Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros











Intervalo de año de publicación
1.
Nat Commun ; 15(1): 8297, 2024 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-39333071

RESUMEN

The cAMP cascade is increasingly recognized to transduce physiological effects locally through spatially limited cAMP gradients. However, little is known about how adenylyl cyclase enzymes that initiate cAMP gradients are localized. Here we address this question in physiologically relevant striatal neurons and investigate how AC localization impacts downstream signaling function. We show that the major striatal AC isoforms are differentially sorted between ciliary and extraciliary domains of the plasma membrane, and that one isoform, AC9, is uniquely concentrated in endosomes. We identify key sorting determinants in the N-terminal cytoplasmic domain responsible for isoform-specific localization. We further show that AC9-containing endosomes accumulate activated dopamine receptors and form an elaborately intertwined network with juxtanuclear PKA stores bound to Golgi membranes. Finally, we provide evidence that endosomal localization enables AC9 to selectively elevate PKA activity in the nucleus relative to the cytoplasm. Together, these results reveal a precise spatial landscape of the cAMP cascade in neurons and a key role of AC localization in directing downstream PKA signaling to the nucleus.


Asunto(s)
Adenilil Ciclasas , Proteínas Quinasas Dependientes de AMP Cíclico , AMP Cíclico , Dopamina , Endosomas , Neuronas , Transducción de Señal , Adenilil Ciclasas/metabolismo , Animales , Dopamina/metabolismo , Neuronas/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Endosomas/metabolismo , Membrana Celular/metabolismo , Ratones , Cuerpo Estriado/metabolismo , Cuerpo Estriado/citología , Receptores Dopaminérgicos/metabolismo , Aparato de Golgi/metabolismo , Núcleo Celular/metabolismo , Humanos , Células HEK293
2.
Nat Chem Biol ; 20(3): 323-332, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37749347

RESUMEN

Many G protein-coupled receptors (GPCRs) initiate a second phase of stimulatory heterotrimeric G protein (Gs)-coupled cAMP signaling after endocytosis. The prevailing current view is that the endosomal signal is inherently ß-arrestin-dependent because ß-arrestin is necessary for receptor internalization and, for some GPCRs, to prolong the endosomal signal. Here we revise this view by showing that the vasoactive intestinal peptide receptor 1 (VIPR1), a secretin-family polypeptide hormone receptor, does not require ß-arrestin to internalize or to generate an endosomal signal. ß-Arrestin instead resolves the plasma membrane and endosomal signaling phases into sequential cAMP peaks by desensitizing the plasma membrane phase without affecting the endosomal phase. This appears to occur through the formation of functionally distinct VIPR1-ß-arrestin complexes at each location that differ in their phosphorylation dependence. We conclude that endosomal GPCR signaling can occur in the absence of ß-arrestin and that ß-arrestin sculpts the spatiotemporal profile of cellular GPCR-G protein signaling through location-specific remodeling of GPCR-ß-arrestin complexes.


Asunto(s)
Hormonas Peptídicas , Transducción de Señal , beta-Arrestinas , beta-Arrestina 1 , Membrana Celular
3.
J Biol Chem ; 299(11): 105293, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37774973

RESUMEN

ß-arrestins play a key role in G protein-coupled receptor (GPCR) internalization, trafficking, and signaling. Whether ß-arrestins act independently of G protein-mediated signaling has not been fully elucidated. Studies using genome-editing approaches revealed that whereas G proteins are essential for mitogen-activated protein kinase activation by GPCRs., ß-arrestins play a more prominent role in signal compartmentalization. However, in the absence of G proteins, GPCRs may not activate ß-arrestins, thereby limiting the ability to distinguish G protein from ß-arrestin-mediated signaling events. We used ß2-adrenergic receptor (ß2AR) and its ß2AR-C tail mutant expressed in human embryonic kidney 293 cells wildtype or CRISPR-Cas9 gene edited for Gαs, ß-arrestin1/2, or GPCR kinases 2/3/5/6 in combination with arrestin conformational sensors to elucidate the interplay between Gαs and ß-arrestins in controlling gene expression. We found that Gαs is not required for ß2AR and ß-arrestin conformational changes, ß-arrestin recruitment, and receptor internalization, but that Gαs dictates the GPCR kinase isoforms involved in ß-arrestin recruitment. By RNA-Seq analysis, we found that protein kinase A and mitogen-activated protein kinase gene signatures were activated by stimulation of ß2AR in wildtype and ß-arrestin1/2-KO cells but absent in Gαs-KO cells. These results were validated by re-expressing Gαs in the corresponding KO cells and silencing ß-arrestins in wildtype cells. These findings were extended to cellular systems expressing endogenous levels of ß2AR. Overall, our results support that Gs is essential for ß2AR-promoted protein kinase A and mitogen-activated protein kinase gene expression signatures, whereas ß-arrestins initiate signaling events modulating Gαs-driven nuclear transcriptional activity.


Asunto(s)
Proteínas de Unión al GTP , Regulación de la Expresión Génica , Receptores Adrenérgicos beta 2 , beta-Arrestinas , Humanos , beta-Arrestina 1/genética , beta-Arrestina 1/metabolismo , Arrestina beta 2/genética , Arrestina beta 2/metabolismo , beta-Arrestinas/genética , beta-Arrestinas/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación de la Expresión Génica/genética , Proteínas de Unión al GTP/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Receptores Adrenérgicos beta 2/química , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Células HEK293 , Subunidades alfa de la Proteína de Unión al GTP/genética , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Estructura Terciaria de Proteína , Isoformas de Proteínas , Activación Enzimática/genética
4.
Cell ; 185(21): 3950-3965.e25, 2022 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-36170854

RESUMEN

The G protein-coupled receptor cascade leading to production of the second messenger cAMP is replete with pharmacologically targetable proteins, with the exception of the Gα subunit, Gαs. GTPases remain largely undruggable given the difficulty of displacing high-affinity guanine nucleotides and the lack of other drug binding sites. We explored a chemical library of 1012 cyclic peptides to expand the chemical search for inhibitors of this enzyme class. We identified two macrocyclic peptides, GN13 and GD20, that antagonize the active and inactive states of Gαs, respectively. Both macrocyclic peptides fine-tune Gαs activity with high nucleotide-binding-state selectivity and G protein class-specificity. Co-crystal structures reveal that GN13 and GD20 distinguish the conformational differences within the switch II/α3 pocket. Cell-permeable analogs of GN13 and GD20 modulate Gαs/Gßγ signaling in cells through binding to crystallographically defined pockets. The discovery of cyclic peptide inhibitors targeting Gαs provides a path for further development of state-dependent GTPase inhibitors.


Asunto(s)
Péptidos , Receptores Acoplados a Proteínas G , GTP Fosfohidrolasas , Nucleótidos de Guanina , Nucleótidos , Péptidos/química , Péptidos Cíclicos/farmacología
5.
Methods Mol Biol ; 2483: 339-349, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35286686

RESUMEN

Nuclear entry of cAMP-dependent protein kinase catalytic subunits is typically inferred from changes in net protein amount or kinase activity in the nucleus. Previous methods to directly assess nuclear entry require kinase subunit overexpression and/or supraphysiological cAMP elevation. We describe a method to detect nuclear entry of catalytic subunits expressed at an endogenous level in living cells, stimulated by cAMP in a physiological range, and in real time.


Asunto(s)
Núcleo Celular , Proteínas Quinasas Dependientes de AMP Cíclico , Bioensayo , Dominio Catalítico , Proyectos de Investigación
6.
J Biol Chem ; 297(1): 100907, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34166681

RESUMEN

Endosomal signaling downstream of G-protein-coupled receptors (GPCRs) has emerged as a novel paradigm with important pharmacological and physiological implications. However, our knowledge of the functional consequences of intracellular signaling is incomplete. To begin to address this gap, we combined an optogenetic approach for site-specific generation of the prototypical second messenger generated by active GPCRs, cyclic AMP (cAMP), with unbiased mass-spectrometry-based analysis of the phosphoproteome. We identified 218 unique, high-confidence sites whose phosphorylation is either increased or decreased in response to cAMP elevation. We next determined that the same amount of cAMP produced from the endosomal membrane led to more robust changes in phosphorylation than the plasma membrane. Remarkably, this was true for the entire repertoire of 218 identified targets and irrespective of their annotated subcellular localizations (endosome, cell surface, nucleus, cytosol). Furthermore, we identified a particularly strong endosome bias for a subset of proteins that are dephosphorylated in response to cAMP. Through bioinformatics analysis, we established these targets as putative substrates for protein phosphatase 2A (PP2A), and we propose compartmentalized activation of PP2A by cAMP-responsive kinases as the likely underlying mechanism. Altogether, our study extends the concept that endosomal signaling is a significant functional contributor to cellular responsiveness to cAMP by establishing a unique role for localized cAMP production in defining categorically distinct phosphoresponses.


Asunto(s)
AMP Cíclico/metabolismo , Endosomas/metabolismo , Fosfoproteínas/metabolismo , Proteoma/metabolismo , Animales , Células HEK293 , Humanos , Fosfoproteínas/química , Fosforilación , Dominios Proteicos , Proteína Fosfatasa 2/metabolismo , Proteoma/química
7.
Nat Chem Biol ; 17(5): 558-566, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33649598

RESUMEN

G-protein-coupled receptor-regulated cAMP production from endosomes can specify signaling to the nucleus by moving the source of cAMP without changing its overall amount. How this is possible remains unknown because cAMP gradients dissipate over the nanoscale, whereas endosomes typically localize micrometers from the nucleus. We show that the key location-dependent step for endosome-encoded transcriptional control is nuclear entry of cAMP-dependent protein kinase (PKA) catalytic subunits. These are sourced from punctate accumulations of PKA holoenzyme that are densely distributed in the cytoplasm and titrated by global cAMP into a discrete metastable state, in which catalytic subunits are bound but dynamically exchange. Mobile endosomes containing activated receptors collide with the metastable PKA puncta and pause in close contact. We propose that these properties enable cytoplasmic PKA to act collectively like a semiconductor, converting nanoscale cAMP gradients generated from endosomes into microscale elevations of free catalytic subunits to direct downstream signaling.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Citoplasma/metabolismo , Endosomas/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal/genética , Animales , Dominio Catalítico , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Cadenas Pesadas de Clatrina/antagonistas & inhibidores , Cadenas Pesadas de Clatrina/genética , Cadenas Pesadas de Clatrina/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Citoplasma/ultraestructura , Dinamina I/genética , Dinamina I/metabolismo , Endosomas/ultraestructura , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Holoenzimas/genética , Holoenzimas/metabolismo , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Unión Proteica , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Receptores Adrenérgicos beta 2/genética
8.
PLoS Genet ; 16(10): e1009103, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33052901

RESUMEN

G protein-coupled receptors (GPCRs) allow cells to respond to chemical and sensory stimuli through generation of second messengers, such as cyclic AMP (cAMP), which in turn mediate a myriad of processes, including cell survival, proliferation, and differentiation. In order to gain deeper insights into the complex biology and physiology of these key cellular pathways, it is critical to be able to globally map the molecular factors that shape cascade function. Yet, to this date, efforts to systematically identify regulators of GPCR/cAMP signaling have been lacking. Here, we combined genome-wide screening based on CRISPR interference with a novel sortable transcriptional reporter that provides robust readout for cAMP signaling, and carried out a functional screen for regulators of the pathway. Due to the sortable nature of the platform, we were able to assay regulators with strong and moderate phenotypes by analyzing sgRNA distribution among three fractions with distinct reporter expression. We identified 45 regulators with strong and 50 regulators with moderate phenotypes not previously known to be involved in cAMP signaling. In follow-up experiments, we validated the functional effects of seven newly discovered mediators (NUP93, PRIM1, RUVBL1, PKMYT1, TP53, SF3A2, and HRAS), and showed that they control distinct steps of the pathway. Thus, our study provides proof of principle that the screening platform can be applied successfully to identify bona fide regulators of GPCR/second messenger cascades in an unbiased and high-throughput manner, and illuminates the remarkable functional diversity among GPCR regulators.


