Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Nat Methods ; 20(5): 682-685, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36973548

RESUMEN

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.


Asunto(s)
Analgésicos Opioides , Receptores Opioides mu , Ratones , Animales , Analgésicos Opioides/farmacología , Receptores Opioides mu/agonistas , Receptores Opioides mu/fisiología , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Área Tegmental Ventral/fisiología , Conducta Animal , Mamíferos
2.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34074761

RESUMEN

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.


Asunto(s)
Analgésicos Opioides/efectos adversos , Morfina/efectos adversos , Neuronas/metabolismo , Receptores Opioides mu/metabolismo , Insuficiencia Respiratoria/inducido químicamente , Insuficiencia Respiratoria/metabolismo , Analgésicos Opioides/farmacología , Animales , Ratones , Ratones Transgénicos , Morfina/administración & dosificación , Morfina/farmacología , Neuronas/patología , Receptores Opioides mu/genética , Insuficiencia Respiratoria/genética , Insuficiencia Respiratoria/patología
3.
J Am Chem Soc ; 145(36): 19611-19621, 2023 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-37649440

RESUMEN

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.


Asunto(s)
Biomimética , Neuropéptidos , Amidas , Aminoácidos , Analgésicos Opioides
4.
Sci Adv ; 10(17): eadj9581, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38669335

RESUMEN

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.


Asunto(s)
Analgésicos Opioides , Locus Coeruleus , Bulbo Raquídeo , Dolor , Sustancia Gris Periacueductal , Locus Coeruleus/metabolismo , Locus Coeruleus/efectos de los fármacos , Sustancia Gris Periacueductal/metabolismo , Sustancia Gris Periacueductal/efectos de los fármacos , Animales , Bulbo Raquídeo/metabolismo , Bulbo Raquídeo/efectos de los fármacos , Dolor/tratamiento farmacológico , Dolor/metabolismo , Analgésicos Opioides/farmacología , Masculino , Neuronas Adrenérgicas/metabolismo , Neuronas Adrenérgicas/efectos de los fármacos , Ratones , Vías Nerviosas/efectos de los fármacos
5.
Nat Neurosci ; 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39009835

RESUMEN

Neuropeptides are ubiquitous in the nervous system. Research into neuropeptides has been limited by a lack of experimental tools that allow for the precise dissection of their complex and diverse dynamics in a circuit-specific manner. Opioid peptides modulate pain, reward and aversion and as such have high clinical relevance. To illuminate the spatiotemporal dynamics of endogenous opioid signaling in the brain, we developed a class of genetically encoded fluorescence sensors based on kappa, delta and mu opioid receptors: κLight, δLight and µLight, respectively. We characterized the pharmacological profiles of these sensors in mammalian cells and in dissociated neurons. We used κLight to identify electrical stimulation parameters that trigger endogenous opioid release and the spatiotemporal scale of dynorphin volume transmission in brain slices. Using in vivo fiber photometry in mice, we demonstrated the utility of these sensors in detecting optogenetically driven opioid release and observed differential opioid release dynamics in response to fearful and rewarding conditions.

6.
bioRxiv ; 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38746090

RESUMEN

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.

7.
Mol Pharmacol ; 84(5): 687-95, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23960100

RESUMEN

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.


Asunto(s)
Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Humanos , Cinética , Ratas , Receptores Opioides mu/efectos de los fármacos
8.
J Am Chem Soc ; 135(42): 15948-54, 2013 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-24117060

RESUMEN

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.


Asunto(s)
Aminocumarinas/química , Color , Cumarinas/química , Luz , AMP Cíclico/química , Hidrólisis , Estructura Molecular , Procesos Fotoquímicos , Ácido gamma-Aminobutírico/química
9.
bioRxiv ; 2023 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-37873091

RESUMEN

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.

10.
Biol Psychiatry ; 94(11): 842-851, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37285896

RESUMEN

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.


Asunto(s)
Núcleo Dorsal del Rafe , Receptores Opioides mu , Neuronas/fisiología , Morfina/farmacología , Analgésicos Opioides , Recompensa
11.
bioRxiv ; 2023 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-36778286

RESUMEN

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.

12.
Neuron ; 111(24): 3926-3940.e10, 2023 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-37848025

RESUMEN

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.


Asunto(s)
Analgésicos Opioides , Oximorfona , Analgésicos Opioides/farmacología , Oximorfona/farmacología , Preparaciones Farmacéuticas , Dopamina/metabolismo , Naloxona/farmacología , Receptores Opioides mu/metabolismo
13.
Front Syst Neurosci ; 16: 1044686, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36591324

RESUMEN

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.

14.
J Neurophysiol ; 106(1): 488-96, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21525363

RESUMEN

Currently available optogenetic tools, including microbial light-activated ion channels and transporters, are transforming systems neuroscience by enabling precise remote control of neuronal firing, but they tell us little about the role of indigenous ion channels in controlling neuronal function. Here, we employ a chemical-genetic strategy to engineer light sensitivity into several mammalian K(+) channels that have different gating and modulation properties. These channels provide the means for photoregulating diverse electrophysiological functions. Photosensitivity is conferred on a channel by a tethered ligand photoswitch that contains a cysteine-reactive maleimide (M), a photoisomerizable azobenzene (A), and a quaternary ammonium (Q), a K(+) channel pore blocker. Using mutagenesis, we identify the optimal extracellular cysteine attachment site where MAQ conjugation results in pore blockade when the azobenzene moiety is in the trans but not cis configuration. With this strategy, we have conferred photosensitivity on channels containing Kv1.3 subunits (which control axonal action potential repolarization), Kv3.1 subunits (which contribute to rapid-firing properties of brain neurons), Kv7.2 subunits (which underlie "M-current"), and SK2 subunits (which are Ca(2+)-activated K(+) channels that contribute to synaptic responses). These light-regulated channels may be overexpressed in genetically targeted neurons or substituted for native channels with gene knockin technology to enable precise optopharmacological manipulation of channel function.


Asunto(s)
Canal de Potasio KCNQ2/química , Canal de Potasio Kv1.3/química , Neuronas/química , Procesos Fotoquímicos , Canales de Potasio Calcio-Activados/química , Ingeniería de Proteínas , Secuencia de Aminoácidos , Compuestos Azo/química , Células HEK293 , Humanos , Activación del Canal Iónico , Canal de Potasio KCNQ2/genética , Canal de Potasio Kv1.3/genética , Maleimidas/química , Datos de Secuencia Molecular , Compuestos de Amonio Cuaternario/química
15.
Nat Methods ; 5(4): 331-8, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18311146

RESUMEN

Light-activated ion channels provide a precise and noninvasive optical means for controlling action potential firing, but the genes encoding these channels must first be delivered and expressed in target cells. Here we describe a method for bestowing light sensitivity onto endogenous ion channels that does not rely on exogenous gene expression. The method uses a synthetic photoisomerizable small molecule, or photoswitchable affinity label (PAL), that specifically targets K+ channels. PALs contain a reactive electrophile, enabling covalent attachment of the photoswitch to naturally occurring nucleophiles in K+ channels. Ion flow through PAL-modified channels is turned on or off by photoisomerizing PAL with different wavelengths of light. We showed that PAL treatment confers light sensitivity onto endogenous K+ channels in isolated rat neurons and in intact neural structures from rat and leech, allowing rapid optical regulation of excitability without genetic modification.


Asunto(s)
Potenciales de Acción/efectos de la radiación , Activación del Canal Iónico/efectos de la radiación , Neuronas , Canales de Potasio/metabolismo , Marcadores de Afinidad/química , Animales , Compuestos Azo/química , Células Cultivadas , Cerebelo/citología , Cerebelo/metabolismo , Cerebelo/efectos de la radiación , Hipocampo/citología , Hipocampo/metabolismo , Hipocampo/efectos de la radiación , Sanguijuelas , Neuronas/metabolismo , Neuronas/efectos de la radiación , Estimulación Luminosa , Fotoquímica , Compuestos de Amonio Cuaternario/química , Ratas
16.
Nano Lett ; 10(1): 250-4, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19904979

RESUMEN

We report a novel and simple proof-of-concept of a nanoparticulate system that targets any tissue selectively upon illumination. Nanoparticles were covalently functionalized with the amino acid sequence YIGSR, which adheres to the beta1 integrins present on most cell surfaces. This peptide was masked with a caging group, rendering it biologically inert. Illumination with UV light released the caging group from the YIGSR, allowing binding to cells.


Asunto(s)
Nanopartículas/química , Nanotecnología/métodos , Fotoquímica/métodos , Animales , Células Cultivadas , Cromatografía Líquida de Alta Presión , Humanos , Nitrobencenos/química , Péptidos/química , Ratas , Ratas Sprague-Dawley , Espectroscopía Infrarroja por Transformada de Fourier , Tirosina/química , Rayos Ultravioleta
17.
Neuron ; 109(11): 1755-1757, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-34081915

RESUMEN

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.


Asunto(s)
Culicidae , Terminales Presinápticos , Animales , Ratones , Neurotransmisores , Optogenética , Rodopsina , Transmisión Sináptica
18.
Elife ; 102021 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-34787079

RESUMEN

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.


Asunto(s)
Hipocampo/fisiología , Interneuronas/metabolismo , Ratones , Parvalbúminas/metabolismo , Receptores Opioides delta/genética , Receptores Opioides mu/genética , Transducción de Señal , Animales , Femenino , Masculino , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo
19.
Nat Cell Biol ; 21(6): 768-777, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31061466

RESUMEN

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.


Asunto(s)
Axones/metabolismo , Mitocondrias/genética , Fotólisis , Multimerización de Proteína/efectos de la radiación , Actinas/antagonistas & inhibidores , Animales , Axones/química , Axones/efectos de la radiación , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Células COS , Chlorocebus aethiops , Cinesinas/química , Luz , Microtúbulos/genética , Microtúbulos/efectos de la radiación , Mitocondrias/química , Mitocondrias/efectos de la radiación , Neuronas/química , Neuronas/efectos de la radiación , Polimerizacion/efectos de los fármacos , Dominios Proteicos/genética , Dominios Proteicos/efectos de la radiación , Multimerización de Proteína/genética , Sinapsis/química , Sinapsis/genética , Sinapsis/efectos de la radiación , Proteínas de Unión a Tacrolimus/química , Proteínas de Unión a Tacrolimus/genética , Tiazolidinas/farmacología , Proteína 1 de Transporte Vesicular de Glutamato/genética
20.
ACS Chem Neurosci ; 9(4): 684-690, 2018 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-29266926

RESUMEN

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.


Asunto(s)
Encefalinas/farmacología , Receptores Opioides delta/efectos de los fármacos , Receptores Opioides mu/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Animales , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Hipocampo/efectos de los fármacos , Neuronas/efectos de los fármacos , Canales de Potasio/efectos de los fármacos , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA