Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38746090

ABSTRACT

The anterior cingulate cortex plays a pivotal role in the cognitive and affective aspects of pain perception. Both endogenous and exogenous opioid signaling within the cingulate mitigate cortical nociception, reducing pain unpleasantness. However, the specific functional and molecular identities of cells mediating opioid analgesia in the cingulate remain elusive. Given the complexity of pain as a sensory and emotional experience, and the richness of ethological pain-related behaviors, we developed a standardized, deep-learning platform for deconstructing the behavior dynamics associated with the affective component of pain in mice-LUPE (Light aUtomated Pain Evaluator). LUPE removes human bias in behavior quantification and accelerated analysis from weeks to hours, which we leveraged to discover that morphine altered attentional and motivational pain behaviors akin to affective analgesia in humans. Through activity-dependent genetics and single-nuclei RNA sequencing, we identified specific ensembles of nociceptive cingulate neuron-types expressing mu-opioid receptors. Tuning receptor expression in these cells bidirectionally modulated morphine analgesia. Moreover, we employed a synthetic opioid receptor promoter-driven approach for cell-type specific optical and chemical genetic viral therapies to mimic morphine's pain-relieving effects in the cingulate, without reinforcement. This approach offers a novel strategy for precision pain management by targeting a key nociceptive cortical circuit with on-demand, non-addictive, and effective analgesia.

2.
Sci Adv ; 10(17): eadj9581, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38669335

ABSTRACT

The supraspinal descending pain modulatory system (DPMS) shapes pain perception via monoaminergic modulation of sensory information in the spinal cord. However, the role and synaptic mechanisms of descending noradrenergic signaling remain unclear. Here, we establish that noradrenergic neurons of the locus coeruleus (LC) are essential for supraspinal opioid antinociception. While much previous work has emphasized the role of descending serotonergic pathways, we find that opioid antinociception is primarily driven by excitatory output from the ventrolateral periaqueductal gray (vlPAG) to the LC. Furthermore, we identify a previously unknown opioid-sensitive inhibitory input from the rostroventromedial medulla (RVM), the suppression of which disinhibits LC neurons to drive spinal noradrenergic antinociception. We describe pain-related activity throughout this circuit and report the presence of prominent bifurcating outputs from the vlPAG to the LC and the RVM. Our findings substantially revise current models of the DPMS and establish a supraspinal antinociceptive pathway that may contribute to multiple forms of descending pain modulation.


Subject(s)
Analgesics, Opioid , Locus Coeruleus , Medulla Oblongata , Pain , Periaqueductal Gray , Locus Coeruleus/metabolism , Locus Coeruleus/drug effects , Periaqueductal Gray/metabolism , Periaqueductal Gray/drug effects , Animals , Medulla Oblongata/metabolism , Medulla Oblongata/drug effects , Pain/drug therapy , Pain/metabolism , Analgesics, Opioid/pharmacology , Male , Adrenergic Neurons/metabolism , Adrenergic Neurons/drug effects , Mice , Neural Pathways/drug effects
3.
bioRxiv ; 2023 Oct 10.
Article in English | MEDLINE | ID: mdl-37873091

ABSTRACT

The supraspinal descending pain modulatory system (DPMS) shapes pain perception via monoaminergic modulation of sensory information in the spinal cord. However, the role and synaptic mechanisms of descending noradrenergic signaling remain unclear. Here, we establish that noradrenergic neurons of the locus coeruleus (LC) are essential for supraspinal opioid antinociception. Unexpectedly, given prior emphasis on descending serotonergic pathways, we find that opioid antinociception is primarily driven by excitatory output from the ventrolateral periaqueductal gray (vlPAG) to the LC. Furthermore, we identify a previously unknown opioid-sensitive inhibitory input from the rostroventromedial medulla (RVM), the suppression of which disinhibits LC neurons to drive spinal noradrenergic antinociception. We also report the presence of prominent bifurcating outputs from the vlPAG to the LC and the RVM. Our findings significantly revise current models of the DPMS and establish a novel supraspinal antinociceptive pathway that may contribute to multiple forms of descending pain modulation.

4.
Neuron ; 111(24): 3926-3940.e10, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-37848025

ABSTRACT

Traditional methods for site-specific drug delivery in the brain are slow, invasive, and difficult to interface with recordings of neural activity. Here, we demonstrate the feasibility and experimental advantages of in vivo photopharmacology using "caged" opioid drugs that are activated in the brain with light after systemic administration in an inactive form. To enable bidirectional manipulations of endogenous opioid receptors in vivo, we developed photoactivatable oxymorphone (PhOX) and photoactivatable naloxone (PhNX), photoactivatable variants of the mu opioid receptor agonist oxymorphone and the antagonist naloxone. Photoactivation of PhOX in multiple brain areas produced local changes in receptor occupancy, brain metabolic activity, neuronal calcium activity, neurochemical signaling, and multiple pain- and reward-related behaviors. Combining PhOX photoactivation with optical recording of extracellular dopamine revealed adaptations in the opioid sensitivity of mesolimbic dopamine circuitry in response to chronic morphine administration. This work establishes a general experimental framework for using in vivo photopharmacology to study the neural basis of drug action.


Subject(s)
Analgesics, Opioid , Oxymorphone , Analgesics, Opioid/pharmacology , Oxymorphone/pharmacology , Pharmaceutical Preparations , Dopamine/metabolism , Naloxone/pharmacology , Receptors, Opioid, mu/metabolism
5.
J Am Chem Soc ; 145(36): 19611-19621, 2023 09 13.
Article in English | MEDLINE | ID: mdl-37649440

ABSTRACT

Photoactivatable neuropeptides offer a robust stimulus-response relationship that can drive mechanistic studies into the physiological mechanisms of neuropeptidergic transmission. The majority of neuropeptides contain a C-terminal amide, which offers a potentially general site for installation of a C-terminal caging group. Here, we report a biomimetic caging strategy in which the neuropeptide C-terminus is extended via a photocleavable amino acid to mimic the proneuropeptides found in large dense-core vesicles. We explored this approach with four prominent neuropeptides: gastrin-releasing peptide (GRP), oxytocin (OT), substance P (SP), and cholecystokinin (CCK). C-terminus extension greatly reduced the activity of all four peptides at heterologously expressed receptors. In cell type-specific electrophysiological recordings from acute brain slices, subsecond flashes of ultraviolet light produced rapidly activating membrane currents via activation of endogenous G protein-coupled receptors. Subsequent mechanistic studies with caged CCK revealed a role for extracellular proteases in shaping the temporal dynamics of CCK signaling, and a striking switch-like, cell-autonomous anti-opioid effect of transient CCK signaling in hippocampal parvalbumin interneurons. These results suggest that C-terminus extension with a photocleavable linker may be a general strategy for photocaging amidated neuropeptides and demonstrate how photocaged neuropeptides can provide mechanistic insights into neuropeptide signaling that are inaccessible using conventional approaches.


Subject(s)
Biomimetics , Neuropeptides , Amides , Amino Acids , Analgesics, Opioid
6.
Biol Psychiatry ; 94(11): 842-851, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37285896

ABSTRACT

BACKGROUND: Mu opioid receptors (MORs) are key for reward processing, mostly studied in dopaminergic pathways. MORs are also expressed in the dorsal raphe nucleus (DRN), which is central for the modulation of reward and mood, but MOR function in the DRN remains underexplored. Here, we investigated whether MOR-expressing neurons of the DRN (DRN-MOR neurons) participate in reward and emotional responses. METHODS: We characterized DRN-MOR neurons anatomically using immunohistochemistry and functionally using fiber photometry in responses to morphine and rewarding/aversive stimuli. We tested the effect of opioid uncaging on the DRN on place conditioning. We examined the effect of DRN-MOR neuron optostimulation on positive reinforcement and mood-related behaviors. We mapped their projections and selected DRN-MOR neurons projecting to the lateral hypothalamus for a similar optogenetic experimentation. RESULTS: DRN-MOR neurons form a heterogeneous neuronal population essentially composed of GABAergic (gamma-aminobutyric acidergic) and glutamatergic neurons. Calcium activity of DRN-MOR neurons was inhibited by rewarding stimuli and morphine. Local photo-uncaging of oxymorphone in the DRN produced conditioned place preference. DRN-MOR neuron optostimulation triggered real-time place preference and was self-administered, promoted social preference, and reduced anxiety and passive coping. Finally, specific optostimulation of DRN-MOR neurons projecting to the lateral hypothalamus recapitulated the reinforcing effects of total DRN-MOR neuron stimulation. CONCLUSIONS: Our data show that DRN-MOR neurons respond to rewarding stimuli and that their optoactivation has reinforcing effects and promotes positive emotional responses, an activity which is partially mediated by their projections to the lateral hypothalamus. Our study also suggests a complex regulation of DRN activity by MOR opioids, involving mixed inhibition/activation mechanisms that fine-tune DRN function.


Subject(s)
Dorsal Raphe Nucleus , Receptors, Opioid, mu , Neurons/physiology , Morphine/pharmacology , Analgesics, Opioid , Reward
7.
Nat Methods ; 20(5): 682-685, 2023 05.
Article in English | MEDLINE | ID: mdl-36973548

ABSTRACT

Photoactivatable drugs and peptides can drive quantitative studies into receptor signaling with high spatiotemporal precision, yet few are compatible with behavioral studies in mammals. We developed CNV-Y-DAMGO-a caged derivative of the mu opioid receptor-selective peptide agonist DAMGO. Photoactivation in the mouse ventral tegmental area produced an opioid-dependent increase in locomotion within seconds of illumination. These results demonstrate the power of in vivo photopharmacology for dynamic studies into animal behavior.


Subject(s)
Analgesics, Opioid , Receptors, Opioid, mu , Mice , Animals , Analgesics, Opioid/pharmacology , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/physiology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Ventral Tegmental Area/physiology , Behavior, Animal , Mammals
8.
bioRxiv ; 2023 Feb 03.
Article in English | MEDLINE | ID: mdl-36778286

ABSTRACT

Traditional methods for site-specific drug delivery in the brain are slow, invasive, and difficult to interface with recordings of neural activity. Here, we demonstrate the feasibility and experimental advantages of in vivo photopharmacology using "caged" opioid drugs that are activated in the brain with light after systemic administration in an inactive form. To enable bidirectional manipulations of endogenous opioid receptors in vivo , we developed PhOX and PhNX, photoactivatable variants of the mu opioid receptor agonist oxymorphone and the antagonist naloxone. Photoactivation of PhOX in multiple brain areas produced local changes in receptor occupancy, brain metabolic activity, neuronal calcium activity, neurochemical signaling, and multiple pain- and reward-related behaviors. Combining PhOX photoactivation with optical recording of extracellular dopamine revealed adaptations in the opioid sensitivity of mesolimbic dopamine circuitry during chronic morphine administration. This work establishes a general experimental framework for using in vivo photopharmacology to study the neural basis of drug action. Highlights: A photoactivatable opioid agonist (PhOX) and antagonist (PhNX) for in vivo photopharmacology. Systemic pro-drug delivery followed by local photoactivation in the brain. In vivo photopharmacology produces behavioral changes within seconds of photostimulation. In vivo photopharmacology enables all-optical pharmacology and physiology.

9.
Front Syst Neurosci ; 16: 1044686, 2022.
Article in English | MEDLINE | ID: mdl-36591324

ABSTRACT

Due to the prevalence of chronic pain worldwide, there is an urgent need to improve pain management strategies. While opioid drugs have long been used to treat chronic pain, their use is severely limited by adverse effects and abuse liability. Neurostimulation techniques have emerged as a promising option for chronic pain that is refractory to other treatments. While different neurostimulation strategies have been applied to many neural structures implicated in pain processing, there is variability in efficacy between patients, underscoring the need to optimize neurostimulation techniques for use in pain management. This optimization requires a deeper understanding of the mechanisms underlying neurostimulation-induced pain relief. Here, we discuss the most commonly used neurostimulation techniques for treating chronic pain. We present evidence that neurostimulation-induced analgesia is in part driven by the release of endogenous opioids and that this endogenous opioid release is a common endpoint between different methods of neurostimulation. Finally, we introduce technological and clinical innovations that are being explored to optimize neurostimulation techniques for the treatment of pain, including multidisciplinary efforts between neuroscience research and clinical treatment that may refine the efficacy of neurostimulation based on its underlying mechanisms.

