Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Addict Biol ; 29(2): e13375, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38380802

RESUMEN

Recent studies found that non-coding RNAs (ncRNAs) played crucial roles in drug addiction through epigenetic regulation of gene expression and underlying drug-induced neuroadaptations. In this study, we characterized lncRNA transcriptome profiles in the nucleus accumbens (NAc) of mice exhibiting morphine-conditioned place preference (CPP) and explored the prospective roles of novel differentially expressed lncRNA, lncLingo2 and its derived miR-876-5p in the acquisition of opioids-associated behaviours. We found that the lncLingo2 was downregulated within the NAc core (NAcC) but not in the NAc shell (NAcS). This downregulation was found to be associated with the development of morphine CPP and heroin intravenous self-administration (IVSA). As Mfold software revealed that the secondary structures of lncLingo2 contained the sequence of pre-miR-876, transfection of LV-lncLingo2 into HEK293 cells significantly upregulated miR-876 expression and the changes of mature miR-876 are positively correlated with lncLingo2 expression in NAcC of morphine CPP trained mice. Delivering miR-876-5p mimics into NAcC also inhibited the acquisition of morphine CPP. Furthermore, bioinformatics analysis and dual-luciferase assay confirmed that miR-876-5p binds to its target gene, Kcnn3, selectively and regulates morphine CPP training-induced alteration of Kcnn3 expression. Lastly, the electrophysiological analysis indicated that the currents of small conductance calcium-activated potassium (SK) channel was increased, which led to low neuronal excitability in NAcC after CPP training, and these changes were reversed by lncLingo2 overexpression. Collectively, lncLingo2 may function as a precursor of miR-876-5p in NAcC, hence modulating the development of opioid-associated behaviours in mice, which may serve as an underlying biomarker and therapeutic target of opioid addiction.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Humanos , Ratones , Animales , Analgésicos Opioides/farmacología , Analgésicos Opioides/metabolismo , Epigénesis Genética , Células HEK293 , Morfina/farmacología , Morfina/metabolismo , Núcleo Accumbens/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo
2.
J Neurosci Res ; 99(2): 649-661, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33094531

RESUMEN

Adolescent alcohol drinking, primarily in the form of binge-drinking episodes, is a serious public health concern. Binge drinking in laboratory animals has been modeled by a procedure involving chronic intermittent ethanol (CIE) administration, as compared with chronic intermittent water (CIW). The prolonged effects of adolescent binge alcohol exposure in adults, such as high risk of developing alcohol use disorder, are severe but available treatments in the clinic are limited. One reason is the lack of sufficient understanding about the associated neuronal alterations. The involvement of the insular cortex, particularly the anterior agranular insula (AAI), has emerged as a critical region to explain neuronal mechanisms of substance abuse. This study was designed to evaluate the functional output of the AAI by measuring the intrinsic excitability of pyramidal neurons from male rats 2 or 21 days after adolescent or adult CIE treatment. Decreases in intrinsic excitability in AAI pyramidal neurons were detected 21 days, relative to 2 days, after adolescent CIE. Interestingly, the decreased intrinsic excitability in the AAI pyramidal neurons was observed 2 days after adult CIE, compared to adult CIW, but no difference was found between 2 versus 21 days after adult CIE. These data indicate that, although the AAI is influenced within a limited period after adult but not adolescent CIE, neuronal alterations in AAI are affected during the prolonged period of withdrawal from adolescent but not adult CIE. This may explain the prolonged vulnerability to mental disorders of subjects with an alcohol binge history during their adolescent stage.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas/fisiopatología , Etanol/toxicidad , Corteza Insular/efectos de los fármacos , Factores de Edad , Animales , Etanol/administración & dosificación , Corteza Insular/crecimiento & desarrollo , Corteza Insular/fisiopatología , Masculino , Técnicas de Placa-Clamp , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Ratas , Ratas Sprague-Dawley , Maduración Sexual
3.
Mol Cell Biochem ; 476(2): 809-818, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33078341

RESUMEN

Silica-induced apoptosis of alveolar macrophages (AMs) is an essential part of silicosis formation. Autophagy tends to present a bidirectional effect on apoptosis. Our previous study found that the blockade of autophagy degradation might aggravate the apoptosis of AMs in human silicosis. We presume that targeting the autophagic pathway is regarded as a promising new strategy for silicosis fibrosis. As a main active component of the Atractylodes rhizome, Atractylenolide III (ATL-III) has been widely applied in clinical anti-inflammation. However, the effect and mechanism of ATL-III on autophagy in AMs of silicosis are unknown. In this study, we found that ATL-III might inhibit autophagy by mTOR-dependent manner, thereby improving the blockage of autophagic degradation in AMs. ATL-III alleviated the apoptosis of AMs in human silicosis. Furthermore, Rapamycin reversed the protective effect of ATL-III in AMs. These results indicate that ATL-III may be a potentially protective ingredient targeting autophagy for workers exposed to silica dust. These findings also suggest that inhibition of autophagy may be an effective way to alleviate the apoptosis of AMs in silicosis.


Asunto(s)
Líquido del Lavado Bronquioalveolar/citología , Lactonas/farmacología , Macrófagos Alveolares/efectos de los fármacos , Sesquiterpenos/farmacología , Silicosis/tratamiento farmacológico , Serina-Treonina Quinasas TOR/metabolismo , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Células Cultivadas , Antagonistas Colinérgicos/farmacología , Humanos , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patología , Silicosis/metabolismo , Silicosis/patología , Serina-Treonina Quinasas TOR/genética
4.
Pharmacol Res ; 170: 105722, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34116208

RESUMEN

A progressive increase in drug craving following drug exposure is an important trigger of relapse. CircularRNAs (CircRNAs), key regulators of gene expression, play an important role in neurological diseases. However, the role of circRNAs in drug craving is unclear. In the present study, we trained mice to morphine conditioned place preference (CPP) and collected the nucleus accumbens (NAc) sections on abstinence day 1 (AD1) and day 14 (AD14) for RNA-sequencing. CircTmeff-1, which was highly expressed in the NAc core, was associated with incubation of context-induced morphine craving. The gain- and loss- of function showed that circTmeff-1 was a positive regulator of incubation. Simultaneously, the expression of miR-541-5p and miR-6934-3p were down-regulated in the NAc core during the incubation period. The dual luciferase reporter, RNA pulldown, and fluorescence insitu hybridization assays confirmed that miR-541-5p and miR-6934-3p bind to circTmeff-1 selectively. Furthermore, bioinformatics and western blot analysis suggested that vesicle-associated membrane protein 1 (VAMP1) and neurofascin (NFASC), both overlapping targets of miR-541-5p and miR-6934-3p, were highly expressed during incubation. Lastly, AAV-induced down-regulation of circTmeff-1 decreased VAMP1 and NFASC expression and incubation of morphine craving. These findings suggested that circTmeff-1, a novel circRNA, promotes incubation of context-induced morphine craving by sponging miR-541/miR-6934 in the NAc core. Thus, circTmeff-1 represents a potential therapeutic target for context-induced opioid craving, following prolonged abstinence.


Asunto(s)
Conducta Animal , Ansia , Comportamiento de Búsqueda de Drogas , Dependencia de Morfina/metabolismo , Núcleo Accumbens/metabolismo , ARN Circular/metabolismo , Animales , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Señales (Psicología) , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Dependencia de Morfina/genética , Dependencia de Morfina/fisiopatología , Dependencia de Morfina/psicología , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Núcleo Accumbens/fisiopatología , ARN Circular/genética , Proteína 1 de Membrana Asociada a Vesículas/genética , Proteína 1 de Membrana Asociada a Vesículas/metabolismo
5.
Acta Biochim Biophys Sin (Shanghai) ; 53(1): 29-35, 2021 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-33201175

RESUMEN

The two-drug combined chemotherapy of platinum and fluorouracil has been reported to efficiently kill tumor cells as the first-line treatment for advanced gastric cancer. However, the effect of these drugs on T cells remains unclear. Here, we showed that T cells including CD4+ T cells and CD8+ T cells of the patients with advanced gastric cancer after platinum and fluorouracil chemotherapy exhibited enhanced ex vivo proliferation ability as compared to that before chemotherapy. In addition, platinum and fluorouracil also promoted the differentiation of human T cells into Th1 and Th9 subtypes and cytotoxic T lymphocytes (CTLs) in vitro and in vivo. Accordingly, the combination therapy greatly suppressed tumor growth with increased tumor infiltration of Th1, Th9, and CTL cells in a mouse tumor model. Moreover, in activated T cells, long-term treatment with these two drugs further facilitates T cell activation along with promoted nuclear factor-κB (NF-κB) activation. Our findings demonstrate a previously unidentified function of platinum and fluorouracil combination chemotherapy in promoting T cell-mediated antitumor immunity.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Fluorouracilo/farmacología , Neoplasias/inmunología , Platino (Metal)/farmacología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Quimioterapia Combinada/métodos , Fluorouracilo/uso terapéutico , Humanos , Melanoma/tratamiento farmacológico , Melanoma/inmunología , Melanoma/patología , Ratones Endogámicos C57BL , Subunidad p50 de NF-kappa B/metabolismo , Oxaliplatino/farmacología , Oxaliplatino/uso terapéutico , Platino (Metal)/uso terapéutico , Neoplasias Gástricas/sangre , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Citotóxicos , Linfocitos T Colaboradores-Inductores/inmunología
6.
Addict Biol ; 25(4): e12793, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31339209

RESUMEN

Postretrieval extinction procedures are effective nonpharmacological interventions for disrupting drug-associated memories. Nonetheless, the conditioned stimulus (CS) memory retrieval-extinction procedure is ineffective in inhibiting drug craving and relapse after prolonged withdrawal, which significantly undermines its therapeutic potential. In the present study, we showed that, unlike the CS memory retrieval-extinction procedure, noncontingent heroin injections (unconditioned stimulus [UCS]) 1 hour before the extinction sessions decreased the heroin-priming-induced reinstatement, renewal, and spontaneous recovery of heroin seeking after 28 days of withdrawal (ie, remote heroin-associated memories) in rats. The UCS retrieval manipulation induced reactivation of the basolateral amygdala (BLA) after prolonged withdrawal, and this reactivation was absent with the CS retrieval manipulation. Chemogenetic inactivation of the BLA abolished the inhibitory effect of the UCS memory retrieval-extinction procedure on heroin-priming-induced reinstatement after prolonged withdrawal. Furthermore, the combination of chemogenetic reactivation of BLA and CS retrieval-extinction procedure resembled the inhibitory effect of UCS retrieval-extinction procedure on heroin seeking after prolonged withdrawal. We also observed that the inhibitory effect of the UCS retrieval-extinction procedure is mediated by regulation of AMPA receptor endocytosis in the BLA. Our results demonstrate critical engagement of the BLA in reconsolidation updating of heroin-associated memory after prolonged withdrawal, extending our knowledge of the boundary conditions of the reconsolidation of drug-associated memories.


Asunto(s)
Complejo Nuclear Basolateral/metabolismo , Comportamiento de Búsqueda de Drogas/fisiología , Extinción Psicológica/fisiología , Dependencia de Heroína/metabolismo , Heroína/farmacología , Consolidación de la Memoria/fisiología , Narcóticos/farmacología , Animales , Complejo Nuclear Basolateral/fisiología , Núcleo Amigdalino Central/metabolismo , Núcleo Amigdalino Central/fisiología , Endocitosis , Dependencia de Heroína/fisiopatología , Masculino , Ratas , Receptores AMPA/metabolismo , Factores de Tiempo
7.
J Neurosci ; 37(37): 8938-8951, 2017 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-28821652

RESUMEN

Exposure to drug-paired cues causes drug memories to be in a destabilized state and interfering with memory reconsolidation can inhibit relapse. Calpain, a calcium-dependent neutral cysteine protease, is involved in synaptic plasticity and the formation of long-term fear memory. However, the role of calpain in the reconsolidation of drug reward memory is still unknown. In the present study, using a conditioned place preference (CPP) model, we found that exposure to drug-paired contextual stimuli induced the activation of calpain and decreased the expression of glutamate receptor interacting protein 1 (GRIP1) in the nucleus accumbens (NAc) core, but not shell, of male rats. Infusions of calpain inhibitors in the NAc core immediately after retrieval disrupted the reconsolidation of cocaine/morphine cue memory and blocked retrieval-induced calpain activation and GRIP1 degradation. The suppressive effect of calpain inhibitors on the expression of drug-induced CPP lasted for at least 14 d. The inhibition of calpain without retrieval 6 h after retrieval or after exposure to an unpaired context had no effects on the expression of reward memory. Calpain inhibition after retrieval also decreased cocaine seeking in a self-administration model and this effect did not recover spontaneously after 28 d. Moreover, the knock-down of GRIP1 expression in the NAc core by lentivirus-mediated short-hairpin RNA blocked disruption of the reconsolidation of drug cue memories that was induced by calpain inhibitor treatment. These results suggest that calpain activity in the NAc core is crucial for the reconsolidation of drug reward memory via the regulation of GRIP1 expression.SIGNIFICANCE STATEMENT Calpain plays an important role in synaptic plasticity and long-term memory consolidation, however, its role in the reconsolidation of drug cue memory remains unknown. Using conditioned place preference and self-administration procedures, we found that exposure to drug-paired cues induced the activation of calpain and decreased glutamate receptor interacting protein 1 (GRIP1) expression in the nucleus accumbens (NAc) core. The inhibition of calpain activity in the NAc core immediately after retrieval disrupted the reconsolidation of cocaine/morphine cue memory that was blocked by prior GRIP1 knock-down. Our findings indicate that calpain-GRIP signaling is essential for the restabilization process that is associated with drug cue memory and the inhibition of calpain activity may be a novel strategy for the prevention of drug relapse.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Calpaína/metabolismo , Señales (Psicología) , Comportamiento de Búsqueda de Drogas/fisiología , Consolidación de la Memoria/fisiología , Proteínas del Tejido Nervioso/metabolismo , Núcleo Accumbens/fisiología , Recompensa , Animales , Expresión Génica/fisiología , Masculino , Ratas , Ratas Sprague-Dawley
8.
J Neurosci ; 35(21): 8308-21, 2015 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-26019344

RESUMEN

Fear extinction forms a new memory but does not erase the original fear memory. Exposure to novelty facilitates transfer of short-term extinction memory to long-lasting memory. However, the underlying cellular and molecular mechanisms are still unclear. Using a classical contextual fear-conditioning model, we investigated the effect of novelty on long-lasting extinction memory in rats. We found that exposure to a novel environment but not familiar environment 1 h before or after extinction enhanced extinction long-term memory (LTM) and reduced fear reinstatement. However, exploring novelty 6 h before or after extinction had no such effect. Infusion of the ß-adrenergic receptor (ßAR) inhibitor propranolol and glucocorticoid receptor (GR) inhibitor RU486 into the CA1 area of the dorsal hippocampus before novelty exposure blocked the effect of novelty on extinction memory. Propranolol prevented activation of the hippocampal PKA-CREB pathway, and RU486 prevented activation of the hippocampal extracellular signal-regulated kinase 1/2 (Erk1/2)-CREB pathway induced by novelty exposure. These results indicate that the hippocampal ßAR-PKA-CREB and GR-Erk1/2-CREB pathways mediate the extinction-enhancing effect of novelty exposure. Infusion of RU486 or the Erk1/2 inhibitor U0126, but not propranolol or the PKA inhibitor Rp-cAMPS, into the CA1 before extinction disrupted the formation of extinction LTM, suggesting that hippocampal GR and Erk1/2 but not ßAR or PKA play critical roles in this process. These results indicate that novelty promotes extinction memory via hippocampal ßAR- and GR-dependent pathways, and Erk1/2 may serve as a behavioral tag of extinction.


Asunto(s)
Extinción Psicológica/fisiología , Miedo/fisiología , Hipocampo/fisiología , Receptores Adrenérgicos beta/fisiología , Receptores de Glucocorticoides/fisiología , Antagonistas Adrenérgicos beta/farmacología , Animales , Extinción Psicológica/efectos de los fármacos , Miedo/efectos de los fármacos , Hipocampo/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Memoria/fisiología , Ratas , Ratas Sprague-Dawley , Receptores de Glucocorticoides/antagonistas & inhibidores
9.
J Neurosci ; 34(19): 6647-58, 2014 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-24806690

RESUMEN

Extinction therapy has been suggested to suppress the conditioned motivational effect of drug cues to prevent relapse. However, extinction forms a new inhibiting memory rather than erasing the original memory trace and drug memories invariably return. Perineuronal nets (PNNs) are a specialized extracellular matrix around interneurons in the brain that have been suggested to be a permissive factor that allows synaptic plasticity in the adolescent brain. The degradation of PNNs caused by chondroitinase ABC (ChABC) may generate induced juvenile-like plasticity (iPlasticity) and promote experience-dependent plasticity in the adult brain. In the present study, we investigated the effect of removing PNNs in the amygdala of rat on the extinction of drug memories. We found that extinction combined with intra-amygdala injections of ChABC (0.01 U/side) prevented the subsequent priming-induced reinstatement of morphine-induced and cocaine-induced, but not food -induced, conditioned place preference (CPP). Intra-amygdala injections of ChABC alone had no effect on the retention, retrieval, or relearning of morphine-induced CPP and storage of acquired food-induced CPP. Moreover, we found that the procedure facilitated the extinction of heroin- and cocaine-seeking behavior and prevented the spontaneous recovery and drug-induced reinstatement of heroin- and cocaine-seeking behavior. We also found that the effect of PNNs degradation combined with extinction may be mediated by the potentiation of several plasticity-related proteins in the amygdala. Altogether, our findings demonstrate that a combination of extinction training with PNNs degradation in the amygdala erases drug memories and suggest that ChABC may be an attractive candidate for the prevention of relapse.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/fisiología , Memoria , Red Nerviosa/fisiología , Trastornos Relacionados con Sustancias/psicología , Animales , Western Blotting , Condroitina ABC Liasa/administración & dosificación , Condroitina ABC Liasa/farmacología , Trastornos Relacionados con Cocaína/psicología , Condicionamiento Operante , Extinción Psicológica , Alimentos , Dependencia de Heroína/psicología , Masculino , Microinyecciones , Dependencia de Morfina/psicología , Plasticidad Neuronal/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Refuerzo en Psicología , Prevención Secundaria
10.
J Neurosci ; 34(30): 10010-21, 2014 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-25057203

RESUMEN

Maladaptive memories elicited by exposure to environmental stimuli associated with drugs of abuse are often responsible for relapse among addicts. Interference with the reconsolidation of drug memory can inhibit drug seeking. Previous studies have indicated that the dephosphorylation of the eukaryotic initiation factor 2 α-subunit (eIF2α) plays an important role in synaptic plasticity and long-term memory consolidation, but its role in the reconsolidation of drug memory remains unknown. The amygdala is required for the reconsolidation of a destabilized drug memory after retrieval of drug-paired stimuli. Here, we used conditioned place preference (CPP) and self-administration procedures to determine whether amygdala eIF2α dephosphorylation is required for the reconsolidation of morphine and cocaine memories in rats. We found that the levels of eIF2α phosphorylation (Ser51) and activating transcription factor 4 (ATF4) were decreased after reexposure to a previously morphine- or cocaine-paired context (i.e., a memory retrieval procedure) in the basolateral amygdala (BLA) but not in the central amygdala. Intra-BLA infusions of Sal003, a selective inhibitor of eIF2α dephosphorylation, immediately after memory retrieval disrupted the reconsolidation of morphine- or cocaine-induced CPP, leading to a long-lasting suppression of drug-paired stimulus-induced craving. Advanced knockdown of ATF4 expression in the BLA by lentivirus-mediated short-hairpin RNA blocked the disruption of the reconsolidation of morphine-induced CPP induced by Sal003 treatment. Furthermore, inhibition of eIF2α dephosphorylation in the BLA immediately after light/tone stimulus retrieval decreased subsequent cue-induced heroin-seeking behavior in the self-administration procedure. These results demonstrate that eIF2α dephosphorylation in the BLA mediates the memory reconsolidation of drug-paired stimuli.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Señales (Psicología) , Comportamiento de Búsqueda de Drogas/fisiología , Factor 2 Eucariótico de Iniciación/metabolismo , Memoria/fisiología , Amígdala del Cerebelo/efectos de los fármacos , Animales , Cocaína/administración & dosificación , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Heroína/administración & dosificación , Masculino , Memoria/efectos de los fármacos , Morfina/administración & dosificación , Fosforilación/fisiología , Ratas , Ratas Sprague-Dawley , Autoadministración
11.
Addict Biol ; 20(3): 513-22, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-24698092

RESUMEN

Time-dependent increases in cue-induced nicotine and methamphetamine craving during abstinence were recently reported in human drug-dependent individuals. In the present study, we sought to determine whether this 'incubation of craving' phenomenon also occurs in alcoholics. Four groups of 80 inpatient adult male alcoholics were assessed in a single session (between-group design) for cue-induced alcohol craving at 7, 14, 30 and 60 days of abstinence. Another group that included 19 patients was repeatedly tested for cue-induced alcohol craving at the same abstinence days as above. Other psychological and physiological measures were assessed at the four abstinence timepoints. Cue-induced alcohol craving measured with visual analogue scales was the highest at 60 days of abstinence both between and within groups. However, heart rate, blood pressure and skin conductance responses did not differ between abstinent groups. These results provide evidence of the incubation of alcohol craving in humans, extending previous reports with smokers and methamphetamine addicts.


Asunto(s)
Abstinencia de Alcohol/psicología , Alcoholismo/psicología , Ansia , Adolescente , Adulto , Análisis de Varianza , Presión Sanguínea/fisiología , Señales (Psicología) , Respuesta Galvánica de la Piel/fisiología , Frecuencia Cardíaca/fisiología , Humanos , Masculino , Persona de Mediana Edad , Síndrome de Abstinencia a Sustancias/psicología , Adulto Joven
12.
Nanomedicine ; 11(2): 391-400, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25461282

RESUMEN

Leukocytes can cross intact blood-brain barrier under healthy conditions and in many neurological diseases, including psychiatric diseases. In present study, a cyclic RGD (cRGD) peptide with high affinity for integrin receptors of leukocytes was used to modify liposomes. The cRGD-modified liposomes (cRGDL) showed high affinity for monocytes in vitro and in vivo and co-migrated across in vitro BBB model with THP-1. The trefoil factor 3 (TFF3), a macromolecular drug, was rapidly and persistently delivered to brain for at least 12 h when loaded into cRGDL while 2.8-fold increase in drug concentration in basolateral amygdala regions related to depression was observed. A systemic administration of cRGDL-TFF3 mimicked antidepressant-like effect of direct intra-basolateral amygdala administration of TFF3 solution in rats subjected to chronic mild stress. The effective dual-brain targeting delivery resulting from the combination and co-migration of cRGDL with leukocyte cross BBB may be a promising strategy for targeted brain delivery. FROM THE CLINICAL EDITOR: In an effort to treat depression, brain targeted delivery via monocyte-cRGD liposome complexes capable of crossing the intact BBB was performed in this study in a murine model. Similar approaches may be helpful in the treatment of other neuropsychiatric conditions.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Depresión/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Péptidos Cíclicos/administración & dosificación , Péptidos/administración & dosificación , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Depresión/patología , Sinergismo Farmacológico , Humanos , Leucocitos/efectos de los fármacos , Liposomas/administración & dosificación , Masculino , Ratones , Ratas , Factor Trefoil-3
13.
Int J Mol Sci ; 16(12): 28386-400, 2015 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-26633367

RESUMEN

The trefoil factors (TFFs) are a family of three polypeptides, among which TFF1 and TFF3 are widely distributed in the central nervous system. Our previous study indicated that TFF3 was a potential rapid-onset antidepressant as it reversed the depressive-like behaviors induced by acute or chronic mild stress. In order to further identify the antidepressant-like effect of TFF3, we applied an olfactory bulbectomy (OB), a classic animal model of depression, in the present study. To elucidate the mechanism underlying the antidepressant-like activity of TFF3, we tested the role of brain-derived neurotrophic factor (BDNF)-extracellular signal-related kinase (ERK)-cyclic adenosine monophosphate response element binding protein (CREB) signaling in the hippocampus in the process. Chronic systemic administration of TFF3 (0.1 mg/kg, i.p.) for seven days not only produced a significant antidepressant-like efficacy in the OB paradigm, but also restored the expression of BDNF, pERK, and pCREB in the hippocampal CA3. Inhibition of BDNF or extracellular signal-related kinase (ERK) signaling in CA3 blocked the antidepressant-like activity of TFF3 in OB rats. Our findings further confirmed the therapeutic effect of TFF3 against depression and suggested that the normalization of the BDNF-ERK-CREB pathway was involved in the behavioral response of TFF3 for the treatment of depression.


Asunto(s)
Conducta Animal/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Depresión/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Neuropéptidos/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Antidepresivos/administración & dosificación , Antidepresivos/farmacología , Depresión/tratamiento farmacológico , Depresión/etiología , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Neuropéptidos/administración & dosificación , Bulbo Olfatorio/cirugía , Fosforilación , Ratas , Receptor trkB/antagonistas & inhibidores , Factor Trefoil-3
14.
Int J Neuropsychopharmacol ; 18(5)2014 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-25522425

RESUMEN

BACKGROUND: Currently approved medications for opioid addiction have shown clinical efficacy, but undesired side effects, dependence induced by the medications themselves, and low treatment compliance necessitate the need for novel therapies. METHODS: A novel morphine-keyhole limpet hemocyanin conjugate vaccine was synthesized with 6-glutarylmorphine as the hapten and a lengthened linker of 6 carbon atoms. The titer and specificity of the triggered antibody were assessed by enzyme-linked immunosorbent assay. The effects of the vaccine on the morphine-induced elevation of dopamine levels in the nucleus accumbens were determined by high-performance liquid chromatography. The effects of the vaccine on morphine-induced locomotor sensitization and heroin-primed reinstatement of heroin self-administration were also assessed. RESULTS: After subcutaneous administration in rats, the vaccine triggered a high antibody titer, with comparable specificity for morphine, 6-acetylmorphine, and heroin, but no interaction with dissimilar therapeutic opioid compounds, including buprenorphine, naloxone, and nalorphine, was observed. The vaccine significantly prevented the elevation of dopamine levels in the nucleus accumbens induced by a single morphine challenge. Moreover, the vaccine prevented the expression of morphine-induced locomotor sensitization and heroin-primed reinstatement of heroin seeking, suggesting its potential for preventing relapse. CONCLUSION: These results demonstrate that active immunization with the present vaccine induces a robust morphine/heroin-specific antibody response in rats and attenuates the behavioral effects of morphine and heroin.


Asunto(s)
Anticuerpos/administración & dosificación , Conducta Animal/efectos de los fármacos , Dopamina/sangre , Morfina/inmunología , Vacunas Conjugadas/administración & dosificación , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/efectos adversos , Animales , Anticuerpos/farmacología , Cromatografía Líquida de Alta Presión , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Heroína/administración & dosificación , Heroína/efectos adversos , Locomoción/efectos de los fármacos , Masculino , Morfina/administración & dosificación , Morfina/efectos adversos , Derivados de la Morfina/administración & dosificación , Derivados de la Morfina/efectos adversos , Derivados de la Morfina/inmunología , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Ratas , Ratas Sprague-Dawley , Refuerzo en Psicología , Autoadministración , Resultado del Tratamiento , Vacunas Conjugadas/farmacología
15.
Int J Neuropsychopharmacol ; 17(11): 1881-94, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24916432

RESUMEN

Repeated exposure to nicotine increases psychomotor activity. Long-lasting neural plasticity changes that contribute to the nicotine-induced development of locomotor sensitization have been identified. The mammalian target of rapamycin complex 1 (mTORC1) signalling pathway is involved in regulating the neuroplasticity of the central nervous system. In this study, we examined the role of mTORC1 in the amygdala in nicotine-induced locomotor sensitization. Rapamycin, an inhibitor of mTORC1, was infused into the basolateral amygdala (BLA) and central amygdala (CeA) or systemically administered to investigate the role of the mTORC1 in the development and expression of nicotine-induced locomotor sensitization. We found that locomotor activity progressively increased during the initiation of nicotine-induced locomotor sensitization and the expression of nicotine sensitization was induced by nicotine challenge injection (0.35 mg/kg s.c.) after five days of withdrawal. The initiation of nicotine-induced locomotor sensitization was accompanied by the increased phosphorylated level of mTORC1 downstream target proteins including p-p70s6k and p-4EBP in the BLA, but not CeA. Intra-BLA infusion or systemic administration of rapamycin blocked locomotor activity. Increased p-p70s6k and p-4EBP were also observed in the expression of nicotine sensitization, which was demonstrated to be inhibited by systemic rapamycin administration. Our findings indicated that mTORC1 activity in the BLA, but not the CeA, mediated the initiation and expression of nicotine-induced locomotor sensitization, and may become a potential target for the treatment of nicotine addiction.


Asunto(s)
Complejo Nuclear Basolateral/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Complejos Multiproteicos/metabolismo , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Animales , Proteínas Portadoras/metabolismo , Esquema de Medicación , Inmunosupresores/farmacología , Péptidos y Proteínas de Señalización Intracelular , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Microinyecciones , Nicotina/administración & dosificación , Agonistas Nicotínicos/administración & dosificación , Fosfoproteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Sirolimus/farmacología
16.
Addict Biol ; 19(6): 996-1005, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23750993

RESUMEN

Cocaine sensitization and reward are reported to be under the influence of diurnal rhythm. However, no previous studies have reported brain areas that play a role as modulators and underlie the mechanism of diurnal variations in cocaine reward. We examined (1) the diurnal rhythm of glycogen synthase kinase-3ß (GSK-3ß) activity in the suprachiasmatic nucleus (SCN) and reward-related brain areas in naive rats; (2) the effect of day and night on the acquisition of cocaine-induced conditioned place preference (CPP); (3) the influence of cocaine-induced CPP on GSK-3ß activity in the SCN and reward-related brain areas; and (4) the effect of the GSK-3ß inhibitor SB216763 microinjected bilaterally into the ventral tegmental area (VTA) on cocaine-induced CPP. A significant diurnal rhythm of GSK-3ß activity was found in the SCN and reward-related brain areas, with diurnal variations in cocaine-induced CPP. GSK-3ß activity in the SCN and reward-related brain areas exhibited marked diurnal variations in rats treated with saline. GSK-3ß activity in rats treated with cocaine exhibited distinct diurnal variations only in the prefrontal cortex and VTA. Cocaine decreased the expression of phosphorylated GSK-3ß (i.e. increased GSK-3ß activity) only in the VTA in rats trained and tested at ZT4 and ZT16. SB216763 microinjected into the VTA bilaterally eliminated the diurnal variations in cocaine-induced CPP, but did not affect the acquisition of cocaine-induced CPP. These findings suggest that the VTA may be a critical area involved in the diurnal variations in cocaine-induced CPP, and GSK-3ß may be a regulator of diurnal variations in cocaine-induced CPP.


Asunto(s)
Cocaína/farmacología , Condicionamiento Psicológico/efectos de los fármacos , Inhibidores de Captación de Dopamina/farmacología , Glucógeno Sintasa Quinasa 3/fisiología , Área Tegmental Ventral/enzimología , Animales , Ritmo Circadiano/efectos de los fármacos , Indoles/farmacología , Masculino , Maleimidas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Ratas Sprague-Dawley , Recompensa , Núcleo Supraquiasmático/enzimología , Área Tegmental Ventral/efectos de los fármacos
17.
CNS Neurosci Ther ; 30(9): e70046, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39295107

RESUMEN

BACKGROUND: Drug addiction, characterized by compulsive drug use and high relapse rates, arises from complex interactions between reward and aversion systems in the brain. The paraventricular nucleus (PVN), located in the anterior hypothalamus, serves as a neuroendocrine center and is a key component of the hypothalamic-pituitary-adrenal axis. OBJECTIVE: This review aimed to explore how the PVN impacts reward and aversion in drug addiction through stress responses and emotional regulation and to evaluate the potential of PVN as a therapeutic target for drug addiction. METHODS: We review the current literature, focusing on three main neuron types in the PVN-corticotropin-releasing factor, oxytocin, and arginine vasopressin neurons-as well as other related neurons, to understand their roles in modulating addiction. RESULTS: Existing studies highlight the PVN as a key mediator in addiction, playing a dual role in reward and aversion systems. These findings are crucial for understanding addiction mechanisms and developing targeted therapies. CONCLUSION: The role of PVN in stress response and emotional regulation suggests its potential as a therapeutic target in drug addiction, offering new insights for addiction treatment.


Asunto(s)
Núcleo Hipotalámico Paraventricular , Recompensa , Trastornos Relacionados con Sustancias , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Humanos , Animales , Trastornos Relacionados con Sustancias/psicología , Trastornos Relacionados con Sustancias/metabolismo , Trastornos Relacionados con Sustancias/fisiopatología , Hormona Liberadora de Corticotropina/metabolismo , Conducta Adictiva/psicología , Estrés Psicológico/psicología , Estrés Psicológico/metabolismo , Reacción de Prevención/fisiología , Reacción de Prevención/efectos de los fármacos , Oxitocina/metabolismo
18.
Neurosci Lett ; 837: 137893, 2024 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-38997082

RESUMEN

Shikonin is an active naphthoquinone with antioxidative, anti-inflammatory, and anticancer properties. In this study, we investigated the effects of shikonin on depressive- and anxiety-like behaviors in lipopolysaccharide- (LPS-) induced depression and chronic unpredictable mild stress (CUMS) rat models and explored the potential mechanism. First, a 14-day intraperitoneal administration of shikonin (10 mg/kg) significantly decreased immobility time in forced swimming test (FST) and increased open arm entries in elevated plus maze (EPM) test, without affecting line crossings in open field test (OFT), indicating that shikonin has anti-depressant- and anxiolytic-like effects. Second, chronic shikonin administration (10 mg/kg) reversed depressive- and anxiety-like behaviors in LPS-induced and CUMS depression models, as shown in the sucrose preference test (SPT), FST, EPM, and novel object recognition test (NORT). Finally, shikonin significantly reduced the levels of interleukin-1ß (IL-1ß), IL-6, and tumor necrosis factor-α (TNF-α) in hippocampus, indicating that the anti-depressant- and anxiolytic-like effects of shikonin are related to the reduction of neuroinflammation in hippocampus. These findings suggest that shikonin exerts anti-depressant- and anxiolytic-like effects via an anti-inflammatory mechanism of shikonin in the hippocampus.


Asunto(s)
Ansiedad , Depresión , Hipocampo , Naftoquinonas , Ratas Sprague-Dawley , Animales , Naftoquinonas/farmacología , Naftoquinonas/uso terapéutico , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Depresión/tratamiento farmacológico , Ansiedad/tratamiento farmacológico , Ratas , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Inflamación/tratamiento farmacológico , Conducta Animal/efectos de los fármacos , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/complicaciones , Estrés Psicológico/psicología , Ansiolíticos/farmacología , Ansiolíticos/uso terapéutico , Lipopolisacáridos
19.
Heliyon ; 10(8): e29481, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38655332

RESUMEN

Addressing the treatment of depression is crucial; nevertheless, the etiology and pathogenesis remain unelucidated. Therefore, this study investigated the effects of teriflunomide (TF) on corticosterone (CORT)-induced depression-like behaviors in mice. Notably, TF administration resulted in a substantial amelioration of anxiety and depression-like behaviors observed in CORT-treated mice. This was evidenced by behavioral assessments conducted via the sucrose preference test (SPT), open-field test (OFT), novelty-suppressed feeding test (NSFT), forced swimming test (FST), and tail suspension test (TST). The administration of CORT inflicts damage upon oligodendrocytes and neurons within the hippocampus. Our findings indicate that TF offers significant protective effects on oligodendrocytes, mitigating apoptosis both invivo and invitro. Additionally, TF was found to counteract the CORT-induced neuronal loss and synaptic damage, as demonstrated by an increase in Nissl-positive cells across hippocampal regions CA1, CA3, and the dentate gyrus (DG) alongside elevated levels of synapse-related proteins including PSD-95 and synaptophysin. Additionally, TF treatment facilitated a reduction in the levels of apoptosis-related proteins while simultaneously augmenting the levels of Bcl2. Our findings indicate that TF administration effectively mitigates CORT-induced depression-like behaviors and reverses damage to oligodendrocytes and neurons in the hippocampus, suggesting TF as a promising candidate for depression.

20.
J Clin Invest ; 134(17)2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39225090

RESUMEN

Liraglutide, a glucagon-like peptide-1 (GLP-1) analog, is approved for obesity treatment, but the specific neuronal sites that contribute to its therapeutic effects remain elusive. Here, we show that GLP-1 receptor-positive (GLP-1R-positive) neurons in the lateral septum (LSGLP-1R) play a critical role in mediating the anorectic and weight-loss effects of liraglutide. LSGLP-1R neurons were robustly activated by liraglutide, and chemogenetic activation of these neurons dramatically suppressed feeding. Targeted knockdown of GLP-1 receptors within the LS, but not in the hypothalamus, substantially attenuated liraglutide's ability to inhibit feeding and lower body weight. The activity of LSGLP-1R neurons rapidly decreased during naturalistic feeding episodes, while synaptic inactivation of LSGLP-1R neurons diminished the anorexic effects triggered by liraglutide. Together, these findings offer critical insights into the functional role of LSGLP-1R neurons in the physiological regulation of energy homeostasis and delineate their instrumental role in mediating the pharmacological efficacy of liraglutide.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón , Liraglutida , Neuronas , Liraglutida/farmacología , Animales , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/genética , Ratones , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Núcleos Septales/metabolismo , Núcleos Septales/efectos de los fármacos , Masculino , Pérdida de Peso/efectos de los fármacos , Depresores del Apetito/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA