Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters











Publication year range
1.
Behav Brain Res ; 462: 114833, 2024 03 28.
Article in English | MEDLINE | ID: mdl-38220059

ABSTRACT

Depression and anxiety are associated with dysfunction of the mesolimbic dopamine system. The rostromedial tegmental nucleus (RMTg) is predominantly composed of GABAergic neurons that exhibit dense projections and strongly inhibit mesolimbic dopaminergic neurons, proposed as a major "brake" for the system. Consequently, the RMTg may be a crucial brain region for regulating these emotions. The central cholinergic system, particularly the muscarinic receptors, plays an important regulatory role in depression and anxiety. M3 muscarinic receptors are distributed on GABAergic neurons in the RMTg, but their involvement in the regulation of depression and anxiety remains uncertain. This study aimed to examine the effects of RMTg M3 muscarinic receptors on regulating depression- and anxiety-like behaviors in adult male Wistar rats, as assessed through the forced swim, tail suspension, and elevated plus maze tests. The results showed that intra-RMTg injections of the M1/M3 muscarinic receptors agonist, pilocarpine (3, 10, and 30 µg/side), or the M3 muscarinic receptors antagonist, 4-DAMP (0.5, 1, and 2 µg/side), did not alter the immobility time in the forced swim and tail suspension tests. Additionally, pilocarpine (30 µg/side) decreased time spent in open arms and increased time in closed arms in the elevated plus maze; while 4-DAMP (1 and 2 µg/side) played the opposite role by increasing time spent in open arms and decreasing time in closed arms. These findings suggest that RMTg M3 muscarinic receptors have differential effects on regulating depression- and anxiety-like behaviors. Enhancing or inhibiting these receptors can produce anxiogenic or anxiolytic effects, but have no impact on depression-like behavior. Therefore, RMTg M3 muscarinic receptors are involved in regulating anxiety and may be a potential therapeutic target for anxiolytic drugs.


Subject(s)
Pilocarpine , Piperidines , Ventral Tegmental Area , Rats , Animals , Male , Pilocarpine/pharmacology , Depression/drug therapy , Rats, Wistar , Receptors, Muscarinic , Anxiety/drug therapy
2.
Neuropharmacology ; 244: 109809, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38048984

ABSTRACT

We recently showed that chemogenetic activation of the locus coeruleus (LC) to the rostromedial tegmental nucleus (RMTg) noradrenergic (NE) pathway significantly blunted binge-like ethanol drinking and induced aversive-like behaviors in mice. The aim of the present study is to determine if silencing this TH + LC → RMTg noradrenergic pathway promotes increased levels of binge-like ethanol intake and reduced ethanol-induced conditioned taste aversion (CTA). To this end, both male and female TH-ires-cre mice on a C57BL/6 J background were cannulated in the RMTg and injected in the LC with rAVV viruses that encode cre-dependent Gi-expressing designer receptor exclusively activated by designer drugs (DREADDs), or its control, to directly control the activity of NE neurons. Inhibition of the LC to RMTg pathway had no effect on the binge-ethanol drinking in a "drinking-in-the-dark" (DID) paradigm. However, when using this paradigm during the light cycle, silencing of this circuit significantly increased ethanol intake without altering sucrose drinking. Moreover, we found that inhibition of this circuit significantly attenuated an ethanol-induced CTA. In addition, when compared to control animals, pairing RMTg-directed Clozapine N-oxide (CNO) with an i.p. injection of 1.5 g/kg ethanol reduced c-Fos activation in the LC, and increased c-Fos expression in the ventral tegmental area (VTA) in Gi-expressing mice. Our data show that inhibition of the TH + LC to the RMTg pathway significantly increased ethanol drinking as well as attenuated ethanol-induced CTA, supporting the involvement of the LC to RMTg noradrenergic circuit as an important protective mechanism against excessive ethanol consumption.


Subject(s)
Ethanol , Locus Coeruleus , Mice , Male , Female , Animals , Ethanol/pharmacology , Photoperiod , Mice, Inbred C57BL , Ventral Tegmental Area , Alcohol Drinking
3.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1045660

ABSTRACT

@#Abstract: Flubromazolam (Flub) is a novel psychoactive substance of benzodiazepines and the mechanism underlying its addiction still remains elusive. This study investigated the reward effect of Flub using conditioned place preference (CPP) mouse model. The neuronal activity was evaluated by c-Fos expression, and the neural circuit was tracked by virus tracing. This study also investigated the regulatory effect of neural circuits on Flub-induced reward effects through chemogenetic approach. The results showed that, at the dose of 3 mg/kg, Flub significantly increased CPP score and c-Fos expression in dopaminergic (DA) neurons of ventral tegmental area (VTA). Inhibition of VTA dopaminergic neuron activity dramatically decreased Flub-induced CPP score. Virus tracing verified GABAergic neuronal projection of medial rostrum tegmental nucleus (RMTg) to VTA dopaminergic neurons. Activation of RMTgGABA→VTADA circuit or blockade of benzodiazepine receptors (BZR) in RMTg significantly decreased Flub-induced CPP score. These results indicate that Flub produced reward effect via BZR-mediated RMTgGABA→VTADA circuit.

4.
Psychopharmacology (Berl) ; 240(10): 2071-2080, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37474756

ABSTRACT

RATIONALE: Opioid drugs indirectly activate dopamine (DA) neurons in the ventral tegmental area (VTA) through a disinhibition mechanism mediated by mu opioid receptors (MORs) present both on the GABA projection neurons located in the medial tegmental nucleus/tail of the VTA (RMTg/tVTA) and on the VTA GABA interneurons. It is well demonstrated that ethanol, like opioid drugs, provokes VTA DA neuron disinhibition by interacting (through its secondary metabolite, salsolinol) with MORs present in VTA GABA interneurons, but it is not known whether ethanol could disinhibit VTA DA neurons through the MORs present in the RMTg/tVTA. OBJECTIVES: The objective of the present study was to determine whether ethanol, directly microinjected into the tVTA/RMTg, is also able to induce VTA DA neurons disinhibition. METHODS: Disinhibition of VTA DA neurons was indirectly assessed through the analysis of the motor activity of rats. Cannulae were placed into the tVTA/RMTg to perform microinjections of DAMGO (0.13 nmol), ethanol (150 or 300 nmol) or acetaldehyde (250 nmol) in animals pre-treated with either aCSF or the irreversible antagonist of MORs, beta-funaltrexamine (beta-FNA; 2.5 nmol). After injections, spontaneous activity was monitored for 30 min. RESULTS: Neither ethanol nor acetaldehyde directly administered into the RMTg/tVTA were able to increase the locomotor activity of rats at doses that, in previous studies performed in the posterior VTA, were effective in increasing motor activities. However, microinjections of 0.13 nmol of DAMGO into the tVTA/RMTg significantly increased the locomotor activity of rats. These activating effects were reduced by local pre-treatment of rats with beta-FNA (2.5 nmol). CONCLUSIONS: The tVTA/RMTg does not appear to be a key brain region for the disinhibiting action of ethanol on VTA DA neurons. The absence of dopamine in the tVTA/RMTg extracellular medium, the lack of local ethanol metabolism or both could explain the present results.


Subject(s)
Analgesics, Opioid , Ethanol , Rats , Animals , Ethanol/pharmacology , Analgesics, Opioid/pharmacology , Dopamine/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Ventral Tegmental Area , Acetaldehyde/metabolism , Acetaldehyde/pharmacology , Receptors, Opioid, mu/metabolism , gamma-Aminobutyric Acid/metabolism
5.
Behav Brain Res ; 446: 114333, 2023 05 28.
Article in English | MEDLINE | ID: mdl-36764486

ABSTRACT

Social interaction is necessary for the development of individuals and society. Social interaction behaviors are rewarding. Similar to exogenous opioids, social interaction behaviors are able to induce rewarding effects that are regulated by the endogenous opioid system as well. As one type of opioid receptor, µ-opioid receptors (MORs), are densely expressed in the rostromedial tegmental nucleus (RMTg), which results in the RMTg being extremely sensitive to rewarding effects induced by exogenous and endogenous opioids. Here, we investigated how RMTg MORs played a role in rewarding effects induced by social interaction behaviors of male Wistar rats, using a conditioned place preference (CPP) model. Results showed that the CPP induced by social interaction behaviors was inhibited when the function of MORs was blocked via injecting CTAP (a selective MOR antagonist) intraperitoneally, and intra-RMTg injections of lower doses of CTAP affected the CPP in the same way. In addition, injecting CTAP intraperitoneally significantly inhibited the expression of pouncing behavior, while intra-RMTg injections of CTAP significantly inhibited the expression of all three types of social behaviors. These results suggest that RMTg MORs may be a crucial target and remain to be further explored in order to better understand the mechanism of the rewarding effects of social interaction behaviors.


Subject(s)
Analgesics, Opioid , Social Interaction , Rats , Male , Animals , Analgesics, Opioid/pharmacology , Rats, Wistar , Tegmentum Mesencephali , Receptors, Opioid/metabolism , Receptors, Opioid, mu/metabolism , Opioid Peptides/metabolism
6.
Neuropsychiatr Dis Treat ; 18: 2957-2965, 2022.
Article in English | MEDLINE | ID: mdl-36573138

ABSTRACT

Objective: There is a relationship between non-rapid eye movement (NREM) sleep and Alzheimer's disease (AD). The rostromedial tegmental nucleus (RMTg) is activated can enhance NREM. Therefore, our experiment was designed to investigate the effects of activation of RMTg by chemical genetic techniques on APP/PS1 mice learning and memory. Materials and Methods: After the AAV-hSyn-hM3Dq-mCherry virus was injected into the RMTg nucleus, CNO solution was intraperitoneally injected to activate RMTg. The new object test and Morris water maze were used to determine the learning and memory level; T2-weighted imaging (T2WI) scanning was performed to analyze the volume of hippocampus and entorhinal cortex of each group; The virus transfection status was determined by laser confocal microscope and use immunohistochemical detection to observe the deposition of Beta Amyloid 1-42 (Aß42). Results: Activation of RMTg by chemical genetic techniques can reduce the escape latency and increase discrimination index (RI) and the number of crossing platform; Activation of RMTg by chemical genetic techniques reduced the atrophy of the entorhinal cortex. Aß42 deposition in the brain was decreased after activation of RMTg. Conclusion: Activation of the RMTg nucleus by chemogenetic techniques can improve the learning and memory impairment in APP/PS1 mice.

7.
Eur J Neurosci ; 2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36479859

ABSTRACT

Drug seeking is associated with the ventral tegmental area (VTA) dopaminergic (DA) activity. Previously, we have shown that brief optogenetic inhibition of VTA DA neurons with 1 s pulses delivered every 9 s attenuates cocaine seeking under extinction conditions in rats without producing overt signs of dysphoria or locomotor sedation. Whether recruitment of neuronal pathways inhibiting VTA neuronal activity would suppress drug seeking remains unknown. Here, we asked if optogenetic stimulation of the lateral habenula (LHb) efferents in the rostromedial tegmental nucleus (RMTg) as well as RMTg efferents in VTA would reduce drug seeking. To investigate this, we measured how recruitment of elements of this inhibitory pathway affects cocaine seeking in male rats under extinction conditions. The effectiveness of brief optogenetic manipulations was confirmed electrophysiologically at the level of electrical activity of VTA DA neurons. Real-time conditioned place aversion (RT-CPA) and open field tests were performed to control for potential dysphoric/sedating effects of brief optogenetic stimulation of LHb-RMTg-VTA circuitry. Optogenetic stimulation of either RMTg or LHb inhibited VTA DAergic neuron firing, whereas similar stimulation of RMTg efferents in VTA or LHb efferents in RMTg reduced cocaine seeking under extinction conditions. Moreover, stimulation of LHb-RMTg efferents produced an effect that was maintained 24 h later, during cocaine seeking test without stimulation. This effect was specific, as brief optogenetic stimulation did not affect locomotor activity and was not aversive. Our results indicate that defined inhibitory pathways can be recruited to inhibit cocaine seeking, providing potential new targets for non-pharmacological treatment of drug craving.

8.
Neurosci Lett ; 786: 136774, 2022 08 24.
Article in English | MEDLINE | ID: mdl-35809878

ABSTRACT

Opioid use disorder mainly results from functional defects in the brain reward loop, which includs the ventral tegmental area (VTA) and nucleus accumbens (NAc; consisting of shell and core, NAcS and NAcC). Reward effects contribute to opioid use disorder. RMTg M3 receptors play a role in opioid reward by regulating the γ-aminobutyric acid (GABA) neuron activity. Dopamine D1 receptors expressed on GABA neurons regulate opioid reward by mediating the dopamine neuron activity in the VTA. Therefore, we investigated the effect of activating M3 receptors by microinjecting pilocarpine into the RMTg along with activating D1 receptors by microinjecting SKF38393 into the VTA on morphine-induced reward effect, using the conditioned place preference (CPP) paradigm (locomotion was also recorded). We also investigated whether the activation of M3 receptors in the RMTg influenced dopamine release in the NAcS. The results showed that the inhibitory role of RMTg pilocarpine (60 µg/rat) infusions in morphine-induced CPP was reversed by VTA SKF38393 (4 µg/rat) infusions. Moreover, morphine (5 mg/kg, i.p.) increased dopamine release in the NAcS, which was blunted by microinjecting pilocarpine (60 µg/rat) into the RMTg. These results indicate that RMTg M3 receptors mediate morphine-induced reward effect, which is probably related to the dopamine activity within the VTA and NAcS. The relationship between RMTg M3 receptors and the mesolimbic dopamine system could be a potential direction for the treatment of opioid use disorder, but further verification through more comprehensive techniques is needed.


Subject(s)
Morphine , Opioid-Related Disorders , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Analgesics, Opioid/pharmacology , Animals , Brain , Cholinergic Agents/pharmacology , Dopamine/pharmacology , Dopaminergic Neurons , Morphine/pharmacology , Nucleus Accumbens , Pilocarpine/pharmacology , Rats , Receptors, Muscarinic , Reward , Ventral Tegmental Area
9.
Behav Brain Res ; 425: 113826, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35247487

ABSTRACT

The tail of the ventral tegmental area (tVTA) or rostromedial tegmental nucleus (RMTg) receives lateral habenula inputs and projects heavily to midbrain dopamine neurons. Midbrain dopamine and lateral habenula neurons participate in learning processes predicting the outcomes of actions, placing the tVTA in a critical location into prediction error pathways. tVTA GABA neurons show electrophysiological inhibition or activation after reward and aversive stimuli, respectively, and their predictive cues. tVTA molecular recruitment, however, is not elicited by all aversive stimuli. Indeed, precipitated opioid withdrawal, repeated footshocks or food restriction raise tVTA Fos expression, whereas various other unpleasant, stressful or painful stimuli does not elicit that molecular response. However, the basis of that difference remains unknown. In the present study, we tried to disentangle whether the tVTA c-Fos induction observed after food restriction was due to the aversive state of food restriction or to procedure-related reward prediction error. To this end, male Sprague-Dawley rats were food-restricted for 7-8 days. During this period, animals were handled and weighed every day before feeding. On the test day, rats underwent several behavioral procedures to explore the impact of food restriction and food-predictive cue exposure on tVTA c-Fos expression. We showed that food restriction per se was not able to recruit c-Fos in the tVTA. On the contrary, the food-predicting cues induced c-Fos locally in the absence of feeding, whereas the food-predicting cues followed by feeding evoked lower c-Fos expression. Overall, our results support the proposed involvement of the tVTA in reward prediction error.


Subject(s)
Habenula , Ventral Tegmental Area , Animals , Dopaminergic Neurons/physiology , Male , Mesencephalon/physiology , Proto-Oncogene Proteins c-fos , Rats , Rats, Sprague-Dawley , Reward , Ventral Tegmental Area/physiology
10.
Neuropharmacology ; 199: 108797, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34547331

ABSTRACT

There is strong evidence that ethanol entails aversive effects that can act as a deterrent to overconsumption. We have found that in doses that support the development of a conditioned taste aversion ethanol increases the activity of tyrosine hydroxylase (TH) positive neurons in the locus coeruleus (LC), a primary source of norepinephrine (NE). Using cre-inducible AAV8-ChR2 viruses in TH-ires-cre mice we found that the LC provides NE projections that innervate the rostromedial tegmental nucleus (RMTg), a brain region that has been implicated in the aversive properties of drugs. Because the neurocircuitry underlying the aversive effects of ethanol is poorly understood, we characterized the role of the LC to RMTg circuit in modulating aversive unconditioned responses and binge-like ethanol intake. Here, both male and female TH-ires-cre mice were cannulated in the RMTg and injected in the LC with rAVV viruses that encode for a Gq-expressing designer receptor exclusively activated by designer drugs (DREADDs) virus, or its control virus, to directly control the activity of NE neurons. A Latin Square paradigm was used to analyze both 20% ethanol and 3% sucrose consumption using the "drinking-in-the-dark" (DID) paradigm. Chemogenetic activation of the LC to RMTg pathway significantly blunted the binge-ethanol drinking, with no effect on the sucrose consumption, increased the emission of mid-frequency vocalizations and induced malaise-like behaviors in mice. The present findings indicate an important involvement of the LC to RMTg pathway in reducing ethanol consumption, and characterize unconditioned aversive reactions induced by activation of this noradrenergic pathway.


Subject(s)
Behavior, Animal/physiology , Binge Drinking/physiopathology , Binge Drinking/therapy , Locus Coeruleus/physiology , Norepinephrine/physiology , Ventral Tegmental Area/physiology , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Disease Models, Animal , Female , Male , Mice , Mice, Transgenic , Signal Transduction/physiology , Vocalization, Animal/drug effects , Vocalization, Animal/physiology
11.
Addict Biol ; 26(6): e13052, 2021 11.
Article in English | MEDLINE | ID: mdl-33969586

ABSTRACT

The rostromedial tegmental nucleus (RMTg), a GABAergic afferent to midbrain dopamine (DA) neurons, has emerged as an integral player in both rewarding and nociceptive responses. While previous studies have demonstrated that acupuncture modulates DA transmission in the mesolimbic reward system originating in the ventral tegmental area (VTA) and projecting to the nucleus accumbens (NAc) and can reduce drug self-administration, the central links between peripheral acupuncture signals and brain reward systems are not well-characterized. Thus, we hypothesised that acupuncture would elicit inhibitory signals from RMTg neurons to brain reward systems. Acupuncture reduced acute cocaine-induced locomotor activity and DA release in a point-specific manner, which was blocked by optogenetic silencing or chemical lesion of the RMTg. The acupuncture effect was mimicked by chemical activation of the RMTg. Acupuncture activated RMTg GABA neurons. In addition, the inhibitory effects of acupuncture on acute cocaine-induced locomotor activity were prevented by electrolytic lesions of the lateral habenula (LHb) or fasciculus retroflexus (FR), areas known to project to the RMTg. These findings suggest that acupuncture recruits the RMTg to reduce the psychomotor responses enhanced by acute cocaine.


Subject(s)
Acupuncture Therapy/methods , Cocaine/pharmacology , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , Tegmentum Mesencephali/metabolism , Animals , GABAergic Neurons/metabolism , Male , Nucleus Accumbens/metabolism , Rats , Rats, Sprague-Dawley , Reward , Ventral Tegmental Area/metabolism
12.
J Neurosci ; 41(18): 4006-4022, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33741724

ABSTRACT

Dopaminergic (DA) neurons of the midbrain are involved in controlling orienting and approach of animals toward relevant external stimuli. The firing of DA neurons is regulated by many brain structures; however, the sensory input is provided predominantly by the ipsilateral superior colliculus (SC). It is suggested that SC also innervates the contralateral rostromedial tegmental nucleus (RMTg)-the main inhibitory input to DA neurons. Therefore, this study aimed to describe the physiology and anatomy of the SC-RMTg pathway. To investigate the anatomic connections within the circuit of interest, anterograde, retrograde, and transsynaptic tract-tracing studies were performed on male Sprague Dawley rats. We have observed that RMTg is monosynaptically innervated predominantly by the lateral parts of the intermediate layer of the contralateral SC. To study the physiology of this neuronal pathway, we conducted in vivo electrophysiological experiments combined with optogenetics; the activity of RMTg neurons was recorded using silicon probes, while either contralateral or ipsilateral SC was optogenetically stimulated. Obtained results revealed that activation of the contralateral SC excites the majority of RMTg neurons, while stimulation of the ipsilateral SC evokes similar proportions of excitatory or inhibitory responses. Consequently, single-unit recordings showed that the activation of RMTg neurons innervated by the contralateral SC, or stimulation of contralateral SC-originating axon terminals within the RMTg, inhibits midbrain DA neurons. Together, the anatomy and physiology of the discovered brain circuit suggest its involvement in the orienting and motivation-driven locomotion of animals based on the direction of external sensory stimuli.SIGNIFICANCE STATEMENT Dopaminergic neurons are the target of predominantly ipsilateral, excitatory innervation originating from the superior colliculus. However, we demonstrate in our study that SC inhibits the activity of dopaminergic neurons on the contralateral side of the brain via the rostromedial tegmental nucleus. In this way, sensory information received by the animal from one hemifield could induce opposite effects on both sides of the dopaminergic system. It was shown that the side to which an animal directs its behavior is a manifestation of asymmetry in dopamine release between left and right striatum. Animals tend to move oppositely to the hemisphere with higher striatal dopamine concentration. This explains how the above-described circuit might guide the behavior of animals according to the direction of incoming sensory stimuli.


Subject(s)
Superior Colliculi/physiology , Ventral Tegmental Area/physiology , Animals , Electric Stimulation , Functional Laterality/physiology , Male , Motivation/physiology , Motor Activity/physiology , Neural Pathways/physiology , Neurons/physiology , Optogenetics , Orientation/physiology , Photic Stimulation , Presynaptic Terminals/physiology , Rats , Rats, Sprague-Dawley , Synapses/physiology
13.
J Comp Neurol ; 529(4): 657-693, 2021 03.
Article in English | MEDLINE | ID: mdl-32621762

ABSTRACT

The parabrachial nucleus (PB) is a complex structure located at the junction of the midbrain and hindbrain. Its neurons have diverse genetic profiles and influence a variety of homeostatic functions. While its cytoarchitecture and overall efferent projections are known, we lack comprehensive information on the projection patterns of specific neuronal subtypes in the PB. In this study, we compared the projection patterns of glutamatergic neurons here with a subpopulation expressing the transcription factor Foxp2 and a further subpopulation expressing the neuropeptide Pdyn. To do this, we injected an AAV into the PB region to deliver a Cre-dependent anterograde tracer (synaptophysin-mCherry) in three different strains of Cre-driver mice. We then analyzed 147 neuroanatomical regions for labeled boutons in every brain (n = 11). Overall, glutamatergic neurons in the PB region project to a wide variety of sites in the cerebral cortex, basal forebrain, bed nucleus of the stria terminalis, amygdala, diencephalon, and brainstem. Foxp2 and Pdyn subpopulations project heavily to the hypothalamus, but not to the cortex, basal forebrain, or amygdala. Among the few differences between Foxp2 and Pdyn cases was a notable lack of Pdyn projections to the ventromedial hypothalamic nucleus. Our results indicate that genetic identity determines connectivity (and therefore, function), providing a framework for mapping all PB output projections based on the genetic identity of its neurons. Using genetic markers to systematically classify PB neurons and their efferent projections will enhance the translation of research findings from experimental animals to humans.


Subject(s)
Enkephalins/biosynthesis , Forkhead Transcription Factors/biosynthesis , Parabrachial Nucleus/metabolism , Protein Precursors/biosynthesis , Repressor Proteins/biosynthesis , Vesicular Glutamate Transport Protein 2/biosynthesis , Animals , Brain Stem/chemistry , Brain Stem/metabolism , Cerebral Cortex/chemistry , Cerebral Cortex/metabolism , Efferent Pathways/chemistry , Efferent Pathways/metabolism , Enkephalins/analysis , Enkephalins/genetics , Female , Forkhead Transcription Factors/analysis , Forkhead Transcription Factors/genetics , Hypothalamus/chemistry , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Parabrachial Nucleus/chemistry , Protein Precursors/analysis , Protein Precursors/genetics , Repressor Proteins/analysis , Repressor Proteins/genetics , Thalamus/chemistry , Thalamus/metabolism , Vesicular Glutamate Transport Protein 2/analysis , Vesicular Glutamate Transport Protein 2/genetics
14.
J Neurosci ; 41(2): 298-306, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33214316

ABSTRACT

The aversive properties associated with drugs of abuse influence both the development of addiction and relapse. Cocaine produces strong aversive effects after rewarding effects wear off, accompanied by increased firing in the lateral habenula (LHb) that contributes to downstream activation of the rostromedial tegmental nucleus (RMTg). However, the sources of this LHb activation are unknown, as the LHb receives many excitatory inputs whose contributions to cocaine aversion remain uncharacterized. Using cFos activation and in vivo electrophysiology in male rats, we demonstrated that the rostral entopeduncular nucleus (rEPN) was the most responsive region to cocaine among LHb afferents examined and that single cocaine infusions induced biphasic responses in rEPN neurons, with inhibition during cocaine's initial rewarding phase transitioning to excitation during cocaine's delayed aversive phase. Furthermore, rEPN lesions reduced cocaine-induced cFos activation by 2-fold in the LHb and by a smaller proportion in the RMTg, while inactivation of the rEPN or the rEPN-LHb pathway attenuated cocaine avoidance behaviors measured by an operant runway task and by conditioned place aversion (CPA). These data show an essential but not exclusive role of rEPN and its projections to the LHb in processing the aversive effects of cocaine, which could serve as a novel target for addiction vulnerability.SIGNIFICANCE STATEMENT Cocaine produces well-known rewarding effects but also strong aversive effects that influence addiction propensity, but whose mechanisms are poorly understood. We had previously reported that the lateral habenula (LHb) is activated by cocaine and contributes to cocaine's aversive effects, and the current findings show that the rostral entopeduncular nucleus (rEPN) is a major contributor to this LHb activation and to conditioned avoidance of cocaine. These findings show a critical, though not exclusive, rEPN role in cocaine's aversive effects, and shed light on the development of addiction.


Subject(s)
Avoidance Learning/drug effects , Cocaine-Related Disorders/psychology , Cocaine/pharmacology , Entopeduncular Nucleus/drug effects , Habenula/drug effects , Animals , Cocaine-Related Disorders/physiopathology , Electrophysiological Phenomena , Entopeduncular Nucleus/physiopathology , Habenula/physiopathology , Male , Neural Pathways/physiopathology , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Reward , Ventral Tegmental Area/physiology
15.
Nat Sci Sleep ; 12: 1215-1223, 2020.
Article in English | MEDLINE | ID: mdl-33380853

ABSTRACT

The rostromedial tegmental nucleus (RMTg), a brake of the dopamine system, is specifically activated by aversive stimuli, such as foot shock. It is principally composed of gamma-aminobutyric acid neurons. However, there is no exact location of the RMTg on the brain stereotaxic atlas. The RMTg can be defined by c-Fos staining elicited by psychostimulants, the position of retrograde-labeled neurons stained by injections into the ventral tegmental area (VTA), the terminal field formed by axons from the lateral habenula, and some molecular markers identified as specifically expressed in the RMTg such as FoxP1. The RMTg receives a broad range of inputs and produces diverse outputs, which indicates that the RMTg has multiple functions. First, the RMTg plays an essential role for non-rapid eye movement sleep. Additionally, the RMTg serves a vital role in response to addiction. Opiates increase the firing rates of dopaminergic neurons in the VTA by acting on µ-opioid receptors on RMTg neurons and their terminals inside the VTA. In this review, we summarize the recent research advances on the anatomical location of the RMTg in rats and mice, its projections, and its regulation of sleep-wake behavior and addiction.

16.
Exp Neurol ; 333: 113433, 2020 11.
Article in English | MEDLINE | ID: mdl-32791155

ABSTRACT

GABAergic neurons in the rostromedial tegmental nucleus (RMTg) receive major input from the lateral habenula (LHb), which conveys negative reward and motivation related information, and project intensively to midbrain dopamine neurons, including those in the ventral tegmental area (VTA) and substantia nigra pars compacta (SNc). The RMTg-VTA circuit has been shown to be linked to the affective behavior, but the role of the RMTg-SNc circuit in aversion and depression has not been well understood. This study demonstrated that exciting or inhibiting VgatRMTg-SNc neurons was sufficient to increase or decrease immobility time in the forced swim test (FST), respectively. Furthermore, exciting the VgatRMTg-SNc pathway caused aversive behavior. Ninety percent of the SNc putative dopamine neurons were inhibited in extracellular recordings. Furthermore, inhibiting the VgatRMTg-SNc pathway reversed behavioral despair in chronic restraint stress (CRS) depression model mice. Manipulations of the pathway did not affect the hedonic value of the reward in the sucrose-preference test (SPT) or general motor function. In conclusion, these results indicate that the VgatRMTg-SNc pathway regulates aversive and despair behavior, which suggests that the RMTg may mediate the role of LHb in negative behaviors through regulating the activity of SNc neurons.


Subject(s)
Avoidance Learning , Pars Compacta/physiopathology , Stress, Psychological/psychology , Ventral Tegmental Area/physiopathology , Anhedonia , Animals , Depression/physiopathology , Depression/psychology , Dopaminergic Neurons , Female , Male , Mice , Motor Activity , Neurons , Restraint, Physical , Reward , Signal Transduction , Swimming/psychology , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
17.
Curr Opin Physiol ; 15: 143-151, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32647777

ABSTRACT

Sleep-wake control is dependent upon multiple brain areas widely distributed throughout the neural axis. Historically, the monoaminergic and cholinergic neurons of the ascending arousal system were the first to be discovered, and it was only relatively recently that GABAergic and glutamatergic wake- and sleep-promoting populations have been identified. Contemporary advances in molecular-genetic tools have revealed both the complexity and heterogeneity of GABAergic NREM sleep-promoting neurons as well as REM sleep-regulating populations in the brainstem such as glutamatergic neurons in the sublaterodorsal nucleus. The sleep-wake cycle progresses from periods of wakefulness to non-rapid eye movement (NREM) sleep and subsequently rapid eye movement (REM) sleep. Each vigilance stage is controlled by multiple neuronal populations, via a complex regulation that is still incompletely understood. In recent years the field has seen a proliferation in the identification and characterization of new neuronal populations involved in sleep-wake control thanks to newer, more powerful molecular genetic tools that are able to reveal neurophysiological functions via selective activation, inhibition and lesion of neuroanatomically defined sub-types of neurons that are widespread in the brain, such as GABAergic and glutamatergic neurons.1,2.

18.
Neurobiol Dis ; 139: 104818, 2020 06.
Article in English | MEDLINE | ID: mdl-32087289

ABSTRACT

Parkinson's disease is a neurodegenerative disorder partly caused by the loss of the dopamine neurons of the nigrostriatal pathway. It is accompanied by motor as well as non-motor symptoms, including pain and depression. The tail of the ventral tegmental area (tVTA) or rostromedial tegmental nucleus (RMTg) is a GABAergic mesopontine structure that acts as a major inhibitory brake for the substantia nigra pars compacta (SNc) dopamine cells, thus controlling their neuronal activity and related motor functions. The present study tested the influence of suppressing this tVTA brake on motor and non-motor symptoms in a rat model of Parkinson's disease. Using behavioral approaches, we showed that male Sprague-Dawley rats with bilateral and partial 6-hydroxydopamine SNc lesion displayed motor impairments in the rotarod test, impairments that were no more present following a co-lesion of the tVTA. Using a larger set of behavioral tests, we then showed that such SNc lesion also led to non-motor symptoms, including lower body weight, lower mechanical nociceptive thresholds in the forceps test and lower thermal nociceptive thresholds in the incremented hot-plate test, and a decreased sucrose preference in a 2-bottle choice paradigm. The excitotoxic co-lesion of the tVTA led to compensation of body weight, mechanical nociceptive thresholds and anhedonia-like behavior. These findings illustrate the major influence that the tVTA exerts on the dopamine system, modulating the motor and non-motor symptoms related to a partial loss of dopamine cells.


Subject(s)
Parkinson Disease/metabolism , Ventral Tegmental Area/metabolism , Anhedonia , Animals , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Male , Models, Theoretical , Neural Pathways/metabolism , Oxidopamine/pharmacology , Pars Compacta/metabolism , Rats , Rats, Sprague-Dawley , Rotarod Performance Test , Substantia Nigra/metabolism
19.
Neurochem Int ; 122: 85-93, 2019 01.
Article in English | MEDLINE | ID: mdl-30448565

ABSTRACT

The GABAergic rostromedial tegmental nucleus (RMTg) has reciprocal connections with the dopaminergic ventral tegmental area and substantia nigra pars compacta (SNc), and is involved in inhibitory control of monoaminergic nuclei. At present, it is not clear whether unilateral 6-hydroxydopamine lesions of the SNc in rats affect AMPA receptor-mediated excitatory transmission in the RMTg. Here we found that lesions of the SNc in rats increased the firing rate of GABAergic neurons and the level of glutamate in the RMTg compared to sham-operated rats. Intra-RMTg injection of AMPA receptor agonist (S)-AMPA increased the firing rate of the GABAergic neurons in both sham-operated and the lesioned rats, while AMPA receptor antagonist NBQX decreased the firing rate of the neurons. Further, intra-RMTg injection of (S)-AMPA decreased the levels of dopamine and serotonin in the medial prefrontal cortex (mPFC) in the two groups of rats; conversely, NBQX increased the levels of dopamine and serotonin. Compared to sham-operated rats, the duration of (S)-AMPA and NBQX action on the firing rate of GABAergic neurons in the RMTg and release of doapmine and serotonin in the mPFC was prolonged in the lesioned rats. In addition, lesions of the SNc in rats increased protein expression of t-GluR1 and p-GluR1-S831 subunits compared to sham-operated rats. Therefore, these changes in the lesioned rats are associated with increased release of glutamate and up-regulated expression of GluR1 subunit-containing AMPA receptors in the RMTg, which suggest that degeneration of the nigrostriatal pathway enhances AMPA receptor-mediated excitatory transmission in the RMTg.


Subject(s)
Neural Pathways/drug effects , Oxidopamine/pharmacology , Receptors, AMPA/metabolism , Synaptic Transmission/drug effects , Action Potentials/drug effects , Action Potentials/physiology , Animals , Dopamine/metabolism , Interneurons/drug effects , Interneurons/metabolism , Male , Neural Pathways/metabolism , Prefrontal Cortex/drug effects , Rats, Sprague-Dawley , Receptors, AMPA/drug effects , Substantia Nigra/drug effects , Ventral Tegmental Area/drug effects
20.
Proc Natl Acad Sci U S A ; 115(22): 5792-5797, 2018 05 29.
Article in English | MEDLINE | ID: mdl-29752382

ABSTRACT

The neural mechanisms conferring reduced motivation, as observed in depressed individuals, is poorly understood. Here, we examine in rodents if reduced motivation to exert effort is controlled by transmission from the lateral habenula (LHb), a nucleus overactive in depressed-like states, to the rostromedial tegmental nucleus (RMTg), a nucleus that inhibits dopaminergic neurons. In an aversive test wherein immobility indicates loss of effort, LHb→RMTg transmission increased during transitions into immobility, driving LHb→RMTg increased immobility, and inhibiting LHb→RMTg produced the opposite effects. In an appetitive test, driving LHb→RMTg reduced the effort exerted to receive a reward, without affecting the reward's hedonic property. Notably, LHb→RMTg stimulation only affected specific aspects of these motor tasks, did not affect all motor tasks, and promoted avoidance, indicating that LHb→RMTg activity does not generally reduce movement but appears to carry a negative valence that reduces effort. These results indicate that LHb→RMTg activity controls the motivation to exert effort and may contribute to the reduced motivation in depression.


Subject(s)
Habenula/physiology , Motivation/physiology , Neural Pathways/physiology , Tegmentum Mesencephali/physiology , Animals , Depression , Humans , Movement/physiology , Optogenetics , Photometry , Rats , Task Performance and Analysis
SELECTION OF CITATIONS
SEARCH DETAIL