10.
Elife ; 102021 11 17.
Article in English | MEDLINE | ID: mdl-34787079

ABSTRACT

Functional interactions between G protein-coupled receptors are poised to enhance neuronal sensitivity to neuromodulators and therapeutic drugs. Mu and delta opioid receptors (MORs and DORs) can interact when overexpressed in the same cells, but whether co-expression of endogenous MORs and DORs in neurons leads to functional interactions is unclear. Here, in mice, we show that both MORs and DORs inhibit parvalbumin-expressing basket cells (PV-BCs) in hippocampal CA1 through partially occlusive signaling pathways that terminate on somato-dendritic potassium channels and presynaptic calcium channels. Using photoactivatable opioid neuropeptides, we find that DORs dominate the response to enkephalin in terms of both ligand sensitivity and kinetics, which may be due to relatively low expression levels of MOR. Opioid-activated potassium channels do not show heterologous desensitization, indicating that MORs and DORs signal independently. In a direct test for heteromeric functional interactions, the DOR antagonist TIPP-Psi does not alter the kinetics or potency of either the potassium channel or synaptic responses to photorelease of the MOR agonist [d-Ala2, NMe-Phe4, Gly-ol5]enkephalin (DAMGO). Thus, aside from largely redundant and convergent signaling, MORs and DORs do not functionally interact in PV-BCs in a way that impacts somato-dendritic potassium currents or synaptic transmission. These findings imply that cross-talk between MORs and DORs, either in the form of physical interactions or synergistic intracellular signaling, is not a preordained outcome of co-expression in neurons.


Subject(s)
Hippocampus/physiology , Interneurons/metabolism , Mice , Parvalbumins/metabolism , Receptors, Opioid, delta/genetics , Receptors, Opioid, mu/genetics , Signal Transduction , Animals , Female , Male , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism
11.
Neuron ; 109(11): 1755-1757, 2021 06 02.
Article in English | MEDLINE | ID: mdl-34081915

ABSTRACT

Reliable optogenetic tools for sustained, projection-specific presynaptic silencing have been elusive. Recently in Neuron, Mahn et al. (2021) and Copits et al. (2021) describe how the light-activated inhibitory GPCRs eOPN3 and PPO can be used to reversibly suppress synaptic transmission in mice.


Subject(s)
Culicidae , Presynaptic Terminals , Animals , Mice , Neurotransmitter Agents , Optogenetics , Rhodopsin , Synaptic Transmission
12.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Article in English | MEDLINE | ID: mdl-34074761

ABSTRACT

Opioid-induced respiratory depression (OIRD) causes death following an opioid overdose, yet the neurobiological mechanisms of this process are not well understood. Here, we show that neurons within the lateral parabrachial nucleus that express the µ-opioid receptor (PBL Oprm1 neurons) are involved in OIRD pathogenesis. PBL Oprm1 neuronal activity is tightly correlated with respiratory rate, and this correlation is abolished following morphine injection. Chemogenetic inactivation of PBL Oprm1 neurons mimics OIRD in mice, whereas their chemogenetic activation following morphine injection rescues respiratory rhythms to baseline levels. We identified several excitatory G protein-coupled receptors expressed by PBL Oprm1 neurons and show that agonists for these receptors restore breathing rates in mice experiencing OIRD. Thus, PBL Oprm1 neurons are critical for OIRD pathogenesis, providing a promising therapeutic target for treating OIRD in patients.


Subject(s)
Analgesics, Opioid/adverse effects , Morphine/adverse effects , Neurons/metabolism , Receptors, Opioid, mu/metabolism , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/metabolism , Analgesics, Opioid/pharmacology , Animals , Mice , Mice, Transgenic , Morphine/administration & dosage , Morphine/pharmacology , Neurons/pathology , Receptors, Opioid, mu/genetics , Respiratory Insufficiency/genetics , Respiratory Insufficiency/pathology
13.
Nat Cell Biol ; 21(6): 768-777, 2019 06.
Article in English | MEDLINE | ID: mdl-31061466

ABSTRACT

Controlling cellular processes with light can help elucidate their underlying mechanisms. Here we present zapalog, a small-molecule dimerizer that undergoes photolysis when exposed to blue light. Zapalog dimerizes any two proteins tagged with the FKBP and DHFR domains until exposure to light causes its photolysis. Dimerization can be repeatedly restored with uncleaved zapalog. We implement this method to investigate mitochondrial motility and positioning in cultured neurons. Using zapalog, we tether mitochondria to constitutively active kinesin motors, forcing them down the axon towards microtubule (+) ends until their instantaneous release via blue light, which results in full restoration of their endogenous motility. We find that one-third of stationary mitochondria cannot be pulled away from their position and that these firmly anchored mitochondria preferentially localize to VGLUT1-positive presynapses. Furthermore, inhibition of actin polymerization with latrunculin A reduces this firmly anchored pool. On release from exogenous motors, mitochondria are preferentially recaptured at presynapses.


Subject(s)
Axons/metabolism , Mitochondria/genetics , Photolysis , Protein Multimerization/radiation effects , Actins/antagonists & inhibitors , Animals , Axons/chemistry , Axons/radiation effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , COS Cells , Chlorocebus aethiops , Kinesins/chemistry , Light , Microtubules/genetics , Microtubules/radiation effects , Mitochondria/chemistry , Mitochondria/radiation effects , Neurons/chemistry , Neurons/radiation effects , Polymerization/drug effects , Protein Domains/genetics , Protein Domains/radiation effects , Protein Multimerization/genetics , Synapses/chemistry , Synapses/genetics , Synapses/radiation effects , Tacrolimus Binding Proteins/chemistry , Tacrolimus Binding Proteins/genetics , Thiazolidines/pharmacology , Vesicular Glutamate Transport Protein 1/genetics
14.
ACS Chem Neurosci ; 9(4): 684-690, 2018 04 18.
Article in English | MEDLINE | ID: mdl-29266926

ABSTRACT

Physiological responses to the opioid neuropeptide enkephalin often involve both mu and delta opioid receptors. To facilitate quantitative studies into opioid signaling, we previously developed a caged [Leu5]-enkephalin that responds to ultraviolet irradiation, but its residual activity at delta receptors confounds experiments that involve both receptors. To reduce residual activity, we evaluated side-chain, N-terminus, and backbone caging sites and further incorporated the dimethoxy-nitrobenzyl moiety to improve sensitivity to ultraviolet light-emitting diodes (LEDs). Residual activity was characterized using an in vitro functional assay, and the power dependence and kinetics of the uncaging response to 355 nm laser irradiation were assayed using electrophysiological recordings of mu opioid receptor-mediated potassium currents in brain slices of rat locus coeruleus. These experiments identified N-MNVOC-LE as an optimal compound. Using ultraviolet LED illumination to photoactivate N-MNVOC-LE in the CA1 region of hippocampus, we found that enkephalin engages both mu and delta opioid receptors to suppress inhibitory synaptic transmission.


Subject(s)
Enkephalins/pharmacology , Receptors, Opioid, delta/drug effects , Receptors, Opioid, mu/drug effects , Synaptic Transmission/drug effects , Animals , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Hippocampus/drug effects , Neurons/drug effects , Potassium Channels/drug effects , Rats, Sprague-Dawley
15.
Elife ; 62017 02 06.
Article in English | MEDLINE | ID: mdl-28165322

ABSTRACT

Much work has explored animal-to-animal variability and compensation in ion channel expression. Yet, little is known regarding the physiological consequences of morphological variability. We quantify animal-to-animal variability in cable lengths (CV = 0.4) and branching patterns in the Gastric Mill (GM) neuron, an identified neuron type with highly-conserved physiological properties in the crustacean stomatogastric ganglion (STG) of Cancer borealis. We examined passive GM electrotonic structure by measuring the amplitudes and apparent reversal potentials (Erevs) of inhibitory responses evoked with focal glutamate photo-uncaging in the presence of TTX. Apparent Erevs were relatively invariant across sites (mean CV ± SD = 0.04 ± 0.01; 7-20 sites in each of 10 neurons), which ranged between 100-800 µm from the somatic recording site. Thus, GM neurons are remarkably electrotonically compact (estimated λ > 1.5 mm). Electrotonically compact structures, in consort with graded transmission, provide an elegant solution to observed morphological variability in the STG.


Subject(s)
Action Potentials , Ganglia, Invertebrate/anatomy & histology , Ganglia, Invertebrate/physiology , Gizzard, Non-avian/innervation , Nerve Net/anatomy & histology , Nerve Net/physiology , Neurons/physiology , Animals , Brachyura
16.
Neuron ; 88(6): 1227-1239, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26671460

ABSTRACT

Opioid neuropeptides and their receptors are evolutionarily conserved neuromodulatory systems that profoundly influence behavior. In dorsal striatum, which expresses the endogenous opioid enkephalin, patches (or striosomes) are limbic-associated subcompartments enriched in mu opioid receptors. The functional implications of opioid signaling in dorsal striatum and the circuit elements in patches regulated by enkephalin are unclear. Here, we examined how patch output is modulated by enkephalin and identified the underlying circuit mechanisms. We found that patches are relatively devoid of parvalbumin-expressing interneurons and exist as self-contained inhibitory microcircuits. Enkephalin suppresses inhibition onto striatal projection neurons selectively in patches, thereby disinhibiting their firing in response to cortical input. The majority of this neuromodulation is mediated by delta, not mu-opioid, receptors, acting specifically on intra-striatal collateral axons of striatopallidal neurons. These results suggest that enkephalin gates limbic information flow in dorsal striatum, acting via a patch-specific function for delta opioid receptors.


Subject(s)
Corpus Striatum/physiology , Enkephalins/pharmacology , Receptors, Opioid, delta/physiology , Receptors, Opioid, mu/physiology , Animals , Corpus Striatum/drug effects , Female , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
17.
J Am Chem Soc ; 135(42): 15948-54, 2013 Oct 23.
Article in English | MEDLINE | ID: mdl-24117060

ABSTRACT

Caged compounds are molecules rendered functionally inert by derivatization with a photochemical protecting group. We describe the design logic behind the development of a diethylaminocoumarin (DEAC) caging chromophore, DEAC450, that absorbs blue light strongly (ε450 = 43,000 M(-1) cm(-1)) and violet light 11-fold more weakly. The absorption minimum is in the wavelength range (340-360 nm) that is traditionally used for photolysis of many widely used nitroaromatic caged compounds (e.g., 4-carboxymethoxy-5,7-dinitroindolinyl(CDNI)-GABA). We used this chromophore to synthesize DEAC450-caged cAMP and found this probe was very stable toward aqueous hydrolysis in the electronic ground state but was photolyzed with a quantum efficiency of 0.78. When DEAC450-cAMP and CDNI-GABA where co-applied to striatal cholinergic interneurons, the caged compounds were photolyzed in an chromatically orthogonal manner using blue and violet light so as to modulate the neuronal firing rate in a bidirectional way.


Subject(s)
Aminocoumarins/chemistry , Color , Coumarins/chemistry , Light , Cyclic AMP/chemistry , Hydrolysis , Molecular Structure , Photochemical Processes , gamma-Aminobutyric Acid/chemistry
18.
Mol Pharmacol ; 84(5): 687-95, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23960100

ABSTRACT

The spatiotemporal dynamics of opioid signaling in the brain remain poorly defined. Photoactivatable opioid ligands provide a means to quantitatively measure these dynamics and their underlying mechanisms in brain tissue. Although activation kinetics can be assessed using caged agonists, deactivation kinetics are obscured by slow clearance of agonist in tissue. To reveal deactivation kinetics of opioid signaling we developed a caged competitive antagonist that can be quickly photoreleased in sufficient concentrations to render agonist dissociation effectively irreversible. Carboxynitroveratryl-naloxone (CNV-NLX), a caged analog of the competitive opioid antagonist NLX, was readily synthesized from commercially available NLX in good yield and found to be devoid of antagonist activity at heterologously expressed opioid receptors. Photolysis in slices of rat locus coeruleus produced a rapid inhibition of the ionic currents evoked by multiple agonists of the µ-opioid receptor (MOR), but not of α-adrenergic receptors, which activate the same pool of ion channels. Using the high-affinity peptide agonist dermorphin, we established conditions under which light-driven deactivation rates are independent of agonist concentration and thus intrinsic to the agonist-receptor complex. Under these conditions, some MOR agonists yielded deactivation rates that are limited by G protein signaling, whereas others appeared limited by agonist dissociation. Therefore, the choice of agonist determines which feature of receptor signaling is unmasked by CNV-NLX photolysis.


Subject(s)
Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Signal Transduction/drug effects , Animals , Brain/drug effects , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Humans , Kinetics , Rats , Receptors, Opioid, mu/drug effects
19.
Methods Mol Biol ; 995: 107-20, 2013.
Article in English | MEDLINE | ID: mdl-23494375

ABSTRACT

Incorporation of photoisomerizable chromophores into small molecule ligands represents a general approach for reversibly controlling protein function with light. Illumination at different wavelengths produces photostationary states (PSSs) consisting of different ratios of photoisomers. Thus optimal implementation of photoswitchable ligands requires knowledge of their wavelength sensitivity. Using an azobenzene-based ion channel blocker as an example, this protocol describes a (1)H NMR assay that can be used to precisely determine the isomeric content of photostationary states (PSSs) as a function of illumination wavelength. Samples of the photoswitchable ligand are dissolved in deuterated water and analyzed by UV/VIS spectroscopy to identify the range of illumination wavelengths that produce PSSs. The PSSs produced by these wavelengths are quantified using (1)H NMR spectroscopy under continuous irradiation through a monochromator-coupled fiber-optic cable. Because aromatic protons of azobenzene trans and cis isomers exhibit sufficiently different chemical shifts, their relative abundances at each PSS can be readily determined by peak integration. Constant illumination during spectrum acquisition is essential to accurately determine PSSs from molecules that thermally relax on the timescale of minutes or faster. This general protocol can be readily applied to any photoswitch that exhibits distinct (1)H NMR signals in each photoisomeric state.


Subject(s)
Azo Compounds/chemistry , Photochemical Processes , Potassium Channel Blockers/chemistry , Quaternary Ammonium Compounds/chemistry , Azo Compounds/pharmacology , Azo Compounds/radiation effects , HEK293 Cells , Humans , Ligands , Light , Magnetic Resonance Spectroscopy/methods , Membrane Potentials/drug effects , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/radiation effects , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Potassium Channels, Voltage-Gated/chemistry , Potassium Channels, Voltage-Gated/metabolism , Protein Binding , Protein Conformation , Quaternary Ammonium Compounds/pharmacology , Quaternary Ammonium Compounds/radiation effects , Spectrophotometry, Ultraviolet , Stereoisomerism
20.
Neuron ; 73(2): 249-59, 2012 Jan 26.
Article in English | MEDLINE | ID: mdl-22284180

ABSTRACT

Neuropeptides activate G protein-coupled receptors to acutely modulate cellular excitability and synaptic transmission. However, due to the lack of reagents for precise delivery of peptides within dense brain tissue, the spatiotemporal scale over which neuropeptides act is unknown. To achieve rapid and spatially delimited delivery of neuropeptides in mammalian brain tissue, we developed photoactivatable analogs of two opioids: [Leu5]-enkephalin (LE) and the 8 amino acid form of Dynorphin A (Dyn-8). These peptides are functionally inactive prior to photolysis, and exposure to ultraviolet (UV) light causes clean release of LE and Dyn-8. Recordings from acute slices of rat locus coeruleus (LC) demonstrated that photorelease of LE activates mu opioid receptor-coupled K+ channels with kinetics that approach the limits imposed by G protein-mediated signaling. Temporally precise and spatially delimited photorelease revealed the kinetics and ionic nature of the mu opioid response and the mechanisms that determine the spatial profile of enkephalinergic volume transmission in LC.


Subject(s)
Analgesics, Opioid/pharmacology , Dynorphins/pharmacology , Enkephalins/pharmacology , Neuropeptides/pharmacology , Analgesics, Opioid/metabolism , Animals , Dynorphins/metabolism , Enkephalins/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Rats , Receptors, Opioid, mu/metabolism , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...