Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
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
2.
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
3.
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.

4.
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
5.
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
6.
J Neurosci ; 34(43): 14304-17, 2014 Oct 22.
Article in English | MEDLINE | ID: mdl-25339743

ABSTRACT

Disruptions in mitochondrial dynamics may contribute to the selective degeneration of dopamine (DA) neurons in Parkinson's disease (PD). However, little is known about the normal functions of mitochondrial dynamics in these neurons, especially in axons where degeneration begins, and this makes it difficult to understand the disease process. To study one aspect of mitochondrial dynamics-mitochondrial fission-in mouse DA neurons, we deleted the central fission protein dynamin-related protein 1 (Drp1). Drp1 loss rapidly eliminates the DA terminals in the caudate-putamen and causes cell bodies in the midbrain to degenerate and lose α-synuclein. Without Drp1, mitochondrial mass dramatically decreases, especially in axons, where the mitochondrial movement becomes uncoordinated. However, in the ventral tegmental area (VTA), a subset of midbrain DA neurons characterized by small hyperpolarization-activated cation currents (Ih) is spared, despite near complete loss of their axonal mitochondria. Drp1 is thus critical for targeting mitochondria to the nerve terminal, and a disruption in mitochondrial fission can contribute to the preferential death of nigrostriatal DA neurons.


Subject(s)
Axons/metabolism , Dopaminergic Neurons/metabolism , Dynamins/deficiency , Mesencephalon/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics/physiology , Animals , Axons/pathology , Dopaminergic Neurons/pathology , Dynamins/genetics , Female , Male , Membrane Potentials/physiology , Mesencephalon/pathology , Mice , Mice, Knockout , Mitochondria/pathology , Organ Culture Techniques
7.
J Neurosci ; 33(15): 6454-9, 2013 Apr 10.
Article in English | MEDLINE | ID: mdl-23575843

ABSTRACT

Activation of mu opioid receptors within the ventral tegmental area (VTA) can produce reward through the inhibition of GABAergic inputs. GABAergic neurons in the ventral pallidum (VP) provide a major input to VTA neurons. To determine the specific VTA neuronal targets of VP afferents and their sensitivity to mu opioid receptor agonists, we virally expressed channel rhodopsin (ChR2) in rat VP neurons and optogenetically activated their terminals in the VTA. Light activation of VP neuron terminals elicited GABAergic IPSCs in both dopamine (DA) and non-DA VTA neurons, and these IPSCs were inhibited by the mu opioid receptor agonist DAMGO. In addition, using a fluorescent retrograde marker to identify VTA-projecting VP neurons, we found them to be hyperpolarized by DAMGO. Both of these actions decrease GABAergic input onto VTA neurons, revealing two mechanisms by which endogenous or exogenous opioids can activate VTA neurons, including DA neurons.


Subject(s)
GABAergic Neurons/physiology , Globus Pallidus/physiology , Receptors, Opioid, mu/physiology , Ventral Tegmental Area/physiology , Analgesics, Opioid/pharmacology , Animals , Channelrhodopsins , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/physiology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , GABAergic Neurons/drug effects , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Male , Neural Pathways/drug effects , Neural Pathways/physiology , Neurons/drug effects , Neurons/physiology , Optogenetics/methods , Photic Stimulation/methods , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/agonists , Ventral Tegmental Area/drug effects
8.
J Neurosci ; 33(45): 17569-76, 2013 Nov 06.
Article in English | MEDLINE | ID: mdl-24198347

ABSTRACT

The mesocorticolimbic system, consisting, at its core, of the ventral tegmental area, the nucleus accumbens, and medial prefrontal cortex, has historically been investigated primarily for its role in positively motivated behaviors and reinforcement learning, and its dysfunction in addiction, schizophrenia, depression, and other mood disorders. Recently, researchers have undertaken a more comprehensive analysis of this system, including its role in not only reward but also punishment, as well as in both positive and negative reinforcement. This focus has been facilitated by new anatomical, physiological, and behavioral approaches to delineate functional circuits underlying behaviors and to determine how this system flexibly encodes and responds to positive and negative states and events, beyond simple associative learning. This review is a summary of topics covered in a mini-symposium at the 2013 Society for Neuroscience annual meeting.


Subject(s)
Association Learning/physiology , Dopaminergic Neurons/physiology , Nerve Net/physiology , Nucleus Accumbens/physiology , Prefrontal Cortex/physiology , Reward , Ventral Tegmental Area/physiology , Animals , Dopamine/physiology
9.
Alcohol Clin Exp Res ; 38(1): 195-203, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24033469

ABSTRACT

BACKGROUND: While there is a growing body of evidence that the delta opioid receptor (DOR) modulates ethanol (EtOH) consumption, development of DOR-based medications is limited in part because there are 2 pharmacologically distinct DOR subtypes (DOR-1 and DOR-2) that can have opposing actions on behavior. METHODS: We studied the behavioral influence of the DOR-1-selective agonist [D-Pen(2) ,D-Pen(5) ]-Enkephalin (DPDPE) and the DOR-2-selective agonist deltorphin microinjected into the ventral tegmental area (VTA) on EtOH consumption and conditioned place preference (CPP) and the physiological effects of these 2 DOR agonists on GABAergic synaptic transmission in VTA-containing brain slices from Lewis rats. RESULTS: Neither deltorphin nor DPDPE induced a significant place preference in EtOH-naïve Lewis rats. However, deltorphin (but not DPDPE) induced a significant CPP in EtOH-drinking rats. In contrast to the previous finding that intra-VTA DOR-1 activity inhibits EtOH consumption and that this inhibition correlates with a DPDPE-induced inhibition of GABA release, here we found no effect of DOR-2 activity on EtOH consumption nor was there a correlation between level of drinking and deltorphin-induced change in GABAergic synaptic transmission. CONCLUSIONS: These data indicate that the therapeutic potential of DOR agonists for alcohol abuse is through a selective action at the DOR-1 form of the receptor.


Subject(s)
Enkephalin, D-Penicillamine (2,5)-/administration & dosage , Ethanol/administration & dosage , Oligopeptides/administration & dosage , Receptors, Opioid, delta/agonists , Reward , Ventral Tegmental Area/drug effects , Alcohol Drinking/psychology , Analgesics, Opioid/administration & dosage , Animals , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Injections, Intraventricular , Male , Rats , Rats, Inbred Lew , Receptors, Opioid, delta/physiology , Ventral Tegmental Area/physiology
10.
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.

11.
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
12.
J Neurosci ; 31(21): 7811-6, 2011 May 25.
Article in English | MEDLINE | ID: mdl-21613494

ABSTRACT

The midbrain ventral tegmental area (VTA) projection to the nucleus accumbens (NAc) is implicated in motivation and reinforcement. A significant number of NAc medium spiny neurons (MSNs) project back to the VTA, although the nature of this projection is essentially unknown. For example, do NAc MSNs directly target accumbens-projecting dopamine neurons and do they act via the GABA(A) or GABA(B) receptor? To address these issues, we expressed the light-sensitive channel rhodopsin-2 in the rat NAc and made electrophysiological recordings from VTA neurons ex vivo. We found that the NAc directly targets non-dopaminergic VTA neurons, including some that project back to the NAc. These MSN GABAergic terminals are opioid sensitive and act via GABA(A) receptors.


Subject(s)
Action Potentials/physiology , Dopamine , Neurons/physiology , Nucleus Accumbens/physiology , Ventral Tegmental Area/physiology , Animals , Dendritic Spines/physiology , Dopamine/physiology , Male , Nerve Net/cytology , Nerve Net/physiology , Neurons/cytology , Nucleus Accumbens/cytology , Rats , Rats, Sprague-Dawley , Ventral Tegmental Area/cytology
13.
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
14.
J Neurosci ; 30(1): 218-29, 2010 Jan 06.
Article in English | MEDLINE | ID: mdl-20053904

ABSTRACT

The ventral tegmental area (VTA) contributes to reward and motivation signaling. In addition to the well established populations of dopamine (DA) or GABA VTA neurons, glutamatergic neurons were recently discovered in the VTA. These glutamatergic neurons express the vesicular glutamate transporter 2, VGluT2. To investigate whether VTA glutamatergic neurons establish local synapses, we tagged axon terminals from resident VTA neurons by intra-VTA injection of Phaseolus vulgaris leucoagglutinin (PHA-L) or an adeno-associated virus encoding wheat germ agglutinin (WGA) and by immunoelectron microscopy determined the presence of VGluT2 in PHA-L- or WGA-positive terminals. We found that PHA-L- or WGA-positive terminals from tagged VTA cells made asymmetric or symmetric synapses within the VTA. VGluT2 immunoreactivity was detected in the vast majority of PHA-L- or WGA-positive terminals forming asymmetric synapses. These results indicate that both VTA glutamatergic and nonglutamatergic (likely GABAergic) neurons establish local synapses. To examine the possible DAergic nature of postsynaptic targets of VTA glutamatergic neurons, we did triple immunolabeling with antibodies against VGluT2, tyrosine hydroxylase (TH), and PHA-L. From triple-labeled tissue, we found that double-labeled PHA-L (+)/VGluT2 (+) axon terminals formed synaptic contacts on dendrites of both TH-positive and TH-negative cells. Consistent with these anatomical observations, in whole-cell slice recordings of VTA neurons we observed that blocking action potential activity significantly decreased the frequency of synaptic glutamatergic events in DAergic and non-DAergic neurons. These observations indicate that resident VTA glutamatergic neurons are likely to affect both DAergic and non-DAergic neurotransmission arising from the VTA.


Subject(s)
Dopamine/physiology , Glutamic Acid/physiology , Neurons/physiology , Synapses/physiology , Ventral Tegmental Area/physiology , Animals , Male , Neurons/ultrastructure , Presynaptic Terminals/physiology , Presynaptic Terminals/ultrastructure , Rats , Rats, Sprague-Dawley , Synapses/ultrastructure , Ventral Tegmental Area/ultrastructure
15.
J Physiol ; 589(17): 4229-42, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21690191

ABSTRACT

Opioid receptors are G-protein-coupled receptors (GPCRs) that modulate synaptic function. Depending upon their nervous system site of action, opioid receptor agonists alter food consumption, pain perception, responses to stress, and drug reward. Opioid receptors signal primarily via G(i/o)-proteins that modulate ion channels to directly inhibit neurons or decrease neurotransmitter release from nerve terminals. Here we report that following stress, activating δ opioid receptors (DORs) on midbrain ventral tegmental area (VTA) neurons causes a novel synaptic effect: the augmentation of GABA(A) receptor (GABA(A)R)-mediated inhibitory postsynaptic currents. Most neurons showing this augmentation were identified as dopaminergic. In addition, in both stressed and unstressed animals, DOR activation decreases GABA(A)R currents in some VTA neurons. Surprisingly, both augmentation and inhibition were also observed when we bypassed the presynaptic terminal by iontophoretically applying GABA, indicating that postsynaptic mechanisms are responsible for both effects. Using a variety of blockers we determined that the augmentation is probably due to insertion of GABA(A)Rs into the synapse by a mechanism that is G-protein independent and mediated by activation of Akt via PI3K. GABA(A)Rs are inserted into the extra-synaptic plasma membrane before trafficking to the synapse, a mechanism consistent with our observation that the DOR-mediated increase in GABA(A)R signalling occurs significantly earlier in iontophoretically applied than in electrically evoked synaptic GABA. This G-protein-independent signalling pathway is not only a novel mechanism of opioid receptor-mediated inhibition, but it also represents the first reported link between activation of a GPCR and insertion of GABA(A)Rs into the plasma membrane.


Subject(s)
Receptors, GABA-A , Ventral Tegmental Area , Animals , Neurons , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Opioid/metabolism
16.
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
17.
J Physiol ; 588(Pt 9): 1541-9, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20231139

ABSTRACT

Substance P (SP) and its receptors are involved in anxiety-related behaviours and regulate the intake of drugs of abuse and alcohol. Within the midbrain ventral tegmental area (VTA), a region that is clearly involved in the control of these behaviours, SP is released by stress and has been shown to trigger relapse. SP activates neurokinin (NK) receptors, which excites midbrain dopamine (DA) neurons and leads to increased DA in target regions. In this study, we have investigated the mechanisms underlying SP actions in the VTA, specifically investigating interactions between SP and GABA(B) receptors. We show that in VTA neurons, NK receptor activation closes an inwardly rectifying potassium channel, and moreover inhibits GABA(B) receptor-mediated transmission through an interaction that depends upon phospholipase C (PLC), intracellular calcium and protein kinase C (PKC).


Subject(s)
GABA Antagonists , GABA-B Receptor Antagonists , Receptors, Neurokinin-2/drug effects , Signal Transduction/drug effects , Substance P/pharmacology , Ventral Tegmental Area/drug effects , Animals , Baclofen/pharmacology , Chelating Agents/pharmacology , Dopamine/physiology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Electrophysiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , GABA Agonists/pharmacology , Immunohistochemistry , In Vitro Techniques , Male , Membrane Potentials/drug effects , Patch-Clamp Techniques , Protein Kinase C/physiology , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-1/drug effects , Tyrosine 3-Monooxygenase/metabolism
18.
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
19.
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
20.
J Neurosci ; 28(48): 12672-81, 2008 Nov 26.
Article in English | MEDLINE | ID: mdl-19036960

ABSTRACT

Alcoholism is a complex and debilitating syndrome affecting approximately 140 million people worldwide. However, not everyone who consumes ethanol develops abuse, raising the possibility that some individuals have a protective mechanism that inhibits elevated alcohol consumption. We tested the hypothesis that the delta-opioid receptor (DOR) plays such a protective role. Here we show that DOR activity in the ventral tegmental area (VTA) robustly decreases ethanol consumption in rats and that these effects depend on baseline ethanol consumption. Intra-VTA microinjection of the DOR agonist DPDPE decreases drinking, particularly in low-drinking animals. Furthermore, VTA microinjection of the DOR selective antagonist TIPP-Psi increases drinking in low, but not high, drinkers and this increase is blocked by comicroinjection of the GABA(A) antagonist bicuculline. Using electrophysiological techniques we found that in VTA brain slices from drinking rats DPDPE presynaptically inhibits GABA(A) receptor mediated IPSCs in low drinkers, but not in high drinkers or naive animals, most likely through activation of DORs on GABA terminals. This DOR-mediated inhibition of IPSCs also correlates inversely with behavioral correlates of anxiety measured in the elevated plus maze. In contrast, presynaptic inhibition of VTA GABA(A) IPSCs by the mu-opioid receptor agonist DAMGO is significantly reduced in both high- and low-drinking rats (<30%) compared with age-matched nondrinking controls (>70%). Together, our findings demonstrate the protective nature of VTA DORs and identify an important new target for therapeutic intervention for alcoholism.


Subject(s)
Alcohol-Induced Disorders, Nervous System/metabolism , Alcoholism/metabolism , Ethanol/pharmacology , Receptors, Opioid, delta/metabolism , Ventral Tegmental Area/metabolism , Alcohol-Induced Disorders, Nervous System/physiopathology , Alcoholism/physiopathology , Analgesics, Opioid/pharmacology , Animals , Anxiety Disorders/chemically induced , Anxiety Disorders/metabolism , Anxiety Disorders/physiopathology , Central Nervous System Depressants/pharmacology , Disease Models, Animal , GABA Antagonists/pharmacology , GABA-A Receptor Antagonists , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Male , Maze Learning/drug effects , Maze Learning/physiology , Microinjections , Narcotic Antagonists/pharmacology , Neural Inhibition/drug effects , Neural Inhibition/physiology , Organ Culture Techniques , Rats , Rats, Inbred Lew , Receptors, GABA-A/metabolism , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Ventral Tegmental Area/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL