Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
J Inorg Biochem ; 237: 112012, 2022 12.
Article En | MEDLINE | ID: mdl-36162209

A significant fraction of patients are affected by persistent fear and anxiety. Currently, there are several anxiolytic drug options, however their clinical outcomes do not fully manage the symptoms. Here, we evaluated the effects of a bromazepam­palladium derivative [2-{(7-bromo-2-oxo-1,3-dihydro-2H-1,4-benzodiazepin-5-il)pyridinyl-κ2-N,N}chloropalladium(II)], [(BMZ)PdCl2], on fear/anxiety and memory-related behavior in mice. For this, female Swiss mice were treated intraperitoneally (i.p.) with saline (NaCl 0.9%) or [(BMZ)PdCl2] (0.5, 5.0, or 50 µg/kg). After 30 min, different tests were performed to evaluate anxiety, locomotion, and memory. We also evaluated the acute toxicity of [(BMZ)PdCl2] using a cell viability assay (neutral red uptake assay), and whether the drugs mechanism of action involves the γ-aminobutyric acid type A (GABAA) receptor complex by pre-treating animals with flumazenil (1.0 mg/kg, i.p., a competitive antagonist of GABAA-binding site). Our results demonstrate that [(BMZ)PdCl2] induces an anxiolytic-like phenotype in the elevated plus-maze test and that this effect can be blocked by flumazenil. Furthermore, there were no behavioral alterations induced by [(BMZ)PdCl2], as evaluated in the light-dark box, open field, and step-down passive avoidance tests. In the acute toxicity assay, [(BMZ)PdCl2] presented IC50 and LD50 values of 218 ± 60 µg/mL and 780 ± 80 mg/kg, respectively, and GSH category 4. Taken together, our results show that the anxiolytic-like effect of acute treatment with [(BMZ)PdCl2] occurs through the modulation of the benzodiazepine site in the GABAA receptor complex. Moreover, we show indications that [(BMZ)PdCl2] does not promote sedation and amnesia and presents the same toxicity as the bromazepam prototype.


Anti-Anxiety Agents , Bromazepam , Animals , Mice , Female , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Flumazenil/pharmacology , Bromazepam/pharmacology , Palladium/pharmacology , gamma-Aminobutyric Acid , Behavior, Animal , Maze Learning
2.
Bioorg Chem ; 119: 105548, 2022 02.
Article En | MEDLINE | ID: mdl-34959174

Epilepsy is a disease that affects millions of people around the globe and has a multifactorial cause. Inflammation is a process that can be involved in the development of seizures. Thus, the present study proposed the design and synthesis of new candidates for antiepileptic drugs that would also control the inflammatory process. Nine new derivatives of the substituted thiazophthalimide hybrid core were obtained with satisfactory purity ≥99% and yields between 27% and 87%. All compounds showed cell viability values greater than 90% in the culture of PBMC cells from healthy volunteers and, therefore, were not considered cytotoxic. These compounds modulated proinflammatory cytokines IFN-y and IL-17A and can mitigate inflammation. Acute toxicity studies of compound 7i in an animal model indicated that the compound has low toxicity and an LD50 greater than 2 g/kg in healthy adult rats. The same compound did not show positive results for anticonvulsant activity through the PTZ test. However, 7i demonstrates the interaction with the target GABA-A receptor in silico, indicating a possible activity as an agonist of that receptor. Thus, further studies are needed to investigate the anticonvulsant activity, in particular, using models in which the inflammatory process triggers epileptic seizures.


Anticonvulsants/therapeutic use , Epilepsy/drug therapy , Phthalimides/therapeutic use , Seizures/drug therapy , Thiazoles/therapeutic use , Animals , Anticonvulsants/chemical synthesis , Anticonvulsants/chemistry , Cells, Cultured , Dose-Response Relationship, Drug , Epilepsy/pathology , Humans , Male , Molecular Docking Simulation , Molecular Structure , Phthalimides/chemical synthesis , Phthalimides/chemistry , Rats , Rats, Wistar , Seizures/pathology , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/chemistry
4.
An Acad Bras Cienc ; 88(3 Suppl): 1993-2004, 2016.
Article En | MEDLINE | ID: mdl-27737339

Spondias tuberosa Arruda, popularly named as umbu, is native from savanna-like vegetation and widely used for medicinal purposes, however, the toxicological profile is not available yet. This study evaluated the phytochemical profile and acute toxicity and citoxicity of Ethanolic Extract of Spondias tuberosa Arruda Bark (EEStb) in hematological, biochemical and histopathological parameters. Female Wistar rats were divided into: control (C) and animal treated single doses of 300mg/Kg (EEStb300) or 2.000mg/kg body weight (ESStb2.000) of the EEStb. After 24 hours and 14 days from gavage, the behavior, hematological, biochemical and histopathological parameters were assayed. Cytotoxicity effect was evaluated on HEp-2 cell lines. Neither EEStb300 nor EEStb2.000 produced mortality nor changes in body weight during the 14-days of observation, but EEStb2.000 reduced quietly the food and water intake as well as locomotor activity at first day. There were no changes in macroscopic, histopathological, biochemical and hematological parameters. EEStb in concentrations of 6.25- 50µg ml-1 on HEp-2 cell did not produce cytotoxic effect. These results suggest that EEStb did not cause acute toxicity and cytotoxic, suggesting a good safety rate for Spondias tuberosa Arruda.


Anacardiaceae/chemistry , Plant Extracts/toxicity , Plants, Medicinal/toxicity , Animals , Ethanol , Female , Plants, Medicinal/chemistry , Rats , Rats, Wistar , Time Factors , Toxicity Tests, Acute
5.
Psychopharmacology (Berl) ; 227(2): 209-19, 2013 May.
Article En | MEDLINE | ID: mdl-23274504

RATIONALE: There is extensive evidence indicating the influence of seizures on emotional responses observed in human and animals, but so far few studies are focusing on the behavioral profile of animals that do not have seizures despite being treated with convulsant agents. OBJECTIVES: We aimed to establish the behavioral profile, biochemical, and electrographic features of rats submitted to the pilocarpine model of temporal lobe epilepsy METHODS: Rats treated with pilocarpine (20 to 350 mg/kg, i.p.) that did not develop status epilepticus or spontaneous recurrent seizures were evaluated 1 month later in the elevated plus maze (EPM), T-maze (ETM), open-field (OF), and step-down avoidance tests. Electroencephalographic (EEG), glutamate uptake, and hippocampal neuronal death assays were also performed RESULTS: Pilocarpine (150 or 350 mg/kg) promoted anxiogenic-like effects in rats evaluated in the EPM, ETM, and OF tests, whereas only the highest dose evoked spike-wave discharges during EEG recordings. Hippocampal theta rhythm was increased by pilocarpine 150 or 350 mg/kg and only the highest dose reduced the L-[(3)H]-glutamate uptake and cell viability on hippocampal slices. CONCLUSIONS: Subconvulsant doses of pilocarpine promote long-lasting alterations on neural circuitry, reflected by an increased theta activity in the hippocampus and an anxiety-like profile of rats evaluated 1 month after the treatment which is independent of seizure occurrence and is not related to changes in glutamate uptake or hippocampal damage. These results prompt us to suggest that a systemic administration of subconvulsant doses of pilocarpine could be useful as a new tool to model trait anxiety in rats.


Anxiety/chemically induced , Behavior, Animal/drug effects , Disease Models, Animal , Pilocarpine/toxicity , Animals , Avoidance Learning , Cell Death/drug effects , Dose-Response Relationship, Drug , Electroencephalography , Epilepsy, Temporal Lobe/chemically induced , Glutamic Acid/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Injections, Intraperitoneal , Male , Maze Learning , Neurons/pathology , Pilocarpine/administration & dosage , Rats , Rats, Wistar , Time Factors
6.
Neurotox Res ; 24(1): 55-62, 2013 Jul.
Article En | MEDLINE | ID: mdl-23184648

Searching for new therapeutic strategies through modulation of glutamatergic transmission using effective neuroprotective agents is essential. Glutamatergic excitotoxicity is a common factor to neurodegenerative diseases and acute events such as cerebral ischemia, traumatic brain injury, and epilepsy. This study aimed to evaluate behavioral and electroencephalographic (EEG) responses of mice cerebral cortex and hippocampus to subconvulsant and convulsant application of NMDA and quinolinic acid (QA), respectively. Moreover, it aimed to evaluate if EEG responses may be related to the neuroprotective effects of NMDA. Mice were preconditioned with NMDA (75 mg/kg, i.p.) and EEG recordings were performed for 30 min. One day later, QA was injected (36.8 nmol/site) and EEG recordings were performed during 10 min. EEG analysis demonstrated NMDA preconditioning promotes spike-wave discharges (SWDs), but it does not display behavioral manifestation of seizures. Animals that were protected by NMDA preconditioning against QA-induced behavioral seizures, presented higher number of SWD after NMDA administration, in comparison to animals preconditioned with NMDA that did display behavioral seizures after QA infusion. No differences were observed in latency for the first seizure or duration of seizures. EEG recordings after QA infusion demonstrated there were no differences in the number of SWD, latency for the first seizure or duration of seizures in animals pretreated with saline or in animals preconditioned by NMDA that received QA. A negative correlation was identified between the number of NMDA-induced SWD and QA-induced seizures severity. These results suggest a higher activation during NMDA preconditioning diminishes mice probability to display behavioral seizures after QA infusion.


Cerebral Cortex/drug effects , Hippocampus/drug effects , N-Methylaspartate/pharmacology , Quinolinic Acid/administration & dosage , Quinolinic Acid/antagonists & inhibitors , Seizures/drug therapy , Animals , Brain Waves/drug effects , Brain Waves/physiology , Cerebral Cortex/physiopathology , Hippocampus/physiopathology , Infusions, Intraventricular , Male , Mice , N-Methylaspartate/therapeutic use , Neuroprotective Agents/pharmacology , Quinolinic Acid/toxicity , Seizures/chemically induced
7.
Behav Brain Res ; 222(1): 206-11, 2011 Sep 12.
Article En | MEDLINE | ID: mdl-21459118

Depression and anxiety disorders present several genetic and neurobiological similarities. Drugs with antidepressant activity are effective in the treatment of a wide spectrum of anxiety disorders. Preclinical results showed that acute and chronic treatment with the NOP antagonist [Nphe(1),Arg(14),Lys(15)]N/OFQ-NH(2) (UFP-101) produced antidepressant-like effects in rodents. Thus, the present study aimed to investigate the effect of central administration of UFP-101 on the anxiety-related behavior in rats as evaluated in the elevated T-maze (ETM) test. Our results showed that UFP-101 reduced the latency of inhibitory avoidance in the ETM, indicating an anxiolytic-like effect. The endogenous peptide N/OFQ prevented this anxiolytic-like action of UFP-101, demonstrating its modulation via central NOP receptors. However, UFP-101 failed to interfere with the latency to escape. No change was observed in locomotor activity after UFP-101 treatment, ruling out any nonspecific motor effect. In conclusion, our results showed that the central administration of UFP-101 presents an anxiolytic-like effect in rats evaluated in the ETM test, providing new insights for drug development to treat anxiety disorders targeting the N/OFQ-NOP receptor system.


Anti-Anxiety Agents/administration & dosage , Anxiety/drug therapy , Maze Learning/drug effects , Analysis of Variance , Animals , Anxiety/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Exploratory Behavior/drug effects , Male , Narcotic Antagonists , Opioid Peptides , Rats , Rats, Wistar , Receptors, Opioid/agonists , Nociceptin Receptor
8.
Exp Neurol ; 226(2): 274-84, 2010 Dec.
Article En | MEDLINE | ID: mdl-20816828

The accumulation of amyloid-beta (Aß) peptides in the brain of human and rodents has been associated with the activation of glial cells, neuroinflammatory and oxidative responses, and cognitive deficits. These oxidative changes leave glutamate transporters more vulnerable and may result in reduction of their functions, resulting in excitotoxic damage. Herein, we evaluated the effects of atorvastatin, a HMG-CoA reductase inhibitor, in molecular and behavioral alterations induced by a single intracerebroventricular injection of aggregated Aß(1-40) (400 pmol) in mice. An increased glial fibrillar acidic protein (GFAP) expression and cyclooxygenase-2 (COX-2) levels, as well as increased lipid peroxidation and impairment in the glutathione antioxidant system and cell degeneration was found in the hippocampus of Aß(1-40)-treated mice. Aß(1-40) also induced a marked decrease in glutamatergic transporters (GLAST and GLT-1) expression and in l-[³H] glutamate uptake in mice hippocampus, in addition to spatial learning and memory deficits. Atorvastatin (10 mg/kg/day v.o.) was administered after Aß(1-40) injection and through seven consecutive days. Atorvastatin treatment was neuroprotective against cell degeneration induced by Aß(1-40), reducing inflammatory and oxidative responses and increasing the expression of glutamatergic transporters. On the other hand, atorvastatin did not reverse the cognitive impairments and failed to alter the hippocampal glutamate uptake in Aß(1-40)-treated mice. These results reinforce and extend the notion of the potential neuroprotective action of atorvastatin against the neuronal toxicity induced by Aß(1-40). In addition, the present findings suggest that the spatial learning and memory deficits induced by Aß peptides in rodents may not be entirely related to neuronal damage.


Amyloid beta-Peptides/toxicity , Encephalitis , Heptanoic Acids/pharmacology , Hippocampus/pathology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neurons/drug effects , Oxidative Stress/drug effects , Peptide Fragments/toxicity , Pyrroles/pharmacology , Amino Acid Transport System X-AG/metabolism , Analysis of Variance , Animals , Animals, Newborn , Astrocytes/drug effects , Atorvastatin , Cell Death/drug effects , Cyclooxygenase 2/metabolism , Encephalitis/chemically induced , Encephalitis/pathology , Encephalitis/prevention & control , Fluoresceins , Gene Expression Regulation/drug effects , Glutamic Acid/metabolism , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Humans , In Vitro Techniques , Learning Disabilities/chemically induced , Learning Disabilities/drug therapy , Male , Maze Learning/drug effects , Memory Disorders/chemically induced , Memory Disorders/drug therapy , Mice , Nerve Tissue Proteins/metabolism , Organic Chemicals , Propidium , Tritium/metabolism
9.
J Pharm Pharmacol ; 62(8): 1061-8, 2010 Aug.
Article En | MEDLINE | ID: mdl-20663041

OBJECTIVES: The aim of the present study was to evaluate the possible neurobehavioural effects in rats of the proanthocyanidin-rich fraction (PRF) isolated from the bark of Croton celtidifolius (Euphorbiaceae). METHODS: Adult Wistar rats were treated with the PRF (0.3-30 mg/kg) and evaluated in different behavioural paradigms classically used for the screening of drugs with psychoactive effects. KEY FINDINGS: Acute intraperitoneal (i.p.) administration of PRF decreased spontaneous locomotor activity (open field arena and activity cage), enhanced the duration of ethyl ether-induced hypnosis, increased the latency to the first convulsion induced by pentylenetetrazole (60 mg/kg, i.p.) and attenuated apomorphine-induced (0.5 mg/kg, i.p.) stereotyped behaviour. In lower doses, PRF (0.3 or 3 mg/kg, i.p.) increased the frequency of open arm entries in the elevated plus-maze test. CONCLUSIONS: The present findings suggest that the systemic administration of PRF induces a wide spectrum of behavioural alterations in rats, consistent with the putative existence of hypnosedative, anticonvulsant and anxiolytic compounds.


Behavior, Animal/drug effects , Central Nervous System Agents/pharmacology , Croton , Plant Extracts/pharmacology , Proanthocyanidins/pharmacology , Anesthetics, Inhalation/pharmacology , Animals , Anti-Anxiety Agents/pharmacology , Anticonvulsants/pharmacology , Antipsychotic Agents/pharmacology , Apomorphine/pharmacology , Central Nervous System Agents/administration & dosage , Central Nervous System Agents/isolation & purification , Consciousness/drug effects , Croton/chemistry , Disease Models, Animal , Dose-Response Relationship, Drug , Ether/pharmacology , Hypnotics and Sedatives/pharmacology , Injections, Intraperitoneal , Male , Motor Activity/drug effects , Pentylenetetrazole , Plant Bark , Plant Extracts/administration & dosage , Proanthocyanidins/administration & dosage , Proanthocyanidins/isolation & purification , Rats , Rats, Wistar , Seizures/chemically induced , Seizures/prevention & control , Stereotyped Behavior/drug effects
10.
Behav Brain Res ; 212(1): 96-102, 2010 Sep 01.
Article En | MEDLINE | ID: mdl-20363258

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the decline in cognitive functions, but it is also related to emotional disturbances. Since pain experience results from a complex integration of sensory, cognitive and affective processes, it is not surprising that AD patients display a distinct pattern of pain responsivity. We evaluated whether mice treated with amyloid beta (Abeta) peptide-thought to be critical in the pathogenesis of AD-exhibit altered pain responses and its relation to altered emotionality. Mice received a single i.c.v. injection of vehicle (PBS) or Abeta fragment (1-40) (400pmol/mice) and after 30 days, they were evaluated in tests of pain (hotplate, footshock-sensitivity), learning/memory (water-maze), emotionality (elevated plus-maze, forced swim) and locomotion (open-field). Abeta(1-40)-treated mice presented similar latencies to the control group in the hotplate test and similar nociceptive flinch threshold in the footshock-sensitivity test. However, they presented an increased jump threshold in footshock-sensitivity, suggesting increased pain tolerance. Altered emotionality was observed in the elevated plus-maze (EPM) and forced-swim tests (FST), suggesting anxiogenic-like and depressive-like states, respectively. A multifactorial principal component analysis (PCA) revealed that jump threshold of the footshock-sensitivity test falls within 'Emotionality' and 'Pain', showing moderate correlation with each one of the components of behavior. Acute treatment with the antidepressant desipramine (10mg/kg, i.p.) reduced the jump threshold (i.e. pain tolerance) and time of immobility in FST (i.e. depressive-like state). Flinch threshold (i.e. pain sensitivity), locomotion and anxiety were not altered with desipramine treatment. These results suggest that Abeta(1-40) peptide increases pain tolerance, but not pain sensitivity in mice, which seems to be linked to alterations in cognitive/emotional components of pain processing.


Amyloid beta-Peptides/pharmacology , Emotions/drug effects , Pain Threshold/drug effects , Pain/drug therapy , Peptide Fragments/pharmacology , Animals , Behavior, Animal/drug effects , Cognition/drug effects , Desipramine/pharmacology , Disease Models, Animal , Drug Interactions , Electroshock/adverse effects , Enzyme Inhibitors/pharmacology , Exploratory Behavior/drug effects , Humans , Immobility Response, Tonic/drug effects , Male , Maze Learning/drug effects , Mice , Motor Activity/drug effects , Pain/etiology , Pain Measurement/drug effects , Pain Measurement/methods
11.
Behav Brain Res ; 209(1): 165-73, 2010 May 01.
Article En | MEDLINE | ID: mdl-20122965

Alzheimer's disease (AD), a chronic degenerative and inflammatory brain disorder characterized by neuronal dysfunction and loss, is linked to accumulation of beta-amyloid (Abeta) peptide. Tumor necrosis factor-alpha (TNF-alpha) and cyclooxygenase-2 (COX-2) are proteins that have key roles in immune cell activation, inflammation and cognitive function in the brain. Here, we evaluated the link between TNF-alpha and COX-2 on the acute responses elicited by Abeta. Behavioral and molecular analyses were performed in mice after an intracerebroventricular (i.c.v.) injection of Abeta(1-40). Genetic and/or pharmacological approaches were used to inhibit TNF-alpha and COX-2. I.c.v. Abeta(1-40) injection in mice activates TNF-alpha signaling pathway resulting in COX-2 upregulation, synaptic loss and cognitive decline. Pharmacological studies revealed that COX-2 is involved in the cognitive impairment mediated by TNF-alpha. However, COX-2 inhibition failed in reducing the synaptophysin loss induced by Abeta(1-40). The COX-2 upregulation induced by Abeta(1-40) was attributed to activation of different protein kinases and transcriptional factors that are greatly regulated by TNF-alpha. Together, these results indicate that Abeta(1-40) induces the activation of several TNF-alpha-dependent intracellular signaling pathways that play a key role in the control of COX-2 upregulation and activation, synaptic loss and cognitive decline in mice. Therefore, selective TNF-alpha inhibitors may be potentially interesting tools for AD drug development.


Amyloid beta-Peptides , Cognition Disorders/chemically induced , Cyclooxygenase 2/metabolism , Peptide Fragments , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/physiology , Analysis of Variance , Animals , Antibodies/pharmacology , CREB-Binding Protein/metabolism , Dinoprostone/metabolism , Disease Models, Animal , Electrophoretic Mobility Shift Assay/methods , Enzyme Inhibitors/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitrobenzenes/pharmacology , Protein Kinase C/metabolism , Receptors, Tumor Necrosis Factor, Type I/deficiency , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Sulfonamides/pharmacology , Tumor Necrosis Factor-alpha/immunology , Up-Regulation/drug effects
12.
Brain Behav Immun ; 24(3): 493-501, 2010 Mar.
Article En | MEDLINE | ID: mdl-20025958

Alzheimer disease (AD) is the most common form of dementia in the elderly, and the neuro-pathological hallmarks of AD include neurofibrillary tangles (NFT), and deposition of beta-amyloid (Abeta) in extracellular plaques. In addition, chronic inflammation due to recruitment of activated glial cells to amyloid plaques are an invariant component in AD, and several studies have reported that the use of non-steroidal anti-inflammatory drugs (NSAIDs) may provide a measure of protection against AD. In this report we have investigated whether phosphoinositide 3-kinase gamma (PI3Kgamma), which is important in inflammatory cell migration, plays a critical role in the neuro-inflammation, synaptic dysfunction, and cognitive deficits induced by intracerebroventricular injection of Abeta(1-40) in mice. We found that the selective inhibitor of PI3Kgamma, AS605240, was able to attenuate the Abeta(1-40)-induced accumulation of activated astrocytes and microglia in the hippocampus, and decrease immuno-staining for p-Akt and cyclooxygenase-2 (COX-2). Interestingly, Abeta(1-40) activated macrophages treated with AS605240 or another PI3Kgamma inhibitor, AS252424, displayed impaired chemotaxis in vitro, but their expression of tumor necrosis factor-alpha (TNF-alpha) and interleukin-1beta (IL-1beta) was unaffected. Finally, AS605240 prevented Abeta(1-40)-induced cognitive deficits and synaptic dysfunction, but failed to modify scopolamine-induced amnesia. Our data suggests that inhibition of PI3Kgamma may represent a novel therapeutic target for treating AD patients.


Amyloid beta-Peptides/toxicity , Cognition/drug effects , Neuritis/enzymology , Neuritis/psychology , Peptide Fragments/toxicity , Phosphatidylinositol 3-Kinases/physiology , Animals , Cell Survival/drug effects , Chemotaxis, Leukocyte/drug effects , Class Ib Phosphatidylinositol 3-Kinase , Cyclooxygenase 2/metabolism , Enzyme Inhibitors/pharmacology , Image Processing, Computer-Assisted , Immunohistochemistry , Interleukin-1beta/metabolism , Isoenzymes/antagonists & inhibitors , Isoenzymes/physiology , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Neuritis/chemically induced , Neuroglia/drug effects , Neuroglia/immunology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Synapses/drug effects , Tumor Necrosis Factor-alpha/metabolism
13.
Peptides ; 29(8): 1404-12, 2008 Aug.
Article En | MEDLINE | ID: mdl-18499303

Nociceptin/orphanin FQ (N/OFQ) and nocistatin are two neuropeptides originated from the same precursor prepronociceptin/orphanin FQ (ppN/OFQ). N/OFQ is the endogenous ligand of the NOP receptor, while the target of action of nocistatin is still unknown. N/OFQ modulates various biological functions, including anxiety. Conversely, nocistatin either behaves as a functional N/OFQ antagonist or evokes per se effects opposite to those of N/OFQ. Here we investigated the interaction between the anxiolytic-like effects of N/OFQ and the anxiogenic-like action of nocistatin with those evoked by GABA(A) receptor ligands in the mouse elevated plus maze. The anxiogenic-like effects of the GABA(A) receptor antagonist pentylenetetrazol (20mg/kg; intraperitoneal, i.p.) were abolished by the co-treatment with N/OFQ (10pmol; intracerebroventricular, i.c.v.) while potentiated by the administration of nocistatin (0.01pmol; i.c.v.). The anxiolytic-like effects of the benzodiazepine receptor agonist diazepam (0.75mg/kg, i.p.) were reversed by nocistatin (0.1pmol; i.c.v.), whereas signs of sedation were observed when mice were co-treated with diazepam and N/OFQ (3pmol). Interesting enough, the i.p. treatment with flumazenil (1mg/kg) blocked the anxiolytic-like effects of N/OFQ (10pmol; i.c.v.), but not the anxiogenic effect elicited by nocistatin. Collectively, our findings suggest that the effects on anxiety elicited by pentylenetetrazol and diazepam can be counteracted or potentiated in the presence of N/OFQ and nocistatin. In addition, the effects on anxiety of N/OFQ, but not nocistatin, appear to be dependent on the benzodiazepine site of the GABA(A) receptor.


Anti-Anxiety Agents/pharmacology , GABA-A Receptor Agonists , Maze Learning/drug effects , Opioid Peptides/pharmacology , Animals , Diazepam/pharmacology , Dose-Response Relationship, Drug , Ligands , Male , Mice , Receptors, GABA-A/drug effects , Signal Transduction/drug effects , Vasodilator Agents/pharmacology , Nociceptin
14.
Behav Brain Res ; 187(1): 140-5, 2008 Feb 11.
Article En | MEDLINE | ID: mdl-17963853

This study aimed to investigate the relationship between substance P (SP) and diazepam (DZP) in the modulation of anxiety and memory in rats as evaluated in the elevated T-maze (ETM). For this purpose, in the first experiment, rats were intraperitoneally (i.p.) pretreated with saline or DZP (1mg/kg) and 25min later they were intracerebroventricularly (i.c.v.) injected with PBS or SP (10 pmol). In the second experiment, rats were i.p. pretreated with saline or DZP (1mg/kg) and 25 min later were i.c.v. injected with FK888 (100 pmol, a NK1 antagonist). After 1 min, animals were i.c.v. injected with vehicle (PBS+ethanol 10%) or SP (10 pmol). Our results show that DZP significantly decreased the latency to leave the enclosed arm of the ETM in the test and re-test session, indicating an anxiolytic and an amnesic effect, respectively. Although the central administration of SP did not significantly alter 'per se' the latency to leave the enclosed arm of the ETM in the test and re-test sessions, there was a trend to increase this parameter in the test session (indicating an anxiogenic-like effect). Furthermore, SP was able to reverse, via NK1 receptors, the effect produced by DZP during the test session. Moreover, none of the treatments interfered in the one-way escape behavior recorded in the test or re-test session in the ETM. In conclusion, our results strengthen and extend previous experimental data showing an interaction between the tachykinergic and benzodiazepine-GABA systems in the modulation of anxiety.


Anti-Anxiety Agents/pharmacology , Anxiety/psychology , Diazepam/pharmacology , Substance P/physiology , Animals , Anxiety/drug therapy , Avoidance Learning/drug effects , Dipeptides/pharmacology , Fear/drug effects , Fear/psychology , Indoles/pharmacology , Injections, Intraperitoneal , Injections, Intraventricular , Male , Memory/drug effects , Neurokinin-1 Receptor Antagonists , Rats , Rats, Wistar , Substance P/antagonists & inhibitors
15.
J Neurosci ; 27(20): 5394-404, 2007 May 16.
Article En | MEDLINE | ID: mdl-17507561

Increased brain deposition of amyloid beta protein (Abeta) and cognitive deficits are classical signals of Alzheimer's disease (AD) that have been highly associated with inflammatory alterations. The present work was designed to determine the correlation between tumor necrosis factor-alpha (TNF-alpha)-related signaling pathways and inducible nitric oxide synthase (iNOS) expression in a mouse model of AD, by means of both in vivo and in vitro approaches. The intracerebroventricular injection of Abeta(1-40) in mice resulted in marked deficits of learning and memory, according to assessment in the water maze paradigm. This cognition impairment seems to be related to synapse dysfunction and glial cell activation. The pharmacological blockage of either TNF-alpha or iNOS reduced the cognitive deficit evoked by Abeta(1-40) in mice. Similar results were obtained in TNF-alpha receptor 1 and iNOS knock-out mice. Abeta(1-40) administration induced an increase in TNF-alpha expression and oxidative alterations in prefrontal cortex and hippocampus. Likewise, Abeta(1-40) led to activation of both JNK (c-Jun-NH2-terminal kinase)/c-Jun and nuclear factor-kappaB, resulting in iNOS upregulation in both brain structures. The anti-TNF-alpha antibody reduced all of the molecular and biochemical alterations promoted by Abeta(1-40). These results provide new insights in mouse models of AD, revealing TNF-alpha and iNOS as central mediators of Abeta action. These pathways might be targeted for AD drug development.


Alzheimer Disease/metabolism , Amyloid beta-Peptides/administration & dosage , Disease Models, Animal , Nitric Oxide Synthase Type II/biosynthesis , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/physiology , Alzheimer Disease/genetics , Amyloid beta-Peptides/physiology , Animals , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Injections, Intraventricular , Male , Memory Disorders/chemically induced , Memory Disorders/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/genetics
16.
Behav Brain Res ; 177(2): 205-13, 2007 Feb 27.
Article En | MEDLINE | ID: mdl-17194489

Considerable evidence supports the role of oxidative stress in the pathogenesis of Alzheimer's disease (AD). Previous studies suggest that the central nervous system (CNS) administration of beta-amyloid peptide, the major constituent of senile plaque in AD, induces oxidative stress in rodents which may contribute to the learning and memory deficits verified in the beta-amyloid model of AD. In the present study, we compared the effects of a single intracerebroventricular (i.c.v.) injection of aggregated beta-amyloid peptide-(1-40) (Abeta(1-40)) (400pmol/mouse) on spatial learning and memory performance, synaptic density and the glutathione (GSH)-dependent antioxidant status in adult male C57BL/6 and Swiss albino mice. Seven days after Abeta(1-40) administration, C57BL/6 and Swiss mice presented similar spatial learning and memory impairments, as evaluated in the water maze task, although these impairments were not found in Abeta(40-1)-treated mice. Moreover, a similar decline of synaptophysin levels was observed in the hippocampus (HC) and prefrontal cortex (PFC) of both Swiss and C57BL/6 mice treated with Abeta(1-40), which suggests synaptic loss. C57BL/6 mice presented lower levels of glutathione-related antioxidant defences (total glutathione (GSH-t) levels, glutathione peroxidase (GPx) and glutathione reductase (GR) activity) in the HC and PFC in comparison to Swiss mice. Despite the reduced basal GSH-dependent antioxidant defences observed in C57BL/6 mice, Abeta(1-40) administration induced significant alterations in the brain antioxidant parameters only in Swiss mice, decreasing GSH-t levels and increasing GPx and GR activity in the HC and PFC 24h after treatment. These results indicate strain differences in the susceptibility to Abeta(1-40)-induced changes in the GSH-dependent antioxidant defences in mice, which should be taken into account in further studies using the Abeta model of AD in mice. In addition, the present findings suggest that the spatial learning and memory deficits induced by beta-amyloid peptides in rodents may not be entirely related to glutathione-dependent antioxidant response.


Amyloid beta-Peptides/administration & dosage , Cognition Disorders/chemically induced , Cognition Disorders/metabolism , Disease Susceptibility , Glutathione/metabolism , Peptide Fragments/administration & dosage , Animals , Behavior, Animal/drug effects , Cognition Disorders/physiopathology , Exploratory Behavior/drug effects , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Immunohistochemistry , Injections, Intraventricular/methods , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Reaction Time/drug effects , Space Perception/drug effects , Species Specificity
17.
Bioorg Med Chem ; 14(15): 5397-401, 2006 Aug 01.
Article En | MEDLINE | ID: mdl-16698270

The present study describes the chemical synthesis and pharmacological evaluation of a new series of eleven compounds stereoisomers of imidobenzenesulfonylaziridines in the forced-swimming test (FST) in mice. The pharmacological results of these compounds show that six of them, given intraperitoneally, reduced the immobility time of mice evaluated in the FST, an antidepressant-like profile of action similar to imipramine, a well-known standard antidepressant drug used for comparison, without compromising the animals' motor performance. The putative antidepressant-like action demonstrated here indicates their viability for the development of new therapeutic options for the treatment of depression.


Antidepressive Agents/chemical synthesis , Antidepressive Agents/pharmacology , Aziridines/chemical synthesis , Aziridines/pharmacology , Depression/prevention & control , Sulfones/chemical synthesis , Sulfones/pharmacology , Swimming , Alkaloids/chemistry , Animals , Antidepressive Agents/chemistry , Aziridines/chemistry , Behavior, Animal/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Imipramine/pharmacology , Immersion , Male , Mice , Molecular Structure , Motor Activity/drug effects , Phyllanthus/chemistry , Piperidones/chemistry , Stereoisomerism , Sulfones/chemistry
18.
Pharmacol Biochem Behav ; 79(1): 1-9, 2004 Sep.
Article En | MEDLINE | ID: mdl-15388277

This study investigated whether the central injection of substance P (SP) promotes differential behavioral [elevated plus-maze (EPM), open-field and hole-board tests] or immunological effects [peripheral blood lymphocyte subsets and nitric oxide (NO) produced by macrophages] in male and female mice. The percentage of time spent on open arms was significantly reduced by SP treatment in female (87%) and male mice (68%). A similar effect was observed in the percentage of entries into open arms (55% and 30%, respectively), as well as in the head-dipping parameter (63% and 27%, respectively), suggesting that SP promoted an anxiogenic-like profile in both sexes, which are detectable only in the plus-maze test. Female mice showed a significant decrease (20%) in the absolute number of lymphocytes and leukocytes comparing with control group and male SP-treated animals (4% of reduction), although only SP-treated males presented an increase (100%) in NO production by macrophages. Thus, our data showed no apparent statistical difference on the anxiogenic-like profile of action induced by SP between female and male mice; notwithstanding, SP, depending on the immune parameter evaluated, differentially influenced both sexes. The implications of these findings, as well as the putative participation of proinflammatory cytokines in this phenomenon, are discussed.


Behavior, Animal/drug effects , Immunity/drug effects , Substance P/pharmacology , Animals , Anxiety/psychology , Female , Grooming/drug effects , Injections, Intraventricular , Leukocyte Count , Lymphocyte Count , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Motor Activity/drug effects , Neutrophils/drug effects , Nitric Oxide/biosynthesis , Sex Characteristics , Substance P/administration & dosage
19.
Behav Brain Res ; 154(2): 501-10, 2004 Oct 05.
Article En | MEDLINE | ID: mdl-15313039

Substance P (SP) and its preferred NK1 receptor are widely expressed throughout the fear-processing pathways of the brain and its role in the modulation of experimental anxiety has been demonstrated. SP, like other peptides, are cleaved by peptidases in two fragments: C-terminal (SP 6-11) and N-terminal (SP 1-7) that could be responsible for its anxiogenic-like response. In this study we investigate the effects of i.c.v. micro-injections of SP free acid (SPfa), which is resistant to enzymatic cleavage, the influence of the pretreatment with peptidase inhibitors (PIs), thiorphan and/or phosphoramidon, as well as the effects of SP 6-11 and SP 1-7 and the participation of NK1 and NK2 receptors on their behavioral effects. Adult male Wistar rats were treated with 10 pmol solutions of SP 6-11, SP 1-7 or 1 and 10 pmol of SPfa and evaluated in the elevated plus maze (EPM) test. Other experimental groups received thiorphan 0.2 pmol, phosphoramidon 2 pmol or both PIs 30 min prior SP 1-11, 10 pmol i.c.v. The C-terminal fragment (SP 6-11, 10 pmol) and SPfa (1 pmol) promoted an anxiogenic-like profile of action similar to 10 pmol of SP 1-11, i.e., a decrease of entries and time spent on the open arms, whereas the N-terminal fragment (SP 1-7) was inactive at the EPM. The effect of SP 6-11 was inhibited by pretreatment (100 pmol) with NK1 (FK 888) and NK2 (SR 48968) antagonists. Moreover, both PIs enhanced the SP effect when used alone, but their combination produced an apparent reversion of anxiogenic-like effect produced by SP. Altogether, our results give further support to the SP role in the modulation of experimental anxiety in rats.


Maze Learning/drug effects , Substance P/analogs & derivatives , Substance P/pharmacology , Analysis of Variance , Animals , Behavior, Animal/drug effects , Benzamides/pharmacology , Dipeptides/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Glycopeptides/pharmacology , Indoles/pharmacology , Injections, Intraventricular , Male , Microinjections/methods , Motor Activity/drug effects , Neurokinin-1 Receptor Antagonists , Peptide Fragments/pharmacology , Piperidines/pharmacology , Protease Inhibitors , Pyrrolidonecarboxylic Acid/analogs & derivatives , Rats , Rats, Wistar , Receptors, Neurokinin-2/antagonists & inhibitors , Thiorphan/pharmacology , Time Factors
...