Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 10(1): 2746, 2019 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-31227712

RESUMEN

Nicotinic acetylcholine receptors (nAChRs) mediate and modulate synaptic transmission throughout the brain, and contribute to learning, memory, and behavior. Dysregulation of α7-type nAChRs in neuropsychiatric as well as immunological and oncological diseases makes them attractive targets for pharmaceutical development. Recently, we identified NACHO as an essential chaperone for α7 nAChRs. Leveraging the robust recombinant expression of α7 nAChRs with NACHO, we utilized genome-wide cDNA library screening and discovered that several anti-apoptotic Bcl-2 family proteins further upregulate receptor assembly and cell surface expression. These effects are mediated by an intracellular motif on α7 that resembles the BH3 binding domain of pro-apoptotic Bcl-2 proteins, and can be blocked by BH3 mimetic Bcl-2 inhibitors. Overexpression of Bcl-2 member Mcl-1 in neurons enhanced surface expression of endogenous α7 nAChRs, while a combination of chemotherapeutic Bcl2-inhibitors suppressed neuronal α7 receptor assembly. These results demonstrate that Bcl-2 proteins link α7 nAChR assembly to cell survival pathways.


Asunto(s)
Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Neuronas/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Secuencias de Aminoácidos/genética , Animales , Benzotiazoles/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Células HEK293 , Humanos , Isoquinolinas/farmacología , Chaperonas Moleculares/metabolismo , Mutación , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/antagonistas & inhibidores , Neuronas/efectos de los fármacos , Agonistas Nicotínicos/farmacología , Cultivo Primario de Células , Unión Proteica/efectos de los fármacos , Piridinas/farmacología , Pirimidinas/farmacología , Ratas , Transmisión Sináptica/efectos de los fármacos , Tiofenos/farmacología , Regulación hacia Arriba , Receptor Nicotínico de Acetilcolina alfa 7/genética
2.
Neurobiol Pain ; 2: 1-12, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29075674

RESUMEN

It has been suggested that the lack of rodent behavioral assays that represent the complexities of human pain contributes to the poor translational record of basic pain research findings. Clinically, chronic pain interferes with patient mobility and physical/social activities, and increases anxiety symptoms, in turn negatively impacting quality of life. To determine whether these behaviors are similarly influenced by putative pain manipulations in rodents, we systematically evaluated wheel running, locomotion, gait, social interaction, and anxiety-like behavior in models of inflammation and nerve injury in adult C57BL6/J male mice. We demonstrate that inflammation and nerve injury differentially affect voluntary behaviors while mice are hypersensitive to mechanical stimuli. Bilateral Complete Freund's Adjuvant (CFA)-induced inflammation transiently suppressed wheel running and locomotion and also induced gait deficits. In contrast, spared nerve injury (SNI) altered gait and impaired gross motor coordination. SNI-induced gait changes were not reversed by the analgesic PD123319, an angiotensin II type 2 receptor antagonist, and are therefore likely to be motor-related rather than pain-related. Neither CFA nor SNI significantly altered social interaction or elicited general anxiety-like behavior. Our findings suggest that in contrast to humans, mobility and physical/social activities are minimally altered, if at all, in mice following inflammation or nerve injury.

3.
Elife ; 62017 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-28708061

RESUMEN

Increased tonic activity of locus coeruleus noradrenergic (LC-NE) neurons induces anxiety-like and aversive behavior. While some information is known about the afferent circuitry that endogenously drives this neural activity and behavior, the downstream receptors and anatomical projections that mediate these acute risk aversive behavioral states via the LC-NE system remain unresolved. Here we use a combination of retrograde tracing, fast-scan cyclic voltammetry, electrophysiology, and in vivo optogenetics with localized pharmacology to identify neural substrates downstream of increased tonic LC-NE activity in mice. We demonstrate that photostimulation of LC-NE fibers in the BLA evokes norepinephrine release in the basolateral amygdala (BLA), alters BLA neuronal activity, conditions aversion, and increases anxiety-like behavior. Additionally, we report that ß-adrenergic receptors mediate the anxiety-like phenotype of increased NE release in the BLA. These studies begin to illustrate how the complex efferent system of the LC-NE system selectively mediates behavior through distinct receptor and projection-selective mechanisms.


Asunto(s)
Neuronas Adrenérgicas/fisiología , Ansiedad , Complejo Nuclear Basolateral/fisiología , Locus Coeruleus/fisiología , Vías Nerviosas/fisiología , Animales , Conducta Animal , Ratones
4.
J Pharmacol Exp Ther ; 362(2): 254-262, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28533287

RESUMEN

Prescription opioids are a mainstay in the treatment of acute moderate to severe pain. However, chronic use leads to a host of adverse consequences including tolerance and opioid-induced hyperalgesia (OIH), leading to more complex treatment regimens and diminished patient compliance. Patients with OIH paradoxically experience exaggerated nociceptive responses instead of pain reduction after chronic opioid usage. The development of OIH and tolerance tend to occur simultaneously and, thus, present a challenge when studying the molecular mechanisms driving each phenomenon. We tested the hypothesis that a G protein-biased µ-opioid peptide receptor (MOPR) agonist would not induce symptoms of OIH, such as mechanical allodynia, following chronic administration. We observed that the development of opioid-induced mechanical allodynia (OIMA), a model of OIH, was absent in ß-arrestin1-/- and ß-arrestin2-/- mice in response to chronic administration of conventional opioids such as morphine, oxycodone and fentanyl, whereas tolerance developed independent of OIMA. In agreement with the ß-arrestin knockout mouse studies, chronic administration of TRV0109101, a G protein-biased MOPR ligand and structural analog of oliceridine, did not promote the development of OIMA but did result in drug tolerance. Interestingly, following induction of OIMA by morphine or fentanyl, TRV0109101 was able to rapidly reverse allodynia. These observations establish a role for ß-arrestins in the development of OIH, independent of tolerance, and suggest that the use of G protein-biased MOPR ligands, such as oliceridine and TRV0109101, may be an effective therapeutic avenue for managing chronic pain with reduced propensity for opioid-induced hyperalgesia.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Proteínas de Unión al GTP/agonistas , Hiperalgesia/tratamiento farmacológico , Dimensión del Dolor/efectos de los fármacos , Receptores Opioides mu/agonistas , Animales , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Proteínas de Unión al GTP/fisiología , Células HEK293 , Humanos , Hiperalgesia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Dimensión del Dolor/métodos , Receptores Opioides mu/fisiología
5.
Cell Rep ; 19(4): 688-696, 2017 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-28445721

RESUMEN

Neuronal nicotinic acetylcholine receptors (nAChRs) participate in diverse aspects of brain function and mediate behavioral and addictive properties of nicotine. Neuronal nAChRs derive from combinations of α and ß subunits, whose assembly is tightly regulated. NACHO was recently identified as a chaperone for α7-type nAChRs. Here, we find NACHO mediates assembly of all major classes of presynaptic and postsynaptic nAChR tested. NACHO acts at early intracellular stages of nAChR subunit assembly and then synergizes with RIC-3 for receptor surface expression. NACHO knockout mice show profound deficits in binding sites for α-bungarotoxin, epibatidine, and conotoxin MII, illustrating essential roles for NACHO in proper assembly of α7-, α4ß2-, and α6-containing nAChRs, respectively. By contrast, GABAA receptors are unaffected consistent with NACHO specifically modulating nAChRs. NACHO knockout mice show abnormalities in locomotor and cognitive behaviors compatible with nAChR deficiency and underscore the importance of this chaperone for physiology and disease associated with nAChRs.


Asunto(s)
Encéfalo/metabolismo , Chaperonas Moleculares/metabolismo , Receptores Nicotínicos/metabolismo , Animales , Sitios de Unión , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Compuestos Bicíclicos Heterocíclicos con Puentes/metabolismo , Bungarotoxinas/química , Bungarotoxinas/metabolismo , Línea Celular , Disfunción Cognitiva/patología , Conotoxinas/química , Conotoxinas/metabolismo , Humanos , Radioisótopos de Yodo/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Chaperonas Moleculares/genética , Neuronas/metabolismo , Nicotina/química , Nicotina/metabolismo , Unión Proteica , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Piridinas/química , Piridinas/metabolismo , Radiofármacos/química , Radiofármacos/metabolismo , Receptores Nicotínicos/genética
6.
Curr Opin Pharmacol ; 32: 77-84, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27936408

RESUMEN

Opioid chemistry and biology occupy a pivotal place in the history of pharmacology and medicine. Morphine offers unmatched efficacy in alleviating acute pain, but is also associated with a host of adverse side effects. The advent of biased agonism at G protein-coupled receptors has expanded our understanding of intracellular signaling and highlighted the concept that certain ligands are able to differentially modulate downstream pathways. The ability to target one pathway over another has allowed for the development of biased ligands with robust clinical efficacy and fewer adverse events. In this review we summarize these concepts with an emphasis on biased mu opioid receptor pharmacology and highlight how far opioid pharmacology has evolved.


Asunto(s)
Analgésicos Opioides/farmacología , Dolor/tratamiento farmacológico , Receptores Opioides mu/agonistas , Analgésicos Opioides/efectos adversos , Animales , Diseño de Fármacos , Humanos , Ligandos , Morfina/efectos adversos , Morfina/farmacología , Dolor/fisiopatología , Receptores Opioides mu/metabolismo , Transducción de Señal/efectos de los fármacos
7.
Neuropsychopharmacology ; 41(8): 2011-23, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26725834

RESUMEN

Anxiety disorders are debilitating psychiatric illnesses with detrimental effects on human health. These heightened states of arousal are often in the absence of obvious threatening cues and are difficult to treat owing to a lack of understanding of the neural circuitry and cellular machinery mediating these conditions. Activation of noradrenergic circuitry in the basolateral amygdala is thought to have a role in stress, fear, and anxiety, and the specific cell and receptor types responsible is an active area of investigation. Here we take advantage of two novel cellular approaches to dissect the contributions of G-protein signaling in acute and social anxiety-like states. We used a chemogenetic approach utilizing the Gαs DREADD (rM3Ds) receptor and show that selective activation of generic Gαs signaling is sufficient to induce acute and social anxiety-like behavioral states in mice. Second, we use a recently characterized chimeric receptor composed of rhodopsin and the ß2-adrenergic receptor (Opto-ß2AR) with in vivo optogenetic techniques to selectively activate Gαs ß-adrenergic signaling exclusively within excitatory neurons of the basolateral amygdala. We found that optogenetic induction of ß-adrenergic signaling in the basolateral amygdala is sufficient to induce acute and social anxiety-like behavior. These findings support the conclusion that activation of Gαs signaling in the basolateral amygdala has a role in anxiety. These data also suggest that acute and social anxiety-like states may be mediated through signaling pathways identical to ß-adrenergic receptors, thus providing support that inhibition of this system may be an effective anxiolytic therapy.


Asunto(s)
Ansiedad/metabolismo , Complejo Nuclear Basolateral/metabolismo , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Animales , Conducta Animal , Células HEK293 , Humanos , Masculino , Ratones Endogámicos C57BL , Neuronas/metabolismo , Optogenética , Transducción de Señal , Conducta Social
8.
Respir Physiol Neurobiol ; 224: 2-10, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26160673

RESUMEN

Medullary motoneurons drive vocalization in many vertebrate lineages including fish, amphibians, birds, and mammals. The developmental history of vocal motoneuron populations in each of these lineages remains largely unknown. The highly conserved transcription factor Paired-like Homeobox 2b (Phox2b) is presumed to be expressed in all vertebrate hindbrain branchial motoneurons, including laryngeal motoneurons essential for vocalization in humans. We used immunohistochemistry and in situ hybridization to examine Phox2b protein and mRNA expression in caudal hindbrain and rostral spinal cord motoneuron populations in seven species across five chordate classes. Phox2b was present in motoneurons dedicated to sound production in mice and frogs (bullfrog, African clawed frog), but not those in bird (zebra finch) or bony fish (midshipman, channel catfish). Overall, the pattern of caudal medullary motoneuron Phox2b expression was conserved across vertebrates and similar to expression in sea lamprey. These observations suggest that motoneurons dedicated to sound production in vertebrates are not derived from a single developmentally or evolutionarily conserved progenitor pool.


Asunto(s)
Evolución Biológica , Neuronas Motoras/fisiología , Vertebrados/fisiología , Vocalización Animal/fisiología , Animales
9.
Nat Commun ; 6: 8480, 2015 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-26412387

RESUMEN

Optogenetics has provided a revolutionary approach to dissecting biological phenomena. However, the generation and use of optically active GPCRs in these contexts is limited and it is unclear how well an opsin-chimera GPCR might mimic endogenous receptor activity. Here we show that a chimeric rhodopsin/ß2 adrenergic receptor (opto-ß2AR) is similar in dynamics to endogenous ß2AR in terms of: cAMP generation, MAP kinase activation and receptor internalization. In addition, we develop and characterize a novel toolset of optically active, functionally selective GPCRs that can bias intracellular signalling cascades towards either G-protein or arrestin-mediated cAMP and MAP kinase pathways. Finally, we show how photoactivation of opto-ß2AR in vivo modulates neuronal activity and induces anxiety-like behavioural states in both fiber-tethered and wireless, freely moving animals when expressed in brain regions known to contain ß2ARs. These new GPCR approaches enhance the utility of optogenetics and allow for discrete spatiotemporal control of GPCR signalling in vitro and in vivo.


Asunto(s)
Optogenética , Quinasas de Receptores Adrenérgicos beta/metabolismo , Animales , Ansiedad/enzimología , AMP Cíclico/metabolismo , Células HEK293 , Humanos , Masculino , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/metabolismo
10.
Neuron ; 87(3): 605-20, 2015 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-26212712

RESUMEN

The locus coeruleus noradrenergic (LC-NE) system is one of the first systems engaged following a stressful event. While numerous groups have demonstrated that LC-NE neurons are activated by many different stressors, the underlying neural circuitry and the role of this activity in generating stress-induced anxiety has not been elucidated. Using a combination of in vivo chemogenetics, optogenetics, and retrograde tracing, we determine that increased tonic activity of the LC-NE system is necessary and sufficient for stress-induced anxiety and aversion. Selective inhibition of LC-NE neurons during stress prevents subsequent anxiety-like behavior. Exogenously increasing tonic, but not phasic, activity of LC-NE neurons is alone sufficient for anxiety-like and aversive behavior. Furthermore, endogenous corticotropin-releasing hormone(+) (CRH(+)) LC inputs from the amygdala increase tonic LC activity, inducing anxiety-like behaviors. These studies position the LC-NE system as a critical mediator of acute stress-induced anxiety and offer a potential intervention for preventing stress-related affective disorders.


Asunto(s)
Neuronas Adrenérgicas/metabolismo , Ansiedad/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Locus Coeruleus/metabolismo , Estrés Psicológico/metabolismo , Neuronas Adrenérgicas/química , Animales , Ansiedad/psicología , Locus Coeruleus/química , Masculino , Ratones , Ratones Endogámicos C57BL , Norepinefrina/metabolismo , Optogenética/métodos , Estrés Psicológico/psicología
11.
J Neural Eng ; 12(5): 056002-56002, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26193450

RESUMEN

OBJECTIVE: Wireless control and power harvesting systems that operate injectable, cellular-scale optoelectronic components provide important demonstrated capabilities in neuromodulatory techniques such as optogenetics. Here, we report a radio frequency (RF) control/harvesting device that offers dramatically reduced size, decreased weight and improved efficiency compared to previously reported technologies. Combined use of this platform with ultrathin, multijunction, high efficiency solar cells allows for hundred-fold reduction of transmitted RF power, which greatly enhances the wireless coverage. APPROACH: Fabrication involves separate construction of the harvester and the injectable µ-ILEDs. To test whether the presence of the implantable device alters behavior, we implanted one group of wild type mice and compared sociability behavior to unaltered controls. Social interaction experiments followed protocols defined by Silverman et al. with minor modifications. MAIN RESULTS: The results presented here demonstrate that miniaturized RF harvesters, and RF control strategies with photovoltaic harvesters can, when combined with injectable µ-ILEDs, offer versatile capabilities in optogenetics. Experimental and modeling studies establish a range of effective operating conditions for these two approaches. Optogenetics studies with social groups of mice demonstrate the utility of these systems. SIGNIFICANCE: The addition of miniaturized, high performance photovoltaic cells significantly expands the operating range and reduces the required RF power. The platform can offer capabilities to modulate signaling path in the brain region of freely-behaving animals. These suggest its potential for widespread use in neuroscience.


Asunto(s)
Suministros de Energía Eléctrica , Iluminación/instrumentación , Optogenética/instrumentación , Estimulación Luminosa/instrumentación , Semiconductores , Tecnología Inalámbrica/instrumentación , Animales , Encéfalo/fisiología , Diseño Asistido por Computadora , Transferencia de Energía , Diseño de Equipo , Análisis de Falla de Equipo , Luz , Masculino , Ratones , Ratones Endogámicos C57BL , Miniaturización , Ondas de Radio
12.
Neuron ; 86(4): 923-935, 2015 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-25937173

RESUMEN

Optogenetics is now a widely accepted tool for spatiotemporal manipulation of neuronal activity. However, a majority of optogenetic approaches use binary on/off control schemes. Here, we extend the optogenetic toolset by developing a neuromodulatory approach using a rationale-based design to generate a Gi-coupled, optically sensitive, mu-opioid-like receptor, which we term opto-MOR. We demonstrate that opto-MOR engages canonical mu-opioid signaling through inhibition of adenylyl cyclase, activation of MAPK and G protein-gated inward rectifying potassium (GIRK) channels and internalizes with kinetics similar to that of the mu-opioid receptor. To assess in vivo utility, we expressed a Cre-dependent viral opto-MOR in RMTg/VTA GABAergic neurons, which led to a real-time place preference. In contrast, expression of opto-MOR in GABAergic neurons of the ventral pallidum hedonic cold spot led to real-time place aversion. This tool has generalizable application for spatiotemporal control of opioid signaling and, furthermore, can be used broadly for mimicking endogenous neuronal inhibition pathways.


Asunto(s)
Analgésicos Opioides/farmacología , Conducta Animal/efectos de los fármacos , Neuronas GABAérgicas/efectos de los fármacos , Optogenética , Receptores Opioides mu/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Células Cultivadas , Neuronas GABAérgicas/metabolismo , Ratas , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/fisiología
13.
Science ; 340(6129): 211-6, 2013 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-23580530

RESUMEN

Successful integration of advanced semiconductor devices with biological systems will accelerate basic scientific discoveries and their translation into clinical technologies. In neuroscience generally, and in optogenetics in particular, the ability to insert light sources, detectors, sensors, and other components into precise locations of the deep brain yields versatile and important capabilities. Here, we introduce an injectable class of cellular-scale optoelectronics that offers such features, with examples of unmatched operational modes in optogenetics, including completely wireless and programmed complex behavioral control over freely moving animals. The ability of these ultrathin, mechanically compliant, biocompatible devices to afford minimally invasive operation in the soft tissues of the mammalian brain foreshadow applications in other organ systems, with potential for broad utility in biomedical science and engineering.


Asunto(s)
Conducta Animal , Mapeo Encefálico , Encéfalo/fisiología , Neuronas/fisiología , Optogenética , Semiconductores , Animales , Mapeo Encefálico/instrumentación , Mapeo Encefálico/métodos , Estimulación Eléctrica , Fenómenos Electrofisiológicos , Células HEK293 , Humanos , Ratones , Microelectrodos , Miniaturización , Estimulación Luminosa
14.
J Neurosci ; 32(44): 15296-308, 2012 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-23115168

RESUMEN

Cerebellar motor coordination and cerebellar Purkinje cell synaptic function require metabotropic glutamate receptor 1 (mGluR1, Grm1). We used an unbiased proteomic approach to identify protein partners for mGluR1 in cerebellum and discovered glutamate receptor δ2 (GluRδ2, Grid2, GluΔ2) and protein kinase Cγ (PKCγ) as major interactors. We also found canonical transient receptor potential 3 (TRPC3), which is also needed for mGluR1-dependent slow EPSCs and motor coordination and associates with mGluR1, GluRδ2, and PKCγ. Mutation of GluRδ2 changes subcellular fractionation of mGluR1 and TRPC3 to increase their surface expression. Fitting with this, mGluR1-evoked inward currents are increased in GluRδ2 mutant mice. Moreover, loss of GluRδ2 disrupts the time course of mGluR1-dependent synaptic transmission at parallel fiber-Purkinje cells synapses. Thus, GluRδ2 is part of the mGluR1 signaling complex needed for cerebellar synaptic function and motor coordination, explaining the shared cerebellar motor phenotype that manifests in mutants of the mGluR1 and GluRδ2 signaling pathways.


Asunto(s)
Neuronas/fisiología , Proteína Quinasa C/fisiología , Células de Purkinje/fisiología , Receptores de Glutamato/fisiología , Receptores de Glutamato Metabotrópico/fisiología , Transmisión Sináptica/fisiología , Canales Catiónicos TRPC/fisiología , Animales , Potenciales Postsinápticos Excitadores/genética , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Mutación/fisiología , Técnicas de Placa-Clamp , Fenotipo , Receptores de Superficie Celular/fisiología , Receptores de Glutamato/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Solubilidad , Fracciones Subcelulares/metabolismo , Fracciones Subcelulares/fisiología
15.
J Biol Chem ; 287(50): 42019-30, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23086955

RESUMEN

We determined the role of carboxyl-terminal regulation of NOPR (nociceptin, orphanin FQ receptor) signaling and function. We mutated C-terminal serine and threonine residues and examined their role in NOPR trafficking, homologous desensitization, and arrestin-dependent MAPK signaling. The NOPR agonist, nociceptin, caused robust NOPR-YFP receptor internalization, peaking at 30 min. Mutation of serine 337, 346, and 351, had no effect on NOPR internalization. However, mutation of C-terminal threonine 362, serine 363, and threonine 365 blocked nociceptin-induced internalization of NOPR. Furthermore, point mutation of only Ser-363 was sufficient to block NOPR internalization. Homologous desensitization of NOPR-mediated calcium channel blockade and inhibition of cAMP were also shown to require Ser-363. Additionally, NOPR internalization was absent when GRK3, and Arrestin3 were knocked down using siRNA, but not when GRK2 and Arrestin2 were knocked down. We also found that nociceptin-induced NOPR-mediated JNK but not ERK signaling requires Ser-363, GRK3, and Arrestin3. Dominant-positive Arrestin3 but not Arrestin2 was sufficient to rescue NOPR-S363A internalization and JNK signaling. These findings suggest that NOPR function may be regulated by GRK3 phosphorylation of Ser-363 and Arrestin3 and further demonstrates the complex nature of G-protein-dependent and -independent signaling in opioid receptors.


Asunto(s)
Arrestinas/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Péptidos Opioides/metabolismo , Receptores Opioides/metabolismo , Transducción de Señal/fisiología , Arrestinas/genética , Quinasa 3 del Receptor Acoplado a Proteína-G/genética , Quinasa 3 del Receptor Acoplado a Proteína-G/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , MAP Quinasa Quinasa 4/genética , Péptidos Opioides/genética , Fosforilación/fisiología , Transporte de Proteínas/fisiología , Receptores Opioides/genética , Receptor de Nociceptina , Nociceptina
16.
Neuron ; 68(6): 1082-96, 2010 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-21172611

RESUMEN

Transmembrane AMPA receptor regulatory proteins (TARPs) and cornichon proteins (CNIH-2/3) independently modulate AMPA receptor trafficking and gating. However, the potential for interactions of these subunits within an AMPA receptor complex is unknown. Here, we find that TARPs γ-4, γ-7, and γ-8, but not γ-2, γ-3, or γ-5, cause AMPA receptors to "resensitize" upon continued glutamate application. With γ-8, resensitization occurs with all GluA subunit combinations; however, γ-8-containing hippocampal neurons do not display resensitization. In recombinant systems, CNIH-2 abrogates γ-8-mediated resensitization and modifies AMPA receptor pharmacology and gating to match that of hippocampal neurons. In hippocampus, γ-8 and CNIH-2 associate in postsynaptic densities and CNIH-2 protein levels are markedly diminished in γ-8 knockout mice. Manipulating neuronal CNIH-2 levels modulates the electrophysiological properties of extrasynaptic and synaptic γ-8-containing AMPA receptors. Thus, γ-8 and CNIH-2 functionally interact with common hippocampal AMPA receptor complexes to modulate synergistically kinetics and pharmacology.


Asunto(s)
Hipocampo/fisiología , Activación del Canal Iónico/fisiología , Proteínas de la Membrana/fisiología , Receptores AMPA/fisiología , Animales , Canales de Calcio , Células Cultivadas , Células HEK293 , Humanos , Activación del Canal Iónico/genética , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Ratas , Ratas Wistar , Potenciales Sinápticos/genética , Potenciales Sinápticos/fisiología
17.
Neuron ; 59(6): 986-96, 2008 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-18817736

RESUMEN

AMPA-type glutamate receptors (GluRs) play major roles in excitatory synaptic transmission. Neuronal AMPA receptors comprise GluR subunits and transmembrane AMPA receptor regulatory proteins (TARPs). Previous studies identified five mammalian TARPs, gamma-2 (or stargazin), gamma-3, gamma-4, gamma-7, and gamma-8, that enhance AMPA receptor function. Here, we classify gamma-5 as a distinct class of TARP that modulates specific GluR2-containing AMPA receptors and displays properties entirely dissimilar from canonical TARPs. Gamma-5 increases peak currents and decreases the steady-state currents selectively from GluR2-containing AMPA receptors. Furthermore, gamma-5 increases rates of GluR2 deactivation and desensitization and decreases glutamate potency. Remarkably, all effects of gamma-5 require editing of GluR2 mRNA. Unlike other TARPs, gamma-5 modulates GluR2 without promoting receptor trafficking. We also find that gamma-7 regulation of GluR2 is dictated by mRNA editing. These data establish gamma-5 and gamma-7 as a separate family of "type II TARPs" that impart distinct physiological features to specific AMPA receptors.


Asunto(s)
Activación del Canal Iónico/fisiología , Potenciales de la Membrana/fisiología , Proteínas de Transporte de Membrana/metabolismo , Neuronas/metabolismo , Receptores AMPA/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Células Cultivadas , Masculino , Proteínas de la Membrana/metabolismo , Isoformas de Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Wistar
18.
J Neurosci ; 27(18): 4969-77, 2007 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-17475805

RESUMEN

AMPA-type glutamate receptors (GluRs) mediate most excitatory signaling in the brain and are composed of GluR principal subunits and transmembrane AMPA receptor regulatory protein (TARP) auxiliary subunits. Previous studies identified four mammalian TARPs, gamma-2 (or stargazin), gamma-3, gamma-4, and gamma-8, that control AMPA receptor trafficking, gating, and pharmacology. Here, we explore roles for the homologous gamma-5 and gamma-7 proteins, which were previously suggested not to serve as TARPs. Western blotting reveals high levels of gamma-5 and gamma-7 in the cerebellum, where gamma-7 is enriched in Purkinje neurons in the molecular layer and glomerular synapses in the granule cell layer. Immunoprecipitation proteomics shows that cerebellar gamma-7 avidly and selectively binds to AMPA receptor GluR subunits and also binds to the AMPA receptor clustering protein, postsynaptic density-95 (PSD-95). Furthermore, gamma-7 occurs together with PSD-95 and AMPA receptor subunits in purified postsynaptic densities. In heterologous cells, gamma-7 but not gamma-5 greatly enhances AMPA receptor glutamate-evoked currents and modulates channel gating. In granule cells from stargazer mice, transfection of gamma-7 but not gamma-5 increases AMPA receptor-mediated currents. Compared with stargazin, gamma-7 differentially modulates AMPA receptor glutamate affinity and kainate efficacy. These studies define gamma-7 as a new member of the TARP family that can differentially influence AMPA receptors in cerebellar neurons.


Asunto(s)
Proteínas de la Membrana/metabolismo , Subunidades de Proteína/metabolismo , Receptores AMPA/metabolismo , Animales , Células Cultivadas , Cerebelo/metabolismo , Cerebelo/fisiología , Humanos , Proteínas de la Membrana/fisiología , Ratones , Ratones Transgénicos , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , Subunidades de Proteína/fisiología , Ratas , Receptores AMPA/fisiología
19.
Methods Mol Biol ; 403: 37-57, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18827986

RESUMEN

Hypofunctioning of glutamate synaptic transmission in the central nervous system (CNS) has been proposed as a factor that may contribute to cognitive deficits associated with various neurological and psychiatric disorders. Positive allosteric modulation of the alpha-amino-3-hydroxy-5-methyl-4-isoxazoleproprionic acid (AMPA) subtype of glutamate receptors has been proposed as a novel therapeutic approach, because these receptors mediate the majority of rapid excitatory neurotransmission and are intimately involved in long-term changes in synaptic plasticity thought to underlie mnemonic processing. By definition, positive allosteric modulators do not affect AMPA receptor activity alone but can markedly enhance ion flux through the ion channel pore in the presence of bound agonist. Despite this commonality, positive allosteric modulators can be segregated on the basis of the preferential effects on AMPA receptor subunits, their alternatively spliced variants and/or their biophysical mechanism of action. This chapter provides a detailed description of the methodologies used to evaluate the potency/efficacy and biophysical mechanism of action of positive allosteric modulators of AMPA receptors.


Asunto(s)
Glutamatos/farmacología , Técnicas de Placa-Clamp/métodos , Receptores AMPA/metabolismo , Regulación Alostérica/efectos de los fármacos , Fenómenos Biofísicos/efectos de los fármacos , Línea Celular , Relación Dosis-Respuesta a Droga , Humanos , Cinética
20.
J Neurosci ; 24(50): 11416-20, 2004 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-15601947

RESUMEN

Flip (i) and flop (o) alternatively spliced variants of the four glutamate AMPA receptor subunits (GluR1-4) are differentially expressed in the CNS and can display distinct rates of desensitization that contribute to the heterogeneity of native AMPA receptor-dependent synaptic responses. In the present study, we initially compared the kinetics of desensitization in response to fast application of glutamate (1 mm) for the eight different homomeric recombinant human AMPA receptors (hGluR1-4i and o) heterologously expressed in mammalian cells. Consistent with previous reports on recombinant rat AMPA receptors, the time constants of desensitization between human GluR1i and GluR1o receptors were the same, whereas the flip isoforms for GluR2-4 receptors exhibited significantly slower rates of desensitization compared with the flop isoforms. To identify the molecular determinants responsible for these functional differences, the effects of exchanging amino acid residues in the flip-flop cassette of GluR2i and GluR2o were investigated. Three amino acid residues in the flip-flop region (Thr765, Pro766, and Ser775 in flip and Asn765, Ala766, and Asn775 in flop) were identified that contribute to splice-variant differences in the rate of desensitization. Recent structural data show that these three residues are located on helix J, which forms part of the intradimer interface of AMPA receptor ligand-binding cores, and that the stability of this interface may regulate desensitization. The present results suggest that these three residues may confer differences in flip and flop receptor desensitization rates by directly and/or indirectly influencing the stability of the interface between adjacent subunits.


Asunto(s)
Receptores AMPA/fisiología , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Línea Celular , Cricetinae , Humanos , Cinética , Potenciales de la Membrana/fisiología , Mesocricetus , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Receptores AMPA/química , Receptores AMPA/genética , Receptores de Ácido Kaínico/fisiología , Proteínas Recombinantes/química , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA