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










Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 44(23)2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38697841

RESUMO

Interneurons in the medial prefrontal cortex (PFC) regulate local neural activity to influence cognitive, motivated, and emotional behaviors. Parvalbumin-expressing (PV+) interneurons are the primary mediators of thalamus-evoked feed-forward inhibition across the mouse cortex, including the anterior cingulate cortex, where they are engaged by inputs from the mediodorsal (MD) thalamus. In contrast, in the adjacent prelimbic (PL) cortex, we find that PV+ interneurons are scarce in the principal thalamorecipient layer 3 (L3), suggesting distinct mechanisms of inhibition. To identify the interneurons that mediate MD-evoked inhibition in PL, we combine slice physiology, optogenetics, and intersectional genetic tools in mice of both sexes. We find interneurons expressing cholecystokinin (CCK+) are abundant in L3 of PL, with cells exhibiting fast-spiking (fs) or non-fast-spiking (nfs) properties. MD inputs make stronger connections onto fs-CCK+ interneurons, driving them to fire more readily than nearby L3 pyramidal cells and other interneurons. CCK+ interneurons in turn make inhibitory, perisomatic connections onto L3 pyramidal cells, where they exhibit cannabinoid 1 receptor (CB1R) mediated modulation. Moreover, MD-evoked feed-forward inhibition, but not direct excitation, is also sensitive to CB1R modulation. Our findings indicate that CCK+ interneurons contribute to MD-evoked inhibition in PL, revealing a mechanism by which cannabinoids can modulate MD-PFC communication.


Assuntos
Colecistocinina , Interneurônios , Inibição Neural , Córtex Pré-Frontal , Animais , Interneurônios/fisiologia , Colecistocinina/metabolismo , Córtex Pré-Frontal/fisiologia , Camundongos , Masculino , Feminino , Inibição Neural/fisiologia , Tálamo/fisiologia , Camundongos Endogâmicos C57BL , Parvalbuminas/metabolismo , Camundongos Transgênicos , Vias Neurais/fisiologia , Optogenética
2.
Cell Rep ; 42(8): 112901, 2023 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-37505982

RESUMO

Individuals with fragile X syndrome (FXS) are frequently diagnosed with autism spectrum disorder (ASD), including increased risk for restricted and repetitive behaviors (RRBs). Consistent with observations in humans, FXS model mice display distinct RRBs and hyperactivity that are consistent with dysfunctional cortico-striatal circuits, an area relatively unexplored in FXS. Using a multidisciplinary approach, we dissect the contribution of two populations of striatal medium spiny neurons (SPNs) in the expression of RRBs in FXS model mice. Here, we report that dysregulated protein synthesis at cortico-striatal synapses is a molecular culprit of the synaptic and ASD-associated motor phenotypes displayed by FXS model mice. Cell-type-specific translational profiling of the FXS mouse striatum reveals differentially translated mRNAs, providing critical information concerning potential therapeutic targets. Our findings uncover a cell-type-specific impact of the loss of fragile X messenger ribonucleoprotein (FMRP) on translation and the sequence of neuronal events in the striatum that drive RRBs in FXS.


Assuntos
Transtorno do Espectro Autista , Síndrome do Cromossomo X Frágil , Animais , Humanos , Camundongos , Síndrome do Cromossomo X Frágil/metabolismo , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/metabolismo , Proteína do X Frágil da Deficiência Intelectual/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Neurônios/metabolismo , Sinapses/metabolismo , Camundongos Knockout , Modelos Animais de Doenças
3.
Nat Neurosci ; 26(1): 92-106, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36522498

RESUMO

The lateral orbitofrontal cortex (lOFC) receives sensory information about food and integrates these signals with expected outcomes to guide future actions, and thus may play a key role in a distributed network of neural circuits that regulate feeding behavior. Here, we reveal a new role for the lOFC in the cognitive control of behavior in obesity. Food-seeking behavior is biased in obesity such that in male obese mice, behaviors are less flexible to changes in the perceived value of the outcome. Obesity is associated with reduced lOFC inhibitory drive and chemogenetic reduction in GABAergic neurotransmission in the lOFC induces obesity-like impairments in goal-directed behavior. Conversely, pharmacological or optogenetic restoration of inhibitory neurotransmission in the lOFC of obese mice reinstates flexible behavior. Our results indicate that obesity-induced disinhibition of the lOFC leads to a failure to update changes in the value of food with satiety, which in turn may influence how individuals make decisions in an obesogenic environment.


Assuntos
Fenômenos Fisiológicos do Sistema Nervoso , Córtex Pré-Frontal , Camundongos , Animais , Masculino , Camundongos Obesos , Córtex Pré-Frontal/fisiologia , Comportamento Animal , Transmissão Sináptica
4.
Cell Rep ; 40(1): 111042, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35793623

RESUMO

Cholinergic interneurons (ChIs) in the nucleus accumbens (NAc) play a central role in motivated behaviors and associated disorders. However, while the activation of ChIs has been well studied in the dorsal striatum, little is known about how they are engaged in the NAc. Here, we find that the ventral hippocampus (vHPC) and the paraventricular nucleus of the thalamus (PVT) are the main excitatory inputs to ChIs in the NAc medial shell. While the PVT activates ChIs, the vHPC evokes a pronounced pause in firing through prominent feedforward inhibition. In contrast to the dorsal striatum, this inhibition reflects strong connections onto ChIs from local parvalbumin interneurons. Our results reveal the mechanisms by which different long-range inputs engage ChIs, highlighting fundamental differences in local connectivity across the striatum.


Assuntos
Interneurônios , Núcleo Accumbens , Colinérgicos , Hipocampo/fisiologia , Interneurônios/fisiologia , Núcleo Accumbens/fisiologia , Parvalbuminas
5.
Neuropsychopharmacology ; 47(3): 728-740, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34663867

RESUMO

Reward and reinforcement processes are critical for survival and propagation of genes. While numerous brain systems underlie these processes, a cardinal role is ascribed to mesolimbic dopamine. However, ventral tegmental area (VTA) dopamine neurons receive complex innervation and various neuromodulatory factors, including input from lateral hypothalamic (LH) orexin/hypocretin neurons which also express and co-release the neuropeptide, dynorphin. Dynorphin in the VTA induces aversive conditioning through the Kappa opioid receptor (KOR) and decreases dopamine when administered intra-VTA. Exogenous application of orexin or orexin 1 receptor (oxR1) antagonists in the VTA bidirectionally modulates dopamine-driven motivation and reward-seeking behaviours, including the attribution of motivational value to primary rewards and associated conditioned stimuli. However, the effect of endogenous stimulation of LH orexin/dynorphin-containing projections to the VTA and the potential contribution of co-released dynorphin on mesolimbic dopamine and reward related processes remains uncharacterised. We combined optogenetic, electrochemical, and behavioural approaches to examine this. We found that optical stimulation of LH orexin/dynorphin inputs in the VTA potentiates mesolimbic dopamine neurotransmission in the nucleus accumbens (NAc) core, produces real time and conditioned place preference, and increases the food cue-directed orientation in a Pavlovian conditioning procedure. LH orexin/dynorphin potentiation of NAc dopamine release and real time place preference was blocked by an oxR1, but not KOR antagonist. Thus, rewarding effects associated with optical stimulation of LH orexin/dynorphin inputs in the VTA are predominantly driven by orexin rather than dynorphin.


Assuntos
Dopamina , Área Tegmentar Ventral , Dopamina/fisiologia , Neurônios Dopaminérgicos/fisiologia , Dinorfinas/farmacologia , Região Hipotalâmica Lateral/fisiologia , Optogenética , Orexinas/farmacologia , Recompensa , Transmissão Sináptica
6.
Cell Rep ; 28(9): 2256-2263.e3, 2019 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-31461643

RESUMO

We examine synaptic connectivity and cocaine-evoked plasticity at specific networks within the nucleus accumbens (NAc). We identify distinct subpopulations of D1+ medium spiny neurons (MSNs) that project to either the ventral pallidum (D1+VP) or the ventral tegmental area (D1+VTA). We show that inputs from the ventral hippocampus (vHPC), but not the basolateral amygdala (BLA), are initially biased onto D1+VTA MSNs. However, repeated cocaine exposure eliminates the bias of vHPC inputs onto D1+VTA MSNs, while strengthening BLA inputs onto D1+VP MSNs. Our results reveal that connectivity and plasticity depend on the specific inputs and outputs of D1+ MSNs and highlight the complexity of cocaine-evoked circuit level adaptations in the NAc.


Assuntos
Cocaína/farmacologia , Inibidores da Captação de Dopamina/farmacologia , Plasticidade Neuronal , Neurônios/efeitos dos fármacos , Núcleo Accumbens/citologia , Animais , Feminino , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/fisiologia , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/fisiologia , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/fisiologia
7.
Front Pharmacol ; 9: 953, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30233365

RESUMO

The trace amine associated receptor 1 (TAAR1) is a G-protein coupled receptor expressed in the monoaminergic regions of the brain, and represents a potential novel therapeutic target for the treatment of neurological disorders. While selective agonists for TAAR1 have been successfully identified, only one high affinity TAAR1 antagonist has been described thus far. We previously identified four potential low potency TAAR1 antagonists through an in silico screen on a TAAR1 homology model. One of the identified antagonists (compound 22) was predicted to have favorable physicochemical properties, which would allow the drug to cross the blood brain barrier. In vivo studies were therefore carried out and showed that compound 22 potentiates amphetamine- and cocaine-mediated locomotor activity. Furthermore, electrophysiology experiments demonstrated that compound 22 increased firing of dopamine neurons similar to EPPTB, the only known TAAR1 antagonist. In order to assess whether the effects of compound 22 were mediated through TAAR1, experiments were carried out on TAAR1-KO mice. The results showed that compound 22 is able to enhance amphetamine- and cocaine-mediated locomotor activity, even in TAAR1-KO mice, suggesting that the in vivo effects of this compound are not mediated by TAAR1. In collaboration with Psychoactive Drug Screening Program, we attempted to determine the targets for compound 22. Psychoactive Drug Screening Program (PDSP) results suggested several potential targets for compound 22 including, the dopamine, norepinephrine and serotonin transporters; as well as sigma 1 and 2 receptors. Our follow-up studies using heterologous cell systems showed that the dopamine transporter is not a target of compound 22. Therefore, the biological target of compound 22 mediating its psychoactive effects still remains unknown.

8.
J Neurosci ; 38(42): 9091-9104, 2018 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-30185462

RESUMO

The nucleus accumbens (NAc) is critical for motivated behavior and is rewired following exposure to drugs of abuse. Medium spiny neurons (MSNs) in the NAc express either D1 or D2 receptors and project to distinct downstream targets. Differential activation of these MSNs depends on both excitation from long-range inputs and inhibition via the local circuit. Assessing how long-range excitatory inputs engage inhibitory circuitry is therefore important for understanding NAc function. Here, we use slice electrophysiology and optogenetics to study ventral hippocampal (vHPC)-evoked feedforward inhibition in the NAc of male and female mice. We find that vHPC-evoked excitation is stronger at D1+ than D1- MSNs, whereas inhibition is unbiased at the two cell types. vHPC inputs contact both parvalbumin-positive (PV+) and somatostatin-positive (SOM+) interneurons, but PV+ cells are preferentially activated. Moreover, suppressing PV+ interneurons indicates they are primarily responsible for vHPC-evoked inhibition. Finally, repeated cocaine exposure alters the excitation of D1+ and D1- MSNs, without concomitant changes to inhibition, shifting the excitation/inhibition balance. Together, our results highlight the contributions of multiple interneuron populations to feedforward inhibition in the NAc. Moreover, they demonstrate that inhibition provides a stable backdrop on which drug-evoked changes to excitation occur within this circuit.SIGNIFICANCE STATEMENT Given the importance of the nucleus accumbens (NAc) in reward learning and drug-seeking behaviors, it is critical to understand what controls the activity of cells in this region. While excitatory inputs to projection neurons in the NAc have been identified, it is unclear how the local inhibitory network becomes engaged. Here, we identify a sparse population of interneurons responsible for feedforward inhibition evoked by ventral hippocampal input and characterize their connections within the NAc. We also demonstrate that the balance of excitation and inhibition that projection neurons experience is altered by exposure to cocaine. Together, this work provides insight into the fundamental circuitry of this region as well as the effects of drugs of abuse.


Assuntos
Cocaína/administração & dosagem , Hipocampo/fisiologia , Inibição Neural , Plasticidade Neuronal , Neurônios/fisiologia , Núcleo Accumbens/fisiologia , Potenciais de Ação , Animais , Feminino , Hipocampo/efeitos dos fármacos , Interneurônios/efeitos dos fármacos , Interneurônios/fisiologia , Locomoção/efeitos dos fármacos , Masculino , Camundongos Transgênicos , Inibição Neural/efeitos dos fármacos , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Optogenética , Parvalbuminas/metabolismo , Receptores de Dopamina D1/fisiologia , Potenciais Sinápticos/efeitos dos fármacos
9.
Br J Pharmacol ; 175(14): 2825-2833, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-28849596

RESUMO

The current view of the midbrain dopaminergic system is evolving towards a complex system of subpopulations of neurons with distinct afferent and efferent connections and, importantly, functionally different intrinsic characteristics. Recent literature on the phenotypic diversity of dopaminergic neurons has outlined that in the ventral tegmental area dopaminergic neurons are not as anatomically or electrophysiologically homogeneous as they were once thought to be. Instead, the midbrain dopaminergic system is now understood to be composed of anatomically and functionally heterogeneous dopaminergic subpopulations receiving specific afferent inputs and with different axonal projections. An additional layer of complexity is the neuromodulation of each of these dopaminergic circuits. This review will examine the distinguishing electrophysiological and neuromodulatory characteristics of the afferent and efferent connections of midbrain dopaminergic neurons. LINKED ARTICLES: This article is part of a themed section on Emerging Areas of Opioid Pharmacology. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.14/issuetoc.


Assuntos
Neurônios Dopaminérgicos/fisiologia , Peptídeos Opioides/fisiologia , Orexinas/fisiologia , Área Tegmentar Ventral/fisiologia , Animais , Humanos
11.
Curr Top Behav Neurosci ; 33: 283-304, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28303403

RESUMO

Dopamine neurons in the ventral tegmental area (VTA) are a critical part of the neural circuits that underlie reward learning and motivation. Dopamine neurons send dense projections throughout the brain and recent observations suggest that both the intrinsic properties and the functional output of dopamine neurons are dependent on projection target and are subject to neuromodulatory influences. Lateral hypothalamic hypocretin (also termed orexin) neurons project to the VTA and contain both hypocretin and dynorphin peptides in the same dense core vesicles suggesting they may be co-released. Hypocretin peptides act at excitatory Gαq protein-coupled receptors and dynorphin acts at inhibitory Gαi/o protein-coupled receptors, which are both expressed on subpopulations of dopamine neurons. This review describes a role for neuromodulation of dopamine neurons and the influence on motivated behaviour in response to natural and drug rewards.


Assuntos
Encéfalo/metabolismo , Plasticidade Neuronal/fisiologia , Orexinas/metabolismo , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Adaptação Fisiológica , Animais , Neurônios Dopaminérgicos/metabolismo , Humanos
12.
Cell Rep ; 18(6): 1346-1355, 2017 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-28178514

RESUMO

Circuit-specific signaling of ventral tegmental area (VTA) dopamine neurons drives different aspects of motivated behavior, but the neuromodulatory control of these circuits is unclear. We tested the actions of co-expressed lateral hypothalamic peptides, orexin A (oxA) and dynorphin (dyn), on projection-target-defined dopamine neurons in mice. We determined that VTA dopamine neurons that project to the nucleus accumbens lateral shell (lAcbSh), medial shell (mAcbSh), and basolateral amygdala (BLA) are largely non-overlapping cell populations with different electrophysiological properties. Moreover, the neuromodulatory effects of oxA and dyn on these three projections differed. OxA selectively increased firing in lAcbSh- and mAcbSh-projecting dopamine neurons. Dyn decreased firing in the majority of mAcbSh- and BLA-projecting dopamine neurons but reduced firing only in a small fraction of those that project to the lAcbSh. In conclusion, the oxA-dyn input to the VTA may drive reward-seeking behavior by tuning dopaminergic output in a projection-target-dependent manner.


Assuntos
Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Dinorfinas/metabolismo , Orexinas/metabolismo , Área Tegmentar Ventral/metabolismo , Animais , Feminino , Hipotálamo/metabolismo , Masculino , Camundongos , Vias Neurais/metabolismo , Neuropeptídeos/metabolismo , Núcleo Accumbens/metabolismo , Recompensa
13.
Nat Med ; 23(3): 355-360, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28134928

RESUMO

Opiates are essential for treating pain, but termination of opiate therapy can cause a debilitating withdrawal syndrome in chronic users. To alleviate or avoid the aversive symptoms of withdrawal, many of these individuals continue to use opiates. Withdrawal is therefore a key determinant of opiate use in dependent individuals, yet its underlying mechanisms are poorly understood and effective therapies are lacking. Here, we identify the pannexin-1 (Panx1) channel as a therapeutic target in opiate withdrawal. We show that withdrawal from morphine induces long-term synaptic facilitation in lamina I and II neurons within the rodent spinal dorsal horn, a principal site of action for opiate analgesia. Genetic ablation of Panx1 in microglia abolished the spinal synaptic facilitation and ameliorated the sequelae of morphine withdrawal. Panx1 is unique in its permeability to molecules up to 1 kDa in size and its release of ATP. We show that Panx1 activation drives ATP release from microglia during morphine withdrawal and that degrading endogenous spinal ATP by administering apyrase produces a reduction in withdrawal behaviors. Conversely, we found that pharmacological inhibition of ATP breakdown exacerbates withdrawal. Treatment with a Panx1-blocking peptide (10panx) or the clinically used broad-spectrum Panx1 blockers, mefloquine or probenecid, suppressed ATP release and reduced withdrawal severity. Our results demonstrate that Panx1-mediated ATP release from microglia is required for morphine withdrawal in rodents and that blocking Panx1 alleviates the severity of withdrawal without affecting opiate analgesia.


Assuntos
Comportamento Animal/efeitos dos fármacos , Conexinas/genética , Microglia/efeitos dos fármacos , Morfina/efeitos adversos , Entorpecentes/efeitos adversos , Proteínas do Tecido Nervoso/genética , Células do Corno Posterior/efeitos dos fármacos , Síndrome de Abstinência a Substâncias/genética , Trifosfato de Adenosina/metabolismo , Animais , Apirase/farmacologia , Western Blotting , Técnicas de Cultura de Células , Técnicas de Cocultura , Conexinas/antagonistas & inibidores , Conexinas/metabolismo , Mefloquina/farmacologia , Camundongos , Microglia/metabolismo , Naloxona/farmacologia , Antagonistas de Entorpecentes/efeitos adversos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Nociceptividade/efeitos dos fármacos , Células do Corno Posterior/metabolismo , Probenecid/farmacologia , Ratos , Síndrome de Abstinência a Substâncias/etiologia , Síndrome de Abstinência a Substâncias/metabolismo
14.
Neuropsychopharmacology ; 42(7): 1480-1490, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28042870

RESUMO

The orbitofrontal cortex (OFC) integrates sensory information with the current value of foods and updates actions based on this information. Obese humans and rats fed a cafeteria diet have impaired devaluation of food rewards, implicating a potential obesity-induced dysfunction of the OFC. We hypothesized that obesity alters OFC pyramidal neuronal structure and function and reduces conditioned suppression of feeding. Rats were given restricted (1 h/day), extended (23 h/day) or no (chow only) access to a cafeteria diet and tested for a conditioned suppression of feeding. Golgi-cox impregnation and whole-cell patch clamp experiments were performed in lateral OFC pyramidal neurons of rats from the 3 feeding groups. Rats with 40 days of extended, but not restricted, access to a cafeteria diet became obese and continued to feed during foot shock-predicting cues. Access to a cafeteria diet induced morphological changes in basilar dendrites of lateral OFC pyramidal neurons. While there were no alterations in excitatory synaptic transmission underlying altered spine density, we observed a more depolarized resting membrane potential. This was accompanied by decreased inhibitory synaptic transmission onto lateral OFC pyramidal neurons due to decreased release probability at GABAergic inputs. These changes could underlie the inability of the OFC to encode changes in the motivation value of food that is observed in obese rodents and humans.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Plasticidade Neuronal/fisiologia , Obesidade/patologia , Córtex Pré-Frontal/patologia , Animais , Ingestão de Energia/fisiologia , Masculino , Obesidade/complicações , Obesidade/fisiopatologia , Técnicas de Cultura de Órgãos , Córtex Pré-Frontal/fisiopatologia , Células Piramidais/fisiologia , Ratos , Ratos Long-Evans
15.
Cereb Cortex ; 26(12): 4524-4539, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-26405054

RESUMO

The orbitofrontal cortex (OFC) integrates information about the environment to guide decision-making. Glutamatergic synaptic transmission mediated through N-methyl-d-aspartate receptors is required for optimal functioning of the OFC. Additionally, abnormal dopamine signaling in this region has been implicated in impulsive behavior and poor cognitive flexibility. Yet, despite the high prevalence of psychostimulants prescribed for attention deficit/hyperactivity disorder, there is little information on how dopamine modulates synaptic transmission in the juvenile or the adult OFC. Using whole-cell patch-clamp recordings in OFC pyramidal neurons, we demonstrated that while dopamine or selective D2-like receptor (D2R) agonists suppress excitatory synaptic transmission of juvenile or adult lateral OFC neurons; in juvenile lateral OFC neurons, higher concentrations of dopamine can target dopamine receptors that couple to a phospholipase C (PLC) signaling pathway to enhance excitatory synaptic transmission. Interfering with the formation of a putative D1R-D2R interaction blocked the potentiation of excitatory synaptic transmission. Furthermore, targeting the putative D1R-D2R complex with a biased agonist, SKF83959, not only enhanced excitatory synaptic transmission in a PLC-dependent manner, but also improved the performance of juvenile rats on a reversal-learning task. Our results demonstrate that dopamine signaling in the lateral OFC differs between juveniles and adults, through potential crosstalk between dopamine receptor subtypes.


Assuntos
Córtex Pré-Frontal/crescimento & desenvolvimento , Córtex Pré-Frontal/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Reversão de Aprendizagem/fisiologia , Animais , Cateteres de Demora , Cognição/efeitos dos fármacos , Cognição/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Função Executiva/efeitos dos fármacos , Função Executiva/fisiologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Neurotransmissores , Técnicas de Patch-Clamp , Córtex Pré-Frontal/efeitos dos fármacos , Células Piramidais/efeitos dos fármacos , Células Piramidais/fisiologia , Ratos Wistar , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/antagonistas & inibidores , Receptores de Dopamina D2/agonistas , Receptores de N-Metil-D-Aspartato/agonistas , Reversão de Aprendizagem/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Técnicas de Cultura de Tecidos
16.
J Neurosci ; 35(18): 7295-303, 2015 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-25948277

RESUMO

Dopamine neurons in the ventral tegmental area (VTA) are a key target of addictive drugs, and neuroplasticity in this region may underlie some of the core features of addiction. From the very first exposure, all drugs of abuse induce synaptic plasticity in the VTA. However, it is not well understood how this diverse group of drugs brings about common synaptic change. Orexin (also known as hypocretin) is a lateral hypothalamic neuropeptide released into the VTA that promotes drug-seeking behaviors and potentiates excitatory synaptic transmission onto VTA dopamine neurons. Here we show that signaling at orexin receptor type 1 (OxR1) in the VTA is required for morphine-induced plasticity of dopamine neurons. Systemic or intra-VTA administration of the OxR1 antagonist SB 334867 in rats blocked a morphine-induced increase in the AMPAR/NMDAR ratio, an increase in presynaptic glutamate release, and a postsynaptic change in AMPAR number or function, including a switch in subunit composition. Furthermore, SB 334867 blocked a morphine-induced decrease in presynaptic GABA release, and a morphine-induced shift in the balance of excitatory and inhibitory synaptic inputs to dopamine neurons. These findings identify a novel role for orexin in morphine-induced plasticity in the VTA and provide a mechanism by which orexin can gate the output of dopamine neurons.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Morfina/farmacologia , Plasticidade Neuronal/fisiologia , Neuropeptídeos/fisiologia , Receptores de Orexina/fisiologia , Transdução de Sinais/fisiologia , Área Tegmentar Ventral/fisiologia , Animais , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/fisiologia , Masculino , Plasticidade Neuronal/efeitos dos fármacos , Orexinas , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos
17.
Br J Pharmacol ; 172(2): 334-48, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24641197

RESUMO

UNLABELLED: Addiction is a devastating disorder that affects 15.3 million people worldwide. While prevalent, few effective treatments exist. Orexin receptors have been proposed as a potential target for anti-craving medications. Orexins, also known as hypocretins, are neuropeptides produced in neurons of the lateral and dorsomedial hypothalamus and perifornical area, which project widely throughout the brain. The absence of orexins in rodents and humans leads to narcolepsy. However, orexins also have an established role in reward seeking. This review will discuss some of the original studies describing the roles of the orexins in reward seeking as well as specific works that were presented at the 2013 International Narcotics Research Conference. Orexin signalling can promote drug-induced plasticity of glutamatergic synapses onto dopamine neurons of the ventral tegmental area (VTA), a brain region implicated in motivated behaviour. Additional evidence suggests that orexin signalling can also promote drug seeking by initiating an endocannabinoid-mediated synaptic depression of GABAergic inputs to the VTA, and thereby disinhibiting dopaminergic neurons. Orexin neurons co-express the inhibitory opioid peptide dynorphin. It has been proposed that orexin in the VTA may not mediate reward per se, but rather occludes the 'anti-reward' effects of dynorphin. Finally, orexin signalling in the prefrontal cortex and the central amygdala is implicated in reinstatement of reward seeking. This review will highlight recent work describing the role of orexin signalling in cellular processes underlying addiction-related behaviours and propose novel hypotheses for the mechanisms by which orexin signalling may impart drug seeking. LINKED ARTICLES: This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neuropeptídeos/metabolismo , Recompensa , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Animais , Dinorfinas/metabolismo , Etanol/administração & dosagem , Humanos , Hipotálamo/metabolismo , Receptores de Orexina/metabolismo , Orexinas , Área Tegmentar Ventral/metabolismo
18.
J Vis Exp ; (70): e3739, 2012 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-23242006

RESUMO

Animal models have been developed to study the reinforcing effects of drugs, including the intravenous self-administration (IVSA) paradigm. The advantages of using an IVSA paradigm to study the reinforcing properties of drugs of abuse such as cocaine include the fact that the drug is self-administered instead of experimenter-administered, the schedule of reinforcement can be altered, and accurate measurement of the quantities of drug consumed as well as the timing and pattern of IV injections can be obtained. Furthermore, the intravenous route of administration avoids potential confounds related to first pass metabolism or taste, and produces rapid increases in blood and brain drug levels. As outlined in this video, intravenous self-administration can be obtained without prior food restriction or prior drug training following careful catheter placement during surgery and meticulous daily catheter flushing and maintenance. Experimental procedures outlined in this paper include a description of animal housing and acclimation methods, operant training using sweetened milk solutions, and catheter implantation surgery.


Assuntos
Injeções Intravenosas/métodos , Modelos Animais , Autoadministração/métodos , Animais , Condicionamento Operante , Feminino , Injeções Intravenosas/instrumentação , Masculino , Camundongos , Autoadministração/instrumentação
19.
Prog Brain Res ; 198: 123-31, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22813972

RESUMO

The ventral tegmental area (VTA) is a brain region centrally involved in the development and expression of a variety of behaviors associated with drug use. Hypocretin (hcrt), also known as orexin, is a lateral hypothalamic neuropeptide that can be released into the VTA. An increasing number of studies show that hcrt in the VTA exerts modulatory effects on a variety of behaviors produced by drugs of abuse. Importantly, at a cellular level, acute application of hcrt in the VTA potentiates N-methyl-D-aspartate receptors expressed in VTA neurons and facilitates the plasticity induced by drugs of abuse. In this review, we discuss evidence that hcrt directly targets dopamine neurons by modulating excitatory synaptic activity and that hcrt action at excitatory synapses onto VTA dopamine neurons plays a central role in motivated behaviors.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Plasticidade Neuronal/fisiologia , Neuropeptídeos/fisiologia , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Área Tegmentar Ventral/fisiopatologia , Animais , Neurônios Dopaminérgicos/metabolismo , Humanos , Motivação/fisiologia , Plasticidade Neuronal/efeitos dos fármacos , Orexinas , Área Tegmentar Ventral/metabolismo
20.
Vitam Horm ; 89: 291-313, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22640620

RESUMO

Hypocretins (also known as orexins) are neuropeptides synthesized in the lateral hypothalamus and perifornical region and projecting widely throughout the brain. They play an important modulatory role in plasticity related to addictive behavior. Hypocretin signaling to the ventral tegmental area (VTA) promotes synaptic plasticity by potentiating glutamatergic inputs to dopamine neurons and is required for the plasticity induced by stimulant drugs like cocaine. Plasticity in the VTA leads to increased output of dopamine neurons and increased release of dopamine in projection areas, which is associated with the development of addiction-related behaviors. Antagonists of hypocretin receptors inhibit some of these behaviors, particularly those with high effort requirements, suggesting a significant role of hypocretin in the motivation to obtain drugs. Furthermore, hypocretin neurons are also targeted by drugs of abuse, such as nicotine. Projections of hypocretin neurons to regions beyond the VTA may also play a significant role in motivation and addiction. Taken together, the hypocretin system may be a prime drug target for treatment of addiction and related disorders.


Assuntos
Estimulantes do Sistema Nervoso Central/farmacologia , Cocaína/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neurônios/efeitos dos fármacos , Neuropeptídeos/metabolismo , Nicotina/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropeptídeos/metabolismo , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Animais , Estimulantes do Sistema Nervoso Central/toxicidade , Cocaína/toxicidade , Humanos , Região Hipotalâmica Lateral/efeitos dos fármacos , Região Hipotalâmica Lateral/metabolismo , Neurônios/metabolismo , Nicotina/toxicidade , Receptores de Orexina , Orexinas , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores de Neuropeptídeos/antagonistas & inibidores , Transmissão Sináptica/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...