Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
J Neuroimmune Pharmacol ; 18(3): 348-365, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37208550

RESUMO

BACKGROUND: Impairment of specific cognitive domains in schizophrenia has been associated with prefrontal cortex (PFC) catecholaminergic deficits. Among other factors, prenatal exposure to infections represents an environmental risk factor for schizophrenia development in adulthood. However, it remains largely unknown whether the prenatal infection-induced changes in the brain may be associated with concrete switches in a particular neurochemical circuit, and therefore, if they could alter behavioral functions. METHODS: In vitro and in vivo neurochemical evaluation of the PFC catecholaminergic systems was performed in offspring from mice undergoing maternal immune activation (MIA). The cognitive status was also evaluated. Prenatal viral infection was mimicked by polyriboinosinic-polyribocytidylic acid (poly(I:C)) administration to pregnant dams (7.5 mg/kg i.p., gestational day 9.5) and consequences were evaluated in adult offspring. RESULTS: MIA-treated offspring showed disrupted recognition memory in the novel object recognition task (t = 2.30, p = 0.031). This poly(I:C)-based group displayed decreased extracellular dopamine (DA) concentrations compared to controls (t = 3.17, p = 0.0068). Potassium-evoked release of DA and noradrenaline (NA) were impaired in the poly(I:C) group (DA: Ft[10,90] = 43.33, p < 0.0001; Ftr[1,90] = 1.224, p = 0.2972; Fi[10,90] = 5.916, p < 0.0001; n = 11); (NA: Ft[10,90] = 36.27, p < 0.0001; Ftr[1,90] = 1.841, p = 0.208; Fi[10,90] = 8.686, p < 0.0001; n = 11). In the same way, amphetamine-evoked release of DA and NA were also impaired in the poly(I:C) group (DA: Ft[8,328] = 22.01, p < 0.0001; Ftr[1,328] = 4.507, p = 0.040; Fi[8,328] = 2.319, p = 0.020; n = 43); (NA: Ft[8,328] = 52.07; p < 0.0001; Ftr[1,328] = 4.322; p = 0.044; Fi[8,398] = 5.727; p < 0.0001; n = 43). This catecholamine imbalance was accompanied by increased dopamine D1 and D2 receptor expression (t = 2.64, p = 0.011 and t = 3.55, p = 0.0009; respectively), whereas tyrosine hydroxylase, DA and NA tissue content, DA and NA transporter (DAT/NET) expression and function were unaltered. CONCLUSIONS: MIA induces in offspring a presynaptic catecholaminergic hypofunction in PFC with cognitive impairment. This poly(I:C)-based model reproduces catecholamine phenotypes reported in schizophrenia and represents an opportunity for the study of cognitive impairment associated to this disorder.


Assuntos
Disfunção Cognitiva , Efeitos Tardios da Exposição Pré-Natal , Gravidez , Feminino , Camundongos , Animais , Humanos , Dopamina/metabolismo , Encéfalo/metabolismo , Córtex Pré-Frontal/metabolismo , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/metabolismo , Anfetamina , Norepinefrina/metabolismo , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Poli I-C/toxicidade , Modelos Animais de Doenças
2.
Neuropsychopharmacology ; 48(2): 341-350, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36088492

RESUMO

Peripheral inputs continuously shape brain function and can influence memory acquisition, but the underlying mechanisms have not been fully understood. Cannabinoid type-1 receptor (CB1R) is a well-recognized player in memory performance, and its systemic modulation significantly influences memory function. By assessing low arousal/non-emotional recognition memory in mice, we found a relevant role of peripheral CB1R in memory persistence. Indeed, the peripherally-restricted CB1R specific antagonist AM6545 showed significant mnemonic effects that were occluded in adrenalectomized mice, and after peripheral adrenergic blockade. AM6545 also transiently impaired contextual fear memory extinction. Vagus nerve chemogenetic inhibition reduced AM6545-induced mnemonic effect. Genetic CB1R deletion in dopamine ß-hydroxylase-expressing cells enhanced recognition memory persistence. These observations support a role of peripheral CB1R modulating adrenergic tone relevant for cognition. Furthermore, AM6545 acutely improved brain connectivity and enhanced extracellular hippocampal norepinephrine. In agreement, intra-hippocampal ß-adrenergic blockade prevented AM6545 mnemonic effects. Altogether, we disclose a novel CB1R-dependent peripheral mechanism with implications relevant for lengthening the duration of non-emotional memory.


Assuntos
Norepinefrina , Receptor CB1 de Canabinoide , Animais , Camundongos , Adrenérgicos/farmacologia , Encéfalo , Hipocampo , Norepinefrina/farmacologia , Receptor CB1 de Canabinoide/antagonistas & inibidores
3.
Front Pharmacol ; 12: 682602, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34054556

RESUMO

Emerging evidence indicates that early-life exposure to environmental factors may increase the risk for schizophrenia via inflammatory mechanisms. Inflammation can alter the metabolism of tryptophan through the oxidative kynurenine pathway to compounds with neurotoxic and neuroprotective activity and compromise serotonin (5-HT) synthesis. Here we investigate the role of serotonergic and kynurenine pathways in the maternal immune activation (MIA) animal model of schizophrenia. The potential reversion exerted by long-term antipsychotic treatment was also evaluated. MIA was induced by prenatal administration of polyinosinic:polycytidylic acid (poly (I:C)) in mice. Expression of different proteins and the content of different metabolites involved in the function of serotonergic and kynurenine pathways was assessed by RT-PCR, immunoblot and ELISA analyses in frontal cortex of the offspring after puberty. MIA decreased tissue 5-HT content and promoted changes in the expression of serotonin transporter, 5-HT2A and 5-HT2C receptors. Expression of indoleamine 2,3-dioxygenase 2 (IDO2) and kynurenine 3-monooxygenase (KMO) was increased by poly (I:C) whereas kynurenine aminotransferase II and its metabolite kynurenic acid were not altered. Long-term paliperidone was able to counteract MIA-induced changes in 5-HT and KMO, and to increase tryptophan availability and tryptophan hydroxylase-2 expression in poly (I:C) mice but not in controls. MIA-induced increase of the cytotoxic risk ratio of kynurenine metabolites (quinolinic/kynurenic acid) was also reversed by paliperidone. MIA induces specific long-term brain effects on serotonergic activity. Such effects seem to be related with alternative activation of the kynurenine metabolic pathway towards a cytotoxic status. Atypical antipsychotic paliperodine partially remediates abnormalities observed after MIA.

4.
Neuropharmacology ; 158: 107731, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31376424

RESUMO

Disruption of the hypothalamic-pituitary-adrenal axis is an established finding in patients with anxiety and/or depression. Chronic corticosterone administration in animals has been proposed as a model for the study of these stress-related disorders and the antidepressant action. Alterations of the central noradrenergic system and specifically of inhibitory α2-adrenoceptors seem to be part of the pathophysiology of depression and contribute to the antidepressant activity. The present study evaluates in male rats the effect of chronic corticosterone treatment during 35 days (16-20 mg kg-1 day-1) on the sensitivity of α2-adrenoceptors expressed in the somatodendritic and terminal noradrenergic areas locus coeruleus (LC) and prefrontal cortex (PFC), respectively. Further, the effect of chronic fluoxetine treatment (5 mg kg-1, i.p., since the 15th day) on the sensitivity of α2-adrenoceptors was examined under control conditions and in corticosterone-treated rats. The α2-adrenoceptor functionality was analysed in vitro by agonist-mediated [35S]GTPγS binding stimulation and in vivo through the modulation of noradrenaline (NA) release evaluated by dual-probe microdialysis. The concentration-effect curves of the [35S]GTPγS binding stimulation by the agonist UK14304 (5-bromo-N-(4,5-dihydro-1H-imidazol-2-yl)-6-quinoxalinamine) demonstrated a desensitization of cortical α2-adrenoceptors induced by corticosterone (-logEC50 = 6.7 ±â€¯0.2 vs 8.2 ±â€¯0.3 in controls) that was reverted by fluoxetine treatment (-logEC50 = 7.5 ±â€¯0.3). Local administration of the α2-adrenoceptor antagonist RS79948 ((8aR,12aS,13aS)-5,8,8a,9,10,11,12,12a,13,13a-decahydro-3-methoxy-12-(ethylsulfonyl)-6H-isoquino[2,1-g][1,6]naphthyridine) (0.1-100 µmol L-1) into the LC induced a concentration-dependent NA increase in the PFC of the control group (Emax = 191 ±â€¯30%) but non-significant effect was observed in corticosterone-treated rats (Emax = 133 ±â€¯46%), reflecting a desensitization of α2-adrenoceptors that control the firing of noradrenergic neurons. Fluoxetine treatment did not alter the corticosterone-induced desensitization in this area (Emax = 136 ±â€¯19%). No effect of fluoxetine on α2-adrenoceptor functionality was observed in control animals (Emax = 223 ±â€¯30%). In PFC, the local administration of RS79948 increased NA in controls (Emax = 226 ±â€¯27%) without effect in the corticosterone group (Emax = 115 ±â€¯26%), suggesting a corticosterone-induced desensitization of terminal α2-adrenoceptors. Fluoxetine administration prevented the desensitization induced by corticosterone in the PFC (Emax = 233 ±â€¯33%) whereas desensitized α2-adrenoceptors in control animals (Emax = -24 ±â€¯10%). These data indicate that chronic corticosterone increases noradrenergic activity by acting at different α2-adrenoceptor subpopulations. Treatment with the antidepressant fluoxetine seems to counteract these changes by acting mainly on presynaptic α2-adrenoceptors expressed in terminal areas.


Assuntos
Neurônios Adrenérgicos/efeitos dos fármacos , Antidepressivos de Segunda Geração/farmacologia , Corticosterona/farmacologia , Fluoxetina/farmacologia , Locus Cerúleo/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Receptores Adrenérgicos alfa 2/efeitos dos fármacos , Neurônios Adrenérgicos/metabolismo , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Antagonistas de Receptores Adrenérgicos alfa 2/farmacologia , Animais , Tartarato de Brimonidina/farmacologia , Corpo Celular/efeitos dos fármacos , Corpo Celular/metabolismo , Dendritos/efeitos dos fármacos , Dendritos/metabolismo , Modelos Animais de Doenças , Guanosina 5'-O-(3-Tiotrifosfato) , Sistema Hipotálamo-Hipofisário/metabolismo , Técnicas In Vitro , Isoquinolinas/farmacologia , Locus Cerúleo/metabolismo , Masculino , Microdiálise , Naftiridinas/farmacologia , Norepinefrina/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Córtex Pré-Frontal/metabolismo , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/metabolismo , Ratos , Receptores Adrenérgicos alfa 2/metabolismo , Estresse Psicológico/metabolismo , Radioisótopos de Enxofre
5.
Mol Neurobiol ; 56(4): 3038-3052, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30088175

RESUMO

Current pharmacological treatments for major depressive disorder (MDD) are severely compromised by both slow action and limited efficacy. RNAi strategies have been used to evoke antidepressant-like effects faster than classical drugs. Using small interfering RNA (siRNA), we herein show that TASK3 potassium channel knockdown in monoamine neurons induces antidepressant-like responses in mice. TASK3-siRNAs were conjugated to cell-specific ligands, sertraline (Ser) or reboxetine (Reb), to promote their selective accumulation in serotonin (5-HT) and norepinephrine (NE) neurons, respectively, after intranasal delivery. Following neuronal internalization of conjugated TASK3-siRNAs, reduced TASK3 mRNA and protein levels were found in the brainstem 5-HT and NE cell groups. Moreover, Ser-TASK3-siRNA induced robust antidepressant-like behaviors, enhanced the hippocampal plasticity, and potentiated the fluoxetine-induced increase on extracellular 5-HT. Similar responses, yet of lower magnitude, were detected for Reb-TASK3-siRNA. These findings provide substantial support for TASK3 as a potential target, and RNAi-based strategies as a novel therapeutic approach to treat MDD.


Assuntos
Depressão/tratamento farmacológico , Técnicas de Silenciamento de Genes , Neurônios/metabolismo , Canais de Potássio/metabolismo , Reboxetina/administração & dosagem , Sertralina/administração & dosagem , Administração Intranasal , Animais , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico , Comportamento Animal , Regulação para Baixo/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Norepinefrina/metabolismo , RNA Interferente Pequeno/metabolismo , Reboxetina/farmacologia , Neurônios Serotoninérgicos/efeitos dos fármacos , Neurônios Serotoninérgicos/metabolismo , Serotonina/metabolismo , Sertralina/farmacologia
6.
Neuropharmacology ; 133: 491-502, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29477299

RESUMO

Activation of serotonin 5-HT3 receptor (5HT3R) in the locus coeruleus (LC), the principal somatodendritic noradrenergic area, decreases LC firing activity and noradrenaline (NA) release in prefrontal cortex (PFC). Blockade of 5HT3R in coadministration with selective serotonin reuptake inhibitors (SSRIs) has been proposed as a potential strategy to accelerate the onset of action of SSRIs. Dual-probe microdialysis in rats was used to evaluate the involvement of 5HT3R in the in vivo effect exerted by the SSRI citalopram on NA release. Besides, forced swimming test (FST) was carried out in mice to evaluate the antidepressant-like effect of citalopram in combination with a 5HT3R antagonist (Y25130). Systemic administration of the 5HT3R agonist SR57227 (10 mg/kg i.p.) increased NA in LC (Emax = 200 ±â€¯27%) and PFC (Emax = 133 ±â€¯2%). The increase in PFC was enhanced in local presence into LC of Y25130 (50 µM) (Emax = 296 ±â€¯41%) suggesting an inhibitory function on NA release exerted by the activation of 5HT3R located in somatodendritic areas. Citalopram administration (10 mg/kg i.p.) increased NA in LC (Emax = 185 ±â€¯11%) and decreased it in PFC (Emax = -35 ±â€¯7%). Intra-LC (50 µM) or systemic co-administration of Y25130 (10 mg/kg i.p.) with citalopram (10 mg/kg i.p.) switched NA release in the PFC from an inhibition to a stimulatory effect. In mice FST, systemic coadministration of citalopram (2.5 mg/kg i.p.) and Y25130 (10 mg/kg i.p.) potentiated the decrease of immobility time through the increase of both swimming and climbing behaviours. These results suggest that the addition of a 5HT3R antagonist to SSRIs could represent a feasible strategy to improve antidepressant response.


Assuntos
Antidepressivos/uso terapêutico , Citalopram/uso terapêutico , Depressão/tratamento farmacológico , Córtex Pré-Frontal/efeitos dos fármacos , Receptores 5-HT3 de Serotonina/metabolismo , Antagonistas da Serotonina/uso terapêutico , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Comportamento Exploratório/efeitos dos fármacos , Locus Cerúleo/efeitos dos fármacos , Locus Cerúleo/metabolismo , Masculino , Camundongos , Microdiálise , Norepinefrina/metabolismo , Piperidinas/farmacologia , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Sprague-Dawley , Agonistas do Receptor de Serotonina/uso terapêutico , Natação
7.
Neuropsychopharmacology ; 43(5): 1041-1051, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28905875

RESUMO

The importance of dopamine (DA) neurotransmission is emphasized by its direct implication in several neurological and psychiatric disorders. The DA transporter (DAT), target of psychostimulant drugs, is the key protein that regulates spatial and temporal activity of DA in the synaptic cleft via the rapid reuptake of DA into the presynaptic terminal. There is strong evidence suggesting that DAT-interacting proteins may have a role in its function and regulation. Performing a two-hybrid screening, we identified snapin, a SNARE-associated protein implicated in synaptic transmission, as a new binding partner of the carboxyl terminal of DAT. Our data show that snapin is a direct partner and regulator of DAT. First, we determined the domains required for this interaction in both proteins and characterized the DAT-snapin interface by generating a 3D model. Using different approaches, we demonstrated that (i) snapin is expressed in vivo in dopaminergic neurons along with DAT; (ii) both proteins colocalize in cultured cells and brain and, (iii) DAT and snapin are present in the same protein complex. Moreover, by functional studies we showed that snapin produces a significant decrease in DAT uptake activity. Finally, snapin downregulation in mice produces an increase in DAT levels and transport activity, hence increasing DA concentration and locomotor response to amphetamine. In conclusion, snapin/DAT interaction represents a direct link between exocytotic and reuptake mechanisms and is a potential target for DA transmission modulation.


Assuntos
Anfetamina/farmacologia , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animais , Sítios de Ligação/efeitos dos fármacos , Encéfalo/metabolismo , Células Cultivadas , Neurônios Dopaminérgicos/metabolismo , Regulação para Baixo , Camundongos , Modelos Moleculares , Atividade Motora/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Ratos , Proteínas de Transporte Vesicular/biossíntese
8.
Neuropharmacology ; 114: 114-122, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-27908769

RESUMO

Selective serotonin reuptake inhibitors (SSRIs) regulate brain noradrenergic neurotransmission both at somatodendritic and nerve terminal areas. Previous studies have demonstrated that noradrenaline (NA) reuptake inhibitors are able to desensitize α2-adrenoceptor-mediated responses. The present study was undertaken to elucidate the effects of repeated treatment with the SSRI citalopram on the α2-adrenoceptor sensitivity in locus coeruleus (LC) and prefrontal cortex (PFC), by using in vivo microdialysis and electrophysiological techniques, and in vitro stimulation of [35S]GTPγS binding autoradiography. Repeated, but not acute, treatment with citalopram (5 mg/kg, i.p., 14 days) increased extracellular NA concentration selectively in PFC. The α2-adrenoceptor agonist clonidine (0.3 mg/kg, i.p.), administered to saline-treated animals (1 ml/kg i.p., 14 days) induced NA decrease in LC (Emax = -44 ± 4%; p < 0.001) and in PFC (Emax = -61 ± 5%, p < 0.001). In citalopram chronically-treated rats, clonidine administration exerted a lower decrease of NA (Emax = -25 ± 7%; p < 0.001) in PFC whereas the effect in LC was not different to controls (Emax = -36 ± 4%). Clonidine administration (0.625-20 µg/kg, i.v.) evoked a dose-dependent decrease of the firing activity of LC noradrenergic neurons in both citalopram- (ED50 = 3.2 ± 0.4 µg/kg) and saline-treated groups (ED50 = 2.6 ± 0.5 µg/kg). No significant differences between groups were found in ED50 values. The α2-adrenoceptor agonist UK14304 stimulated specific [35S]GTPγS binding in brain sections containing LC (144 ± 14%) and PFC (194 ± 32%) of saline-treated animals. In citalopram-treated animals, this increase did not differ from controls in LC (146 ± 22%) but was lower in PFC (141 ± 8%; p < 0.05). Taken together, long-term citalopram treatment induces a desensitization of α2-adrenoceptors acting as axon terminal autoreceptors in PFC without changes in somatodendritic α2-adrenoceptor sensitivity.


Assuntos
Citalopram/administração & dosagem , Locus Cerúleo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Receptores Adrenérgicos alfa 2/fisiologia , Inibidores Seletivos de Recaptação de Serotonina/administração & dosagem , Neurônios Adrenérgicos/efeitos dos fármacos , Neurônios Adrenérgicos/fisiologia , Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Tartarato de Brimonidina/administração & dosagem , Citalopram/farmacocinética , Locus Cerúleo/metabolismo , Locus Cerúleo/fisiologia , Masculino , Neurônios/fisiologia , Norepinefrina/metabolismo , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/fisiologia , Ratos , Ratos Sprague-Dawley , Inibidores Seletivos de Recaptação de Serotonina/farmacocinética
9.
Psychopharmacology (Berl) ; 233(23-24): 3861-3867, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27592231

RESUMO

RATIONALE: Noradrenergic system plays a critical role in the hypothalamic-pituitary-adrenal (HPA) axis regulation and the stress response. A dysregulated HPA axis may be indicative of an increased biological vulnerability for depression. In addition, a variety of studies have focused on specific alterations of α2-adrenoceptors as a mechanism involved in the pathogenesis of mood disorders and antidepressant response. OBJECTIVES: This study aimed to evaluate the effect of subchronic corticosterone administration on rat brain α2-adrenoceptor functionality by in vitro [35S]GTPγS binding stimulation assays and in vivo dual-probe microdialysis determination of extracellular noradrenaline concentrations. RESULTS: Implantation of a time release corticosterone pellet during 14 days induced sustained changes in endocrine function. However, there were no differences in α2-adrenoceptor agonist UK14304-induced stimulation of [35S]GTPγS binding in prefrontal cortex (PFC) between corticosterone-treated and control rats. In the same way, the in vivo evaluation of α2-adrenoceptor-mediated noradrenaline release responses to the α2-adrenoceptor agonist clonidine local administration into the locus coeruleus (LC), and the PFC did not show differences between the groups. CONCLUSIONS: The present results show that subchronic corticosterone administration does not induce changes on functionality of α2-adrenoceptors neither in the LC nor in noradrenergic cortical terminal areas.


Assuntos
Anti-Inflamatórios/farmacologia , Corticosterona/farmacologia , Receptores Adrenérgicos/efeitos dos fármacos , Animais , Clonidina/farmacologia , Corticosterona/sangue , Modelos Animais de Doenças , Sistema Hipotálamo-Hipofisário/metabolismo , Locus Cerúleo/efeitos dos fármacos , Masculino , Microdiálise , Norepinefrina/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Sprague-Dawley
10.
J Infect Dev Ctries ; 10(5): 512-7, 2016 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-27249527

RESUMO

INTRODUCTION: Liver cirrhosis (LC) constitutes one of the main 10 causes of death worldwide. LC has a characteristic asymptomatic compensated phase followed by a progressive decompensated phase, in which diverse complications are presented. LC patients are highly prone to bacterial infections. The neutrophils in these patients present defects in the production of oxygen radicals, which are essential for bacteria elimination as in the activation of neutrophil extracellular traps (NETs). The main objective of this work was to determine the NETs and neutrophil activation markers in LC patients. METHODOLOGY: Neutrophil purification was done with Ficoll Histopaque from a sample of the peripheral blood of patients with compensated and decompensated LC. Neutrophils were activated with Phorbol 12-myristate 13-acetate to evaluate the release of NETs by means of fluorescence microscopy and fluorometry, while expression of activation markers (CD69, CD80, perforin, and CAP-18) was evaluated by flow cytometry. RESULTS: A significant decrease in the release capability of NETs was observed as the level of LC in the patient increased. When comparing serum levels in inflammatory cytokines among the different study groups, significant differences were observed. No significant differences were detected on neutrophil activation markers; nevertheless, there was a correlation between diminution of CD69 and CD80 expression in decompensated patients. CONCLUSIONS: We demonstrated that LC patients with neutrophil extracellular trap release deficiencies could have an increased rate of complications.


Assuntos
Armadilhas Extracelulares/metabolismo , Cirrose Hepática/imunologia , Cirrose Hepática/patologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Citometria de Fluxo , Fluorometria , Humanos , Microscopia de Fluorescência
12.
PLoS One ; 10(3): e0119717, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25768308

RESUMO

Vitronectin, a multifunctional glycoprotein, is involved in coagulation, inhibition of the formation of the membrane attack complex (MAC), cell adhesion and migration, wound healing, and tissue remodeling. The primary cellular source of vitronectin is hepatocytes; it is not known whether resident cells of airways produce vitronectin, even though the glycoprotein has been found in exhaled breath condensate and bronchoalveolar lavage from healthy subjects and patients with interstitial lung disease. It is also not known whether vitronectin expression is altered in subjects with asthma and COPD. In this study, bronchial tissue from 7 asthmatic, 10 COPD and 14 control subjects was obtained at autopsy and analyzed by immunohistochemistry to determine the percent area of submucosal glands occupied by vitronectin. In a separate set of experiments, quantitative colocalization analysis was performed on tracheobronchial tissue sections obtained from donor lungs (6 asthmatics, 4 COPD and 7 controls). Vitronectin RNA and protein expressions in bronchial surface epithelium were examined in 12 subjects who undertook diagnostic bronchoscopy. Vitronectin was found in the tracheobronchial epithelium from asthmatic, COPD, and control subjects, although its expression was significantly lower in the asthmatic group. Colocalization analysis of 3D confocal images indicates that vitronectin is expressed in the glandular serous epithelial cells and in respiratory surface epithelial cells other than goblet cells. Expression of the 65-kDa vitronectin isoform was lower in bronchial surface epithelium from the diseased subjects. The cause for the decreased vitronectin expression in asthma is not clear, however, the reduced concentration of vitronectin in the epithelial/submucosal layer of airways may be linked to airway remodeling.


Assuntos
Asma/metabolismo , Brônquios/metabolismo , Regulação da Expressão Gênica , Doença Pulmonar Obstrutiva Crônica/metabolismo , Vitronectina/metabolismo , Adulto , Idoso , Asma/genética , Asma/patologia , Brônquios/patologia , Estudos de Casos e Controles , Células Epiteliais/metabolismo , Glândulas Exócrinas/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Doença Pulmonar Obstrutiva Crônica/genética , Doença Pulmonar Obstrutiva Crônica/patologia , Vitronectina/genética , Adulto Jovem
13.
J Med Chem ; 58(2): 963-77, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25490760

RESUMO

We have previously identified phenylguanidine and phenyl-2-aminoimidazoline compounds as high affinity ligands with conflicting functional activity at the α2-adrenoceptor, a G-protein-coupled receptor with relevance in several neuropsychiatric conditions. In this paper we describe the design, synthesis, and pharmacological evaluation of a new series of pyridine derivatives [para substituted 2- and 3-guanidino and 2- and 3-(2-aminoimidazolino)pyridines, disubstituted 2-guanidinopyridines and N-substituted-2-amino-1,4-dihydroquinazolines] that were found to be antagonists/inverse agonists of the α2-adrenoceptor. Furthermore, the compounds exert their effects at the α2-adrenoceptor both in vitro in human prefrontal cortex tissue and in vivo in rat brain as shown by microdialysis experiments. We also provide a docking study at the α2A- and α2C-adrenoceptor subtypes demonstrating the structural features required for high affinity binding to the receptor.


Assuntos
Antagonistas de Receptores Adrenérgicos alfa 2/síntese química , Antagonistas de Receptores Adrenérgicos alfa 2/química , Antagonistas de Receptores Adrenérgicos alfa 2/farmacologia , Animais , Guanidinas/síntese química , Guanidinas/farmacologia , Humanos , Imidazolinas/síntese química , Imidazolinas/farmacologia , Masculino , Modelos Moleculares , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade
14.
J Inflamm (Lond) ; 11(1): 39, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25493077

RESUMO

INTRODUCTION: Obesity is the world's most important public health problem. Adipose tissue contributes significantly to increase pro-inflammatory mediators whose cascade begins with the union of TLR4 to its microbial ligands (TLR: Toll Like Receptors). It has been reported recently that NEFAs (Non-Esterified Fatty Acids) bind to this receptor as agonists. The purpose of our study was to determine the levels of expression of TLR4-CD14, the pro-inflammatory cytokines, the metabolic markers and the NEFAs in a group of adult, non-diabetic obese Mexicans. METHOD: A group of 210 adult middle-class Mexican non-diabetic obese patients was evaluated: 105 normal weight individuals, and 105 non-diabetic obese. On both groups, the following was tested in each patient: TLR4-CD14 receptors on monocytes in peripheral blood, inflammatory profile, HOMA-IR (Homeostasis Model Assessment-Insulin Resistance), NEFAs and each individual's anthropometric profile. RESULTS: Obesity is strongly associated with the expression of TLR4 (77%, MFI (Mean Fluorescence Index) 7.70) and CD14 (86% MFI 1.61) with 66% double positives (p = 0.000). These figures contrast with those for the normal weight individuals that constituted the control group: TLR4 (70% MFI 6.41) and CD14 (84% MFI 1.25) with 59% double positives. As for cytokine concentration, non- diabetic obese individuals vs the normal weight/thin, the numbers were: IL-1ß = 2.0 vs 2.5 pg/ml (p = NS), IL-6 = 36 vs 28 pg/ml (p = 0.030), IL-8 = 27 vs 27 pg/ml (p = NS), IL-10 = 8.4 vs 6.8 pg/ml (p = NS), TNF-α =31 vs 15 pg/ml (p = 0.000) respectively. Insulin levels were 12.1 vs 19.7 mcU/ml (p = 0.000) and the NEFAs were much higher in the obese vs normal weight/thin individuals (p = 0.000). CONCLUSION: Adipose tissue used to be thought of as mere storage of fats and energy, but it has been revealed to be an important neuro-immune-endocrine organ. Immune cells, stimulated by NEFAs, produce pro-inflammatory cytokines, which have a direct effect on oxidating radicals that directly target the release of noradrenalin. This in turn, reactivates the vicious cycle of low-grade chronic inflammation, as is now described in obesity.

15.
J Neuroinflammation ; 11: 128, 2014 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-25069615

RESUMO

BACKGROUND: An accumulating body of evidence points to the significance of neuroinflammation and immunogenetics in schizophrenia, and an imbalance of cytokines in the central nervous system (CNS) has been suggested to be associated with the disorder. Munc18-overexpressing mice (Munc18-OE) have provided a model for the study of the alterations that may underlie the symptoms of subjects with schizophrenia. The aim of the present study was to elucidate the involvement of neuroinflammation and cytokine imbalance in this model. METHODS: Cytokines were evaluated in the cortex and the striatum of Munc18-OE and wild-type (WT) mice by enzyme-linked immunosorbent assay (ELISA). Protein levels of specific microglia and macrophage, astrocytic and neuroinflammation markers were quantified by western blot in the cortex and the striatum of Munc18-OE and WT mice. RESULTS: Each cytokine evaluated (Interferon-gamma (IFN-γ), Tumor Necrosis Factor-alpha (TNF-α), Interleukin-2 (IL-2) and CCL2 chemokine) was present at higher levels in the striatum of Munc18-OE mice than WT. Cortical TNF-α and IL-2 levels were significantly lower in Munc18-OE mice than WT mice. The microglia and macrophage marker CD11b was lower in the cortexes of Munc18-OE mice than WT, but no differences were observed in the striatum. Glial Fibrillary Acidic Protein (GFAP) and Nuclear Factor-kappaB (NF-κB)p65 levels were not different between the groups. Interleukin-1beta (IL-1ß) and IL-6 levels were beneath detection limits. CONCLUSIONS: The disrupted levels of cytokines detected in the brain of Munc18-OE mice was found to be similar to clinical reports and endorses study of this type for analysis of this aspect of the disorder. The lower CD11b expression in the cortex but not in the striatum of the Munc18-OE mice may reflect differences in physiological activity. The cytokine expression pattern observed in Munc18-OE mice is similar to a previously published model of schizophrenia caused by maternal immune activation. Together, these data suggest a possible role for an immune imbalance in this disorder.


Assuntos
Citocinas/metabolismo , Proteínas Munc18/metabolismo , Esquizofrenia/metabolismo , Transdução de Sinais/fisiologia , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Expressão Gênica , Proteína Glial Fibrilar Ácida , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Munc18/genética , NF-kappa B/metabolismo , Esquizofrenia/genética , Esquizofrenia/patologia , Transdução de Sinais/efeitos dos fármacos
16.
J Interferon Cytokine Res ; 33(12): 717-27, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23869900

RESUMO

Obesity is a world health problem that increases the risk for developing type 2 diabetes, cardiovascular disease, fatty liver, and some types of cancer. In postmenopausal women, it represents an important risk factor for the development of breast cancer (BC). Leptin is an adipokine that is secreted by fatty tissue, and high leptin levels are observed both in mouse models of obesity and in obese subjects. High levels of leptin promote the proliferation and progression of various types of cancer, including BC. This review provides a general overview of the biology of leptin, important laboratory studies, and animal and clinical models that have provided evidence for an active role of leptin in the proliferation, progression, and survival of mammary tumors. Finally, this review addresses the most recent studies on the use of leptin receptor antagonists as a novel therapeutic treatment for BC.


Assuntos
Neoplasias da Mama/metabolismo , Leptina/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/terapia , Modelos Animais de Doenças , Metabolismo Energético , Feminino , Homeostase , Humanos , Leptina/genética , Camundongos , Terapia de Alvo Molecular , Receptores para Leptina/antagonistas & inibidores , Receptores para Leptina/química , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Fatores de Risco , Transdução de Sinais
17.
Psychopharmacology (Berl) ; 229(2): 331-44, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23636303

RESUMO

RATIONALE: Selective serotonin reuptake inhibitors (SSRIs), in addition to being able to enhance serotonergic neurotransmission, are able to modulate other brain systems involved in depression. OBJECTIVES: This study evaluates the neurochemical effect of the SSRI citalopram on brain noradrenergic activity and the serotonin receptor involved in this effect. METHODS: Dual-probe microdialysis in the locus coeruleus (LC) and prefrontal cortex (PFC) was performed in freely awake rats. RESULTS: Systemic citalopram (10 mg/kg, i.p.) increased noradrenaline (NA) in the LC (E max = 141 ± 13%) and simultaneously decreased NA in the PFC (Emax = -46 ± 7%). In the local presence into the LC of the α2-adrenoceptor antagonist RS79948 (1 µM), systemic citalopram increased NA in the LC (Emax = 157 ± 25%) and PFC (Emax = 175 ± 24%). Local citalopram (0.1-100 µM) into the LC induced NA increase in the LC (Emax = 210 ± 25%) and decrease in the PFC (Emax = -38 ± 9%). Local LC citalopram effect was abolished by LC presence of the 5-HT3 receptor antagonist MDL72222 (1 µM) but not the 5-HT1/2 receptor antagonist methiothepin (1 µM). Systemic citalopram in the LC presence of MDL72222 did not modify NA in the LC but increased NA in the PFC (Emax = 158 ± 26%). Local citalopram into the PFC enhanced NA (Emax = 376 ± 18%) in the area, which was prevented by MDL72222. CONCLUSIONS: The SSRI citalopram modulates central noradrenergic neurotransmission by activation, through endogenous serotonin, of 5-HT3 receptors expressed in the somatodendritic (LC) and terminal (PFC) areas, which subsequently promote an enhancement of local NA. Therefore, 5-HT3 receptors and somatodendritic α2-adrenoceptors in the LC play an important role in the global effect of SSRIs.


Assuntos
Locus Cerúleo/metabolismo , Norepinefrina/metabolismo , Córtex Pré-Frontal/metabolismo , Receptores 5-HT3 de Serotonina/metabolismo , Análise de Variância , Animais , Área Sob a Curva , Cromatografia Líquida de Alta Pressão , Relação Dose-Resposta a Droga , Interações Medicamentosas , Eletroquímica , Isoquinolinas/farmacologia , Locus Cerúleo/efeitos dos fármacos , Masculino , Microdiálise , Naftiridinas/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Serotoninérgicos/farmacologia , Vigília
18.
Biol Psychiatry ; 73(1): 54-62, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22854119

RESUMO

BACKGROUND: Patients suffering chronic pain are at high risk of suffering long-lasting emotional disturbances characterized by persistent low mood and anxiety. We propose that this might be the result of a functional impairment in noradrenergic circuits associated with locus coeruleus (LC) and prefrontal cortex, where emotional and sensorial pain processes overlap. METHODS: We used a chronic constriction injury of sciatic nerve as a model of neuropathic pain in male Sprague-Dawley rats to assess the time-dependent changes that might potentially precipitate mood disorders (2, 7, 14, and 28 days after injury). This was measured through a combination of behavioral, electrophysiological, microdialysis, immunohistochemical, and Western blot assays. RESULTS: As expected, nerve injury produced an early and stable decrease in sensorial pain threshold over the testing period. By contrast, long-term neuropathic pain (28 days after injury) resulted in an inability to cope with stressful situations, provoking depressive and anxiogenic-like behaviors, even more intense than the aversiveness associated with pain perception. The onset of these behavioral changes coincided with irruption of noradrenergic dysfunction, evident as: an increase in LC bursting activity; in tyrosine hydroxylase expression and that of the noradrenaline transporter; and enhanced expression and sensitivity of α2-adrenoceptors in the LC. CONCLUSIONS: Long-term neuropathic pain leads to anxio-depressive-like behaviors that are more predominant than the aversion of a painful experience. These changes are consistent with the impairment of noradrenergic system described in depressive disorders.


Assuntos
Neurônios Adrenérgicos/fisiologia , Dor Crônica/fisiopatologia , Dor Crônica/psicologia , Transtornos do Humor/fisiopatologia , Transtornos do Humor/psicologia , Potenciais de Ação/fisiologia , Neurônios Adrenérgicos/metabolismo , Animais , Comportamento Animal/fisiologia , Dor Crônica/complicações , Modelos Animais de Doenças , Líquido Extracelular/metabolismo , Locus Cerúleo/metabolismo , Locus Cerúleo/fisiologia , Masculino , Transtornos do Humor/complicações , Norepinefrina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/biossíntese , Medição da Dor/métodos , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos alfa 2/fisiologia , Tirosina 3-Mono-Oxigenase/biossíntese
19.
Neuropharmacology ; 62(8): 2472-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22401957

RESUMO

A functional interaction between serotonergic and noradrenergic systems has been shown in the locus coeruleus (LC). Noradrenaline (NA) levels in the prefrontal cortex (PFC) are dependent on the firing rate of LC neurons, which is controlled by α(2) adrenoceptors (α2ADR). The aim of the present study was to investigate the role of 5-HT(3) receptors (5HT3R) in the modulation of central noradrenergic activity. We measured extracellular NA concentrations in the LC and PFC by dual-probe microdialysis in awake rats and the firing rate of LC neurons by electrophysiological techniques in vitro. Administration of the 5HT3R agonists SR57227 (1-100 µM) and m-chlorophenylbiguanide (mCPBG, 1-100 µM) into the LC increased NA in this nucleus (E(max) = 675 ± 121% and E(max) = 5575 ± 1371%, respectively) and decreased NA in the PFC (E(max) = -49 ± 6% and E(max) = -25 ± 11%, respectively). Administration of the 5HT3R antagonist Y25130 (50 µM) into LC attenuated SR57227 effect in the LC (E(max) = 323 ± 28%) and PFC (E(max) = -37 ± 7%). The α2ADR antagonist RS79948 (1 µM) blocked the SR57227 effect in the PFC but it did not change the effect in the LC (E(max) = 677 ± 202%). In electrophysiological assays, both mCPBG (1-10 µM) and SR57227 (1-10 µM) reduced the firing rate of about 50% of tested LC neurons (maximal effect = -37 ± 2% and -31 ± 4%, respectively); this effect was partially blocked by Y25130 (50 µM). Administration of RS79948 (1 µM) reversed the inhibition induced by mCPBG. Competition radioligand assays against [(3)H]UK14304 and [(3)H]RX821002 (α2ADR selective drugs) in the rat brain cortex showed a very weak affinity of SR57227 for α2ADR, whereas the affinity of mCPBG for α2ADR was 17-fold higher than that of SR57227 for α2ADR. The present results suggest that 5HT3R stimulate NA release in the LC, which promotes simultaneously a decrease in the firing rate of LC neurons through α2ADR and then a decrease of NA release in terminal areas such as the PFC.


Assuntos
Locus Cerúleo/metabolismo , Neurônios/metabolismo , Norepinefrina/metabolismo , Receptores 5-HT3 de Serotonina/metabolismo , Antagonistas Adrenérgicos alfa/farmacologia , Animais , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Relação Dose-Resposta a Droga , Isoquinolinas/farmacologia , Locus Cerúleo/efeitos dos fármacos , Masculino , Microdiálise , Naftiridinas/farmacologia , Neurônios/efeitos dos fármacos , Oxazinas/farmacologia , Ratos , Ratos Sprague-Dawley , Antagonistas da Serotonina/farmacologia , Agonistas do Receptor de Serotonina/farmacologia
20.
Neuropharmacology ; 62(5-6): 1944-53, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22245561

RESUMO

The objective of the present work was to study the effects of an early-life stress (maternal separation, MS) in the excitatory/inhibitory ratio as a potential factor contributing to the ageing process, and the purported normalizing effects of chronic treatment with the antidepressant venlafaxine. MS induced depressive-like behaviour in the Porsolt forced swimming test that was reversed by venlafaxine, and that persisted until senescence. Aged MS rats showed a downregulation of vesicular glutamate transporter 1 and 2 (VGlut1 and VGlut2) and GABA transporter (VGAT) and increased expression of excitatory amino acid transporter 2 (EAAT2) in the hippocampus. Aged rats showed decreased expression of glutamic acid decarboxylase 65 (GAD65), while the excitatory amino acid transporter 1 (EAAT1) was affected only by stress. Glutamate receptor subunits NR1 and NR2A and GluR4 were upregulated in stressed rats, and this effect was reversed by venlafaxine. NR2B, GluR1 and GluR2/3 were not affected by either stress or age. MS, both in young and aged rats, induced an increase in the circulating levels of corticosterone. Corticosterone induced an increase glutamate and a decrease in GABA release in hippocampal slices, which was reversed by venlafaxine. Chronic treatment with corticosterone recapitulated the main biochemical findings observed in MS. The different effects that chronic stress exerts in young and adult animals on expression of proteins responsible for glutamate/GABA cycling may explain the involvement of glucocorticoids in ageing-related diseases. Modulation of glutamate/GABA release may be a relevant component of the therapeutic action of antidepressants, such as venlafaxine.


Assuntos
Depressão/metabolismo , Ácido Glutâmico/metabolismo , Hipocampo/metabolismo , Privação Materna , Rede Nervosa/metabolismo , Neurônios/metabolismo , Ácido gama-Aminobutírico/metabolismo , Fatores Etários , Animais , Antidepressivos de Segunda Geração/farmacologia , Antidepressivos de Segunda Geração/uso terapêutico , Corticosterona/sangue , Cicloexanóis/farmacologia , Cicloexanóis/uso terapêutico , Depressão/tratamento farmacológico , Depressão/fisiopatologia , Transportador 1 de Aminoácido Excitatório/metabolismo , Feminino , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Glutamato Descarboxilase/metabolismo , Hipocampo/fisiopatologia , Masculino , Rede Nervosa/fisiopatologia , Ratos , Cloridrato de Venlafaxina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...