Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Psychiatry ; 27(4): 2136-2145, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35079125

RESUMO

Relapse remains a major challenge to the treatment of cocaine addiction. Recent studies suggested that the trace amine-associated receptor 1 (TAAR1) could be a promising target to treat cocaine addiction and relapse; however, the underlying mechanism remains unclear. Here, we aimed to investigate the neural mechanism underlying the role of TAAR1 in the drug priming-induced reinstatement of cocaine-seeking behavior in rats, an animal model of cocaine relapse. We focused on the shell subregion of nucleus accumbens (NAc), a key brain region of the brain reward system. We found that activation of TAAR1 by systemic and intra-NAc shell administration of the selective TAAR1 agonist RO5166017 attenuated drug-induced reinstatement of cocaine-seeking and prevented drug priming-induced CaMKIIα activity in the NAc shell. Activation of TAAR1 dampened the CaMKIIα/GluR1 signaling pathway in the NAc shell and reduced AMPAR-EPSCs on the NAc slice. Microinjection of the selective TAAR1 antagonist EPPTB into the NAc shell enhanced drug-induced reinstatement as well as potentiated CaMKIIα activity in the NAc shell. Furthermore, viral-mediated expression of CaMKIIα in the NAc shell prevented the behavioral effects of TAAR1 activation. Taken together, our findings indicate that TAAR1 regulates drug-induced reinstatement of cocaine-seeking by negatively regulating CaMKIIα activity in the NAc. Our findings elucidate a novel mechanism of TAAR1 in regulating drug-induced reinstatement of cocaine-seeking and further suggests that TAAR1 is a promising target for the treatment of cocaine relapse.


Assuntos
Transtornos Relacionados ao Uso de Cocaína , Cocaína , Animais , Cocaína/farmacologia , Transtornos Relacionados ao Uso de Cocaína/tratamento farmacológico , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Comportamento de Procura de Droga , Núcleo Accumbens/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G , Recidiva , Autoadministração
2.
Brain Behav Immun ; 101: 37-48, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34958862

RESUMO

Opioid addiction remains a severe health problem. While substantial insights underlying opioid addiction have been yielded from neuron-centric studies, the contribution of non-neuronal mechanisms to opioid-related behavioral adaptations has begun to be recognized. Toll-like receptor 4 (TLR4), a pattern recognition receptor, has been widely suggested in opioid-related behaviors. Interleukin-1 receptor-associated kinase 4 (IRAK4) is a kinase essential for TLR4 responses, However, the potential role of IRAK4 in opioid-related responses has not been examined. Here, we explored the role of IRAK4 in cue-induced opioid-seeking behavior in male rats. We found that morphine self-administration increased the phosphorylation level of IRAK4 in the nucleus accumbens (NAc) in rats; the IRAK4 signaling remained activated after morphine extinction and cue-induced reinstatement test. Both systemic and local inhibition of IRAK4 in the NAc core attenuated cue-induced morphine-seeking behavior without affecting the locomotor activity and cue-induced sucrose-seeking. In addition, inhibition of IRAK4 also reduced the cue-induced reinstatement of fentanyl-seeking. Our findings suggest an important role of IRAK4 in opioid relapse-like behaviors and provide novel evidence in the association between innate immunity and drug addiction.


Assuntos
Comportamento de Procura de Droga , Quinases Associadas a Receptores de Interleucina-1 , Núcleo Accumbens , Transtornos Relacionados ao Uso de Opioides , Analgésicos Opioides/farmacologia , Animais , Sinais (Psicologia) , Extinção Psicológica , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Masculino , Morfina/farmacologia , Proteínas Serina-Treonina Quinases , Ratos , Ratos Sprague-Dawley , Receptor 4 Toll-Like
3.
Addict Biol ; 27(1): e13075, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34170054

RESUMO

Nicotine addiction is a leading avoidable brain disorder globally. Although nicotine induces a modest reinforcing effect, which is important for the initial drug use, the transition from nicotine use to nicotine addiction involves the mechanisms responsible for the negative consequences of drug abstinence. Recent study suggested that trace amine-associated receptor 1 (TAAR1) is a promising pharmacological target for the modulation of positive reinforcing effects of nicotine. However, whether TAAR1 plays a part in the negative reinforcement of nicotine withdrawal remains to be determined. Here, using a long-access (LA) self-administration model, we investigated whether LA rats show increased nicotine intake and withdrawal symptoms in comparison with saline and ShA rats and then tested the effect of TAAR1 partial agonist RO5263397 on nicotine withdrawal effects. We found that rats from long-access group showed significant abstinence-induced anxiety-like behaviour, mechanic hypersensitivity, increased number of precipitated withdrawal signs and higher motivation for the drug, while rats from short-access did not differ from saline group. TAAR1 partial agonist RO5263397 significantly reduced the physical and motivational withdrawal effects of nicotine in LA rats, as reflected by increased time spent on the open arm in the elevated plus maze (EPM) test, normalized paw withdrawal threshold, decreased withdrawal signs and motivation to self-administer nicotine. This study indicates that activation of TAAR1 attenuates the negative-reinforcing effects of nicotine withdrawal and further suggests TAAR1 as a promising target to treat nicotine addiction.


Assuntos
Nicotina/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Síndrome de Abstinência a Substâncias/metabolismo , Tabagismo/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Masculino , Oxazóis , Ratos , Reforço Psicológico , Autoadministração
4.
Handb Exp Pharmacol ; 276: 275-290, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35434747

RESUMO

Drug addiction is a chronic brain disease characterized by compulsive drug-seeking and drug-taking behaviors despite the major negative consequences. Current well-established neuronal underpinnings of drug addiction have promoted the substantial progress in understanding this disorder. However, non-neuronal mechanisms of drug addiction have long been underestimated. Fortunately, increased evidence indicates that neuroimmune system, especially Toll-like receptor 4 (TLR4) signaling, plays an important role in the different stages of drug addiction. Drugs like opioids, psychostimulants, and alcohol activate TLR4 signaling and enhance the proinflammatory response, which is associated with drug reward-related behaviors. While extensive studies have shown that inhibition of TLR4 attenuated drug-related responses, there are conflicting findings implicating that TLR4 signaling may not be essential to drug addiction. In this chapter, preclinical and clinical studies will be discussed to further evaluate whether TLR4-based neuroimmune pharmacotherapy can be used to treat drug addiction. Furthermore, the possible mechanisms underlying the effects of TLR4 inhibition in modulating drug-related behaviors will also be discussed.


Assuntos
Comportamento de Procura de Droga , Transtornos Relacionados ao Uso de Substâncias , Receptor 4 Toll-Like , Analgésicos Opioides/farmacologia , Comportamento de Procura de Droga/efeitos dos fármacos , Etanol/farmacologia , Humanos , Transdução de Sinais , Transtornos Relacionados ao Uso de Substâncias/tratamento farmacológico , Transtornos Relacionados ao Uso de Substâncias/genética , Receptor 4 Toll-Like/antagonistas & inibidores
5.
J Neurosci ; 39(50): 10071-10080, 2019 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-31704786

RESUMO

The claustrum connects with a broad range of cortical areas including the prefrontal cortex (PFC). However, the function of the claustrum (CLA) and its neural projections remains largely unknown. Here, we elucidated the role of the neural projections from the CLA to the PFC in regulating impulsivity in male rats. We first identified the CLA-PFC pathway by retrograde tracer and virus expression. By using immunofluorescent staining of the c-Fos-positive neurons, we showed that chemogenetic activation and inhibition of the CLA-PFC pathway reduced and increased overall activity of the PFC, respectively. In the 5-choice serial reaction time task (5-CSRTT), we found that chemogenetic activation and inhibition of the CLA-PFC pathway increased and reduced the impulsive-like behavior (i.e., premature responses), respectively. Furthermore, chemogenetic inhibition of the CLA-PFC pathway prevented methamphetamine-induced impulsivity, without affecting methamphetamine-induced hyperactivity. In contrast to the role of CLA-PFC pathway in selectively regulating impulsivity, activation of the claustrum disrupted attention in the 5-CSRTT. These results indicate that the CLA-PFC pathway is essential for impulsivity. This study may shed light on the understanding of impulsivity-related disorders such as drug addiction.SIGNIFICANCE STATEMENT The claustrum is one of the most mysterious brain regions. Although extensive anatomical studies demonstrated that the claustrum connects with many cortical areas, the function of the neural projections between the claustrum and cortical areas remain largely unknown. Here, we showed that the neural projections from the claustrum to the prefrontal cortex regulates impulsivity by using the designer drugs (DREADDs)-based chemogenetic tools. Interestingly, the claustrum-prefrontal cortex pathway also regulates methamphetamine-induced impulsivity, suggesting a critical role of this neural pathway in regulating impulsivity-related disorders such as drug addiction. Our results provided preclinical evidence that the claustrum-prefrontal cortex regulates impulsivity. The claustrum-prefrontal cortex pathway may be a novel target for the treatment of impulsivity-related brain disorders.


Assuntos
Comportamento de Escolha/fisiologia , Claustrum/fisiologia , Comportamento Impulsivo/fisiologia , Córtex Pré-Frontal/fisiologia , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Estimulantes do Sistema Nervoso Central/farmacologia , Comportamento de Escolha/efeitos dos fármacos , Claustrum/efeitos dos fármacos , Comportamento Impulsivo/efeitos dos fármacos , Masculino , Metanfetamina/farmacologia , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Córtex Pré-Frontal/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/fisiologia
6.
Small ; 16(32): e2001963, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32613757

RESUMO

Poor reversibility and high desorption temperature restricts the practical use of lithium borohydride (LiBH4 ) as an advanced hydrogen store. Herein, a LiBH4 composite confined in unique double-layered carbon nanobowls prepared by a facile melt infiltration process is demonstrated, thanks to powerful capillary effect under 100 bar of H2 pressure. The gradual formation of double-layered carbon nanobowls is witnessed by transmission electron microscopy (TEM) observation. Benefiting from the nanoconfinement effect and catalytic function of carbon, this composite releases hydrogen from 225 °C and peaks at 353 °C, with a hydrogen release amount up to 10.9 wt%. The peak temperature of dehydriding is lowered by 112 °C compared with bulk LiBH4 . More importantly, the composite readily desorbs and absorbs ≈8.5 wt% of H2 at 300 °C and 100 bar H2 , showing a significant reversibility of hydrogen storage. Such a high reversible capacity has not ever been observed under the identical conditions. The usable volumetric energy density reaches as high as 82.4 g L-1 with considerable dehydriding kinetics. The findings provide insights in the design and development of nanosized complex hydrides for on-board applications.

7.
Cell Mol Neurobiol ; 40(2): 229-238, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31974906

RESUMO

Trace amine-associated receptor 1 is one of the best-characterized receptors of trace amines. Growing evidence shows that TAAR1 negatively regulates the monoaminergic activity, including dopamine transmission in the mesocorticolimbic system. Neurochemical assays demonstrated that selective TAAR1 full and partial agonists were effective to prevent psychostimulants-induced dopamine transmission in vitro and in vivo. In the last decade, many preclinical models of psychostimulant addiction such as drug-induced behavioral sensitization, drug-induced conditioned place preference, drug self-administration, drug discrimination, and relapse models were used to assess the effects of TAAR1 agonists on psychostimulants' behavioral effects. In general, activation of TAAR1 attenuated while knockout of TAAR1 potentiated psychostimulant abuse-related behaviors. Here, we review the advances in TAAR1 and its agonists in modulating psychostimulant addiction. We discuss the similarities and differences between the neurochemical and behavioral effects of TAAR1 full and partial agonists. We also discuss several concerns including the abuse liability, sleep reduction, and species-dependent effects that might affect the successful translation of TAAR1 agonists from preclinical studies to clinical application. In conclusion, although further investigations are in need to address certain concerns and the underlying neural mechanisms, TAAR1 agonists appear to be a promising pharmacotherapy to treat psychostimulant addiction and prevent relapse.


Assuntos
Estimulantes do Sistema Nervoso Central/metabolismo , Psicotrópicos/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Animais , Comportamento Aditivo/metabolismo , Comportamento Aditivo/prevenção & controle , Estimulantes do Sistema Nervoso Central/farmacologia , Dopamina/metabolismo , Humanos , Psicotrópicos/farmacologia
8.
Addict Biol ; 22(6): 1475-1485, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27151647

RESUMO

Alcohol dependence is a complex disease involving polygenes, environment and their interactions. Inadequate consideration of these interactions may have hampered the progress on genome-wide association studies of alcohol dependence. By using the dataset of the Study of Addiction: Genetics and Environment with 3838 subjects, we conducted a genome-wide association studies of alcohol dependence symptom count (ADSC) with a full genetic model considering additive, dominance, epistasis and their interactions with ethnicity, as well as conditions of co-morbid substance dependence. Twenty quantitative trait single nucleotide polymorphisms (QTSs) showed highly significant associations with ADSC, including four previously reported genes (ADH1C, PKNOX2, CPE and KCNB2) and the reported intergenic rs1363605, supporting the overall validity of the analysis. Two QTSs within or near ADH1C showed very strong association in a dominance inheritance mode and increased the phenotype value of ADSC when the effect of co-morbid opiate or marijuana dependence was controlled. Highly significant association was also identified in variants within four novel genes (RGS6, FMN1, NRM and BPTF), two non-coding RNA and two epistasis loci. QTS rs7616413, located near PTPRG encoding a protein tyrosine phosphatase receptor, interacted with rs10090742 within ANGPT1 encoding a protein tyrosine phosphatase in an additive × additive or dominance × additive manner. The detected QTSs contributed to about 20 percent of total heritability, in which dominance and epistasis effects accounted for over 50 percent. These results demonstrated that perturbations arising from gene-gene interaction and conditions of co-morbidity substantially influence the genetic architecture of complex trait.


Assuntos
Alcoolismo/epidemiologia , Epistasia Genética/genética , Estudo de Associação Genômica Ampla/métodos , Adolescente , Adulto , Alcoolismo/genética , Comorbidade , Feminino , Estudo de Associação Genômica Ampla/estatística & dados numéricos , Humanos , Masculino , Pessoa de Meia-Idade , Transtornos Relacionados ao Uso de Substâncias/epidemiologia , Transtornos Relacionados ao Uso de Substâncias/genética , Estados Unidos/epidemiologia , Adulto Jovem
9.
BMC Genomics ; 17(1): 897, 2016 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-27829354

RESUMO

BACKGROUND: Waterlily (Nymphaea spp.), a perennial herbaceous aquatic plant, is divided into two ecological groups: hardy waterlily and tropical waterlily. Although the hardy waterlily has no attractive blue flower cultivar, its adaptability is stronger than tropical waterlily because it can survive a cold winter. Thus, breeding hardy waterlily with real blue flowers has become an important target for breeders. Molecular breeding may be a useful way. However, molecular studies on waterlily are limited due to the lack of sequence data. RESULTS: In this study, six cDNA libraries generated from the petals of two different coloring stages of blue tropical waterlily cultivar Nymphaea 'King of Siam' were sequenced using the Illumina HiSeq™ 2500 platform. Each library produced no less than 5.65 Gb clean reads. Subsequently, de novo assembly generated 112,485 unigenes, including 26,206 unigenes annotated to seven public protein databases. Then, 127 unigenes could be identified as putative homologues of color-related genes in other species, including 28 up-regulated and 5 down-regulated unigenes. In petals, 16 flavonoids (4 anthocyanins and 12 flavonols) were detected in different contents during the color development due to the different expression levels of color-related genes, and four flavonols were detected in waterlily for the first time. Furthermore, UA3GTs were selected as the most important candidates involved in the flavonoid metabolic pathway, UA3GTs induced blue petal color formation in Nymphaea 'King of Siam'. CONCLUSIONS: This study will improve our understanding of the molecular mechanism of blue flowers in waterlily and provide the basis for molecular breeding of blue hardy waterlily cultivars.


Assuntos
Flores/genética , Flores/metabolismo , Metaboloma , Nymphaea/genética , Nymphaea/metabolismo , Transcriptoma , Biologia Computacional/métodos , Perfilação da Expressão Gênica , Metabolômica , Fenótipo
10.
Cell Mol Neurobiol ; 36(7): 1189-96, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26825573

RESUMO

Accumulating evidence indicated that N-methyl-D-aspartate (NMDA) receptors are involved in the pathophysiology of depression and implicated in therapeutic targets. NMDA antagonists, such as ketamine, displayed fast-onset and long-lasting antidepressant activity in preclinical and clinical studies. Previous studies showed that Yueju pill exerts antidepressant effects similar to ketamine. Here, we focused on investigating the association of acute and lasting antidepressant responses of Yueju with time course changes of NMDA receptor subunits NR1, NR2A, and NR2B expressions in the hippocampus, a key region regulating depression response. As a result, Yueju reduced immobility time in the forced swimming test from 30 min to 5 days post a single administration. Yueju acutely decreased NR1 and NR2B protein expression in the hippocampus, with NR2A expression unaltered. NR1 expression remained down-regulated 5 days post Yueju administration, whereas NR2B returned to normal level in 24 h. Yueju and ketamine similarly ameliorated the depression-like symptoms at least for 72 h in learned helplessness test. They both reversed the up-regulated expression of NR1 in the learned helpless mice 1 or 3 days post administration. Different from ketamine, the antidepressant effects of Yueju were not influenced by blockade of amino-3-hydroxy-5-methyl-4-isoxazole propionate receptor. These findings served as preclinical evidence that Yueju may confer acute and long-lasting antidepressant effects by favorably modulating NMDA function in the hippocampus.


Assuntos
Antidepressivos/farmacologia , Comportamento Animal/efeitos dos fármacos , Depressão/tratamento farmacológico , Hipocampo/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Transtorno Depressivo/metabolismo , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Hipocampo/metabolismo , Ketamina/farmacologia , Camundongos
11.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 38(2): 187-91, 2016 Apr.
Artigo em Zh | MEDLINE | ID: mdl-27181896

RESUMO

OBJECTIVE: To study the effect of postpartum depression (PPD) on adolescent depression of mice offspring. METHODS: Totally 48 Balb/c female mice were equally randomized into control group and stress group. Control group was not given any stress, whereas stress group were given chronic stress: constraining (6 h/d) combined with light stimulation for 24 hours (twice a week). The stress group was divided into 3 groups to measue the animals' behaviors immediately after modeling, three weeks after modeling, and three weeks after delivery to test whether the PPD models were successfully constructed. The first generation (F1) of normal mothers and PPD-born F1 were as follows: control group (CTL-F1) and PPD offspring group (PPD-F1). The 3-4-week-old male CTL-F1 and PPD-F1 mice (n=8 each) were weighed, and received sucrose preference test, forced swimming test, and novelty-supressed feeding test to measure the depression-like behaviors. RESULTS: The 3-and 4-week-old PPD-F1 had significantly lower body mass than CTL-F1 (P=0.000, P=0.002). Also, the sucrose preference significantly decreased (P=0.000), the forced swimming immobility time significantly increased (P=0.001), the latency to feed significantly increased (P=0.000), while food intake significantly decreased (P=0.005). CONCLUSION: PPD offspring may be more susceptible to depression,with a possible eary onset in adolescence.


Assuntos
Depressão Pós-Parto , Depressão , Estresse Psicológico , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Gravidez , Fatores de Risco
12.
Pharmacol Ther ; 253: 108580, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38142862

RESUMO

Trace amines, a group of amines expressed at the nanomolar level in the mammalian brain, can modulate monoamine transmission. The discovery of and the functional research on the trace amine-associated receptors (TAARs), especially the most well-characterized TAAR1, have largely facilitated our understanding of the function of the trace amine system in the brain. TAAR1 is expressed in the mammalian brain at a low level and widely distributed in the monoaminergic system, including the ventral tegmental area and substantial nigra, where the dopamine neurons reside in the mammalian brain. Growing in vitro and in vivo evidence has demonstrated that TAAR1 could negatively modulate monoamine transmission and play a crucial role in many psychiatric disorders, including schizophrenia, substance use disorders, sleep disorders, depression, and anxiety. Notably, in the last two decades, many studies have repeatedly confirmed the pharmacological effects of the selective TAAR1 ligands in various preclinical models of psychiatric disorders. Recent clinical trials of the dual TAAR1 and serotonin receptor agonist ulotaront also revealed a potential efficacy for treating schizophrenia. Here, we review the current understanding of the TAAR1 system and the recent advances in the elucidation of behavioral and physiological properties of TAAR1 agonists evaluated both in preclinical animal models and clinical trials. We also discuss the potential TAAR1-dependent signaling pathways and the cellular mechanisms underlying the inhibitory effects of TAAR1 activation on drug addiction. We conclude that TAAR1 is an emerging target for the treatment of psychiatric disorders.


Assuntos
Transtornos Mentais , Transtornos Relacionados ao Uso de Substâncias , Animais , Humanos , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Transtornos Mentais/tratamento farmacológico , Transtornos Mentais/metabolismo , Encéfalo/metabolismo , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Aminas/metabolismo , Mamíferos/metabolismo
13.
Chem Commun (Camb) ; 60(9): 1104-1107, 2024 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-38132846

RESUMO

The introduction of local tensile strain in Ni(OH)2 nanosheets accelerates the Ni(OH)2-to-NiOOH transition and boosts the electrocatalytic ammonia oxidation reaction (EAOR), i.e., reducing the onset potential by 80 mV, doubling both the current density and N2 faradaic efficiency, and enabling 1000 hours of operation at 160 mA cm-2.

14.
J Adv Res ; 43: 219-231, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36585110

RESUMO

INTRODUCTION: Adult hippocampal neurogenesis (AHN) is acknowledged to play a critical role in depression. Emerging evidence suggests that the Wnt/ß-catenin pathway can modulate hippocampal neurogenesis. Crocin, a natural carotenoid, possesses antidepressant property. Yet, how it affects neurogenesis and exerts antidepressant response remains unknown. OBJECTIVE: To explore the role of AHN and Wnt/ß-catenin in the antidepressant action of crocin. METHODS: Depressive-related behaviors, including sucrose preference test (SPT), tail suspension test (TST), forced swimming test (FST), and sexual behaviors were performed following crocin treatment. Neurogenesis was characterized via immunohistochemistry, immunofluorescence, Golgi staining and electrophysiology approach. Wnt/ß-catenin signaling was examined with western blot analysis. The role of AHN Wnt/ß-catenin cascade in crocin's antidepressant response was assessed by conditional removal of glial fibrillary acidic protein (GFAP)-expressing newborn neural cells, temozolomide administration, microinfusion of Dkk1 or viral-mediated shRNA of Wnt3a. RESULTS: Crocin decreased the immobility duration in TST and FST without impairing the performance in sexual behaviors. Crocin boosted the proliferation and differentiation of progenitors, and promoted dendritic maturation and functional integration of hippocampal newborn neurons. Conditional removal of GFAP-expressing neural cells or temozolomide administration impaired the antidepressant response of crocin. Additionally, Wnt/ß-catenin signaling was promoted following crocin treatment. In chronic unpredictable mild stress (CUMS) murine model, crocin treatment displayed antidepressant response in SPT, FST and TST, and restored the neurogenesis levels and Wnt/ß-catenin signaling impaired by CUMS. Infusion of Dickkopf-1 (DKK1) or knockdown of Wnt3a in the hippocampus impaired the antidepressant response of crocin. CONCLUSION: Crocin exerted antidepressant response, which was dependent on enhancement of AHN and activation of the Wnt/ß-catenin pathway.


Assuntos
Carotenoides , Hipocampo , Neurogênese , Animais , Camundongos , Antidepressivos/farmacologia , Antidepressivos/metabolismo , beta Catenina/efeitos dos fármacos , beta Catenina/metabolismo , beta Catenina/farmacologia , Carotenoides/metabolismo , Carotenoides/farmacologia , Carotenoides/uso terapêutico , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Neurogênese/efeitos dos fármacos , Neurogênese/fisiologia , Temozolomida/metabolismo , Temozolomida/farmacologia
15.
Adv Pharmacol ; 93: 373-401, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35341572

RESUMO

Trace amine-associated receptor 1 (TAAR1) is the best characterized receptor selectively activated by trace amines. It is broadly expressed in the monoaminergic system in the brain including ventral tegmental area (VTA), nucleus accumbens (NAc), dorsal raphe (DR) and substantial nigra (SN). Extensive studies have suggested that TAAR1 plays an important role in the modulation of monoaminergic system, especially dopamine (DA) transmission which may underlie the mechanisms by which TAAR1 interventions affect drug abuse-like behaviors. TAAR1 activation inhibits the rewarding and reinforcing effects of drugs from different classes including psychostimulants, opioid and alcohol as well as drug-induced increase in DA accumulation. The mechanisms of TAAR1's function in mediating drug abuse-like behaviors are not clear. However, it is hypothesized that TAAR1 interaction with DA transporter (DAT) and dopamine D2 receptor (D2) and the subsequent modulation of cellular cascades may contribute to the effects of TAAR1 in regulating drug abuse. Further studies are needed to investigate the role of TAAR1 in other drugs of abuse-related behaviors and its safety and efficacy for prolonged medications. Together, TAAR1 inhibits drug-induced DA transmission and drug abuse-related behaviors. Therefore, TAAR1 may be a promising therapeutic target for the treatment of drug addiction.


Assuntos
Estimulantes do Sistema Nervoso Central , Transtornos Relacionados ao Uso de Substâncias , Estimulantes do Sistema Nervoso Central/efeitos adversos , Dopamina , Humanos , Receptores Acoplados a Proteínas G , Transtornos Relacionados ao Uso de Substâncias/tratamento farmacológico
16.
Psychopharmacology (Berl) ; 239(10): 3345-3353, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36056214

RESUMO

RATIONALE: Trace amine-associated receptor 1 (TAAR1) is the best-studied receptor of trace amines, a group of biogenic amines expressed at a relatively low level in the mammalian brain. Growing evidence suggests that TAAR1 plays a critical role in various neuropsychiatric disorders. Given that selective TAAR1 agonists were shown to produce pro-cognition and antipsychotic-like effects as well as to suppress drug use and relapse, they have been proposed to be novel treatments for mental disorders such as schizophrenia and addiction. However, the aversive effects of selective TAAR1 agonists remain largely unknown. OBJECTIVES: Here, we evaluated whether the selective TAAR1 full agonist RO5166017 and partial agonist RO5263397 could induce conditioned taste aversion (CTA). RESULTS: We found that RO5166017 and RO5263397 produced significant aversions to both saccharin and NaCl taste novelty. Furthermore, RO5166017 produced CTA to saccharin in TAAR1 heterozygous knockout (taar1±) and wild-type rats but not in TAAR1 homozygous knockout rats (taar1-/-), suggesting that TAAR1 was sufficient for the taste aversive stimulus property of RO5166017. CONCLUSIONS: Taken together, our data indicate that selective TAAR1 agonists could produce strong CTA. Our study urges careful evaluations of the aversive effects of TAAR1 agonists before translating them to clinical use for the treatment of mental disorders.


Assuntos
Antipsicóticos , Receptores Acoplados a Proteínas G , Percepção Gustatória , Animais , Antipsicóticos/química , Antipsicóticos/farmacologia , Agentes Aversivos/química , Agentes Aversivos/farmacologia , Humanos , Mamíferos , Oxazóis , Fenetilaminas/farmacologia , Ratos , Receptores Acoplados a Proteínas G/agonistas , Sacarina/farmacologia , Cloreto de Sódio , Paladar/efeitos dos fármacos , Percepção Gustatória/efeitos dos fármacos
17.
CNS Drugs ; 35(12): 1239-1248, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34766253

RESUMO

Trace amines, including ß-phenylethylamine (ß-PEA), p-tyramine (TYR), tryptamine (TRP), and p-octopamine (OCT), represent a group of amines expressed at low levels in the mammalian brain. Given the close structural similarities to traditional monoamines, links between trace amines and the monoaminergic system have long been suspected. Trace amine-associated receptor 1 (TAAR1), the most well characterized receptor in the TAAR family, has been shown to be potently activated by trace amines such as TYR and PEA. Further, catecholamine metabolites and amphetamine analogs are also potent agonists of TAAR1, implicating the receptor in mediating the monoaminergic system and in substance use disorders. In the central nervous system, TAAR1 is expressed in brain regions involved in dopaminergic, serotonergic, and glutamatergic transmission, and genetic animal models and electrophysiological studies have revealed that TAAR1 is a potent modulator of the monoaminergic system. Selective and potent engineered TAAR1 ligands, including full (RO5166017 and RO5256390) and partial (RO5203648, RO5263397 and RO5073012) agonists and the antagonist EPPTB (N-(3-ethoxyphenyl)-4-(1-pyrrolidinyl)-3-(trifluoromethyl) benzamide, RO5212773), serve as invaluable tools for the investigation of TAAR1 functions and display significant potential for the development of TAAR1-based pharmacotherapies for the treatment of substance use disorders. Despite a number of advances that have been made, more clinical studies are warranted in order to test the potential and efficacy of TAAR1 ligands in the treatment of psychiatric disorders, including substance use disorders.


Assuntos
Receptores Acoplados a Proteínas G/uso terapêutico , Transtornos Relacionados ao Uso de Substâncias/tratamento farmacológico , Animais , Humanos , Ligantes , Modelos Animais , Resultado do Tratamento
18.
Psychopharmacology (Berl) ; 238(11): 3221-3228, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34291306

RESUMO

RATIONALE: Trace amine-associated receptor 1 (TAAR1) is a G protein-coupled receptor that has a particular role in regulating dopaminergic, serotonergic, and glutamatergic transmission. TAAR1 agonists have shown pro-cognitive activities. However, it remains largely unknown of the effects of TAAR1 agonists on memory performance. OBJECTIVES: Here, by using the mice novel object recognition (NOR) test, we examined the effects of the selective TAAR1 partial agonist RO5263397 on recognition memory. RESULTS: We found that RO5263397 significantly enhanced the retrieval of short-term memory (STM; 20 min after training) both in male and female mice. RO5263397 promoted the retrieval of STM in the wild-type (WT) littermates but not TAAR1-KO mice, indicating that the effects of RO5263397 were dependent on TAAR1. Interestingly, compared to their WT litters, TAAR1-KO mice showed similar levels of STM, suggesting that genetic deletion of taar1 gene did not affect the STM retrieval. Furthermore, RO5263397 also promoted the retrieval of long-term NOR memory (24 h after training). CONCLUSIONS: These results indicate that TAAR1 activation promotes NOR memory retrieval. Consistent with previous studies, our finding further suggests that TAAR1 agonists have pro-cognitive properties.


Assuntos
Oxazóis , Receptores Acoplados a Proteínas G , Animais , Dopamina , Feminino , Masculino , Camundongos
19.
Br J Pharmacol ; 178(4): 933-945, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33247948

RESUMO

BACKGROUND AND PURPOSE: Trace amine-associated TA1 receptors play critical roles in regulating dopamine transmission. Previous studies showed that pharmacologically or genetically manipulating the activity of TA1 receptors modulates addiction-like behaviours associated with psychostimulants. However, little is known about whether TA1 receptor modulation would regulate the behavioural effects of opioids. EXPERIMENTAL APPROACH: Effects of the selective TA1 receptor partial agonist RO5263397 on the addiction-related and antinociceptive effects of morphine were systematically assessed in male rats and mice. KEY RESULTS: RO5263397 attenuated the expression of morphine-induced behavioural sensitization in wildtype but not TA1 receptor knockout mice. RO5263397 shifted the dose-effect curve of morphine self-administration downward and reduced the breakpoint in a progressive ratio schedule of reinforcement but did not affect food self-administration in rats. RO5263397 decreased the cue- and drug-induced reinstatement of morphine-seeking behaviour in rats. RO5263397 alone did not trigger reinstatement of morphine-seeking behaviour or change locomotor activity in rats with a history of morphine self-administration. However, RO5263397 did not affect the expression of morphine-induced conditioned place preference in mice or rats. RO5263397 did not affect naltrexone-precipitated jumping behaviour or naltrexone-induced conditioned place aversion in morphine-dependent mice. Furthermore, RO5263397 did not affect the analgesic effects of morphine in an acute nociception model in mice and a chronic pain model in rats. CONCLUSION AND IMPLICATIONS: These results indicated that TA1 receptor activation selectively attenuated the reinforcing, but not withdrawal or antinociceptive effects of morphine, suggesting that selective TA1 receptor agonists might be useful to combat opioid addiction, while sparing the analgesic effects.


Assuntos
Dependência de Morfina , Morfina , Animais , Relação Dose-Resposta a Droga , Masculino , Camundongos , Ratos , Receptores Acoplados a Proteínas G , Reforço Psicológico
20.
Front Pharmacol ; 11: 603445, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33424612

RESUMO

The emphasis of neuronal alterations and adaptations have long been the main focus of the studies of the mechanistic underpinnings of drug addiction. Recent studies have begun to appreciate the role of innate immune system, especially toll-like receptor 4 (TLR4) signaling in drug reward-associated behaviors and physiology. Drugs like opioids, alcohol and psychostimulants activate TLR4 signaling and subsequently induce proinflammatory responses, which in turn contributes to the development of drug addiction. Inhibition of TLR4 or its downstream effectors attenuated the reinforcing effects of opioids, alcohol and psychostimulants, and this effect is also involved in the withdrawal and relapse-like behaviors of different drug classes. However, conflicting results also argue that TLR4-related immune response may play a minimal part in drug addiction. This review discussed the preclinical evidence that whether TLR4 signaling is involved in multiple drug classes action and the possible mechanisms underlying this effect. Moreover, clinical studies which examined the potential efficacy of immune-base pharmacotherapies in treating drug addiction are also discussed.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA