricular Function, Left/drug effects
16.
Anesth Analg ; 133(5): 1140-1151, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34673725

ABSTRACT

BACKGROUND: Parkinson disease is a chronic and progressive movement disorder characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). The causes of Parkinson disease are not clear but may involve genetic susceptibilities and environmental factors. As in other neurodegenerative disorders, individuals predisposed to Parkinson disease may have an accelerated onset of symptoms following perioperative stress such as anesthesia, surgery, pain, and inflammation. We hypothesized that anesthesia alone accelerates the onset of Parkinson disease-like pathology and symptoms. METHODS: A presymptomatic Parkinson rat model (the protein, DJ-1, encoded by the Park7 gene [DJ-1], PARK7 knockout) was exposed to a surgical plane of isoflurane or 20% oxygen balanced with nitrogen for 2 hours on 3 occasions between 6 and 7 months of age. Acute and long-term motor and neuropathological effects were examined from 7 to 12 months of age in male DJ-1 rats, using the ladder rung, rotarod, and novel object recognition assays, as well as the immunohistochemical localization of tyrosine hydroxylase in dopaminergic neurons in the substantia nigra and ionized calcium-binding adaptor protein-1 (Iba-1) microglial activation in the substantia nigra and hippocampus. RESULTS: In the acute group, after the third anesthetic exposure at 7 months of age, the isoflurane group had a significant reduction in the density of dopaminergic neurons in the SNpc compared to controls. However, this reduction was not associated with increased microglial activation in the hippocampus or substantia nigra. With the ladder rung motor skills test, there was no effect of anesthetic exposure on the total number of foot faults or the ladder rung pattern in the acute group. The rotarod test also detected no differences before and after the third exposure in controls. For the long-term group, immunohistochemical analyses detected no differences in the density of dopaminergic neurons or microglial cells compared to unexposed DJ-1 rats from 8 to 12 months of age. The ladder rung test in the long-term group showed no differences in the total number of foot faults with time and exposure or between ladder rung patterns. The rotarod test detected no significant effect of exposure with time or between groups at any time point. The novel object recognition task in the long-term group revealed no differences in short- or long-term memory or in the number of rearings as a function of exposure. CONCLUSIONS: Multiple isoflurane exposures in this rat model of Parkinson disease transiently enhanced dopaminergic neurodegeneration in the SNpc that resolved over time and had no effects on progression in this Parkinson disease-like phenotype.


Subject(s)
Anesthetics, Inhalation/toxicity , Dopaminergic Neurons/drug effects , Isoflurane/toxicity , Nerve Degeneration , Parkinsonian Disorders/chemically induced , Pars Compacta/drug effects , Protein Deglycase DJ-1/genetics , Animals , Behavior, Animal/drug effects , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Gene Knockout Techniques , Male , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Motor Activity/drug effects , Open Field Test/drug effects , Parkinsonian Disorders/genetics , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Pars Compacta/metabolism , Pars Compacta/pathology , Protein Deglycase DJ-1/deficiency , Rats, Long-Evans , Rats, Transgenic , Rotarod Performance Test , Time Factors , Tyrosine 3-Monooxygenase/metabolism
17.
Biomed Pharmacother ; 144: 112280, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34628167

ABSTRACT

Metformin is currently used as a first-line drug to treat patients with type 2 diabetes. Previous studies have demonstrated that metformin has antioxidant properties and reduces neuroinflammation and hippocampal neuronal cell loss, which eventually improves memory. Methotrexate (MTX) is an antimetabolite chemotherapeutic agent reported to activate cognitive impairment found in many patients. Moreover, MTX negatively affects the spatial working memory, related to neurogenesis reduction in animal models. Therefore, the present study aimed to investigate the antioxidant effect of metformin on the reduction of memory and neurogenesis caused by MTX. Male Sprague-Dawley rats were divided into four groups: control, MTX, metformin, and MTX+metformin. MTX (75 mg/kg, i.v.) was administered on days 7 and 14. Rats were administered metformin (200 mg/kg, i.p.) for 14 days. Memory was determined using novel object location (NOL) and novel object recognition (NOR) tests. Furthermore, cell cycle arrest was quantified by p21 immunostaining. Levels of neuronal protein expression, scavenging enzymes activity, and malondialdehyde (MDA) level changes in the hippocampus and prefrontal cortex were investigated. Rats receiving only MTX showed memory impairment. Decreases in scavenging enzyme activity and BDNF, DCX, and Nrf2 protein expressions levels were detected in the MTX-treated rats. In addition, MTX significantly increased p21-positive cell numbers and MDA levels. However, these adverse MTX effects were counteracted by co-administration with metformin. These results demonstrate that metformin can improve memory impairments, increase BDNF, DCX and Nrf2 protein expressions and antioxidant capacities, and decrease MDA levels in MTX-treated rats.


Subject(s)
Behavior, Animal/drug effects , Hippocampus/drug effects , Memory Disorders/prevention & control , Memory/drug effects , Metformin/pharmacology , Neurogenesis/drug effects , Nootropic Agents/pharmacology , Oxidative Stress/drug effects , Prefrontal Cortex/drug effects , Animals , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Doublecortin Protein/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Male , Memory Disorders/chemically induced , Memory Disorders/metabolism , Memory Disorders/pathology , Methotrexate , NF-E2-Related Factor 2/metabolism , Open Field Test/drug effects , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Rats, Sprague-Dawley
18.
Biomed Pharmacother ; 144: 112266, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34634555

ABSTRACT

BACKGROUND: In the present study, we aimed to investigate the effects of probucol on aging-related hippocampus-dependent cognitive impairment and explore the potential mechanisms. METHODS: D-galactose (100 mg/kg, once daily for 6 weeks) was subcutaneously injected to induce aging in mice. Then the mice were administered with probucol or vehicle once a day for 2 weeks. The hippocampus-related cognition was evaluated with Morris water maze test, novel object recognition test, and contextual fear conditioning test. Moreover, synaptic plasticity was assessed, and RNA-sequencing was applied to further explore the molecular mechanisms. RESULTS: Aging mice induced by D-galactose showed conspicuous learning and memory impairment, which was significantly ameliorated by probucol. Meanwhile, probucol enhanced the spine density and dendritic branches, improved long-term potentiation, and increased the expression of PSD95 of aging mice. Probucol regulated 70 differentially expressed genes compared to D-galactose group, of which 38 genes were upregulated and 32 genes were downregulated. At last, RNA-sequencing results were verified by quantitative reverse transcription-polymerase chain reaction. CONCLUSIONS: Probucol improved learning and memory in aging mice through enhancing synaptic plasticity and regulating gene expression, indicating the potential application of probucol to prevent and treat aging-related disorders.


Subject(s)
Behavior, Animal/drug effects , Cognition/drug effects , Cognitive Dysfunction/drug therapy , Hippocampus/drug effects , Neurons/drug effects , Nootropic Agents/pharmacology , Probucol/pharmacology , Age Factors , Animals , Cellular Senescence/drug effects , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/psychology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Models, Animal , Disks Large Homolog 4 Protein/metabolism , Fear/drug effects , Gene Expression Regulation , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Long-Term Potentiation/drug effects , Male , Mice, Inbred C57BL , Morris Water Maze Test/drug effects , Neurons/metabolism , Neurons/pathology , Open Field Test/drug effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
19.
Neurotoxicology ; 87: 156-166, 2021 12.
Article in English | MEDLINE | ID: mdl-34597708

ABSTRACT

The growing concern surrounding bisphenol A (BPA) has led to increased industrial production and application of its analog bisphenol S (BPS). The goals of this study were: (1) To examine the generational effects in the nematode C. elegans for up to three generations following developmental exposure to BPS (0.1, 1.0, 5.0 and 10.0 µM), and (2) To examine the neurotoxicity and metabolic toxicity in NODEF mouse offspring exposed to BPS (3 µg/kg BW) in utero throughout gestation once/day via oral pipette. First, worms were exposed to BPS developmentally for a single period of 48 hours and then propagated for 2 additional generations. Exposure to 0.1 and 1.0 µM BPS decreased lifespan and the number of progeny with an ability to recover in subsequent generations. In contrast, worms exposed to 5.0 or 10.0 µM BPS exhibited a continuous effect in the second generation, e.g., decreased lifespan and reduced number of progeny. Only worms exposed to 10.0 µM BPS continued to have a significant long-term effect (e.g., decreased lifespan) through the third generation. In addition, worms developmentally exposed to BPS at 5.0 µM and 10.0 µM also showed decreases in body bends. In contrast, worms exposed to 0.1 µM BPS exhibited a significant increase in head thrashes. When the multigenerational effects were examined by exposing worms to BPS for 48 hours developmentally at each generation for three generations, an accumulative effect was observed in worms treated with 0.1 or 1.0 µM BPS for two generations, but not for three generations, suggesting a threshold existed. Worms exposed to either 5.0 or 10.0 µM BPS demonstrated accumulative effects through two and three generations. When the developmental effects of BPS were studied in NODEF mice, offspring exposed gestationally exhibited behavioral deficits at 12, but not at 3, weeks of age. Specifically, female offspring had decreases in working and short-term memories while male offspring showed increases in hyperactivity and anxiety-like behaviors. In summary, this study demonstrates the sex-related effects of BPS in NODEF mouse offspring exposed in utero, along with the generational effects observed in C. elegans.


Subject(s)
Caenorhabditis elegans/drug effects , Phenols/toxicity , Prenatal Exposure Delayed Effects/chemically induced , Sulfones/toxicity , Animals , Behavior, Animal/drug effects , Blood Glucose/analysis , Blood Glucose/drug effects , Body Weight/drug effects , Caenorhabditis elegans/growth & development , Female , Fertility/drug effects , Glucose Tolerance Test , Hindlimb Suspension , Longevity/drug effects , Male , Maze Learning/drug effects , Mice/growth & development , Open Field Test/drug effects , Phenols/administration & dosage , Pregnancy , Prenatal Exposure Delayed Effects/psychology , Sulfones/administration & dosage
20.
Biomed Pharmacother ; 144: 112369, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34715446

ABSTRACT

As an N-methyl-D-aspartate (NMDA) receptor inhibitor, ketamine has become a popular recreational substance and currently is used to address treatment-resistant depression. Since heavy ketamine use is associated with persisting psychosis, cognitive impairments, and neuronal damage, the safety of ketamine treatment for depression should be concerned. The nutrient supplement betaine has been shown to counteract the acute ketamine-induced psychotomimetic effects and cognitive dysfunction through modulating NMDA receptors. This study aimed to determine whether the adjunctive or subsequent betaine treatment would improve the enduring behavioral disturbances and hippocampal synaptic abnormality induced by repeated ketamine exposure. Mice received ketamine twice daily for 14 days, either combined with betaine co-treatment or subsequent betaine post-treatment for 7 days. Thereafter, three-chamber social approach test, reciprocal social interaction, novel location/object recognition test, forced swimming test, and head-twitch response induced by serotonergic hallucinogen were monitored. Data showed that the enduring behavioral abnormalities after repeated ketamine exposure, including disrupted social behaviors, recognition memory impairments, and increased depression-like and hallucinogen-induced head-twitch responses, were remarkably improved by betaine co-treatment or post-treatment. Consistently, betaine protected and reversed the reduced hippocampal synaptic activity, such as decreases in field excitatory post-synaptic potentiation (fEPSP), long-term potentiation (LTP), and PSD-95 levels, after repeated ketamine treatment. These results demonstrated that both co-treatment and post-treatment with betaine could effectively prevent and reverse the adverse behavioral manifestations and hippocampal synaptic plasticity after repeated ketamine use, suggesting that betaine can be used as a novel adjunct therapy with ketamine for treatment-resistant depression and provide benefits for ketamine use disorders.


Subject(s)
Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Betaine/pharmacology , Hippocampus/drug effects , Neuronal Plasticity/drug effects , Psychoses, Substance-Induced/prevention & control , Synaptic Transmission/drug effects , Animals , Cognition/drug effects , Disease Models, Animal , Disks Large Homolog 4 Protein/metabolism , Excitatory Amino Acid Antagonists , Excitatory Postsynaptic Potentials/drug effects , Female , Hippocampus/metabolism , Hippocampus/physiopathology , Ketamine , Locomotion/drug effects , Male , Mice, Inbred ICR , Open Field Test/drug effects , Psychoses, Substance-Induced/etiology , Psychoses, Substance-Induced/physiopathology , Psychoses, Substance-Induced/psychology , Recognition, Psychology/drug effects , Social Behavior , Swimming
SELECTION OF CITATIONS
SEARCH DETAIL