Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 May 13.
Article in English | MEDLINE | ID: mdl-38645054

ABSTRACT

Parkinson's disease (PD) is characterized by the death of substantia nigra (SNc) dopamine (DA) neurons, but the pathophysiological mechanisms that precede and drive their death remain unknown. The activity of DA neurons is likely altered in PD, but we understand little about if or how chronic changes in activity may contribute to degeneration. To address this question, we developed a chemogenetic (DREADD) mouse model to chronically increase DA neuron activity, and confirmed this increase using ex vivo electrophysiology. Chronic hyperactivation of DA neurons resulted in prolonged increases in locomotor activity during the light cycle and decreases during the dark cycle, consistent with chronic changes in DA release and circadian disturbances. We also observed early, preferential degeneration of SNc projections, recapitulating the PD hallmarks of selective vulnerability of SNc axons and the comparative resilience of ventral tegmental area axons. This was followed by eventual loss of midbrain DA neurons. Continuous DREADD activation resulted in a sustained increase in baseline calcium levels, supporting an important role for increased calcium in the neurodegeneration process. Finally, spatial transcriptomics from DREADD mice examining midbrain DA neurons and striatal targets, and cross-validation with human patient samples, provided insights into potential mechanisms of hyperactivity-induced toxicity and PD. Our results thus reveal the preferential vulnerability of SNc DA neurons to increased neural activity, and support a potential role for increased neural activity in driving degeneration in PD.

2.
Neuropharmacology ; 225: 109376, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36516892

ABSTRACT

The opioid G-protein coupled receptors (GPCRs) strongly modulate many of the central nervous system structures that contribute to neurological and psychiatric disorders including pain, major depressive disorder, and substance use disorders. To better treat these and related diseases, it is essential to understand the signaling of their endogenous ligands. In this review, we focus on what is known and unknown about the regulation of the over two dozen endogenous peptides with high affinity for one or more of the opioid receptors. We briefly describe which peptides are produced, with a particular focus on the recently proposed possible synthesis pathways for the endomorphins. Next, we describe examples of endogenous opioid peptide expression organization in several neural circuits and how they appear to be released from specific neural compartments that vary across brain regions. We discuss current knowledge regarding the strength of neural activity required to drive endogenous opioid peptide release, clues about how far peptides diffuse from release sites, and their extracellular lifetime after release. Finally, as a translational example, we discuss the mechanisms of action of naltrexone (NTX), which is used clinically to treat alcohol use disorder. NTX is a synthetic morphine analog that non-specifically antagonizes the action of most endogenous opioid peptides developed in the 1960s and FDA approved in the 1980s. We review recent studies clarifying the precise endogenous activity that NTX prevents. Together, the works described here highlight the challenges and opportunities the complex opioid system presents as a therapeutic target.


Subject(s)
Alcoholism , Depressive Disorder, Major , Opioid-Related Disorders , Humans , Alcoholism/drug therapy , Analgesics, Opioid/therapeutic use , Narcotic Antagonists/therapeutic use , Narcotic Antagonists/pharmacology , Depressive Disorder, Major/drug therapy , Opioid Peptides/metabolism , Naltrexone/pharmacology , Opioid-Related Disorders/drug therapy
3.
Nat Commun ; 13(1): 764, 2022 02 09.
Article in English | MEDLINE | ID: mdl-35140231

ABSTRACT

Mu opioid receptor (MOR) agonists are potent analgesics, but also cause sedation, respiratory depression, and addiction risk. The epithalamic lateral habenula (LHb) signals aversive states including pain, and here we found that it is a potent site for MOR-agonist analgesia-like responses in rats. Importantly, LHb MOR activation is not reinforcing in the absence of noxious input. The LHb receives excitatory inputs from multiple sites including the ventral tegmental area, lateral hypothalamus, entopeduncular nucleus, and the lateral preoptic area of the hypothalamus (LPO). Here we report that LHb-projecting glutamatergic LPO neurons are excited by noxious stimulation and are preferentially inhibited by MOR selective agonists. Critically, optogenetic stimulation of LHb-projecting LPO neurons produces an aversive state that is relieved by LHb MOR activation, and optogenetic inhibition of LHb-projecting LPO neurons relieves the aversiveness of ongoing pain.


Subject(s)
Analgesics, Opioid , Receptors, Opioid, mu/agonists , Reinforcement, Psychology , Analgesia , Animals , Female , Habenula/physiology , Hyperalgesia , Hypothalamic Area, Lateral , Male , Neural Pathways/physiology , Neurons/physiology , Preoptic Area , Rats , Ventral Tegmental Area/physiology
4.
J Med Chem ; 64(18): 13873-13892, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34505767

ABSTRACT

Mitragynine and 7-hydroxymitragynine (7OH) are the major alkaloids mediating the biological actions of the psychoactive plant kratom. To investigate the structure-activity relationships of mitragynine/7OH templates, we diversified the aromatic ring of the indole at the C9, C10, and C12 positions and investigated their G-protein and arrestin signaling mediated by mu opioid receptors (MOR). Three synthesized lead C9 analogs replacing the 9-OCH3 group with phenyl (4), methyl (5), or 3'-furanyl [6 (SC13)] substituents demonstrated partial agonism with a lower efficacy than DAMGO or morphine in heterologous G-protein assays and synaptic physiology. In assays limiting MOR reserve, the G-protein efficacy of all three was comparable to buprenorphine. 6 (SC13) showed MOR-dependent analgesia with potency similar to morphine without respiratory depression, hyperlocomotion, constipation, or place conditioning in mice. These results suggest the possibility of activating MOR minimally (G-protein Emax ≈ 10%) in cell lines while yet attaining maximal antinociception in vivo with reduced opioid liabilities.


Subject(s)
Analgesics, Opioid/pharmacology , Receptors, Opioid, mu/agonists , Secologanin Tryptamine Alkaloids/pharmacology , Analgesics, Opioid/adverse effects , Analgesics, Opioid/chemical synthesis , Analgesics, Opioid/metabolism , Animals , Male , Mice, Inbred C57BL , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Rats, Sprague-Dawley , Receptors, Opioid, mu/metabolism , Secologanin Tryptamine Alkaloids/adverse effects , Secologanin Tryptamine Alkaloids/chemical synthesis , Secologanin Tryptamine Alkaloids/metabolism , Structure-Activity Relationship
5.
Eur J Neurosci ; 2021 Feb 22.
Article in English | MEDLINE | ID: mdl-33619763

ABSTRACT

The ventral tegmental area (VTA) contains dopamine neurons intermixed with GABA-releasing (expressing vesicular GABA transporter, VGaT), glutamate-releasing (expressing vesicular glutamate transporter 2, VGluT2), and glutamate-GABA co-releasing (co-expressing VGluT2 and VGaT) neurons. By delivering INTRSECT viral vectors into the VTA of double vglut2-Cre/vgat-Flp transgenic mice, we targeted specific VTA cell populations for ex vivo recordings. We found that VGluT2+ VGaT- and VGluT2+ VGaT+ neurons on average had relatively hyperpolarized resting membrane potential, greater rheobase, and lower spontaneous firing frequency compared to VGluT2- VGaT+ neurons, suggesting that VTA glutamate-releasing and glutamate-GABA co-releasing neurons require stronger excitatory drive to fire than GABA-releasing neurons. In addition, we detected expression of Oprm1mRNA (encoding µ opioid receptors, MOR) in VGluT2+ VGaT- and VGluT2- VGaT+ neurons, and that the MOR agonist DAMGO hyperpolarized neurons with these phenotypes. Collectively, we demonstrate the utility of the double transgenic mouse to access VTA glutamate, glutamate-GABA, and GABA neurons to determine their electrophysiological properties. SIGNIFICANT STATEMENT: Some physiological properties of VTA glutamate-releasing and glutamate-GABA co-releasing neurons are distinct from those of VTA GABA-releasing neurons. µ-opioid receptor activation hyperpolarizes some VTA glutamate-releasing and some GABA-releasing neurons.

6.
PLoS One ; 15(12): e0232864, 2020.
Article in English | MEDLINE | ID: mdl-33373369

ABSTRACT

Activation of the kappa opioid receptor (KOR) contributes to the aversive properties of stress, and modulates key neuronal circuits underlying many neurobehavioral disorders. KOR agonists directly inhibit ventral tegmental area (VTA) dopaminergic neurons, contributing to aversive responses (Margolis et al. 2003, 2006); therefore, selective KOR antagonists represent a novel therapeutic approach to restore circuit function. We used whole cell electrophysiology in acute rat midbrain slices to evaluate pharmacological properties of four novel KOR antagonists: BTRX-335140, BTRX-395750, PF-04455242, and JNJ-67953964. Each compound concentration-dependently reduced the outward current induced by the KOR selective agonist U-69,593. BTRX-335140 and BTRX-395750 fully blocked U-69,593 currents (IC50 = 1.2 ± 0.9 and 1.2 ± 1.3 nM, respectively). JNJ-67953964 showed an IC50 of 3.0 ± 4.6 nM. PF-04455242 exhibited partial antagonist activity asymptoting at 55% blockade (IC50 = 6.7 ± 15.1 nM). In 3/8 of neurons, 1 µM PF-04455242 generated an outward current independent of KOR activation. BTRX-335140 (10 nM) did not affect responses to saturating concentrations of the mu opioid receptor (MOR) agonist DAMGO or the delta opioid receptor (DOR) agonist DPDPE, while JNJ-67953964 (10 nM) partially blocked DAMGO and DPDPE responses. Importantly, BTRX-335140 (10 nM) rapidly washed out with complete recovery of U-69,593 responses within 10 min. Collectively, we show electrophysiological evidence of key differences amongst KOR antagonists that could impact their therapeutic potential and have not been observed using recombinant systems. The results of this study demonstrate the value of characterizing compounds in native neuronal tissue and within circuits implicated in the neurobehavioral disorders of interest.


Subject(s)
Dopaminergic Neurons/drug effects , Membrane Potentials/drug effects , Receptors, Opioid, kappa/antagonists & inhibitors , Analgesics, Opioid/pharmacology , Animals , Benzamides/pharmacology , Biphenyl Compounds/pharmacology , Dopaminergic Neurons/metabolism , Electrophysiology , Enkephalin, D-Penicillamine (2,5)-/pharmacology , Male , Mesencephalon/metabolism , Narcotic Antagonists/pharmacology , Oxadiazoles/pharmacology , Patch-Clamp Techniques/methods , Piperidines/pharmacology , Pyrrolidines/pharmacology , Quinolines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Sulfonamides/pharmacology , Ventral Tegmental Area/drug effects
7.
eNeuro ; 7(5)2020.
Article in English | MEDLINE | ID: mdl-32747458

ABSTRACT

The neuropeptide nociceptin/orphanin FQ (N/OFQ) can be released by stressors and is associated with disorders of emotion regulation and reward processing. N/OFQ and its receptor, NOP, are enriched in dopaminergic pathways, and intra-ventricular agonist delivery decreases dopamine levels in the dorsal striatum, nucleus accumbens (NAc), and ventral tegmental area (VTA). We used whole-cell electrophysiology in acute rat midbrain slices to investigate synaptic actions of N/OFQ. N/OFQ was primarily inhibitory, causing outward currents in both immunocytochemically identified dopaminergic (tyrosine hydroxylase positive (TH(+))) and non-dopaminergic (TH(-)) VTA neurons; effect at 1 µm: 20 ± 4 pA. Surprisingly, this effect was mediated by augmentation of postsynaptic GABAAR currents, unlike the substantia nigra pars compacta (SNc), where the N/OFQ-induced outward currents were K+ channel dependent. A smaller population, 17% of all VTA neurons, responded to low concentrations of N/OFQ with inward currents (10 nm: -11 ± 2 pA). Following 100 nm N/OFQ, the response to a second N/OFQ application was markedly diminished in VTA neurons (14 ± 10% of first response) but not in SNc neurons (90 ± 20% of first response). N/OFQ generated outward currents in medial prefrontal cortex (mPFC)-projecting VTA neurons, but inward currents in a subset of posterior anterior cingulate cortex (pACC)-projecting VTA neurons. While N/OFQ inhibited NAc-projecting VTA cell bodies, it had little effect on electrically or optogenetically evoked terminal dopamine release in the NAc measured ex vivo with fast scan cyclic voltammetry (FSCV). These results extend our understanding of the N/OFQ system in brainstem circuits implicated in many neurobehavioral disorders.


Subject(s)
Receptors, Opioid , Ventral Tegmental Area , Animals , Dopamine , Opioid Peptides , Rats , Receptors, Opioid/metabolism , Ventral Tegmental Area/metabolism , Nociceptin
8.
Mol Pharmacol ; 98(2): 96-108, 2020 08.
Article in English | MEDLINE | ID: mdl-32487735

ABSTRACT

In the mid-1970s, an intense race to identify endogenous substances that activated the same receptors as opiates resulted in the identification of the first endogenous opioid peptides. Since then, >20 peptides with opioid receptor activity have been discovered, all of which are generated from three precursors, proenkephalin, prodynorphin, and proopiomelanocortin, by sequential proteolytic processing by prohormone convertases and carboxypeptidase E. Each of these peptides binds to all three of the opioid receptor types (µ, δ, or κ), albeit with differing affinities. Peptides derived from proenkephalin and prodynorphin are broadly distributed in the brain, and mRNA encoding all three precursors are highly expressed in some peripheral tissues. Various approaches have been used to explore the functions of the opioid peptides in specific behaviors and brain circuits. These methods include directly administering the peptides ex vivo (i.e., to excised tissue) or in vivo (in animals), using antagonists of opioid receptors to infer endogenous peptide activity, and genetic knockout of opioid peptide precursors. Collectively, these studies add to our current understanding of the function of endogenous opioids, especially when similar results are found using different approaches. We briefly review the history of identification of opioid peptides, highlight the major findings, address several myths that are widely accepted but not supported by recent data, and discuss unanswered questions and future directions for research. SIGNIFICANCE STATEMENT: Activation of the opioid receptors by opiates and synthetic drugs leads to central and peripheral biological effects, including analgesia and respiratory depression, but these may not be the primary functions of the endogenous opioid peptides. Instead, the opioid peptides play complex and overlapping roles in a variety of systems, including reward pathways, and an important direction for research is the delineation of the role of individual peptides.


Subject(s)
Opioid Peptides/genetics , Opioid Peptides/metabolism , Receptors, Opioid/metabolism , Animals , Brain/metabolism , Carboxypeptidase H/metabolism , Enkephalins/chemistry , Enkephalins/genetics , Humans , Pro-Opiomelanocortin/chemistry , Pro-Opiomelanocortin/genetics , Proprotein Convertases/metabolism , Protein Precursors/chemistry , Protein Precursors/genetics
9.
Proc Natl Acad Sci U S A ; 117(21): 11820-11828, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32393639

ABSTRACT

Opioids, such as morphine and fentanyl, are widely used for the treatment of severe pain; however, prolonged treatment with these drugs leads to the development of tolerance and can lead to opioid use disorder. The "Opioid Epidemic" has generated a drive for a deeper understanding of the fundamental signaling mechanisms of opioid receptors. It is generally thought that the three types of opioid receptors (µ, δ, κ) are activated by endogenous peptides derived from three different precursors: Proopiomelanocortin, proenkephalin, and prodynorphin. Posttranslational processing of these precursors generates >20 peptides with opioid receptor activity, leading to a long-standing question of the significance of this repertoire of peptides. Here, we address some aspects of this question using a technical tour de force approach to systematically evaluate ligand binding and signaling properties ([35S]GTPγS binding and ß-arrestin recruitment) of 22 peptides at each of the three opioid receptors. We show that nearly all tested peptides are able to activate the three opioid receptors, and many of them exhibit agonist-directed receptor signaling (functional selectivity). Our data also challenge the dogma that shorter forms of ß-endorphin do not exhibit receptor activity; we show that they exhibit robust signaling in cultured cells and in an acute brain slice preparation. Collectively, this information lays the groundwork for improved understanding of the endogenous opioid system that will help in developing more effective treatments for pain and addiction.


Subject(s)
Opioid Peptides , Receptors, Opioid/metabolism , Signal Transduction/physiology , Animals , Cell Line, Tumor , Humans , Male , Opioid Peptides/agonists , Opioid Peptides/metabolism , Pro-Opiomelanocortin/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley
10.
Cell Rep ; 28(11): 2739-2747.e4, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31509737

ABSTRACT

Migraines are a major health burden, but treatment is limited because of inadequate understanding of neural mechanisms underlying headache. Imaging studies of migraine patients demonstrate changes in both pain-modulatory circuits and reward-processing regions, but whether these changes contribute to the experience of headache is unknown. Here, we demonstrate a direct connection between the ventrolateral periaqueductal gray (vlPAG) and the ventral tegmental area (VTA) that contributes to headache aversiveness in rats. Many VTA neurons receive monosynaptic input from the vlPAG, and cranial nociceptive input increases Fos expression in VTA-projecting vlPAG neurons. Activation of PAG inputs to the VTA induces avoidance behavior, while inactivation of these projections induces a place preference only in animals with headache. This work identifies a distinct pathway that mediates cranial nociceptive aversiveness.


Subject(s)
Headache/metabolism , Neural Pathways/metabolism , Neurons/metabolism , Periaqueductal Gray/metabolism , Ventral Tegmental Area/metabolism , Animals , Headache/genetics , Male , Migraine Disorders/genetics , Migraine Disorders/metabolism , Oncogene Proteins v-fos/genetics , Oncogene Proteins v-fos/metabolism , Periaqueductal Gray/cytology , Periaqueductal Gray/radiation effects , Rats , Rats, Sprague-Dawley , Synapses/metabolism , Time Factors , Ventral Tegmental Area/radiation effects
11.
Neurochem Int ; 129: 104504, 2019 10.
Article in English | MEDLINE | ID: mdl-31301327

ABSTRACT

Neural circuits that enable an organism to protect itself by promoting escape from immediate threat and avoidance of future injury are conceptualized to carry an "aversive" signal. One of the key molecular elements of these circuits is the kappa opioid receptor (KOR) and its endogenous peptide agonist, dynorphin. In many cases, the aversive response to an experimental manipulation can be eliminated by selective blockade of KOR function, indicating its necessity in transmitting this signal. The dopamine system, through its contributions to reinforcement learning, is also involved in processing of aversive stimuli, and KOR control of dopamine in the context of aversive behavioral states has been intensely studied. In this review, we have discussed the multiple ways in which the KORs regulate dopamine dynamics with a central focus on dopamine neurons and projections from the ventral tegmental area. At the neuronal level, KOR agonists inhibit dopamine neurons both in the somatodendritic region as well as at terminal release sites, through various signaling pathways and ion channels, and these effects are specific to different synaptic sites. While the dominant hypotheses are that aversive states are driven by decreases in dopamine and increases in dynorphin, reported exceptions to these patterns indicate these ideas require refinement. This is critical given that KOR is being considered as a target for development of new therapeutics for anxiety, depression, pain, and other psychiatric disorders.


Subject(s)
Avoidance Learning/physiology , Dopamine/physiology , Dopaminergic Neurons/physiology , Receptors, Opioid, kappa/physiology , Amygdala/metabolism , Animals , Avoidance Learning/drug effects , Dynorphins/physiology , Forecasting , Learning/physiology , Nucleus Accumbens/metabolism , Punishment , Reinforcement, Psychology , Signal Transduction/physiology , Ventral Tegmental Area/metabolism
12.
J Comp Neurol ; 527(5): 916-941, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30393861

ABSTRACT

The ventral tegmental area (VTA) is a heterogeneous midbrain structure that contains dopamine (DA), GABA, and glutamate neurons that project to many different brain regions. Here, we combined retrograde tracing with immunocytochemistry against tyrosine hydroxylase (TH) or glutamate decarboxylase (GAD) to systematically compare the proportion of dopaminergic and GABAergic VTA projections to 10 target nuclei: anterior cingulate, prelimbic, and infralimbic cortex; nucleus accumbens core, medial shell, and lateral shell; anterior and posterior basolateral amygdala; ventral pallidum; and periaqueductal gray. Overall, the non-dopaminergic component predominated VTA efferents, accounting for more than 50% of all projecting neurons to each region except the nucleus accumbens core. In addition, GABA neurons contributed no more than 20% to each projection, with the exception of the projection to the ventrolateral periaqueductal gray, where the GABAergic contribution approached 50%. Therefore, there is likely a significant glutamatergic component to many of the VTA's projections. We also found that VTA cell bodies retrogradely labeled from the various target brain regions had distinct distribution patterns within the VTA, including in the locations of DA and GABA neurons. Despite this patterned organization, VTA neurons comprising these different projections were intermingled and never limited to any one subregion. These anatomical results are consistent with the idea that VTA neurons participate in multiple distinct, parallel circuits that differentially contribute to motivation and reward. While attention has largely focused on VTA DA neurons, a better understanding of VTA subpopulations, especially the contribution of non-DA neurons to projections, will be critical for future work.


Subject(s)
Dopaminergic Neurons/cytology , GABAergic Neurons/cytology , Ventral Tegmental Area/cytology , Animals , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Dopaminergic Neurons/metabolism , GABAergic Neurons/metabolism , Glutamate Decarboxylase/metabolism , Immunohistochemistry , Male , Neural Pathways/cytology , Neural Pathways/metabolism , Neuroanatomical Tract-Tracing Techniques , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/metabolism
13.
Nat Neurosci ; 21(8): 1072-1083, 2018 08.
Article in English | MEDLINE | ID: mdl-30038277

ABSTRACT

Environmental cues, through Pavlovian learning, become conditioned stimuli that guide animals toward the acquisition of rewards (for example, food) that are necessary for survival. We tested the fundamental role of midbrain dopamine neurons in conferring predictive and motivational properties to cues, independent of external rewards. We found that brief phasic optogenetic excitation of dopamine neurons, when presented in temporal association with discrete sensory cues, was sufficient to instantiate those cues as conditioned stimuli that subsequently both evoked dopamine neuron activity on their own and elicited cue-locked conditioned behavior. Notably, we identified highly parcellated functions for dopamine neuron subpopulations projecting to different regions of striatum, revealing dissociable dopamine systems for the generation of incentive value and conditioned movement invigoration. Our results indicate that dopamine neurons orchestrate Pavlovian conditioning via functionally heterogeneous, circuit-specific motivational signals to create, gate, and shape cue-controlled behaviors.


Subject(s)
Conditioning, Classical/physiology , Dopaminergic Neurons/physiology , Motivation/physiology , Nerve Net/physiology , Animals , Animals, Genetically Modified , Cues , Female , Male , Mesencephalon/cytology , Mesencephalon/physiology , Neostriatum/cytology , Neostriatum/physiology , Optogenetics , Rats , Rats, Sprague-Dawley , Reward , Self Stimulation , Substantia Nigra/cytology , Substantia Nigra/physiology , Ventral Tegmental Area/cytology , Ventral Tegmental Area/physiology
14.
Neuropharmacology ; 123: 420-432, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28645621

ABSTRACT

The mu and delta opioid receptors (MOR and DOR) are highly homologous members of the opioid family of GPCRs. There is evidence that MOR and DOR interact, however the extent to which these interactions occur in vivo and affect synaptic function is unknown. There are two stable DOR subtypes: DPDPE sensitive (DOR1) and deltorphin II sensitive (DOR2); both agonists are blocked by DOR selective antagonists. Robust motivational effects are produced by local actions of both MOR and DOR ligands in the ventral tegmental area (VTA). Here we demonstrate that a majority of both dopaminergic and non-dopaminergic VTA neurons express combinations of functional DOR1, DOR2, and/or MOR, and that within a single VTA neuron, DOR1, DOR2, and MOR agonists can differentially couple to downstream signaling pathways. As reported for the MOR agonist DAMGO, DPDPE and deltorphin II produced either a predominant K+ dependent hyperpolarization or a Cav2.1 mediated depolarization in different neurons. In some neurons DPDPE and deltorphin II produced opposite responses. Excitation, inhibition, or no effect by DAMGO did not predict the response to DPDPE or deltorphin II, arguing against a MOR-DOR interaction generating DOR subtypes. However, in a subset of VTA neurons the DOR antagonist TIPP-Ψ augmented DAMGO responses; we also observed DPDPE or deltorphin II responses augmented by the MOR selective antagonist CTAP. These findings directly support the existence of two independent, stable forms of the DOR, and show that MOR and DOR can interact in some neurons to alter downstream signaling.


Subject(s)
Neurons/metabolism , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Ventral Tegmental Area/metabolism , Animals , Calcium Channels, N-Type/metabolism , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/drug effects , Neurotransmitter Agents/pharmacology , Patch-Clamp Techniques , Potassium Channels/metabolism , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/antagonists & inhibitors , Tissue Culture Techniques , Ventral Tegmental Area/drug effects
15.
Nat Rev Neurosci ; 18(2): 73-85, 2017 02.
Article in English | MEDLINE | ID: mdl-28053327

ABSTRACT

Dopamine-releasing neurons of the ventral tegmental area (VTA) have central roles in reward-related and goal-directed behaviours. VTA dopamine-releasing neurons are heterogeneous in their afferent and efferent connectivity and, in some cases, release GABA or glutamate in addition to dopamine. Recent findings show that motivational signals arising from the VTA can also be carried by non-dopamine-releasing projection neurons, which have their own specific connectivity. Both dopamine-releasing and non-dopamine-releasing VTA neurons integrate afferent signals with local inhibitory or excitatory inputs to generate particular output firing patterns. Various individual inputs, outputs and local connections have been shown to be sufficient to generate reward- or aversion-related behaviour, indicative of the impressive contribution of this small population of neurons to behaviour.


Subject(s)
Neural Pathways/anatomy & histology , Neural Pathways/physiology , Reward , Ventral Tegmental Area/anatomy & histology , Ventral Tegmental Area/physiology , Animals , Dopaminergic Neurons/physiology , GABAergic Neurons/physiology , Glutamic Acid/physiology , Models, Neurological , Motivation/physiology , Synaptic Transmission/physiology
16.
Neuropharmacology ; 110(Pt A): 386-395, 2016 11.
Article in English | MEDLINE | ID: mdl-27475082

ABSTRACT

Enhanced dopamine (DA) neurotransmission from the ventral tegmental area (VTA) to the ventral striatum is thought to drive drug self-administration and mediate positive reinforcement. We examined neuronal firing rates in slices of mouse midbrain following adolescent binge-like alcohol drinking and find that prior alcohol experience greatly enhanced the sensitivity to excitation by ethanol itself (10-50 mM) in a subset of ventral midbrain DA neurons located in the medial VTA. This enhanced response after drinking was not associated with alterations of firing rate or other measures of intrinsic excitability. In addition, the phenomenon appears to be specific to adolescent drinking, as mice that established a drinking preference only after the onset of adulthood showed no change in alcohol sensitivity. Here we demonstrate not only that drinking during adolescence induces enhanced alcohol sensitivity, but also that this DA neuronal response occurs over a range of alcohol concentrations associated with social drinking in humans.


Subject(s)
Binge Drinking/physiopathology , Central Nervous System Depressants/toxicity , Dopaminergic Neurons/drug effects , Ethanol/toxicity , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/growth & development , Action Potentials/drug effects , Alcohol Drinking/pathology , Alcohol Drinking/physiopathology , Animals , Binge Drinking/pathology , Choice Behavior , Disease Models, Animal , Dopaminergic Neurons/pathology , Dopaminergic Neurons/physiology , Dose-Response Relationship, Drug , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Substance Withdrawal Syndrome/pathology , Substance Withdrawal Syndrome/physiopathology , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/pathology , Ventral Tegmental Area/physiopathology , Volition
17.
PLoS One ; 11(7): e0159097, 2016.
Article in English | MEDLINE | ID: mdl-27427945

ABSTRACT

Increased activity of lateral habenula (LHb) neurons is correlated with aversive states including pain, opioid abstinence, rodent models of depression, and failure to receive a predicted reward. Agonists at the mu opioid receptor (MOR) are among the most powerful rewarding and pain relieving drugs. Injection of the MOR agonist morphine directly into the habenula produces analgesia, raising the possibility that MOR acts locally within the LHb. Consequently, we examined the synaptic actions of MOR agonists in the LHb using whole cell patch clamp recording. We found that the MOR selective agonist DAMGO inhibits a subset of LHb neurons both directly and by inhibiting glutamate release onto these cells. Paradoxically, DAMGO also presynaptically inhibited GABA release onto most LHb neurons. The behavioral effect of MOR activation will thus depend upon both the level of intrinsic neuronal activity in the LHb and the balance of activity in glutamate and GABA inputs to different LHb neuronal populations.


Subject(s)
Analgesics, Opioid/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Habenula/drug effects , Habenula/physiology , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Animals , Glutamic Acid/metabolism , Habenula/cytology , Male , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Patch-Clamp Techniques , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/metabolism
18.
Sci Signal ; 9(425): ra43, 2016 04 26.
Article in English | MEDLINE | ID: mdl-27117253

ABSTRACT

PEN is an abundant peptide in the brain that has been implicated in the regulation of feeding. We identified a receptor for PEN in mouse hypothalamus and Neuro2A cells. PEN bound to and activated GPR83, a G protein (heterotrimeric guanine nucleotide)-binding protein)-coupled receptor (GPCR). Reduction of GPR83 expression in mouse brain and Neuro2A cells reduced PEN binding and signaling, consistent with GPR83 functioning as the major receptor for PEN. In some brain regions, GPR83 colocalized with GPR171, a GPCR that binds the neuropeptide bigLEN, another neuropeptide that is involved in feeding and is generated from the same precursor protein as is PEN. Coexpression of these two receptors in cell lines altered the signaling properties of each receptor, suggesting a functional interaction. Our data established PEN as a neuropeptide that binds GPR83 and suggested that these two ligand-receptor systems-PEN-GPR83 and bigLEN-GPR171-may be functionally coupled in the regulation of feeding.


Subject(s)
Hypothalamus/metabolism , Neuropeptide Y/metabolism , Receptors, G-Protein-Coupled/metabolism , Adenosine Triphosphate/metabolism , Animals , Appetite Regulation/physiology , Blotting, Western , CHO Cells , Cell Membrane/metabolism , Cells, Cultured , Cricetulus , HEK293 Cells , Humans , Male , Mice , Phosphorylation , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/genetics
19.
Trends Neurosci ; 38(4): 217-25, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25637939

ABSTRACT

Opioids are the most potent analgesics in clinical use; however, their powerful rewarding properties can lead to addiction. The scientific challenge is to retain analgesic potency while limiting the development of tolerance, dependence, and addiction. Both rewarding and analgesic actions of opioids depend upon actions at the mu opioid (MOP) receptor. Systemic opioid reward requires MOP receptor function in the midbrain ventral tegmental area (VTA) which contains dopaminergic neurons. VTA dopaminergic neurons are implicated in various aspects of reward including reward prediction error, working memory, and incentive salience. It is now clear that subsets of VTA neurons have different pharmacological properties and participate in separate circuits. The degree to which MOP receptor agonists act on different VTA circuits depends upon the behavioral state of the animal, which can be altered by manipulations such as food deprivation or prior exposure to MOP receptor agonists.


Subject(s)
Analgesics, Opioid/pharmacology , Receptors, Opioid, mu/drug effects , Reward , Animals , Dopaminergic Neurons/drug effects , Humans , Receptors, Opioid/drug effects , Reinforcement, Psychology , Ventral Tegmental Area/drug effects
20.
J Neurosci ; 34(44): 14707-16, 2014 Oct 29.
Article in English | MEDLINE | ID: mdl-25355223

ABSTRACT

The ventral tegmental area (VTA) is required for the rewarding and motivational actions of opioids and activation of dopamine neurons has been implicated in these effects. The canonical model posits that opioid activation of VTA dopamine neurons is indirect, through inhibition of GABAergic inputs. However, VTA dopamine neurons also express postsynaptic µ-opioid peptide (MOP) receptors. We report here that in Sprague Dawley rat, the MOP receptor-selective agonist DAMGO (0.5-3 µM) depolarized or increased the firing rate of 87 of 451 VTA neurons (including 22 of 110 dopamine neurons). This DAMGO excitation occurs in the presence of GABAA receptor blockade and its EC50 value is two orders of magnitude lower than for presynaptic inhibition of GABA release on to VTA neurons. Consistent with a postsynaptic channel opening, excitations were accompanied by a decrease in input resistance. Excitations were blocked by CdCl2 (100 µM, n = 5) and ω-agatoxin-IVA (100 nM, n = 3), nonselective and Cav2.1 Ca(2+) channel blockers, respectively. DAMGO also produced a postsynaptic inhibition in 233 of 451 VTA neurons, including 45 of 110 dopamine neurons. The mean reversal potential of the inhibitory current was -78 ± 7 mV and inhibitions were blocked by the K(+) channel blocker BaCl2 (100 µM, n = 7). Blockade of either excitation or inhibition unmasked the opposite effect, suggesting that MOP receptors activate concurrent postsynaptic excitatory and inhibitory processes in most VTA neurons. These results provide a novel direct mechanism for MOP receptor control of VTA dopamine neurons.


Subject(s)
Analgesics, Opioid/pharmacology , Dopaminergic Neurons/drug effects , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Mesencephalon/drug effects , Receptors, Opioid, mu/agonists , Animals , Dopaminergic Neurons/physiology , Male , Mesencephalon/physiology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...