Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neurosci Biobehav Rev ; 163: 105748, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38857667

RESUMO

Corticotropin-releasing factor (CRF, corticoliberin) is a neuromodulatory peptide activating the hypothalamic-pituitary-adrenal (HPA) axis, widely distributed in the central nervous system (CNS) in mammals. In addition to its neuroendocrine effects, CRF is essential in regulating many functions under physiological and pathophysiological conditions through CRF1 and CRF2 receptors (CRF1R, CRF2R). This review aims to present selected examples of the diverse and sometimes opposite effects of CRF and its receptor ligands in various pathophysiological states, including stress/anxiety, depression, and processes associated with brain injury. It seems interesting to draw particular attention to the fact that CRF and its receptor ligands exert different effects depending on the brain structures or subregions, likely stemming from the varied distribution of CRFRs in these regions and interactions with other neurotransmitters. CRFR-mediated region-specific effects might also be related to brain site-specific ligand binding and the associated activated signaling pathways. Intriguingly, different types of CRF molecules can also influence the diverse actions of CRF in the CNS.


Assuntos
Ansiedade , Hormônio Liberador da Corticotropina , Receptores de Hormônio Liberador da Corticotropina , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Humanos , Animais , Hormônio Liberador da Corticotropina/metabolismo , Ansiedade/metabolismo , Ansiedade/fisiopatologia , Lesões Encefálicas/metabolismo , Lesões Encefálicas/fisiopatologia , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia , Depressão/metabolismo , Depressão/fisiopatologia , Encéfalo/metabolismo , Encéfalo/fisiopatologia
2.
Pharmacol Rep ; 74(1): 67-83, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34762280

RESUMO

BACKGROUND: Prolonged inflammation, oxidative stress, and protein aggregation are important factors contributing to Parkinson's disease (PD) pathology. A known ROS generator, pesticide paraquat (PQ), was indicated as an environmental substance potentially increasing the incidence of PD and is used to model this disease. We investigated if a combination of inflammation and oxidative stress in subthreshold doses would exacerbate the modelled neuropathology. METHODS: We examined the late effects of acute or repeated peripheral inflammation induced by low dose of LPS (10 µg/kg, ip) on PQ toxicity in the rat nigrostriatal dopaminergic pathway, microglial activation markers and expression of major Lewy bodies proteins, α-synuclein and synphilin-1. RESULTS: We observed that LPS increased, while PQ decreased body temperature and microglia CD11b expression in the SN. Single LPS pretreatment, 3 h before repeated weekly PQ injections (4×) slightly aggravated neuronal degeneration in the SN. Moreover, degeneration of dopaminergic neurons after weekly repeated inflammation itself (4×) was observed. Interestingly, repeated LPS administration combined with each PQ dose counteracted such effect. The expression of α-synuclein decreased after repeated LPS injections, while only combined, repeated LPS and PQ treatment lowered the levels of synphilin-1. Therefore, α-synuclein and synphilin-1 expression change was influenced by different mechanisms. Concomitantly, decreased levels of the two proteins correlated with decreased degeneration of dopaminergic neurons and with a normalized microglia activation marker. CONCLUSIONS: Our results indicate that both oxidative insult triggered by PQ and inflammation caused by peripheral LPS injection can individually induce neurotoxicity. Those factors act through different mechanisms that are not additive and not selective towards dopaminergic neurons, probably implying microglia. Repeated, but small insults from oxidative stress and inflammation when administered in significant time intervals can counteract each other and even act protective as a preconditioning effect. The timing of such repetitive insults is also of essence.


Assuntos
Proteínas de Transporte/metabolismo , Lipopolissacarídeos/farmacologia , Microglia , Proteínas do Tecido Nervoso/metabolismo , Doença de Parkinson/metabolismo , Substância Negra , alfa-Sinucleína/metabolismo , Animais , Modelos Animais de Doenças , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Relação Dose-Resposta a Droga , Exposição Ambiental , Herbicidas/toxicidade , Inflamação/metabolismo , Microglia/efeitos dos fármacos , Microglia/metabolismo , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Paraquat/toxicidade , Substâncias Protetoras/farmacologia , Ratos , Substância Negra/efeitos dos fármacos , Substância Negra/metabolismo
3.
Neuropeptides ; 88: 102162, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34062382

RESUMO

Corticotropin releasing factor (CRF) is a neuropeptide widely distributed in the brain as a hormonal modulator and neurotransmitter. The best known behavioral function of CRF is activation of stress and anxiety via the hypothalamus and limbic structures but the role of CRF in the cortex is still poorly understood. Our previous studies have shown anxiolytic-like effects of high doses of CRF injected into the Fr2 frontal cortex and involvement of CRF1 receptors (R) in that effect. These results seemed to be controversial as most other studies suggested anxiogenic and not anxiolytic effects of CRF1R stimulation. Since stress is associated with adrenergic system, in the present study, we focused on participation of alpha1 and alpha2 or beta adrenergic receptors in the anxiolytic-like effect of CRF. Moreover, we verified whether these effects of CRF in the Fr2 were really connected with CRF1R. Male Wistar rats were bilaterally microinjected with CRF in a dose of 0.2 µg/1 µl/site or with the specific agonist of CRF1R, stressin 1 (0.2-0.0125 µg/1 µl/site) into the Fr2 area. The elevated plus maze (EPM) test was performed 30 min later to assess the anxiolysis. An involvement of noradrenergic receptors in the CRF induced anxiolytic-like effect in the Fr2 was studied by pretreatment with the alpha1 antagonist prazosin, alpha2 agonist clonidine, alpha2 antagonist RS 79948 or beta antagonist propranolol, 20-30 min before CRF. The influence on anxiety was assessed in the EPM test. The results show that anxiolytic behavior after CRF microinjection into the Fr2 area seems to be mainly connected with the CRF1R activation because a similar effect was observed after stressin 1 administration and it was blocked by CRF1R antagonist. The results observed after administration of noradrenergic ligands indicated that anxiolytic effects of CRF in the Fr2 engaged the alpha1 and alpha2 adrenergic receptors but not beta receptors.


Assuntos
Ansiolíticos/farmacologia , Ansiedade/tratamento farmacológico , Comportamento Animal/efeitos dos fármacos , Hormônio Liberador da Corticotropina/farmacologia , Lobo Frontal/efeitos dos fármacos , Animais , Hormônio Liberador da Corticotropina/administração & dosagem , Isoquinolinas/farmacologia , Naftiridinas/farmacologia , Ratos , Receptores de Hormônio Liberador da Corticotropina/efeitos dos fármacos
4.
ACS Chem Neurosci ; 10(8): 3454-3463, 2019 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-31267743

RESUMO

This paper discusses the biological and three-dimensional molecular structure of the novel, nonpeptide Y2R antagonist, SF-11 [N-(4-ethoxyphenyl)-4-(hydroxydiphenylmethyl)-1-piperidinecarbothioamide]. Pharmacokinetic studies in a rat model indicated that, following intraperitoneal dosing, SF-11 crossed the blood-brain barrier and was able to penetrate the brain, making it a suitable tool for behavioral studies. We showed for the first time that SF-11 decreased the immobility time in the forced swim test (FST) after acute peripheral administration (10 and 20 mg/kg), indicating that it has antidepressant potential. Inhibitors of the mitogen-activated protein kinase/extracellular signal regulated kinase (MAPK/ERK) and phosphatidylinositol 3-kinase (PI3K) signaling pathways blocked the anti-immobility effect of SF-11, suggesting that these pathways are involved in the antidepressant-like activity of SF-11 in the FST. The results of locomotor activity of rats indicate that the effects observed in the FST are specific and due to the antidepressant-like activity of SF-11. These findings provide further evidence for the antidepressant potential of Y2R antagonists. Also, the application of Fourier transform infrared absorption (FT-IR) and Raman spectroscopy (RS) methods combined with theoretical density functional theory (DFT) calculations allowed us to present the optimized spatial orientation of the investigated drug. Structural characterization of SF-11 based on vibrational spectroscopic data is of great importance and will aid in understanding its biological activity and pave the way for its development as a new antidepressant agent.


Assuntos
Antidepressivos/farmacologia , Receptores de Neuropeptídeo Y/antagonistas & inibidores , Animais , Comportamento Animal/efeitos dos fármacos , Masculino , Atividade Motora/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
5.
Prog Neuropsychopharmacol Biol Psychiatry ; 84(Pt A): 93-101, 2018 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-29438731

RESUMO

Our previous studies have shown that ACPT-I [(1S, 3R,4S)-1-aminocyclopentane-1,2,4-tricarboxylic acid], a blood-brain barrier permeable agonist of group III metabotropic glutamate (mGlu) receptors, was neuroprotective against middle cerebral artery occlusion/reperfusion (MCAO/R) in normotensive rats. Preclinical studies are typically performed on healthy animals, whereas stroke patients predominately exhibit comorbidities, such as hypertension; therefore, in the present study, we investigated the effect of ACPT-I in spontaneously hypertensive rats (SHR) after MCAO/R. We examined the potential neuroprotective action of ACPT-I (30 mg/kg) when administered during occlusion or reperfusion via the assessment of not only the brain infarction volume but also motor (CatWalk gait analysis and open field test) and sensorimotor (vibrissae-evoked forelimb-placing test) functions following MCAO/R. We determined that ACPT-I not only reduced the cortico-striatal infarction but also improved several gait parameters (run speed, run and stand durations, swing speed and stride length) and mobility when administered 30 min after the start of the occlusion or 30 min after the start of reperfusion. Moreover, the sensorimotor function was improved in hypertensive rats treated with ACPT-I during occlusion. In conclusion, the current findings provide further evidence for the neuroprotective effects of ACPT-I against ischemic damage. These findings may have clinical implications because hypertension is an important risk factor for ischemic stroke.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Ciclopentanos/farmacologia , Hipertensão Essencial/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Acidente Vascular Cerebral/tratamento farmacológico , Ácidos Tricarboxílicos/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Isquemia Encefálica/complicações , Isquemia Encefálica/patologia , Hipertensão Essencial/complicações , Hipertensão Essencial/patologia , Marcha/efeitos dos fármacos , Masculino , Distribuição Aleatória , Ratos Endogâmicos SHR , Receptores de Glutamato Metabotrópico/agonistas , Sensação/efeitos dos fármacos , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/patologia
6.
Neuroscience ; 344: 305-325, 2017 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-28057538

RESUMO

It was postulated that neuropeptide Y (NPY)-ergic system could be involved in the ischemic pathophysiology, however, the role of particular subtypes of NPY receptors (YRs) in neuroprotection against ischemia is still not well known. Therefore, we investigated the effect of NPY and YR ligands using in vitro and in vivo experimental ischemic stroke models. Our in vitro findings showed that NPY (0.5-1µM) and specific agonists of Y2R (0.1-1µM) and Y5R (0.5-1µM) but not that of Y1R produced neuroprotective effects against oxygen-glucose deprivation (OGD)-induced neuronal cell death, being also effective when given 30min after the end of OGD. The neuroprotective effects of Y2R and Y5R agonists were reversed by appropriate antagonists. Neuroprotection mediated by NPY, Y2R and Y5R agonists was accompanied by the inhibition of both OGD-induced calpain activation and glutamate release. Data from in vivo studies demonstrated that Y2R agonist (10µg/6µl; i.c.v.) not only diminished the infarct volume in rats subjected to transient middle cerebral artery occlusion (MCAO) but also improved selected gait parameters in CatWalk behavioral test, being also effective after delayed treatment. Moreover, we found that a Y5R agonist (10µg/6µl; i.c.v.) did not reduce MCAO-evoked brain damage but improved stride length, when it was given 30min after starting the occlusion. In conclusion, our studies indicate that Y5 and especially Y2 receptors may be promising targets for neuroprotection against ischemic damage.


Assuntos
Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Receptores de Neuropeptídeo Y/agonistas , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Calpaína/metabolismo , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Marcha/efeitos dos fármacos , Marcha/fisiologia , Glucose/deficiência , Ácido Glutâmico/metabolismo , Ataque Isquêmico Transitório/tratamento farmacológico , Ataque Isquêmico Transitório/metabolismo , Ataque Isquêmico Transitório/patologia , Masculino , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Neuroproteção/efeitos dos fármacos , Neuroproteção/fisiologia , Distribuição Aleatória , Ratos Sprague-Dawley , Receptores de Neuropeptídeo Y/antagonistas & inibidores , Receptores de Neuropeptídeo Y/metabolismo
7.
Psychopharmacology (Berl) ; 234(4): 631-645, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27975125

RESUMO

RATIONALE: It has recently been found that chronic treatment with the highly selective, brain-penetrating Y5 receptor antagonist, Lu AA33810 [N-[[trans-4-[(4,5-dihydro [1] benzothiepino[5,4-d] thiazol-2-yl) amino] cyclohexyl]methyl]-methanesulfonamide], produces antidepressant-like effects in the rat chronic mild stress model. OBJECTIVE: In the present study, we investigated the possible antidepressant-like activity of Lu AA33810 in rats subjected to glial ablation in the prefrontal cortex (PFC) by the gliotoxin L-AAA, which is an astroglial degeneration model of depression. RESULTS: We observed that Lu AA33810 administered intraperitoneally at a single dose of 10 mg/kg both reversed depressive-like behavioral changes in the forced swim test (FST) and prevented degeneration of astrocytes in the mPFC. The mechanism of antidepressant and glioprotective effects of Lu AA33810 has not been studied, so far. We demonstrated the contribution of the noradrenergic rather than the serotonergic pathway to the antidepressant-like action of Lu AA33810 in the FST. Moreover, we found that antidepressant-like effect of Lu AA33810 was connected with the influence on brain-derived neurotrophic factor (BDNF) protein expression. We also demonstrated the antidepressant-like effect of Lu AA33810 in the FST in rats which did not receive the gliotoxin. We found that intracerebroventricular injection of the selective MAPK/ERK inhibitor U0126 (5 µg/2 µl) and the selective PI3K inhibitor LY294002 (10 nmol/2 µl) significantly inhibited the anti-immobility effect of Lu AA33810 in the FST in rats, suggesting that MAPK/ERK and PI3K signaling pathways could be involved in the antidepressant-like effect of Lu AA33810. CONCLUSION: Our results indicate that Lu AA33810 exerts an antidepressant-like effect and suggest the Y5 receptors as a promising target for antidepressant therapy.


Assuntos
Antidepressivos/farmacologia , Benzotiepinas/farmacologia , Depressão/tratamento farmacológico , Sulfonamidas/farmacologia , Animais , Antidepressivos/uso terapêutico , Comportamento Animal/efeitos dos fármacos , Benzotiepinas/uso terapêutico , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Cromonas/farmacologia , Depressão/metabolismo , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Masculino , Morfolinas/farmacologia , Neuropeptídeo Y/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/uso terapêutico , Natação
8.
Neuropharmacology ; 102: 276-94, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26647070

RESUMO

In the present study, we investigated the effect of ACPT-I [(1S, 3R,4S)-1-aminocyclopentane-1,2,4-tricarboxylic acid], a blood-brain-barrier permeable agonist of group III mGlu receptor, against oxygen-glucose deprivation (OGD)-evoked neuronal cell death in primary neuronal cell cultures and in the model of transient middle cerebral artery occlusion (MCAO) in rats. We found that ACPT-I (1-200 µM) in a concentration- and time-dependent way attenuated the OGD-induced neuronal cell damage, being also effective after a delayed application (30 min after OGD). The neuroprotective effects of ACPT-I were blocked by the group III mGlu receptor antagonist, (RS)-alpha-cyclopropyl-4-phosphonophenyl glycine (CPPG), and by the activator of cAMP-dependent PKA, 8-Bromo-cAMP, but not by an inhibitor of PI-3-K signaling pathway. Moreover, ACPT-I attenuated the OGD-induced calpain activity and glutamate release. In the in vitro study, we also demonstrated the neuroprotective potential of mGluR4 positive allosteric modulators (PAMs), PHCCC (30 µM) and VU0155041 (10 and 30 µM) and synergism in neuroprotective action of low concentrations of ACPT-I and mGluR4 PAMs suggesting an important role of mGluR4 activation in prevention of ischemic neuronal cell death. In the rat MCAO model, we demonstrated that ACPT-I (30 mg/kg) injected intraperitoneally either 30 min after starting MCAO or 30 min after beginning reperfusion not only diminished the infarction volume by about 30%, but also improved selected gait parameters (CatWalk analysis) and the mobility of animals in the open field test. In conclusion, our results indicate that ACPT-I may be not only neuroprotective against ischemic neuronal damage but may also diminish the postischemic functional deficits.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Ciclopentanos/uso terapêutico , Agonistas de Aminoácidos Excitatórios/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Receptores de Glutamato Metabotrópico/agonistas , Acidente Vascular Cerebral/tratamento farmacológico , Ácidos Tricarboxílicos/uso terapêutico , Animais , Isquemia Encefálica/metabolismo , Células Cultivadas , Ciclopentanos/farmacologia , Modelos Animais de Doenças , Agonistas de Aminoácidos Excitatórios/farmacologia , L-Lactato Desidrogenase/metabolismo , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/metabolismo , Ácidos Tricarboxílicos/farmacologia
9.
Neurochem Int ; 88: 110-23, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25576184

RESUMO

Although numerous studies demonstrated a neuroprotective potency of unspecific group III mGluR agonists in in vitro and in vivo models of excitotoxicity, little is known about the protective role of group III mGlu receptor activation against neuronal cell injury evoked by ischemic conditions. The aim of the present study was to assess neuroprotective potential of the allosteric agonist of mGlu7 receptor, N,N'-Bis(diphenylmethyl)-1,2-ethanediamine dihydrochloride (AMN082) against oxygen-glucose deprivation (OGD)- and kainate (KA)-evoked neuronal cell damage in primary neuronal cultures, with special focus on its efficacy after delayed application. We demonstrated that in cortical neuronal cultures exposed to a 180 min OGD, AMN082 (0.01-1 µM) in a concentration- and time-dependent way attenuated the OGD-induced changes in the LDH release and MTT reduction assays. AMN082 (0.5 and 1 µM) produced also neuroprotective effects against KA-evoked neurotoxicity both in cortical and hippocampal cultures. Of particular importance was the finding that AMN082 attenuated excitotoxic neuronal injury after delayed application (30 min after OGD, or 30 min-1 h after KA). In both models of neurotoxicity, namely OGD- and KA-induced injury, the neuroprotective effects of AMN082 (1 µM) were reversed by the selective mGlu7 antagonist, 6-(4-Methoxyphenyl)-5-methyl-3-(4-pyridinyl)-isoxazolo[4,5-c]pyridin-4(5H)-one hydrochloride (MMPIP, 1 µM), suggesting the mGlu7-dependent mechanism of neuroprotective effects of AMN082. Next, we showed that AMN082 (0.5 and 1 µM) attenuated the OGD-induced increase in the number of necrotic nuclei as well inhibited the OGD-evoked calpain activation, suggesting the participation of these processes in the mechanism of AMN082-mediated protection. Additionally, we showed that protection evoked by AMN082 (1 µM) in KA model was connected with the inhibition of toxin-induced caspase-3 activity, and this effect was abolished by the mGlu7 receptor antagonist. The obtained results indicated that the activation of mGlu7 receptors may be a promising target for neuroprotection against ischemic and excitotoxic insults.


Assuntos
Compostos Benzidrílicos/farmacologia , Glucose/deficiência , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Oxigênio/metabolismo , Receptores de Glutamato Metabotrópico/agonistas , Regulação Alostérica , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Ácido Caínico/toxicidade , Camundongos , Neurônios/efeitos dos fármacos
10.
Behav Brain Res ; 273: 23-33, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25043733

RESUMO

The glutamatergic predominance in the excitatory-inhibitory balance is postulated to be involved in the pathogenesis of depression. Such imbalance may be induced by astrocyte ablation which reduces glutamate uptake and increases glutamate level in the synaptic cleft. In the present study, we tried to ascertain whether astroglial degeneration in the prefrontal cortex could serve as an animal model of depression and whether inhibition of glutamatergic transmission by the mGluR5 antagonist MTEP could have antidepressant potential. Astrocytic toxins l-or dl-alpha-aminoadipic acid (AAA), 100µg/2µl, were microinjected, bilaterally into the rat medial prefrontal cortex (PFC) on the first and second day of experiment. MTEP (10mg/kg) or imipramine (30mg/kg) were administered on the fifth day. Following administration of MTEP or imipramine the forced swim test (FST) was performed for assessment of depressive-like behavior. The brains were taken out for analysis on day eight. The astrocytic marker, glial fibrillary acidic protein (GFAP) was quantified in PFC by Western blot method and by stereological counting of immunohistochemically stained sections. Both l-AAA and dl-AAA induced a significant increase in immobility time in the FST. This effect was reversed by imipramine, which indicates depressive-like effects of these toxins. A significant decrease in GFAP (about 50%) was found after l-AAA. Both the behavioral and GFAP level changes were prevented by MTEP injection. The obtained results indicate that the degeneration of astrocytes in the PFC by l-AAA may be a useful animal model of depression and suggest antidepressant potential of MTEP.


Assuntos
Antidepressivos/farmacologia , Astrócitos/patologia , Depressão/metabolismo , Depressão/patologia , Piridinas/farmacologia , Tiazóis/farmacologia , Ácido 2-Aminoadípico/toxicidade , Animais , Astrócitos/efeitos dos fármacos , Modelos Animais de Doenças , Masculino , Atividade Motora/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/patologia , Ratos , Ratos Sprague-Dawley , Receptor de Glutamato Metabotrópico 5/antagonistas & inibidores
11.
Neurotox Res ; 26(3): 255-73, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24842651

RESUMO

A number of studies suggest that the ubiquitin-proteasome system (UPS) impairment may underlie neuronal death in Parkinson's disease. Celastrol is a neuroprotective agent with anti-inflammatory and antioxidant properties. The aim of this study was to determine whether celastrol may exert neuroprotective effects both in vitro and in vivo under conditions of the lactacystin-induced UPS inhibition. In the in vitro study, mouse primary cortical neurons and neuroblastoma SH-SY5Y cells were incubated with lactacystin for 48 h (2.5 and 10 µg/ml, respectively). The animal study was performed on male Wistar rats injected unilaterally with lactacystin (5 µg/2 µl) into the substantia nigra (SN) pars compacta. In the in vitro study, we did not found any protective effects of celastrol, given either in the pre- or co-treatment mode. Moreover, in the higher concentrations, celastrol itself reduced cell viability, and enhanced the lactacystin-induced cell death in both types of cells. In the in vivo study, none of the celastrol doses (0.3-3 mg/kg) attenuated the lactacystin-induced decrease in the level of dopamine (DA) and its metabolites or protected nigral dopaminergic neurons against the lactacystin-induced degeneration. The highest celastrol dose potentiated the lactacystin-induced decrease in the level of DA and its metabolites in the lesioned striatum, and accelerated the lactacystin-induced increase in the oxidative and total metabolism of DA. Moreover, when given alone, this dose of celastrol bilaterally decreased the number and/or density of dopaminergic neurons in the SN. Our results demonstrate that celastrol does not induce neuroprotective effects under conditions of UPS inhibition.


Assuntos
Acetilcisteína/análogos & derivados , Fármacos Neuroprotetores/farmacologia , Doença de Parkinson/metabolismo , Inibidores de Proteassoma/toxicidade , Triterpenos/farmacologia , Acetilcisteína/toxicidade , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Dopamina/metabolismo , Humanos , Masculino , Camundongos , Neuroblastoma , Neurônios/efeitos dos fármacos , Triterpenos Pentacíclicos , Ratos , Ratos Wistar , Substância Negra/efeitos dos fármacos , Substância Negra/patologia
12.
Neurotox Res ; 26(1): 99-113, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24402869

RESUMO

Many evidence suggest that metabotropic glutamate receptors (mGluRs) may modulate glutamatergic transmission, hence, these receptors are regarded as potential targets for neuroprotective drugs. Since group III mGlu receptor agonists are known to reduce glutamatergic transmission by inhibiting glutamate release, we decided to investigate the neuroprotective potential of the group III mGlu receptor agonist, (1S,3R,4S)-1-aminocyclopentane-1,2,4-tricarboxylic acid (ACPT-I) against kainate (KA)-induced excitotoxicity in vitro and in vivo. In primary neuronal cell cultures ACPT-I (1-200 µM), applied 30 min-3 h after starting the exposure to KA (150 µM), significantly attenuated the KA-induced LDH release, increased cell viability, and inhibited caspase-3 activity both in cortical and hippocampal cell cultures. The effects were dose-, time- and structure-dependent. The neuroprotective effects of ACPT-I were reversed by (RS)-alpha-cyclopropyl-4-phosphonophenyl glycine, a group III mGluR antagonist. In the in vivo studies, KA (2.5 nmol/1 µl) was unilaterally injected into the rat dorsal CA1 hippocampal region and the size of degeneration was examined by stereological counting of surviving neurons in the CA pyramidal layer. It was found that ACPT-I (7.5 or 15 nmol/1 µl), injected into the dorsal hippocampus 30 min, 1 or 3 h after KA in dose-dependent manner prevented the KA-induced neuronal damage. Moreover, in vivo microdialysis studies in the rat hippocampus showed that ACPT-I (200 µM) given simultaneously with KA (50 µM) significantly diminished the KA-induced glutamate release in the hippocampus. This mechanism seems to play a role in mediating the neuroprotective effect of ACPT-I.


Assuntos
Córtex Cerebral/efeitos dos fármacos , Ciclopentanos/farmacologia , Hipocampo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Receptores de Glutamato Metabotrópico/agonistas , Ácidos Tricarboxílicos/farmacologia , Animais , Células Cultivadas , Córtex Cerebral/fisiologia , Agonistas de Aminoácidos Excitatórios/toxicidade , Espaço Extracelular/metabolismo , Ácido Glutâmico/metabolismo , Hipocampo/patologia , Hipocampo/fisiopatologia , Ácido Caínico/toxicidade , Masculino , Camundongos , Neurônios/patologia , Neurônios/fisiologia , Ratos Wistar , Receptores de Glutamato Metabotrópico/metabolismo
13.
Brain Res ; 1541: 92-105, 2013 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-24129225

RESUMO

Recently, it has been strongly suggested that reciprocal interactions between nitrergic and dopaminergic systems play a crucial role in the control of the nigrostriatal pathway. Degeneration of dopaminergic neurons in the substantia nigra (SN) in Parkinson's disease leads to disturbances in the nitrergic transmission in the basal ganglia. In the present study, we aimed to compare regional distribution of nNOS immunoreactivity and NADPH-diaphorase activity in the SN and subthalamic nucleus (STN) of unilaterally 6-OHDA-lesioned rats treated chronically with l-DOPA (25mg/kg) and the nitric oxide donor, molsidomine (2 or 4mg/kg). Our results showed that degeneration of dopaminergic neurons in the ipsilateral SN resulted in a 25% decrease in the number of nNOS-immunoreactive neurons in that structure and in nNOS protein level determined by Western blot. We also found that nNOS was present in about 70% of all SN neurons. NADPH-d histochemistry did not reveal nNOS activity in the SN of any studied groups. Furthermore, the stereological analysis of the SN volume showed that chronic administration of l-DOPA evoked a hypertrophy of the ipsilateral SN when compared to the contralateral side. Such difference between sides was abolished in the group receiving l-DOPA in combination with molsidomine. Degeneration of the nigrostriatal pathway had no influence on the number of nNOS-ir neurons in the STN. NADPH-histochemistry revealed nNOS activity only in a part of neurons of that structure. Our results make an essential contribution to the research on the role of nitric oxide in the regulation of basal ganglia function.


Assuntos
Óxido Nítrico Sintase Tipo I/biossíntese , Transtornos Parkinsonianos/metabolismo , Substância Negra/enzimologia , Núcleo Subtalâmico/enzimologia , Adrenérgicos/toxicidade , Animais , Antiparkinsonianos/farmacologia , Western Blotting , Imuno-Histoquímica , Levodopa/farmacologia , Masculino , Molsidomina/farmacologia , NADPH Desidrogenase/metabolismo , Neurônios/enzimologia , Doadores de Óxido Nítrico/farmacologia , Oxidopamina/toxicidade , Ratos , Ratos Wistar , Substância Negra/efeitos dos fármacos , Núcleo Subtalâmico/efeitos dos fármacos
14.
Pharmacol Rep ; 65(6): 1572-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24553005

RESUMO

Major depression (MD) is a common and disabling disorder but knowledge of its pathophysiology is still incomplete. In the last years, degenerations or dysfunctions of glial cells, especially astrocytes, have been postulated to play a critical role in the pathogenesis of depression. Glial loss in prefrontal and limbic brain regions was observed in depressed patients and in animal models of stress and depression. Degeneration of astrocytes resulted in an excess glutamate in the synaptic cleft and glutamate/GABA imbalance in the affected structures. This review presents an up-to-date information concerning the role of glial cells in maintenance of glutamate/GABA balance in the brain tripartite glutamatergic synapses; discusses the importance of glial pathology and presents models of depression based on astrocyte impairment. The model of degeneration of astrocytes in the medial prefrontal cortex of the rat, induced by the specific astrocytic toxin α-aminoadipic acid, is presented as a valuable model for studying antidepressant compounds.


Assuntos
Depressão/patologia , Degeneração Neural/patologia , Neuroglia/patologia , Animais , Astrócitos/patologia , Encéfalo/patologia , Humanos
15.
Neurotox Res ; 21(4): 379-92, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22144346

RESUMO

Abundant evidence suggests that indirect inhibitory modulation of glutamatergic transmission, via metabotropic glutamatergic receptors (mGluR), may induce neuroprotection. The present study was designed to determine whether the selective antagonist of mGluR1 (3-ethyl-2-methyl-quinolin-6-yl)-(4-methoxy-cyclohexyl)-methanone methanesulfonate (EMQMCM), showed neuroprotection against the kainate (KA)-induced excitotoxicity in vitro and in vivo. In in vitro studies on mouse primary cortical and hippocampal neuronal cultures, incubation with KA (150 µM) induced strong degeneration [measured as lactate dehydrogenase (LDH) efflux] and apoptosis (measured as caspase-3 activity). EMQMCM (0.1-100 µM) added 30 min to 6 h after KA, significantly attenuated the KA-induced LDH release and prevented the increase in caspase-3 activity in the cultures. Those effects were dose- and time-dependent. In in vivo studies KA (2.5 nmol/1 µl) was unilaterally injected into the rat dorsal CA1 hippocampal region. Degeneration was calculated by counting surviving neurons in the CA pyramidal layer using stereological methods. It was found that EMQMCM (5-10 nmol/1 µl) injected into the dorsal hippocampus 30 min, 1 h, or 3 h (the higher dose only) after KA significantly prevented the KA-induced neuronal degeneration. In vivo microdialysis studies in rat hippocampus showed that EMQMCM (100 µM) significantly increased γ-aminobutyric acid (GABA) and decreased glutamate release. When perfused simultaneously with KA, EMQMCM substantially increased GABA release and prevented the KA-induced glutamate release. The obtained results indicate that the mGluR1 antagonist, EMQMCM, may exert neuroprotection against excitotoxicity after delayed treatment (30 min to 6 h). The role of enhanced GABAergic transmission in the neuroprotection is postulated.


Assuntos
Apoptose/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Ácido Caínico/antagonistas & inibidores , Fármacos Neuroprotetores/farmacologia , Quinolinas/farmacologia , Animais , Caspase 3/metabolismo , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Ácido Glutâmico/metabolismo , Hipocampo/patologia , Ácido Caínico/administração & dosagem , Ácido Caínico/toxicidade , L-Lactato Desidrogenase/metabolismo , Masculino , Camundongos , Microinjeções , Degeneração Neural/induzido quimicamente , Degeneração Neural/patologia , Cultura Primária de Células , Quinolinas/administração & dosagem , Ratos , Ratos Wistar , Ácido gama-Aminobutírico/metabolismo
16.
Neurochem Int ; 58(7): 839-49, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21419185

RESUMO

Impairment of the ubiquitin-proteasome system, responsible for clearing of misfolded and unwanted proteins, has been implicated in the loss of nigrostriatal dopaminergic neurons characteristic of Parkinson's disease (PD). Recently, proteasome inhibitors have been used to model parkinsonian-like changes in animals. In the present study, the effects of intrastriatal and intranigral injections of the selective proteasome inhibitor lactacystin on key markers of PD were examined in Wistar rats. Comparisons of these two different routes of lactacystin administration revealed that only a unilateral, intranigral injection of lactacystin at a dose of 0.5, 1, 2.5 and 5 µg/2 µl produced after 7 days distinct decreases in the concentrations of dopamine (DA) and its metabolites (DOPAC, 3-MT, HVA) in the ipsilateral striatum. The used doses of lactacystin (except for 0.5 µg/2 µl) significantly accelerated DA catabolism, i.e. the total, oxidative MAO-dependent and COMT-catalyzed pathways, as assessed by HVA/DA, DOPAC/DA and 3-MT/DA ratios, respectively, in the ipsilateral striatum. Such alterations were not observed in the striatal DA content and catabolism either 7, 14 or 21 days after a unilateral, intrastriatal high-dose lactacystin injection (5 and 10 µg/2 µl). Intranigrally administered lactacystin (1 µg/2 µl) caused a marked decline of tyrosine hydroxylase (TH) and α-synuclein protein levels in that structure. Neither TH nor α-synuclein protein levels in the substantia nigra (SN) were affected by high lactacystin doses injected intrastriatally. Moreover, stereological counting of TH-immunoreactive neurons and autoradiographic analysis of [(3)H]GBR 12,935 binding to dopamine transporter confirmed a loss of nigrostriatal dopaminergic neurons after an intranigral lactacystin (1 and 2.5 µg/2 µl) injection. An appearance of cardinal neurochemical and histological changes of parkinsonian type only after intranigral lactacystin injection indicates that DA cell bodies in the SN, but not DA terminals in the striatum are susceptible to proteasome inhibition.


Assuntos
Acetilcisteína/análogos & derivados , Corpo Estriado/efeitos dos fármacos , Doença de Parkinson/metabolismo , Inibidores de Proteassoma/farmacologia , Substância Negra/efeitos dos fármacos , Acetilcisteína/administração & dosagem , Acetilcisteína/farmacologia , Animais , Western Blotting , Relação Dose-Resposta a Droga , Masculino , Inibidores de Proteassoma/administração & dosagem , Ratos , Ratos Wistar
17.
Pharmacol Rep ; 62(6): 1051-61, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21273662

RESUMO

Extensive research into glutamate receptors in the central nervous system has shown important role of metabotropic glutamate receptors (mGluR) as potential targets for neuroprotective drugs. The aim of the present study was to investigate neuroprotective potential of the highly selective mGlu5 antagonist 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]-pyridine (MTEP) against kainate (KA)-induced excitotoxicity in vivo. Our attention was focused mainly on the effectiveness of delayed treatment. In order to evoke neuronal injury, rats were unilaterally injected with kainic acid (KA; 2.5 nmol/1 µl) into the CA1 region of the hippocampus. MTEP (1, 5 or 10 nmol/1 µl) was administered into CA1 30 min, 1, 3 and 6 h after KA. Additionally, other rats were injected intraperitoneally (i.p.) with MTEP in a dose of 1 mg/kg, once daily for 7 days. The first injection of MTEP was 1 h after KA. Seven days after treatment, the brains were taken out and analyzed histologically to estimate the total number of neurons in CA region of dorsal hippocampus using stereological methods. The study was also aimed at determining a possible influence of MTEP on neuronal glutamate release induced by KA in the hippocampus, using microdialysis method. The obtained results showed that MTEP had neuroprotective effect after both intrahippocampal and intraperitoneal injection. It was found that MTEP could prevent excitotoxic neuronal damage even when it was applied 1-6 h after the toxin. Moreover, it was observed that MTEP significantly reduced the KA-induced glutamate release in the hippocampus. It seems to play a role in mediating neuroprotective effects of MTEP.


Assuntos
Região CA1 Hipocampal/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/farmacologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Piridinas/farmacologia , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Tiazóis/farmacologia , Animais , Região CA1 Hipocampal/metabolismo , Tamanho Celular/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Ácido Caínico/farmacologia , Masculino , Microdiálise , Neurônios/metabolismo , Ratos , Ratos Wistar , Receptor de Glutamato Metabotrópico 5 , Receptores de Glutamato Metabotrópico/metabolismo , Fatores de Tempo
18.
Neuropeptides ; 43(3): 235-49, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19318226

RESUMO

It is generally assumed that neurodegeneration is connected with glutamatergic hyperactivity, and that neuropeptide Y (NPY) inhibits glutamate release. Some earlier studies indicated that NPY may have neuroprotective effect; however, the results obtained so far are still divergent, and the role of different Y receptors remains unclear. Therefore in the presented study we investigated the neuroprotective potential of NPY and its Y2, Y5 or Y1 receptor (R) ligands against the kainate (KA)-induced excitotoxicity in neuronal cultures in vitro, as well as in vivo after intrahippocampal KA injection and also in an ischemic middle cerebral artery occlusion model after intraventricular injection of Y2R agonist. NPY compounds were applicated 30 min, 1, 3 or 6 h after the start of the exposure to KA, or 30 min after the onset of ischemia. Our results indicate the neuroprotective activity of NPY and its Y2R and Y5R ligands against the kainate-induced excitotoxicity in primary cortical and hippocampal cultures. Importantly, NPY was effective when given as late as 6 h, while Y2R or Y5R agonists 3 h, after starting the exposure to KA. In in vitro studies those protective effects were inhibited by the respective receptor antagonists. Neuroprotection was also observed in vivo after intrahippocampal injection of Y2R and Y5R agonists 30 min or 1 h after KA. No protection was found either in vitro or in vivo after the Y1R agonist. The Y2R agonist also showed neuroprotective activity in the ischemic model. The obtained results indicate that neuropeptide Y produces neuroprotective effect via Y2 and Y5 receptors, and that the compounds may be effective after delayed application.


Assuntos
Fármacos Neuroprotetores/farmacologia , Receptores de Neuropeptídeo Y/agonistas , Animais , Células Cultivadas , Agonistas de Aminoácidos Excitatórios/farmacologia , Hipocampo/citologia , Ácido Caínico/farmacologia , Ligantes , Masculino , Camundongos , Neurônios/citologia , Ratos , Ratos Wistar
19.
Neuropeptides ; 43(1): 31-9, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19012964

RESUMO

Preconditioning with sublethal ischemia or hypoxia may reduce the high susceptibility of CA1 pyramidal neurons to ischemic injury. In this study, we tested the hypothesis that enhanced level of neuropeptide Y (NPY) might play a role in the mechanisms responsible for this induced tolerance. Changes in NPY immunoreactivity in the hippocampal formation of preconditioned Mongolian gerbils were compared with the level of tolerance to test ischemia. Tolerance was induced by preconditioning with 2-min of ischemia or with three trials of mild hypobaric hypoxia (360 Torr, 2 h), separated by 24 h, that were completed 48 h before the 3-min test ischemia. The number of NPY-positive neurons in the gerbil hippocampal formation was assessed 2, 4 and 7 days after preconditioning. Survival of the CA1 pyramidal neurons was examined 14 days after the insult. Our experiments demonstrated that ischemic and hypoxic preconditioning produced equal attenuation of the damage evoked by 3-min ischemia, although the pattern of NPY immunoreactivity in the hippocampus differed. Preconditioning ischemia resulted in a 20% rise in the number of NPY-positive neurons 2 days later that disappeared 4 days after the ischemic episode, while mild hypobaric hypoxia induced a twofold increase in the number of NPY-positive neurons that lasted for at least 7 days. Although induced tolerance to ischemia 2 days after ischemic or hypoxic preconditioning was accompanied by increased immunoreactivity of NPY, there was no correlation between its intensity and the level of neuroprotection.


Assuntos
Gerbillinae , Hipocampo/metabolismo , Hipóxia Encefálica/metabolismo , Precondicionamento Isquêmico , Neuropeptídeo Y/metabolismo , Animais , Sobrevivência Celular , Hipocampo/citologia , Masculino , Neurônios/citologia , Neurônios/metabolismo
20.
Neuropeptides ; 42(5-6): 513-23, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18617263

RESUMO

Corticotropin releasing factor (CRF) is a neuropeptide widely distributed in the brain. The role of CRF in the behavioural activity and modulation of anxiety states in several brain structures has been well documented, but its function in the cerebral cortex still remains unknown. The aim of our study was to investigate the effect of CRF injected bilaterally into rat frontal cortex on the locomotor and exploratory activity and anxiety of rats. We also examined the effect of CRF on extracellularly recorded field potentials in rat frontal cortical slices in vitro. Behavioural experiments showed that CRF in doses of 0.05, 0.1, 0.2 microg/1 microl/site decreased locomotor and exploratory activity during a 40-min session in the open field test. In the elevated plus-maze test, CRF in a dose of 0.2 microg/1 microl/site produced a significant anxiolytic-like effect, which was prevented by CRF receptor antagonists (alpha-helicalCRF(9-41) and NBI 27914). Electrophysiological experiments showed that CRF-induced a transient depression of field potentials in slices partly disinhibited by GABA(A) and GABA(B) receptors antagonists. The blockade of NMDA receptors prevented the occurrence of that effect. The obtained results suggest that CRF may have anxiolytic-like effects in the frontal cortex. Moreover, the peptide inhibits locomotor and exploratory activity and depresses excitatory synaptic transmission in a NMDA receptor-dependent manner.


Assuntos
Comportamento Animal/efeitos dos fármacos , Hormônio Liberador da Corticotropina/farmacologia , Lobo Frontal/efeitos dos fármacos , Atividade Motora/efeitos dos fármacos , Animais , Ansiolíticos/farmacologia , Eletrofisiologia , Lobo Frontal/fisiologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores de Hormônio Liberador da Corticotropina/agonistas , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Receptores de Hormônio Liberador da Corticotropina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...