Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 157
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(7): 1465-1477.e18, 2023 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-37001505

RESUMEN

Receptor activity-modifying proteins (RAMPs) modulate the activity of many Family B GPCRs. We show that RAMP2 directly interacts with the glucagon receptor (GCGR), a Family B GPCR responsible for blood sugar homeostasis, and broadly inhibits receptor-induced downstream signaling. HDX-MS experiments demonstrate that RAMP2 enhances local flexibility in select locations in and near the receptor extracellular domain (ECD) and in the 6th transmembrane helix, whereas smFRET experiments show that this ECD disorder results in the inhibition of active and intermediate states of the intracellular surface. We determined the cryo-EM structure of the GCGR-Gs complex at 2.9 Å resolution in the presence of RAMP2. RAMP2 apparently does not interact with GCGR in an ordered manner; however, the receptor ECD is indeed largely disordered along with rearrangements of several intracellular hallmarks of activation. Our studies suggest that RAMP2 acts as a negative allosteric modulator of GCGR by enhancing conformational sampling of the ECD.


Asunto(s)
Glucagón , Receptores de Glucagón , Membrana Celular/metabolismo , Glucagón/metabolismo , Receptores de Glucagón/metabolismo , Proteína 2 Modificadora de la Actividad de Receptores/metabolismo
2.
Nature ; 629(8013): 951-956, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38632403

RESUMEN

Metabotropic glutamate receptors belong to a family of G protein-coupled receptors that are obligate dimers and possess a large extracellular ligand-binding domain that is linked via a cysteine-rich domain to their 7-transmembrane domain1. Upon activation, these receptors undergo a large conformational change to transmit the ligand binding signal from the extracellular ligand-binding domain to the G protein-coupling 7-transmembrane domain2. In this manuscript, we propose a model for a sequential, multistep activation mechanism of metabotropic glutamate receptor subtype 5. We present a series of structures in lipid nanodiscs, from inactive to fully active, including agonist-bound intermediate states. Further, using bulk and single-molecule fluorescence imaging, we reveal distinct receptor conformations upon allosteric modulator and G protein binding.


Asunto(s)
Ligandos , Dominios Proteicos , Receptor del Glutamato Metabotropico 5 , Humanos , Regulación Alostérica/efectos de los fármacos , Fluorescencia , Modelos Moleculares , Unión Proteica , Receptor del Glutamato Metabotropico 5/agonistas , Receptor del Glutamato Metabotropico 5/química , Receptor del Glutamato Metabotropico 5/metabolismo , Imagen Individual de Molécula , Proteínas de Unión al GTP Heterotriméricas/metabolismo
3.
Proc Natl Acad Sci U S A ; 120(43): e2311131120, 2023 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-37844228

RESUMEN

Many neurons in the central nervous system produce a single primary cilium that serves as a specialized signaling organelle. Several neuromodulatory G-protein-coupled receptors (GPCRs) localize to primary cilia in neurons, although it is not understood how GPCR signaling from the cilium impacts circuit function and behavior. We find that the vertebrate ancient long opsin A (VALopA), a Gi-coupled GPCR extraretinal opsin, targets to cilia of zebrafish spinal neurons. In the developing 1-d-old zebrafish, brief light activation of VALopA in neurons of the central pattern generator circuit for locomotion leads to sustained inhibition of coiling, the earliest form of locomotion. We find that a related extraretinal opsin, VALopB, is also Gi-coupled, but is not targeted to cilia. Light-induced activation of VALopB also suppresses coiling, but with faster kinetics. We identify the ciliary targeting domains of VALopA. Retargeting of both opsins shows that the locomotory response is prolonged and amplified when signaling occurs in the cilium. We propose that ciliary localization provides a mechanism for enhancing GPCR signaling in central neurons.


Asunto(s)
Receptores Acoplados a Proteínas G , Pez Cebra , Animales , Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal/fisiología , Opsinas , Opsinas de Bastones , Neuronas , Cilios/fisiología
5.
J Am Chem Soc ; 145(34): 18778-18788, 2023 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-37586061

RESUMEN

Dopamine D2-like receptors (D2R, D3R, and D4R) control diverse physiological and behavioral functions and are important targets for the treatment of a variety of neuropsychiatric disorders. Their complex distribution and activation kinetics in the brain make it difficult to target specific receptor populations with sufficient precision. We describe a new toolkit of light-activatable, fast-relaxing, covalently taggable chemical photoswitches that fully activate, partially activate, or block D2-like receptors. This technology combines the spatiotemporal precision of a photoswitchable ligand (P) with cell type and spatial specificity of a genetically encoded membrane anchoring protein (M) to which the P tethers. These tools set the stage for targeting endogenous D2-like receptor signaling with molecular, cellular, and spatiotemporal precision using only one wavelength of light.


Asunto(s)
Dopamina , Receptores de Dopamina D2 , Dopamina/metabolismo , Receptores de Dopamina D2/metabolismo , Encéfalo/metabolismo
6.
Proc Natl Acad Sci U S A ; 117(34): 20898-20907, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32788354

RESUMEN

The voltage-gated proton channel Hv1 is a member of the voltage-gated ion channel superfamily, which stands out in design: It is a dimer of two voltage-sensing domains (VSDs), each containing a pore pathway, a voltage sensor (S4), and a gate (S1) and forming its own ion channel. Opening of the two channels in the dimer is cooperative. Part of the cooperativity is due to association between coiled-coil domains that extend intracellularly from the S4s. Interactions between the transmembrane portions of the subunits may also contribute, but the nature of transmembrane packing is unclear. Using functional analysis of a mutagenesis scan, biochemistry, and modeling, we find that the subunits form a dimer interface along the entire length of S1, and also have intersubunit contacts between S1 and S4. These interactions exert a strong effect on gating, in particular on the stability of the open state. Our results suggest that gating in Hv1 is tuned by extensive VSD-VSD interactions between the gates and voltage sensors of the dimeric channel.


Asunto(s)
Canales Iónicos/metabolismo , Secuencia de Aminoácidos , Humanos , Activación del Canal Iónico , Canales Iónicos/química , Modelos Moleculares , Conformación Proteica , Multimerización de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Protones
7.
J Am Chem Soc ; 143(24): 8951-8956, 2021 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-34115935

RESUMEN

G protein-coupled receptors (GPCRs) are the most common targets of drug discovery. However, the similarity between related GPCRs combined with the complex spatiotemporal dynamics of receptor activation in vivo has hindered drug development. Photopharmacology offers the possibility of using light to control the location and timing of drug action by incorporating a photoisomerizable azobenzene into a GPCR ligand, enabling rapid and reversible switching between an inactive and active configuration. Recent advances in this area include (i) photoagonists and photoantagonists that directly control receptor activity but are nonselective because they bind conserved sites, and (ii) photoallosteric modulators that bind selectively to nonconserved sites but indirectly control receptor activity by modulating the response to endogenous ligand. In this study, we designed a photoswitchable allosteric agonist that targets a nonconserved allosteric site for selectivity and activates the receptor on its own to provide direct control. This work culminated in the development of aBINA, a photoswitchable allosteric agonist that selectively activates the Gi/o-coupled metabotropic glutamate receptor 2 (mGluR2). aBINA is the first example of a new class of precision drugs for GPCRs and other clinically important signaling proteins.


Asunto(s)
Derivados del Benceno/farmacología , Receptores Acoplados a Proteínas G/agonistas , Regulación Alostérica/efectos de los fármacos , Derivados del Benceno/síntesis química , Derivados del Benceno/química , Humanos , Ligandos , Procesos Fotoquímicos
8.
Nat Chem Biol ; 15(6): 623-631, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31036923

RESUMEN

Sphingosine-1-phosphate (S1P) plays important roles as a signaling lipid in a variety of physiological and pathophysiological processes. S1P signals via a family of G-protein-coupled receptors (GPCRs) (S1P1-5) and intracellular targets. Here, we report on photoswitchable analogs of S1P and its precursor sphingosine, respectively termed PhotoS1P and PhotoSph. PhotoS1P enables optical control of S1P1-3, shown through electrophysiology and Ca2+ mobilization assays. We evaluated PhotoS1P in vivo, where it reversibly controlled S1P3-dependent pain hypersensitivity in mice. The hypersensitivity induced by PhotoS1P is comparable to that induced by S1P. PhotoS1P is uniquely suited for the study of S1P biology in cultured cells and in vivo because it exhibits prolonged metabolic stability compared to the rapidly metabolized S1P. Using lipid mass spectrometry analysis, we constructed a metabolic map of PhotoS1P and PhotoSph. The formation of these photoswitchable lipids was found to be light dependent, providing a novel approach to optically probe sphingolipid biology.


Asunto(s)
Lisofosfolípidos/metabolismo , Esfingosina/análogos & derivados , Animales , Lisofosfolípidos/química , Ratones , Modelos Moleculares , Estructura Molecular , Imagen Óptica , Procesos Fotoquímicos , Esfingosina/química , Esfingosina/metabolismo
9.
Nature ; 524(7566): 497-501, 2015 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-26258295

RESUMEN

G-protein-coupled receptors (GPCRs) constitute the largest family of membrane receptors in eukaryotes. Crystal structures have provided insight into GPCR interactions with ligands and G proteins, but our understanding of the conformational dynamics of activation is incomplete. Metabotropic glutamate receptors (mGluRs) are dimeric class C GPCRs that modulate neuronal excitability, synaptic plasticity, and serve as drug targets for neurological disorders. A 'clamshell' ligand-binding domain (LBD), which contains the ligand-binding site, is coupled to the transmembrane domain via a cysteine-rich domain, and LBD closure seems to be the first step in activation. Crystal structures of isolated mGluR LBD dimers led to the suggestion that activation also involves a reorientation of the dimer interface from a 'relaxed' to an 'active' state, but the relationship between ligand binding, LBD closure and dimer interface rearrangement in activation remains unclear. Here we use single-molecule fluorescence resonance energy transfer to probe the activation mechanism of full-length mammalian group II mGluRs. We show that the LBDs interconvert between three conformations: resting, activated and a short-lived intermediate state. Orthosteric agonists induce transitions between these conformational states, with efficacy determined by occupancy of the active conformation. Unlike mGluR2, mGluR3 displays basal dynamics, which are Ca(2+)-dependent and lead to basal protein activation. Our results support a general mechanism for the activation of mGluRs in which agonist binding induces closure of the LBDs, followed by dimer interface reorientation. Our experimental strategy should be widely applicable to study conformational dynamics in GPCRs and other membrane proteins.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia , Receptores de Glutamato Metabotrópico/química , Receptores de Glutamato Metabotrópico/clasificación , Animales , Sitios de Unión , Agonismo Parcial de Drogas , Humanos , Ligandos , Modelos Biológicos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Ratas , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo
10.
J Am Chem Soc ; 142(24): 10612-10616, 2020 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-32469525

RESUMEN

Lysophosphatidic acid (LPA) is a phospholipid that acts as an extracellular signaling molecule and activates the family of lysophosphatidic acid receptors (LPA1-6). These G protein-coupled receptors (GPCRs) are broadly expressed and are particularly important in development as well as in the nervous, cardiovascular, reproductive, gastrointestinal, and pulmonary systems. Here, we report on a photoswitchable analogue of LPA, termed AzoLPA, which contains an azobenzene photoswitch embedded in the acyl chain. AzoLPA enables optical control of LPA receptor activation, shown through its ability to rapidly control LPA-evoked increases in intracellular Ca2+ levels. AzoLPA shows greater activation of LPA receptors in its light-induced cis-form than its dark-adapted (or 460 nm light-induced) trans-form. AzoLPA enabled the optical control of neurite retraction through its activation of the LPA2 receptor.


Asunto(s)
Lisofosfolípidos/metabolismo , Humanos , Lisofosfolípidos/química , Procesos Fotoquímicos , Receptores del Ácido Lisofosfatídico/química , Receptores del Ácido Lisofosfatídico/metabolismo , Transducción de Señal
11.
Nat Chem Biol ; 14(7): 655-663, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29867144

RESUMEN

The unusually high demand for metals in the brain, along with insufficient understanding of how their dysregulation contributes to neurological diseases, motivates the study of how inorganic chemistry influences neural circuitry. We now report that the transition metal copper is essential for regulating rest-activity cycles and arousal. Copper imaging and gene expression analysis in zebrafish identifies the locus coeruleus-norepinephrine (LC-NE) system, a vertebrate-specific neuromodulatory circuit critical for regulating sleep, arousal, attention, memory and emotion, as a copper-enriched unit with high levels of copper transporters CTR1 and ATP7A and the copper enzyme dopamine ß-hydroxylase (DBH) that produces NE. Copper deficiency induced by genetic disruption of ATP7A, which loads copper into DBH, lowers NE levels and hinders LC function as manifested by disruption in rest-activity modulation. Moreover, LC dysfunction caused by copper deficiency from ATP7A disruption can be rescued by restoring synaptic levels of NE, establishing a molecular CTR1-ATP7A-DBH-NE axis for copper-dependent LC function.


Asunto(s)
Cobre/metabolismo , Locus Coeruleus/metabolismo , Norepinefrina/metabolismo , Animales , Cobre/química , Locus Coeruleus/química , Norepinefrina/química , Pez Cebra
12.
Chem Rev ; 118(21): 10748-10773, 2018 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-29874052

RESUMEN

Degenerative retinal diseases such as retinitis pigmentosa (RP) and age-related macular degeneration (AMD) affect millions of people around the world and lead to irreversible vision loss if left untreated. A number of therapeutic strategies have been developed over the years to treat these diseases or restore vision to already blind patients. In this Review, we describe the development and translational application of light-sensitive chemical photoswitches to restore visual function to the blind retina and compare the translational potential of photoswitches with other vision-restoring therapies. This therapeutic strategy is enabled by an efficient fusion of chemical synthesis, chemical biology, and molecular biology and is broadly applicable to other biological systems. We hope this Review will be of interest to chemists as well as neuroscientists and clinicians.


Asunto(s)
Compuestos Azo/uso terapéutico , Ceguera/terapia , Degeneración Retiniana/terapia , Animales , Ceguera/patología , Regulación de la Expresión Génica , Humanos , Luz , Optogenética , Procesos Fotoquímicos , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Neuronas Retinianas/efectos de los fármacos , Neuronas Retinianas/patología , Neuronas Retinianas/efectos de la radiación , Transducción de Señal
13.
Proc Natl Acad Sci U S A ; 114(17): E3546-E3554, 2017 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-28396447

RESUMEN

G protein-coupled receptor (GPCR) signaling occurs in complex spatiotemporal patterns that are difficult to probe using standard pharmacological and genetic approaches. A powerful approach for dissecting GPCRs is to use light-controlled pharmacological agents that are tethered covalently and specifically to genetically engineered receptors. However, deficits in our understanding of the mechanism of such photoswitches have limited application of this approach and its extension to other GPCRs. In this study, we have harnessed the power of bioorthogonal tethering to SNAP and CLIP protein tags to create a family of light-gated metabotropic glutamate receptors (mGluRs). We define the mechanistic determinants of photoswitch efficacy, including labeling efficiency, dependence on photoswitch structure, length dependence of the linker between the protein tag and the glutamate ligand, effective local concentration of the glutamate moiety, and affinity of the receptor for the ligand. We improve the scheme for photoswitch synthesis as well as photoswitch efficiency, and generate seven light-gated group II/III mGluRs, including variants of mGluR2, 3, 6, 7, and 8. Members of this family of light-controlled receptors can be used singly or in specifically labeled, independently light-controlled pairs for multiplexed control of receptor populations.


Asunto(s)
Activación del Canal Iónico , Luz , Receptores de Glutamato Metabotrópico , Transducción de Señal , Animales , Células HEK293 , Humanos , Activación del Canal Iónico/genética , Activación del Canal Iónico/efectos de la radiación , Ratas , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Transducción de Señal/genética , Transducción de Señal/efectos de la radiación
14.
J Am Chem Soc ; 141(29): 11522-11530, 2019 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-31291105

RESUMEN

G protein-coupled receptors (GPCRs) are membrane proteins that play important roles in biology. However, our understanding of their function in complex living systems is limited because we lack tools that can target individual receptors with sufficient precision. State-of-the-art approaches, including DREADDs, optoXRs, and PORTL gated-receptors, control GPCR signaling with molecular, cell type, and temporal specificity. Nonetheless, these tools are based on engineered non-native proteins that may (i) express at nonphysiological levels, (ii) localize and turnover incorrectly, and/or (iii) fail to interact with endogenous partners. Alternatively, membrane-anchored ligands (t-toxins, DARTs) target endogenous receptors with molecular and cell type specificity but cannot be turned on and off. In this study, we used a combination of chemistry, biology, and light to control endogenous metabotropic glutamate receptor 2 (mGluR2), a Family C GPCR, in primary cortical neurons. mGluR2 was rapidly, reversibly, and selectively activated with photoswitchable glutamate tethered to a genetically targeted-plasma membrane anchor (membrane anchored Photoswitchable Orthogonal Remotely Tethered Ligand; maPORTL). Photoactivation was tuned by adjusting the length of the PORTL as well as the expression level and geometry of the membrane anchor. Our findings provide a template for controlling endogenous GPCRs with cell type specificity and high spatiotemporal precision.


Asunto(s)
Biología Molecular/métodos , Receptores de Glutamato Metabotrópico/genética , Aminoácidos/farmacología , Animales , Compuestos Azo/química , Membrana Celular/metabolismo , Ácido Glutámico/química , Células HEK293 , Humanos , Ligandos , Luz , Neuronas/metabolismo , Procesos Fotoquímicos , Ingeniería de Proteínas/métodos , Ratas , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Receptores de Glutamato Metabotrópico/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Xantenos/farmacología
15.
EMBO Rep ; 18(5): 677-692, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28396573

RESUMEN

Membrane receptors and ion channels respond to various stimuli and relay that information across the plasma membrane by triggering specific and timed processes. These include activation of second messengers, allowing ion permeation, and changing cellular excitability, to name a few. Gaining control over equivalent processes is essential to understand neuronal physiology and pathophysiology. Recently, new optical techniques have emerged proffering new remote means to control various functions of defined neuronal populations by light, dubbed optogenetics. Still, optogenetic tools do not typically address the activity of receptors and channels native to neurons (or of neuronal origin), nor gain access to their signaling mechanisms. A related method-synthetic optogenetics-bridges this gap by endowing light sensitivity to endogenous neuronal receptors and channels by the appending of synthetic, light-receptive molecules, or photoswitches. This provides the means to photoregulate neuronal receptors and channels and tap into their native signaling mechanisms in select regions of the neurons, such as the synapse. This review discusses the development of synthetic optogenetics as a means to study neuronal receptors and channels remotely, in their natural environment, with unprecedented spatial and temporal precision, and provides an overview of tool design, mode of action, potential clinical applications and insights and achievements gained.


Asunto(s)
Canales Iónicos/metabolismo , Optogenética , Receptores de Neurotransmisores/metabolismo , Transducción de Señal , Sinapsis/fisiología , Animales , Luz , Ratones , Neuronas/fisiología
16.
Proc Natl Acad Sci U S A ; 113(15): 4194-9, 2016 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-27035963

RESUMEN

Twik-related K(+) channel 1 (TREK1), TREK2, and Twik-related arachidonic-acid stimulated K(+) channel (TRAAK) form the TREK subfamily of two-pore-domain K(+) (K2P) channels. Despite sharing up to 78% sequence homology and overlapping expression profiles in the nervous system, these channels show major differences in their regulation by physiological stimuli. For instance, TREK1 is inhibited by external acidification, whereas TREK2 is activated. Here, we investigated the ability of the members of the TREK subfamily to assemble to form functional heteromeric channels with novel properties. Using single-molecule pull-down (SiMPull) from HEK cell lysate and subunit counting in the plasma membrane of living cells, we show that TREK1, TREK2, and TRAAK readily coassemble. TREK1 and TREK2 can each heterodimerize with TRAAK, but do so less efficiently than with each other. We functionally characterized the heterodimers and found that all combinations form outwardly rectifying potassium-selective channels but with variable voltage sensitivity and pH regulation. TREK1-TREK2 heterodimers show low levels of activity at physiological external pH but, unlike their corresponding homodimers, are activated by both acidic and alkaline conditions. Modeling based on recent crystal structures, along with mutational analysis, suggests that each subunit within a TREK1-TREK2 channel is regulated independently via titratable His. Finally, TREK1/TRAAK heterodimers differ in function from TRAAK homodimers in two critical ways: they are activated by both intracellular acidification and alkalinization and are regulated by the enzyme phospholipase D2. Thus, heterodimerization provides a means for diversifying functionality through an expansion of the channel types within the K2P channels.


Asunto(s)
Canales de Potasio de Dominio Poro en Tándem/metabolismo , Línea Celular , Dimerización , Humanos , Concentración de Iones de Hidrógeno , Canales de Potasio de Dominio Poro en Tándem/química
17.
Nat Methods ; 12(9): 852-8, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26167640

RESUMEN

Circuit mapping requires knowledge of both structural and functional connectivity between cells. Although optical tools have been made to assess either the morphology and projections of neurons or their activity and functional connections, few probes integrate this information. We have generated a family of photoactivatable genetically encoded Ca(2+) indicators that combines attributes of high-contrast photolabeling with high-sensitivity Ca(2+) detection in a single-color protein sensor. We demonstrated in cultured neurons and in fruit fly and zebrafish larvae how single cells could be selected out of dense populations for visualization of morphology and high signal-to-noise measurements of activity, synaptic transmission and connectivity. Our design strategy is transferrable to other sensors based on circularly permutated GFP (cpGFP).


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Proteínas Luminiscentes/metabolismo , Neuronas/citología , Neuronas/fisiología , Optogenética/métodos , Animales , Rastreo Celular/métodos , Células Cultivadas , Drosophila , Luz , Proteínas Luminiscentes/genética , Microscopía Fluorescente/métodos , Ingeniería de Proteínas/métodos , Ratas , Pez Cebra
18.
Nat Chem Biol ; 12(4): 261-7, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26878552

RESUMEN

Allostery provides a critical control over enzyme activity, biasing the catalytic site between inactive and active states. We found that the Ciona intestinalis voltage-sensing phosphatase (Ci-VSP), which modifies phosphoinositide signaling lipids (PIPs), has not one but two sequential active states with distinct substrate specificities, whose occupancy is allosterically controlled by sequential conformations of the voltage-sensing domain (VSD). Using fast fluorescence resonance energy transfer (FRET) reporters of PIPs to monitor enzyme activity and voltage-clamp fluorometry to monitor conformational changes in the VSD, we found that Ci-VSP switches from inactive to a PIP3-preferring active state when the VSD undergoes an initial voltage-sensing motion and then into a second PIP2-preferring active state when the VSD activates fully. This two-step allosteric control over a dual-specificity enzyme enables voltage to shape PIP concentrations in time, and provides a mechanism for the complex modulation of PIP-regulated ion channels, transporters, cell motility, endocytosis and exocytosis.


Asunto(s)
Fosfatidilinositoles/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Regulación Alostérica , Sitio Alostérico , Animales , Dominio Catalítico , Ciona intestinalis/enzimología , Transferencia Resonante de Energía de Fluorescencia , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Oocitos , Técnicas de Placa-Clamp , Monoéster Fosfórico Hidrolasas/genética , Especificidad por Sustrato , Xenopus laevis
19.
Proc Natl Acad Sci U S A ; 112(37): 11690-5, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26324944

RESUMEN

Caenorhabditis elegans senses gentle touch in the six touch receptor neurons (TRNs) using a mechanotransduction complex that contains the pore-forming degenerin/epithelial sodium channel (DEG/ENaC) proteins MEC-4 and MEC-10. Past work has suggested these proteins interact with the paraoxonase-like MEC-6 and the cholesterol-binding stomatin-like MEC-2 proteins. Using single molecule optical imaging in Xenopus oocytes, we found that MEC-4 forms homotrimers and MEC-4 and MEC-10 form 4:4:10 heterotrimers. MEC-6 and MEC-2 do not associate tightly with these trimers and do not influence trimer stoichiometry, indicating that they are not part of the core channel transduction complex. Consistent with the in vitro data, MEC-10, but not MEC-6, formed puncta in TRN neurites that colocalize with MEC-4 when MEC-4 is overexpressed in the TRNs.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Caenorhabditis elegans/fisiología , Mecanorreceptores/fisiología , Proteínas de la Membrana/fisiología , Neuronas/fisiología , Animales , Animales Modificados Genéticamente , Arildialquilfosfatasa/química , Proteínas de Caenorhabditis elegans/química , Electrofisiología , Canales Epiteliales de Sodio/química , Mecanotransducción Celular/fisiología , Proteínas de la Membrana/química , Oocitos/citología , Unión Proteica , Multimerización de Proteína , Xenopus laevis
20.
Proc Natl Acad Sci U S A ; 112(7): E776-85, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25653339

RESUMEN

Mammalian neurotransmitter-gated receptors can be conjugated to photoswitchable tethered ligands (PTLs) to enable photoactivation, or photoantagonism, while preserving normal function at neuronal synapses. "MAG" PTLs for ionotropic and metabotropic glutamate receptors (GluRs) are based on an azobenzene photoswitch that is optimally switched into the liganding state by blue or near-UV light, wavelengths that penetrate poorly into the brain. To facilitate deep-tissue photoactivation with near-infrared light, we measured the efficacy of two-photon (2P) excitation for two MAG molecules using nonlinear spectroscopy. Based on quantitative characterization, we find a recently designed second generation PTL, L-MAG0460, to have a favorable 2P absorbance peak at 850 nm, enabling efficient 2P activation of the GluK2 kainate receptor, LiGluR. We also achieve 2P photoactivation of a metabotropic receptor, LimGluR3, with a new mGluR-specific PTL, D-MAG0460. 2P photoswitching is efficiently achieved using digital holography to shape illumination over single somata of cultured neurons. Simultaneous Ca(2+)-imaging reports on 2P photoswitching in multiple cells with high temporal resolution. The combination of electrophysiology or Ca(2+) imaging with 2P activation by optical wavefront shaping should make second generation PTL-controlled receptors suitable for studies of intact neural circuits.


Asunto(s)
Compuestos Azo/química , Optogenética , Fotones , Receptores de Glutamato/química , Animales , Células Cultivadas , Isomerismo , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato/metabolismo , Receptores de Glutamato/efectos de la radiación , Análisis Espectral/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA