Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(1)2023 Dec 23.
Article in English | MEDLINE | ID: mdl-38203414

ABSTRACT

The HSP70 and HSP90 family members belong to molecular chaperones that exhibit protective functions during the cellular response to stressful agents. We investigated whether the exposure of rats to chronic mild stress (CMS), a validated model of depression, affects the expression of HSP70 and HSP90 in the prefrontal cortex (PFC), hippocampus (HIP) and thalamus (Thal). Male Wistar rats were exposed to CMS for 3 or 8 weeks. The antidepressant imipramine (IMI, 10 mg/kg, i.p., daily) was introduced in the last five weeks of the long-term CMS procedure. Depressive-like behavior was verified by the sucrose consumption test. The expression of mRNA and protein was quantified by real-time PCR and Western blot, respectively. In the 8-week CMS model, stress alone elevated HSP72 and HSP90B mRNA expression in the HIP. HSP72 mRNA was increased in the PFC and HIP of rats not responding to IMI treatment vs. IMI responders. The CMS exposure increased HSP72 protein expression in the cytosolic fraction of the PFC and HIP, and this effect was diminished by IMI treatment. Our results suggest that elevated levels of HSP72 may serve as an important indicator of neuronal stress reactions accompanying depression pathology and could be a potential target for antidepressant strategy.


Subject(s)
Imipramine , Molecular Chaperones , Male , Rats , Animals , Imipramine/pharmacology , Rats, Wistar , HSP70 Heat-Shock Proteins , Hippocampus , HSP90 Heat-Shock Proteins/genetics , Prefrontal Cortex , RNA, Messenger/genetics , Antidepressive Agents/pharmacology
2.
Psychopharmacology (Berl) ; 239(12): 3847-3857, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36278982

ABSTRACT

RATIONALE: We have discovered that rats at the age of 18 months begin to twitch their heads spontaneously (spontaneous head twitching, SHT). To date, no one has described this phenomenon. OBJECTIVES: The purpose of this study was to characterize SHT pharmacologically and to assess some possible mechanisms underlying SHT. METHODS: Wistar male rats were used in the study. Animals at the age of 18 months were qualified as HSHT (SHT ≥ 7/10 min observations) or LSHT (SHT < 7/10 min observations). Quantitative real-time PCR with TaqMan low-density array (TLDA) approach was adopted to assess the mRNA expression of selected genes in rat's hippocampus. RESULTS: HSHT rats did not differ from LSHT rats in terms of survival time, general health and behavior, water intake, and spontaneous locomotor activity. 2,5-dimethoxy-4-iodoamphetamine (DOI) at a dose of 2.5 mg/kg increased the SHT in HSHT and LSHT rats, while ketanserin dose-dependently abolished the SHT in the HSHT rats. The SHT was reduced or abolished by olanzapine, clozapine, risperidone, and pimavanserin. All these drugs have strong 5-HT2A receptor-inhibiting properties. Haloperidol and amisulpride, as antipsychotic drugs with a mostly dopaminergic mechanism of action, did not influence SHT. Similarly, escitalopram did not affect SHT. An in-depth gene expression analysis did not reveal significant differences between the HSHT and the LSHT rats. CONCLUSIONS: SHT appears in some aging rats (about 50%) and is permanent over time and specific to individuals. The 5-HT2A receptor strongly controls SHT. HSHT animals can be a useful animal model for studying 5-HT2A receptor ligands.


Subject(s)
Antipsychotic Agents , Clozapine , Rats , Animals , Male , Rats, Wistar , Receptor, Serotonin, 5-HT2A , Ketanserin/pharmacology , Antipsychotic Agents/pharmacology
3.
Psychopharmacology (Berl) ; 238(11): 3167-3181, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34333674

ABSTRACT

RATIONALE: Pterostilbene is the 3,5-dimethoxy derivative of resveratrol with numerous beneficial effects including neuroprotective properties. Experimental studies revealed its anticonvulsant action in the acute seizure tests. OBJECTIVES: The purpose of the present study was to evaluate the effect of pterostilbene in the pentetrazol (PTZ)-induced kindling model of epilepsy in mice as well as to assess some possible mechanisms of its anticonvulsant action in this model. METHODS: Mice were repeatedly treated with pterostilbene (50-200 mg/kg) and its effect on the development of seizure activity in the PTZ kindling was estimated. Influence of pterostilbene on the locomotor activity and anxiety- and depression-like behavior in the PTZ-kindled mice was also assessed. To understand the possible mechanisms of anticonvulsant activity of pterostilbene, γ-aminobutyric acid (GABA) and glutamate concentrations in the prefrontal cortex and hippocampus of the PTZ-kindled mice were measured using LC-MS/MS method. Moreover, mRNA expression of BDNF, TNF-α, IL-1ß, IL-6, GABRA1A, and GRIN2B was determined by RT-qPCR technique. RESULTS: We found that pterostilbene at a dose of 200 mg/kg considerably reduced seizure activity but did not influence the locomotor activity and depression- and anxiety-like behavior in the PTZ-kindled mice. In the prefrontal cortex and hippocampus, pterostilbene reversed the kindling-induced decrease of GABA concentration. Neither in the prefrontal cortex nor hippocampus pterostilbene affected mRNA expression of IL-1ß, IL-6, GABRA1A, and GRIN2B augmented by PTZ kindling. Pterostilbene at a dose of 100 mg/kg significantly decreased BDNF and TNF-α mRNA expression in the hippocampus of the PTZ-kindled mice. CONCLUSIONS: Although further studies are necessary to understand the mechanism of anticonvulsant properties of pterostilbene, our findings suggest that it might be considered a candidate for a new antiseizure drug.


Subject(s)
Anticonvulsants , Kindling, Neurologic , Animals , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Anxiety/drug therapy , Chromatography, Liquid , Depression/drug therapy , Mice , Pentylenetetrazole/pharmacology , Stilbenes , Tandem Mass Spectrometry
4.
Pharmacol Rep ; 73(4): 1179-1187, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34117630

ABSTRACT

BACKGROUND: Evidence indicates that Gα12, Gα13, and its downstream effectors, RhoA and Rac1, regulate neuronal morphology affected by stress. This study was aimed at investigating whether repeated stress influences the expression of proteins related to the Gα12/13 intracellular signaling pathway in selected brain regions sensitive to the effects of stress. Furthermore, the therapeutic impact of ß(1)adrenergic receptors (ß1AR) blockade was assessed. METHODS: Restraint stress (RS) model in mice (2 h/14 days) was used to assess prolonged stress effects on the mRNA expression of Gα12, Gα13, RhoA, Rac1 in the prefrontal cortex (PFC), hippocampus (HIP) and amygdala (AMY). In a separate study, applying RS model in rats (3-4 h/1 day or 14 days), we evaluated stress effects on the expression of Gα12, Gα11, Gαq, RhoA, RhoB, RhoC, Rac1/2/3 in the HIP. Betaxolol (BET), a selective ß1AR antagonist, was introduced (5 mg/kg/p.o./8-14 days) in the rat RS model to assess the role of ß1AR in stress effects. RT-qPCR and Western Blot were used for mRNA and protein assessments, respectively. RESULTS: Chronic RS decreased mRNA expression of Gα12 and increased mRNA for Rac1 in the PFC of mice. In the mice AMY, decreased mRNA expression of Gα12, Gα13 and RhoA was observed. Fourteen days of RS exposure increased RhoA protein level in the rats' HIP in the manner dependent on ß1AR activity. CONCLUSIONS: Together, these results suggest that repeated RS affects the expression of genes and proteins known to be engaged in neural plasticity, providing potential targets for further studies aimed at unraveling the molecular mechanisms of stress-related neuropsychiatric diseases.


Subject(s)
Brain/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Neurons/metabolism , Restraint, Physical/physiology , Signal Transduction/physiology , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Adrenergic beta-1 Receptor Antagonists/pharmacology , Animals , Brain/drug effects , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Adrenergic, beta-1/metabolism , Signal Transduction/drug effects
5.
Biomolecules ; 11(2)2021 02 16.
Article in English | MEDLINE | ID: mdl-33669305

ABSTRACT

This study demonstrates how exposure to psychosocial crowding stress (CS) for 3, 7, and 14 days affects glutamate synapse functioning and signal transduction in the frontal cortex (FC) of rats. CS effects on synaptic activity were evaluated in FC slices of the primary motor cortex (M1) by measuring field potential (FP) amplitude, paired-pulse ratio (PPR), and long-term potentiation (LTP). Protein expression of GluA1, GluN2B mGluR1a/5, VGLUT1, and VGLUT2 was assessed in FC by western blot. The body's response to CS was evaluated by measuring body weight and the plasma level of plasma corticosterone (CORT), adrenocorticotropic hormone (ACTH), and interleukin 1 beta (IL1B). CS 3 14d increased FP and attenuated LTP in M1, while PPR was augmented in CS 14d. The expression of GluA1, GluN2B, and mGluR1a/5 was up-regulated in CS 3d and downregulated in CS 14d. VGLUTs expression tended to increase in CS 7d. The failure to blunt the effects of chronic CS on FP and LTP in M1 suggests the impairment of habituation mechanisms by psychosocial stressors. PPR augmented by chronic CS with increased VGLUTs level in the CS 7d indicates that prolonged CS exposure changed presynaptic signaling within the FC. The CS bidirectional profile of changes in glutamate receptors' expression seems to be a common mechanism evoked by stress in the FC.


Subject(s)
Frontal Lobe/metabolism , Receptors, Glutamate/biosynthesis , Adrenocorticotropic Hormone/biosynthesis , Animals , Body Weight , Corticosterone/biosynthesis , Crowding , Electrophysiology , Glutamic Acid , Interleukin-1beta/biosynthesis , Long-Term Potentiation , Male , Models, Animal , Motor Cortex , Organ Size , Rats , Rats, Wistar , Receptors, AMPA/biosynthesis , Receptors, Metabotropic Glutamate/biosynthesis , Receptors, N-Methyl-D-Aspartate/biosynthesis , Spleen/pathology , Stress, Psychological , Synaptic Transmission/drug effects , Vesicular Glutamate Transport Protein 1/biosynthesis , Vesicular Glutamate Transport Protein 2/biosynthesis
6.
Article in English | MEDLINE | ID: mdl-30953677

ABSTRACT

Disturbances in fear-evoked signal transduction in the hippocampus (HP), the nuclei of the amygdala (AMY), and the prefrontal cortex (PFC) underlie anxiety-related disorders. However, the molecular mechanisms underlying these effects remain elusive. Heterotrimeric G proteins (GPs) are divided into the following four families based on the intracellular activity of their alpha subunit (Gα): Gα(s) proteins stimulate cyclic AMP (cAMP) generation, Gα(i/o) proteins inhibit the cAMP pathway, Gα(q/11) proteins increase the intracellular Ca++ concentration and the inositol trisphosphate level, and Gα(12/13) proteins activate monomeric GP-Rho. In the present study, we assessed the effects of a fear memory procedure on the mRNA expression of the Gα subunits of all four GP families in the HP, AMY and PFC. C57BL/6 J mice were subjected to a fear conditioning (FC) procedure followed by a contextual or cued fear memory test (CTX-R and CS-R, respectively). Morphine (MOR, 1 mg/kg/ip) was injected immediately after FC to prevent the fear consolidation process. Real-time quantitative PCR was used to measure the mRNA expression levels of Gα subunits at 1 h after FC, 24 h after FC, and 1 h after the CTX-R or CS-R. In the HP, the mRNA levels of Gα(s), Gα(12) and Gα(11) were higher at 1 h after training. Gα(s) levels were slightly lower when consolidation was stabilized and after the CS-R. The mRNA levels of Gα(12) were increased at 1 h after FC, returned to control levels at 24 h after FC and increased again with the CTX-R. The increase in the Gα(11) level persisted at 24 h after FC and after CTX-R. In the AMY, no specific changes were induced by FC. In the PFC, CTX-R was accompanied by a decrease in Gα(i/o) mRNA levels; however, only Gα(i2) downregulation was prevented by MOR treatment. Hence, the FC-evoked changes in Gα mRNA expression were observed mainly in the HP and connected primarily to contextual learning. These results suggest that the activation of signaling pathways by Gα(s) and Gα(12) is required to begin the fear memory consolidation process in the HP, while signal transduction via Gα(11) is implicated in the maintenance of fear consolidation. In the PFC, the downregulation of Gα(i2) appears to be related to the contextual learning of fear.


Subject(s)
Brain/metabolism , Fear , GTP-Binding Proteins/metabolism , Memory/drug effects , Morphine/pharmacology , Animals , Brain/drug effects , Conditioning, Classical/drug effects , Cues , Fear/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism
7.
Neurotox Res ; 34(3): 706-716, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30129004

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder of the central nervous system (CNS) caused by a progressive loss of nigrostriatal dopaminergic neurons. Dysfunction of the ubiquitin-proteasome system (UPS) plays an important role in the pathogenesis of PD. Intranigral administration of the UPS inhibitor lactacystin is used to obtain a valuable animal model for investigating putative neuroprotective treatments for PD. 1-Methyl-1,2,3,4-tetrahydroisoquinoline (1MeTIQ) is an endogenous amine that displays neuroprotective properties. This compound acts as a reversible monoamine oxidase (MAO) inhibitor and a natural free radical scavenger. In the present experiment, we investigated the effect of acute and chronic treatment with 1MeTIQ on locomotor activity and the release of dopamine as well as its metabolites in the striatum of unilaterally lactacystin-lesioned and sham-operated rats using in vivo microdialysis. Additionally, changes in the level of tyrosine hydroxylase (TH) in the substantia nigra were measured. Unilateral lactacystin injection into the substantia nigra caused significant impairment of dopamine release (approx. 45%) and a marked decline in the TH level. These effects were completely antagonized by multiple treatments with 1MeTIQ. The results obtained from the in vivo microdialysis study as well as from the ex vivo experiments suggest that multiple administration of 1MeTIQ protects dopaminergic neurons against the lactacystin-induced decline in TH concentration in the substantia nigra and prevents disturbances of dopamine release in the striatum. We have demonstrated that 1MeTIQ is capable of maintaining the physiological functions of the striatal dopamine neurons damaged by unilateral lactacystin lesion.


Subject(s)
Brain Injuries/metabolism , Brain Injuries/prevention & control , Brain/drug effects , Dopamine/metabolism , Neuroprotective Agents/therapeutic use , Tetrahydroisoquinolines/therapeutic use , 3,4-Dihydroxyphenylacetic Acid/metabolism , Acetylcysteine/analogs & derivatives , Acetylcysteine/toxicity , Analysis of Variance , Animals , Brain/metabolism , Brain Injuries/chemically induced , Brain Injuries/pathology , Cysteine Proteinase Inhibitors/toxicity , Exploratory Behavior/drug effects , Functional Laterality/drug effects , Locomotion/drug effects , Male , Microdialysis , Rats , Rats, Wistar
8.
Neurotox Res ; 33(3): 523-531, 2018 04.
Article in English | MEDLINE | ID: mdl-29076060

ABSTRACT

Parkinson's disease (PD) represents one of the neurodegenerative disorders which are caused by degeneration of dopaminergic neurons in the nigrostriatal pathway. Different toxins, e.g., 6-hydroxydopamine (6-OHDA), are used to model PD in animals. 6-OHDA is a neurotoxin which damages catecholaminergic neurons via production of oxygen radicals. Tetrahydroisoquinolines (TIQs) are endogenous amines which are present in the mammalian brain. Some of them, like TIQ and 1-methyl-1,2,3,4-tetrahydroisoquinoline (1MeTIQ), demonstrate neuroprotective properties. These compounds act as reversible MAO inhibitors and this way block free radical formation. To continue our previous experiments, we evaluated the effect of acute and chronic treatment with TIQ and 1MeTIQ on locomotor/exploratory activity and the release of dopamine as well as its metabolite 3-methoxytyramine (3-MT) in the striatum of unilaterally 6-OHDA-lesioned and sham-operated rats using in vivo microdialysis methodology. Additionally, the changes in the concentration of tyrosine hydroxylase in the substantia nigra were measured. A unilateral 6-OHDA lesion in the substantia nigra produces a strong reduction in the release of dopamine (approx. 70%) and 3-MT (approx. 50%) in the rat striatum. This effect was completely inhibited by multiple administration of TIQ and 1MeTIQ. The results obtained from the in vivo microdialysis study suggest that multiple treatment with both endogenous amines, TIQ and 1MeTIQ, protects dopaminergic neurons against a 6-OHDA-induced deficit of dopamine release. Furthermore, these amines were able to maintain physiological functions of striatal dopamine neurons damaged by a unilateral 6-OHDA lesion.


Subject(s)
Corpus Striatum/drug effects , Dopamine/metabolism , Neuroprotective Agents/pharmacology , Tetrahydroisoquinolines/pharmacology , Analysis of Variance , Animals , Corpus Striatum/metabolism , Exploratory Behavior/drug effects , Functional Laterality/drug effects , Male , Microdialysis/methods , Motor Activity/drug effects , Oxidopamine/toxicity , Rats , Rats, Wistar , Sympatholytics/toxicity , Time Factors , Tyrosine 3-Monooxygenase/metabolism
9.
Int Immunopharmacol ; 48: 35-42, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28460354

ABSTRACT

Some antidepressants show a significantly lower efficacy in elderly patients, particularly in women. Previous studies have shown that antidepressants administered to young animals reduced depression-like symptoms induced by lipopolysaccharide (LPS). The aim of this study was to find out whether the antidepressant and anti-inflammatory properties of fluoxetine (FLU) can be observed also in old female C57BL/6J mice. A depression-like state was evoked by the administration of LPS (100µg/kg for 4 consecutive days) which was followed by reduction of sucrose preference (anhedonia) and enhancement of immobility-time in the forced swim test (FST). Animals, which received FLU (10mg/kg, 11days) exhibited a decreased LPS-induced expression of some inflammatory cytokines in the hippocampus and spleen but this effect was not accompanied by beneficial changes in animals' behavior. Despite the lack of antidepressant-properties of FLU in this model, our studies have proven significant profound anti-inflammatory properties of chronic FLU treatment which may suggest its suitability for fending off inflammatory processes in the elderly.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antidepressive Agents/pharmacology , Cytokines/immunology , Depression/immunology , Fluoxetine/pharmacology , Aging/immunology , Animals , Antidepressive Agents/therapeutic use , Behavior, Animal/drug effects , Cytokines/genetics , Depression/drug therapy , Female , Fluoxetine/therapeutic use , Hippocampus/drug effects , Hippocampus/immunology , Lipopolysaccharides , Mice, Inbred C57BL , RNA, Messenger/metabolism , Spleen/cytology , Spleen/drug effects , Spleen/immunology
10.
J Steroid Biochem Mol Biol ; 171: 94-109, 2017 07.
Article in English | MEDLINE | ID: mdl-28263910

ABSTRACT

Several lines of evidence suggest that exposures to Endocrine Disrupting Chemicals (EDCs) such as pesticides increase the risks of neuropsychiatric disorders. Despite extended residual persistence of dichlorodiphenyltrichloroethane (DDT) in the environment, the mechanisms of perinatal actions of DDT that could account for adult-onset of depression are largely unknown. This study demonstrated the isomer-specific induction of depressive-like behavior and impairment of Htr1a/serotonin signaling in one-month-old mice that were prenatally exposed to DDT. The effects were reversed by the antidepressant citalopram as evidenced in the forced swimming (FST) and tail suspension (TST) tests in the male and female mice. Prenatally administered DDT accumulated in mouse brain as determined with gas chromatography and tandem mass spectrometry, led to global DNA hypomethylation, and altered the levels of methylated DNA in specific genes. The induction of depressive-like behavior and impairment of Htr1a/serotonin signaling were accompanied by p,p'-DDT-specific decrease in the levels of estrogen receptors i.e. ESR1 and/or GPER1 depending on sex. In contrast, o,p'-DDT did not induce depressive-like effects and exhibited quite distinct pattern of biochemical alterations that was related to aryl hydrocarbon receptor (AHR), its nuclear translocator ARNT, and ESR2. Exposure to o,p'-DDT increased AHR expression in male and female brains, and reduced expression levels of ARNT and ESR2 in the female brains. The evolution of p,p'-DDT-induced depressive-like behavior was preceded by attenuation of Htr1a and Gper1/GPER1 expression as observed in the 7-day-old mouse pups. Because p,p'-DDT caused sex- and age-independent attenuation of GPER1, we suggest that impairment of GPER1 signaling plays a key role in the propagation of DDT-induced depressive-like symptoms.


Subject(s)
Brain/drug effects , DDT/toxicity , DNA Methylation/drug effects , Endocrine Disruptors/toxicity , Estrogen Receptor alpha/antagonists & inhibitors , Prenatal Exposure Delayed Effects , Receptors, Estrogen/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Antidepressive Agents, Second-Generation/therapeutic use , Behavior, Animal/drug effects , Brain/metabolism , Citalopram/therapeutic use , DDT/chemistry , Depression/chemically induced , Depression/drug therapy , Depression/metabolism , Endocrine Disruptors/chemistry , Estrogen Receptor alpha/metabolism , Female , Isomerism , Male , Mice , Neurons/drug effects , Neurons/metabolism , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/metabolism , Pregnancy , Random Allocation , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Sex Characteristics , Signal Transduction/drug effects
12.
Behav Pharmacol ; 27(4): 397-401, 2016 06.
Article in English | MEDLINE | ID: mdl-26588212

ABSTRACT

The α1-adrenergic receptors (α1-ARs), which belong to a G protein-coupled receptor family, consist of three highly homologous subtypes known as α1A-ARs, α1B-ARs, and α1D-ARs. Our previous findings suggested that α1A-ARs are an important target for imipramine and electroconvulsive therapy. The current study sought to evaluate whether S-(+)-niguldipine and B8805-033, two selective antagonists of α1A-ARs, can evoke antidepressant-like effects in the forced swim test in rats. Both compounds were administered at three time points (24, 5, and 1 h before testing), and the effects of three doses (2, 5, and 10 mg/kg) of each compound were investigated. S-(+)-Niguldipine produced no antidepressant-like effects other than a 14% reduction in immobility time at the highest dose. Although B8805-033 at a dose of 2 mg/kg did not influence the rats' behavior, higher B8805-033 doses (5 and 10 mg/kg) produced significant reductions in immobility time (approximately 42 and 44% vs. controls, respectively; P<0.01). However, this effect was abolished by the concomitant administration of WAY100135, a serotonin receptor antagonist, suggesting that the observed antidepressant-like effects of B8805-033 are unrelated to α1A-ARs. Nevertheless, given the current dearth of selective α1A-AR agonists, the question of whether this particular subtype could be involved in antidepressant therapy mechanisms remains unresolved.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/pharmacology , Antidepressive Agents/pharmacology , Dihydropyridines/pharmacology , Dioxins/pharmacology , Pyrimidinones/pharmacology , Adrenergic alpha-1 Receptor Antagonists/administration & dosage , Animals , Antidepressive Agents/administration & dosage , Dihydropyridines/administration & dosage , Dioxins/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Male , Piperazines/pharmacology , Pyrimidinones/administration & dosage , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-1/drug effects , Receptors, Adrenergic, alpha-1/metabolism , Swimming , Time Factors
13.
Neurotox Res ; 29(3): 351-63, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26303030

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder that is hallmarked by pathological changes associated with the death of dopaminergic neurons, particularly in the extrapyramidal system (substantia nigra pars compacta, striatum) of the brain. Although the causes of slow neuronal death in PD are unknown, both genetic and environmental factors are likely involved. Endogenous isoquinolines, such as 1-benzyl-1,2,3,4-tetrahydroisoquinoline (1BnTIQ), present in the human brain have been previously reported to participate in the pathogenesis of PD. The chronic administration of 1BnTIQ induced parkinsonism in primates, and this effect might be associated with idiopathic PD. However, another endogenous derivative of tetrahydroisoquinoline, 1-methyl-1,2,3,4-tetrahydroisoquinoline (1MeTIQ), displays clear neuroprotective properties in the brain. In the present study, we investigated the neuroprotective effects of 1MeTIQ (25 and 50 mg/kg) in an animal model of PD after the chronic administration of 1BnTIQ (25 mg/kg). Behavioral analyses demonstrate that both acute and repeated treatment with 1MeTIQ completely antagonized 1BnTIQ-induced changes in rat locomotor activity. Neurochemical experiments indicate that 1MeTIQ co-administered with 1BnTIQ completely antagonized 1BnTIQ-induced reduction in the dopamine (DA) concentration in rat brain structures. In conclusion, the results demonstrate that 1MeTIQ possesses important neuroprotective properties in the animal model of PD and that the rats did not develop tolerance after its chronic administration.


Subject(s)
Brain/drug effects , Brain/metabolism , Dopamine/metabolism , Neuroprotective Agents/administration & dosage , Parkinsonian Disorders/metabolism , Tetrahydroisoquinolines/administration & dosage , Tetrahydroisoquinolines/toxicity , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Hyperkinesis/chemically induced , Male , Parkinson Disease , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/physiopathology , Parkinsonian Disorders/prevention & control , Rats , Rats, Wistar
14.
Pharmacol Biochem Behav ; 137: 69-77, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26261018

ABSTRACT

Recently, we have demonstrated that conditional inactivation of glucocorticoid receptors (GRs) in the noradrenergic system, may evoke depressive-like behavior in female but not male mutant mice (GR(DBHCre) mice). The aim of the current study was to dissect how selective ablation of glucocorticoid signaling in the noradrenergic system influences the previously reported depressive-like phenotype and whether it might be linked to neurotrophic alterations or secondary changes in the serotonergic system. We demonstrated that selective depletion of GRs enhances brain derived neurotrophic factor (BDNF) expression in female but not male GR(DBHCre) mice on both the mRNA and protein levels. The possible impact of the mutation on brain noradrenergic and serotonergic systems was addressed by investigating the tissue neurotransmitter levels under basal conditions and after acute restraint stress. The findings indicated a stress-provoked differential response in tissue noradrenaline content in the GR(DBHCre) female but not male mutant mice. An analogous gender-specific effect was identified in the diminished content of 5-hydroxyindoleacetic acid, the main metabolite of serotonin, in the prefrontal cortex, which suggests down-regulation of this monoamine system in female GR(DBHCre) mice. The lack of GR also resulted in an up-regulation of alpha2-adrenergic receptor (α2-AR) density in the female but not male mutants in the locus coeruleus. We have also confirmed the utility of the investigated model in pharmacological studies, which demonstrates that the depressive-like phenotype of GR(DBHCre) female mice can be reversed by antidepressant treatment with desipramine or fluoxetine, with the latter drug evoking more pronounced effects. Overall, our study validates the use of female GR(DBHCre) mice as an interesting and novel genetic tool for the investigation of the cross-connected mechanisms of depression that is not only based on behavioral phenotypes.


Subject(s)
Adrenergic Neurons/metabolism , Brain-Derived Neurotrophic Factor/biosynthesis , Depression/metabolism , Phenotype , Receptors, Glucocorticoid/metabolism , Serotonergic Neurons/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Depression/genetics , Depression/psychology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Glucocorticoid/genetics , Up-Regulation/physiology
15.
Int J Neuropsychopharmacol ; 18(1)2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25522393

ABSTRACT

BACKGROUND: Dysregulation in the prefrontal cortex-nucleus accumbens pathway has been implicated in cocaine addiction. We have previously demonstrated that one intra-dorsomedial prefrontal cortex brain-derived neurotrophic factor (BDNF) infusion immediately following the last cocaine self-administration session caused a long-lasting inhibition of cocaine-seeking and normalized the cocaine-induced disturbance of glutamate transmission in the nucleus accumbens after extinction and a cocaine prime. However, the molecular mechanism mediating the brain-derived neurotrophic factor effect on cocaine-induced alterations in extracellular glutamate levels is unknown. METHODS: In the present study, we determined the effects of brain-derived neurotrophic factor on cocaine-induced changes in the phosphorylation of synapsin (p-synapsin), a family of presynaptic proteins that mediate synaptic vesicle mobilization, in the nucleus accumbens during early withdrawal. RESULTS: Two hours after cocaine self-administration, p-synapsin Ser9 and p-synapsin Ser62/67, but not p-synapsin Ser603, were increased in the nucleus accumbens. At 22 hours, only p-synapsin Ser9 was still elevated. Elevations at both time points were attenuated by an intra-dorsomedial prefrontal cortex brain-derived neurotrophic factor infusion immediately after the end of cocaine self-administration. Brain-derived neurotrophic factor also reduced cocaine self-administration withdrawal-induced phosphorylation of the protein phosphatase 2A C-subunit, suggesting that brain-derived neurotrophic factor disinhibits protein phosphatase 2A C-subunit, consistent with p-synapsin Ser9 dephosphorylation. Further, co-immunoprecipitation demonstrated that protein phosphatase 2A C-subunit and synapsin are associated in a protein-protein complex that was reduced after 2 hours of withdrawal from cocaine self-administration and reversed by brain-derived neurotrophic factor. CONCLUSIONS: Taken together, these findings demonstrate that brain-derived neurotrophic factor normalizes the cocaine self-administration-induced elevation of p-synapsin in nucleus accumbens that may underlie a disturbance in the probability of neurotransmitter release or represent a compensatory neuroadaptation in response to the hypofunction within the prefrontal cortex-nucleus accumbens pathway during cocaine withdrawal.


Subject(s)
Brain-Derived Neurotrophic Factor/administration & dosage , Central Nervous System Agents/administration & dosage , Cocaine-Related Disorders/drug therapy , Nucleus Accumbens/metabolism , Substance Withdrawal Syndrome/drug therapy , Synapsins/metabolism , Animals , Cocaine/administration & dosage , Cocaine-Related Disorders/metabolism , Disease Models, Animal , Dopamine Uptake Inhibitors/administration & dosage , Male , Nucleus Accumbens/drug effects , Phosphorylation/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats, Sprague-Dawley , Self Administration , Substance Withdrawal Syndrome/metabolism
16.
Eur Neuropsychopharmacol ; 24(9): 1546-56, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25106693

ABSTRACT

It has been shown that stressful events occurring in early life have a powerful influence on the development of the central nervous system. Insulin-like growth factor-1 (IGF-1) promotes the growth, differentiation and survival of both neurons and glial cells and is thought to exert antidepressant-like activity. Thus, it is possible that disturbances in the function of the IGF-1 system may be responsible for disturbances observed over the course of depression. Prenatal stress was used as a valid model of depression. Adult male offspring of control and stressed rat dams were subjected to behavioural testing (forced swim test). The level of IGF-1 in the blood and the expression of IGF-1, IGF-1R, and IRS-1/2 in the hippocampus and frontal cortex using RT-PCR, ELISA and western blotting were measured. In addition the effect of intracerebroventricularly administered IGF-1 and/or the IGF-1R receptor antagonist JB1 in the forced swim test was studied. Prenatally stressed rats showed depressive like behaviour, including increased immobility time as well as decreased mobility and climbing. Intracerebroventricular administration of IGF-1 reversed these effects in stressed animals, whereas concomitant administration of the IGF-1R antagonist JB1 completely blocked the effects. Biochemical analysis of homogenates from the hippocampus and frontal cortex revealed decreases in IGF-1 level and IGF-1R phosphorylation along with disturbances in IRS-1 phosphorylation. These findings reveal that prenatal stress alters IGF-1 signalling, which may contribute to the behavioural changes observed in depression.


Subject(s)
Brain/metabolism , Insulin-Like Growth Factor I/metabolism , Prenatal Exposure Delayed Effects/physiopathology , Receptor, IGF Type 1/metabolism , Stress, Psychological/etiology , Stress, Psychological/pathology , Analysis of Variance , Animals , Brain/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Injections, Intraventricular , Insulin-Like Growth Factor I/antagonists & inhibitors , Insulin-Like Growth Factor I/pharmacology , Male , Peptides/pharmacology , Phosphorylation/drug effects , Pregnancy , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Swimming/psychology , Time Factors
17.
Addict Biol ; 19(1): 77-86, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23461423

ABSTRACT

Abstinence from cocaine self-administration (SA) is associated with neuroadaptations in the prefrontal cortex (PFC) and nucleus accumbens (NAc) that are implicated in cocaine-induced neuronal plasticity and relapse to drug-seeking. Alterations in cAMP-dependent protein kinase A (PKA) signaling are prominent in medium spiny neurons in the NAc after repeated cocaine exposure but it is unknown whether similar changes occur in the PFC. Because cocaine SA induces disturbances in glutamatergic transmission in the PFC-NAc pathway, we examined whether dysregulation of PKA-mediated molecular targets in PFC-NAc neurons occurs during abstinence and, if so, whether it contributes to cocaine-seeking. We measured the phosphorylation of cAMP response element binding protein (Ser133) and GluA1 (Ser845) in the dorsomedial (dm) PFC and the presynaptic marker, synapsin I (Ser9, Ser62/67, Ser603), in the NAc after 7 days of abstinence from cocaine SA with or without cue-induced cocaine-seeking. We also evaluated whether infusion of the PKA inhibitor, 8-bromo-Rp-cyclic adenosine 3', 5'-monophosphorothioate (Rp-cAMPs), into the dmPFC after abstinence would affect cue-induced cocaine-seeking and PKA-regulated phosphoprotein levels. Seven days of forced abstinence increased the phosphorylation of cAMP response element binding protein and GluA1 in the dmPFC and synapsin I (Ser9) in the NAc. Induction of these phosphoproteins was reversed by a cue-induced relapse test of cocaine-seeking. Bilateral intra-dmPFC Rp-cAMPs rescued abstinence-elevated PKA-mediated phosphoprotein levels in the dmPFC and NAc and suppressed cue-induced relapse. Thus, by inhibiting abstinence-induced PKA molecular targets, relapse reverses abstinence-induced neuroadaptations in the dmPFC that are responsible, in part, for the expression of cue-induced cocaine-seeking.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Drug-Seeking Behavior/physiology , Nucleus Accumbens/metabolism , Prefrontal Cortex/metabolism , Analysis of Variance , Animals , Blotting, Western , CREB-Binding Protein/metabolism , Cocaine/administration & dosage , Cocaine/pharmacology , Cues , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Dopamine Uptake Inhibitors/administration & dosage , Dopamine Uptake Inhibitors/pharmacology , Drug-Seeking Behavior/drug effects , Male , Neuronal Plasticity/drug effects , Phosphoproteins/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Receptors, AMPA/metabolism , Recurrence , Self Administration , Synapsins/metabolism , Thionucleotides/pharmacology
18.
Article in English | MEDLINE | ID: mdl-24128685

ABSTRACT

Exposure to adversity during early life is a risk factor for the development of different mood and psychiatric disorders, including depressive-like behaviors. Here, neonatal mice were temporarily but repeatedly (day 1 to day 13) separated from mothers and placed in a testing environment containing a layer of odorless clean bedding (CB). We assessed in adult animals the impact of this early experience on binding sites and mRNA expression of α1-adrenergic receptor subtypes, heat shock proteins (HSPs) and proapoptotic and antiapoptotic members of the Bcl-2 family proteins in different brain regions involved in processing of olfactory information and rewarding stimuli. We found that repeated exposure to CB experience produced anhedonic-like behavior in terms of reduced saccharin intake and α1-adrenoceptor downregulation in piriform and somatosensory cortices, hippocampus, amygdala and discrete thalamic nuclei. We also found a selective decrease of α1B-adrenoceptor binding sites in the cingulate cortex and hippocampus and an increase of hippocampal α1A and α1B receptor, but not of α1D-adrenoceptor, mRNA levels. Moreover, while a significant decrease of antiapoptotic heat shock proteins Hsp72 and Hsp90 was identified in the prefrontal cortex, a parallel increase of antiapoptotic members of Bcl-2 family proteins was found at the hippocampal level. Together, these data provide evidence that the early exposure to CB experience produced enduring downregulation of α1-adrenoceptors in the prefrontal-limbic forebrain/limbic midbrain network, which plays a key role in the processing of olfactory information and reaction to rewarding stimuli. Finally, these data show that CB experience can "prime" the hippocampal circuitry and promote the expression of antiapoptotic factors that can confer potential neuroprotection to subsequent adversity.


Subject(s)
Aging/pathology , Apoptosis/physiology , Brain/metabolism , Maternal Deprivation , Receptors, Adrenergic, alpha-1/metabolism , Signal Transduction/physiology , Adrenergic alpha-1 Receptor Antagonists/pharmacokinetics , Animals , Cyclin D1/genetics , Cyclin D1/metabolism , Dioxanes/pharmacokinetics , Female , Food Preferences , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Hippocampus/metabolism , Male , Mice , Prazosin/pharmacokinetics , Receptors, Adrenergic, alpha-1/genetics , Saccharin/administration & dosage , Sweetening Agents/administration & dosage , Time Factors
19.
Psychopharmacology (Berl) ; 229(4): 603-13, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23624776

ABSTRACT

RATIONALE: Dephosphorylation of extracellular signal-regulated kinase (ERK) and cyclic AMP response element binding protein (CREB) in the dorsomedial prefrontal cortex (dmPFC) at the end of short access (ShA) cocaine self-administration is implicated in cocaine seeking. However, what receptors and phosphatases mediate this effect and whether ERK/CREB and related phospho-proteins in the dmPFC react similarly during early withdrawal from long access (LgA) cocaine self-administration are unknown. OBJECTIVES: The effects of ShA vs. LgA cocaine self-administration on the phosphorylation of protein phosphatase 2A (PP2A) and striatal-enriched protein tyrosine phosphatase (STEP), as well as GluN and GluA receptor subtype expression in the dmPFC during early withdrawal, were compared. METHODS: Rats self-administered cocaine or received saline during 2- or 6-h daily sessions for 10-11 days. Two hours after the final session, the dmPFC was dissected out and processed for immunoblotting. RESULTS: Similar to previous findings after ShA cocaine, phospho-ERK and phospho-CREB in the dmPFC were decreased after LgA cocaine. Cocaine elevated phospho-PP2A (deactivation) and decreased phospho-STEP (activation) in both ShA and LgA cocaine rats. GluN1, GluN2B, and phospho-GluN2B Tyr1472 in the dmPFC were decreased after ShA and LgA cocaine. Further, a significant reduction of GluA2, GluA1, and phospho-GluA1 Ser845 was found only in LgA rats. CONCLUSIONS: Activation of phospho-STEP may underlie ERK and CREB dephosphorylation in the dmPFC as well as internalization and degradation of GluN complexes during early withdrawal from both ShA and LgA cocaine self-administration, whereas differential alteration of AMPA receptor subunits after ShA and LgA cocaine self-administration depends on cocaine intake.


Subject(s)
Cocaine/administration & dosage , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Drug-Seeking Behavior , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Male , Phosphorylation/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Protein Phosphatase 2/drug effects , Protein Phosphatase 2/metabolism , Rats , Rats, Sprague-Dawley , Receptors, AMPA/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Self Administration , Time Factors
20.
Pharmacol Rep ; 64(3): 546-57, 2012.
Article in English | MEDLINE | ID: mdl-22814008

ABSTRACT

BACKGROUND: The conditioned place preference (CPP) test is an animal model serving to assess addictive potential of drugs in which environmental cues become associated with the subjective effects of drugs of abuse. Morphine, a known addictive drug, is an agonist of opioid receptors that couple to the G(i/o) family of guanine nucleotide-binding proteins (GP). We have recently found that chronic treatment with morphine affects mRNA levels of GPs that are not coupled to opioid receptors (OR). Therefore, in this study, we investigated the influence of morphine-induced CPP on mRNA expression of the Gα subunits, G(i/o), G(s), G(q/11), and G(12), in the rat prefrontal cortex (PFC) and nucleus accumbens (NAc) using standard PCR techniques. METHODS: CPP and NO-CPP experiments were conducted; Wistar rats were either subjected to the standard CPP procedure or were injected with morphine (or saline) in their home cage. All rats were decapitated 24 h after the last injection. RESULTS: We found that mRNA levels of Gα(q), Gα(11) and Gα(12) were increased after morphine in non-conditioned treatment in the PFC but remained unchanged in the NAc. In rats showing conditioned place preference to morphine, levels of Gα(i2) in the PFC and levels of Gα(oA) in the NAc were diminished by ≈58% and ≈30%, respectively (p < 0.05 vs. saline), but levels of Gα(s-l) in NAc were increased (≈60%, p = 0.05). CONCLUSION: Our data indicate that only G(i/o) and G(s) were specifically changed in animals after morphine-induced CPP, thus suggesting that the effect was related to learning environmental cues associated with morphine.


Subject(s)
Analgesics, Opioid/pharmacology , GTP-Binding Protein alpha Subunits/genetics , Gene Expression Regulation/drug effects , Morphine/pharmacology , Animals , Behavior, Animal/drug effects , Conditioning, Psychological/drug effects , Cues , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Polymerase Chain Reaction , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , RNA, Messenger , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...