Asunto(s)
Sistemas CRISPR-Cas/genética , Proliferación Celular/genética , AMP Cíclico/genética , Receptores Acoplados a Proteínas G/genética , ATPasas Asociadas con Actividades Celulares Diversas/genética , Proteínas Portadoras/genética , Diferenciación Celular/genética , Células Cultivadas , ADN Helicasas/genética , ADN Primasa/genética , Células HEK293 , Humanos , Proteínas de la Membrana/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Factores de Empalme de ARN/genética , Transducción de Señal/genética , Proteína p53 Supresora de Tumor/genética
9.
Elife ; 92020 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-32515353

RESUMEN

GPCRs are increasingly recognized to initiate signaling via heterotrimeric G proteins as they move through the endocytic network, but little is known about how relevant G protein effectors are localized. Here we report selective trafficking of adenylyl cyclase type 9 (AC9) from the plasma membrane to endosomes while adenylyl cyclase type 1 (AC1) remains in the plasma membrane, and stimulation of AC9 trafficking by ligand-induced activation of Gs-coupled GPCRs. AC9 transits a similar, dynamin-dependent early endocytic pathway as ligand-activated GPCRs. However, unlike GPCR traffic control which requires ß-arrestin but not Gs, AC9 traffic control requires Gs but not ß-arrestin. We also show that AC9, but not AC1, mediates cAMP production stimulated by endogenous receptor activation in endosomes. These results reveal dynamic and isoform-specific trafficking of adenylyl cyclase in the endocytic network, and a discrete role of a heterotrimeric G protein in regulating the subcellular distribution of a relevant effector.


Cells sense changes in their chemical environment using proteins called receptors. These proteins often sit on the cell surface, detecting molecules outside the cell and relaying messages across the membrane to the cell interior. The largest family of receptors is formed of 'G protein-coupled receptors' (or GPCRs for short), so named because they relay messages through so-called G proteins, which then send information into the cell by interacting with other proteins called effectors. Next, the receptors leave the cell surface, travelling into the cell in compartments called endosomes. Researchers used to think that this switched the receptors off, stopping the signaling process, but it is now clear that this is not the case. Some receptors continue to signal from inside the cell, though the details of how this works are unclear. For signals to pass from a GPCR to a G protein to an effector, all three proteins need to be in the same place. This is certainly happening at the cell surface, but whether all three types of proteins come together inside endosomes is less clear. One way to find out is to look closely at the location of effector proteins when GPCRs are receiving signals. One well-studied effector of GPCR signaling is called adenylyl cyclase, a protein that makes a signal molecule called cAMP. Some G proteins switch adenylyl cyclase on, increasing cAMP production, while others switch it off. To find out how GPCRs send signals from inside endosomes, Lazar et al tracked adenylyl cyclase proteins inside human cells. This revealed that a type of adenylyl cyclase, known as adenylyl cyclase 9, follows receptors as they travel into the cell. Under the influence of active G proteins, activated adenylyl cyclase 9 left the cell surface and entered the endosomes. Once inside the cell, adenylyl cyclase 9 generated the signal molecule cAMP, allowing the receptors to send messages from inside the cell. Other types of adenylyl cyclase behaved differently. Adenylyl cyclase 1, for example, remained on the cell surface even after its receptors had left, and did not signal from inside the cell at all. Which cell behaviors are triggered from the membrane, and which are triggered from inside the cell is an important question in drug design. Understanding where effector proteins are active is a step towards finding the answers. This could help research into diseases of the heart, the liver and the lungs, all of which use adenylyl cyclase 9 to send signals.


Asunto(s)
Adenilil Ciclasas/metabolismo , Endosomas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adenilil Ciclasas/genética , Membrana Celular/genética , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Endosomas/genética , Humanos , Transporte de Proteínas , Receptores Acoplados a Proteínas G/genética , beta-Arrestinas/genética , beta-Arrestinas/metabolismo
10.
Cell ; 174(3): 505-520, 2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-30053424

RESUMEN

Although gene discovery in neuropsychiatric disorders, including autism spectrum disorder, intellectual disability, epilepsy, schizophrenia, and Tourette disorder, has accelerated, resulting in a large number of molecular clues, it has proven difficult to generate specific hypotheses without the corresponding datasets at the protein complex and functional pathway level. Here, we describe one path forward-an initiative aimed at mapping the physical and genetic interaction networks of these conditions and then using these maps to connect the genomic data to neurobiology and, ultimately, the clinic. These efforts will include a team of geneticists, structural biologists, neurobiologists, systems biologists, and clinicians, leveraging a wide array of experimental approaches and creating a collaborative infrastructure necessary for long-term investigation. This initiative will ultimately intersect with parallel studies that focus on other diseases, as there is a significant overlap with genes implicated in cancer, infectious disease, and congenital heart defects.


Asunto(s)
Mapeo Cromosómico/métodos , Trastornos del Neurodesarrollo/genética , Biología de Sistemas/métodos , Redes Reguladoras de Genes/genética , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo/métodos , Genómica/métodos , Humanos , Neurobiología/métodos , Neuropsiquiatría
11.
Neuron ; 98(5): 963-976.e5, 2018 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-29754753

RESUMEN

Opioid receptors (ORs) precisely modulate behavior when activated by native peptide ligands but distort behaviors to produce pathology when activated by non-peptide drugs. A fundamental question is how drugs differ from peptides in their actions on target neurons. Here, we show that drugs differ in the subcellular location at which they activate ORs. We develop a genetically encoded biosensor that directly detects ligand-induced activation of ORs and uncover a real-time map of the spatiotemporal organization of OR activation in living neurons. Peptide agonists produce a characteristic activation pattern initiated in the plasma membrane and propagating to endosomes after receptor internalization. Drugs produce a different activation pattern by additionally driving OR activation in the somatic Golgi apparatus and Golgi elements extending throughout the dendritic arbor. These results establish an approach to probe the cellular basis of neuromodulation and reveal that drugs distort the spatiotemporal landscape of neuronal OR activation.


Asunto(s)
Analgésicos Opioides/metabolismo , Membrana Celular/metabolismo , Dendritas/metabolismo , Endosomas/metabolismo , Aparato de Golgi/metabolismo , Neuronas/metabolismo , Péptidos/metabolismo , Receptores Opioides/metabolismo , Animales , Técnicas Biosensibles , Encefalina Ala(2)-MeFe(4)-Gli(5)/metabolismo , Encefalina D-Penicilamina (2,5)/metabolismo , Leucina Encefalina-2-Alanina/metabolismo , Células HEK293 , Células HeLa , Humanos , Espacio Intracelular , Microscopía Fluorescente , Morfina/metabolismo , Naloxona , Antagonistas de Narcóticos , Ratas , Análisis Espacio-Temporal
12.
Cell Rep ; 22(10): 2593-2600, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29514089

RESUMEN

Growth factor binding to EGFR drives conformational changes that promote homodimerization and transphosphorylation, followed by adaptor recruitment, oligomerization, and signaling through Ras. Whether specific receptor conformations and oligomerization states are necessary for efficient activation of Ras is unclear. We therefore evaluated the sufficiency of a phosphorylated EGFR dimer to activate Ras without growth factor by developing a chemical-genetic strategy to crosslink and "trap" full-length EGFR homodimers on cells. Trapped dimers become phosphorylated and recruit adaptor proteins at stoichiometry equivalent to that of EGF-stimulated receptors. Surprisingly, these phosphorylated dimers do not activate Ras, Erk, or Akt. In the absence of EGF, phosphorylated dimers do not further oligomerize or reorganize on cell membranes. These results suggest that a phosphorylated EGFR dimer loaded with core signaling adapters is not sufficient to activate Ras and that EGFR ligands contribute to conformational changes or receptor dynamics necessary for oligomerization and efficient signal propagation through the SOS-Ras-MAPK pathway.


Asunto(s)
Receptores ErbB/metabolismo , Multimerización de Proteína , Proteínas ras/metabolismo , Vesículas Cubiertas por Clatrina/efectos de los fármacos , Vesículas Cubiertas por Clatrina/metabolismo , Reactivos de Enlaces Cruzados/química , Reactivos de Enlaces Cruzados/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/química , Células HEK293 , Humanos , Ligandos , Fosforilación/efectos de los fármacos , Fosfotirosina/metabolismo , Conformación Proteica , Transducción de Señal/efectos de los fármacos
13.
Mol Pharmacol ; 91(1): 65-73, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27821547

RESUMEN

G protein-coupled receptors (GPCRs), the largest family of signaling receptors, are critically regulated by endosomal trafficking, suggesting that endosomes might provide new strategies for manipulating GPCR signaling. Here we test this hypothesis by focusing on class III phosphatidylinositol 3-kinase (Vps34), which is an essential regulator of endosomal trafficking. We verify that Vps34 is required for recycling of the ß2-adrenoceptor (ß2AR), a prototypical GPCR, and then investigate the effects of Vps34 inhibition on the canonical cAMP response elicited by ß2AR activation. Vps34 inhibition impairs the ability of cells to recover this response after prolonged activation, which is in accord with the established role of recycling in GPCR resensitization. In addition, Vps34 inhibition also attenuates the short-term cAMP response, and its effect begins several minutes after initial agonist application. These results establish Vps34 as an essential determinant of both short-term and long-term canonical GPCR signaling, and support the potential utility of the endosomal system as a druggable target for signaling.


Asunto(s)
Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Endosomas/enzimología , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal , AMP Cíclico/metabolismo , Células HEK293 , Humanos , Modelos Biológicos , Fosfatos de Fosfatidilinositol/metabolismo
14.
Proc Natl Acad Sci U S A ; 113(39): E5721-30, 2016 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-27621449

RESUMEN

We previously showed that guanine nucleotide-binding (G) protein α subunit (Gα)-interacting vesicle-associated protein (GIV), a guanine-nucleotide exchange factor (GEF), transactivates Gα activity-inhibiting polypeptide 1 (Gαi) proteins in response to growth factors, such as EGF, using a short C-terminal motif. Subsequent work demonstrated that GIV also binds Gαs and that inactive Gαs promotes maturation of endosomes and shuts down mitogenic MAPK-ERK1/2 signals from endosomes. However, the mechanism and consequences of dual coupling of GIV to two G proteins, Gαi and Gαs, remained unknown. Here we report that GIV is a bifunctional modulator of G proteins; it serves as a guanine nucleotide dissociation inhibitor (GDI) for Gαs using the same motif that allows it to serve as a GEF for Gαi. Upon EGF stimulation, GIV modulates Gαi and Gαs sequentially: first, a key phosphomodification favors the assembly of GIV-Gαi complexes and activates GIV's GEF function; then a second phosphomodification terminates GIV's GEF function, triggers the assembly of GIV-Gαs complexes, and activates GIV's GDI function. By comparing WT and GIV mutants, we demonstrate that GIV inhibits Gαs activity in cells responding to EGF. Consequently, the cAMP→PKA→cAMP response element-binding protein signaling axis is inhibited, the transit time of EGF receptor through early endosomes are accelerated, mitogenic MAPK-ERK1/2 signals are rapidly terminated, and proliferation is suppressed. These insights define a paradigm in G-protein signaling in which a pleiotropically acting modulator uses the same motif both to activate and to inhibit G proteins. Our findings also illuminate how such modulation of two opposing Gα proteins integrates downstream signals and cellular responses.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proliferación Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Quinasa 5 Dependiente de la Ciclina/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Subunidades beta de la Proteína de Unión al GTP , Subunidades gamma de la Proteína de Unión al GTP , Guanosina Trifosfato/metabolismo , Células HeLa , Humanos , Proteínas de Microfilamentos/química , Proteínas Mutantes/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica , Proteína Quinasa C-theta/metabolismo , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad , Proteínas de Transporte Vesicular/química
15.
J Biol Chem ; 290(11): 6689-96, 2015 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-25605726

RESUMEN

Some G protein-coupled receptors (GPCRs), in addition to activating heterotrimeric G proteins in the plasma membrane, appear to elicit a "second wave" of G protein activation after ligand-induced internalization. We briefly summarize evidence supporting this view and then discuss what is presently known about the functional significance of GPCR-G protein activation in endosomes. Endosomal activation can shape the cellular response temporally by prolonging its overall duration, and may shape the response spatially by moving the location of intracellular second messenger production relative to effectors.


Asunto(s)
Endosomas/metabolismo , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Animales , Técnicas Biosensibles/métodos , AMP Cíclico/metabolismo , Endocitosis , Humanos
16.
Nat Chem Biol ; 10(12): 1061-5, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25362359

RESUMEN

G protein-coupled receptors (GPCRs) are well known to signal via cyclic AMP (cAMP) production at the plasma membrane, but it is now clear that various GPCRs also signal after internalization. Apart from its temporal impact through prolonging the cellular response, we wondered whether the endosome-initiated signal encodes any discrete spatial information. Using the ß2-adrenoceptor (ß2-AR) as a model, we show that endocytosis is required for the full repertoire of downstream cAMP-dependent transcriptional control. Next, we describe an orthogonal optogenetic approach to definitively establish that the location of cAMP production is indeed the critical variable determining the transcriptional response. Finally, our results suggest that this spatial encoding scheme helps cells functionally discriminate chemically distinct ß2-AR ligands according to differences in their ability to promote receptor endocytosis. These findings reveal a discrete principle for achieving cellular signaling specificity based on endosome-mediated spatial encoding of intracellular second messenger production and 'location-aware' downstream transcriptional control.


Asunto(s)
Adenilil Ciclasas/genética , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Endosomas/metabolismo , Receptores Adrenérgicos beta 2/genética , Adenilil Ciclasas/metabolismo , Antagonistas de Receptores Adrenérgicos beta 2/farmacología , Membrana Celular/efectos de los fármacos , Dinaminas/antagonistas & inhibidores , Dinaminas/genética , Dinaminas/metabolismo , Endocitosis , Endosomas/efectos de los fármacos , Regulación de la Expresión Génica , Células HEK293 , Humanos , Hidrazonas/farmacología , Naftoles/farmacología , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Transporte de Proteínas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal , Transcripción Genética
17.
PLoS One ; 8(8): e70857, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23936473

RESUMEN

A number of G protein-coupled receptors (GPCRs) localize to primary cilia but the functional significance of cilia to GPCR signaling remains incompletely understood. We investigated this question by focusing on the D1 dopamine receptor (D1R) and beta-2 adrenergic receptor (B2AR), closely related catecholamine receptors that signal by stimulating production of the diffusible second messenger cyclic AMP (cAMP) but differ in localization relative to cilia. D1Rs robustly concentrate on cilia of IMCD3 cells, as shown previously in other ciliated cell types, but disrupting cilia did not affect D1R surface expression or ability to mediate a concentration-dependent cAMP response. By developing a FRET-based biosensor suitable for resolving intra- from extra- ciliary cAMP changes, we found that the D1R-mediated cAMP response is not restricted to cilia and extends into the extra-ciliary cytoplasm. Conversely the B2AR, which we show here is effectively excluded from cilia, also generated a cAMP response in both ciliary and extra-ciliary compartments. We identified a distinct signaling effect of primary cilia through investigating GPR88, an orphan GPCR that is co-expressed with the D1R in brain, and which we show here is targeted to cilia similarly to the D1R. In ciliated cells, mutational activation of GPR88 strongly reduced the D1R-mediated cAMP response but did not affect the B2AR-mediated response. In marked contrast, in non-ciliated cells, GPR88 was distributed throughout the plasma membrane and inhibited the B2AR response. These results identify a discrete 'insulating' function of primary cilia in conferring selectivity on integrated catecholamine signaling through lateral segregation of receptors, and suggest a cellular activity of GPR88 that might underlie its effects on dopamine-dependent behaviors.


Asunto(s)
Cilios/metabolismo , AMP Cíclico/metabolismo , Embrión de Mamíferos/metabolismo , Neuronas/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Técnicas Biosensibles , Membrana Celular/metabolismo , Células Cultivadas , Embrión de Mamíferos/citología , Femenino , Transferencia Resonante de Energía de Fluorescencia , Humanos , Técnicas para Inmunoenzimas , Ratones , Neuronas/citología , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Receptor Cross-Talk , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética
18.
Nature ; 495(7442): 534-8, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23515162

RESUMEN

A long-held tenet of molecular pharmacology is that canonical signal transduction mediated by G-protein-coupled receptor (GPCR) coupling to heterotrimeric G proteins is confined to the plasma membrane. Evidence supporting this traditional view is based on analytical methods that provide limited or no subcellular resolution. It has been subsequently proposed that signalling by internalized GPCRs is restricted to G-protein-independent mechanisms such as scaffolding by arrestins, or GPCR activation elicits a discrete form of persistent G protein signalling, or that internalized GPCRs can indeed contribute to the acute G-protein-mediated response. Evidence supporting these various latter hypotheses is indirect or subject to alternative interpretation, and it remains unknown if endosome-localized GPCRs are even present in an active form. Here we describe the application of conformation-specific single-domain antibodies (nanobodies) to directly probe activation of the ß2-adrenoceptor, a prototypical GPCR, and its cognate G protein, Gs (ref. 12), in living mammalian cells. We show that the adrenergic agonist isoprenaline promotes receptor and G protein activation in the plasma membrane as expected, but also in the early endosome membrane, and that internalized receptors contribute to the overall cellular cyclic AMP response within several minutes after agonist application. These findings provide direct support for the hypothesis that canonical GPCR signalling occurs from endosomes as well as the plasma membrane, and suggest a versatile strategy for probing dynamic conformational change in vivo.


Asunto(s)
Técnicas Biosensibles/métodos , Endosomas/metabolismo , Receptores Adrenérgicos beta 2/química , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal , Agonistas de Receptores Adrenérgicos beta 2/farmacología , Membrana Celular/química , Membrana Celular/metabolismo , Vesículas Cubiertas por Clatrina , AMP Cíclico/metabolismo , Endocitosis , Endosomas/química , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Isoproterenol/farmacología , Modelos Biológicos , Conformación Proteica , Receptores Adrenérgicos beta 2/inmunología , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/inmunología
19.
Adv Pharmacol ; 62: 279-314, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21907913

RESUMEN

PDZ proteins, named for the common structural domain shared by the postsynaptic density protein (PSD95), Drosophila disc large tumor suppressor (DlgA), and zonula occludens-1 protein (ZO-1), constitute a family of 200-300 recognized members. These cytoplasmic adapter proteins are capable of assembling a variety of membrane-associated proteins and signaling molecules in short-lived functional units. Here, we review PDZ proteins that participate in the regulation of signaling, trafficking, and function of G protein-coupled receptors. Salient structural features of PDZ proteins that allow them to recognize targeted GPCRs are considered. Scaffolding proteins harboring PDZ domains may contain single or multiple PDZ modules and may also include other protein-protein interaction modules. PDZ proteins may impact receptor signaling by diverse mechanisms that include retaining the receptor at the cell membrane, thereby increasing the duration of ligand binding, as well as importantly influencing GPCR internalization, trafficking, recycling, and intracellular sorting. PDZ proteins are also capable of modifying the assembled complex of accessory proteins such as ß-arrestins that themselves regulate GPCR signaling. Additionally, PDZ proteins may modulate GPCR signaling by altering the G protein to which the receptor binds, or affect other regulatory proteins that impact GTPase activity, protein kinase A, phospholipase C, or modify downstream signaling events. Small molecules targeting the PDZ protein-GPCR interaction are being developed and may become important and selective drug candidates.


Asunto(s)
Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Secuencias de Aminoácidos , Animales , Humanos , Estructura Terciaria de Proteína , Transporte de Proteínas
20.
Neuron ; 71(2): 278-90, 2011 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-21791287

RESUMEN

D(1) dopamine receptors are primary mediators of dopaminergic signaling in the CNS. These receptors internalize rapidly following agonist-induced activation, but the functional significance of this process is unknown. We investigated D(1) receptor endocytosis and signaling in HEK293 cells and cultured striatal neurons using real-time fluorescence imaging and cAMP biosensor technology. Agonist-induced activation of D(1) receptors promoted endocytosis of receptors with a time course overlapping that of acute cAMP accumulation. Inhibiting receptor endocytosis blunted acute D(1) receptor-mediated signaling in both dissociated cells and striatal slice preparations. Although endocytic inhibition markedly attenuated acute cAMP accumulation, inhibiting the subsequent recycling of receptors had no effect. Further, D(1) receptors localized in close proximity to endomembrane-associated trimeric G protein and adenylyl cyclase immediately after endocytosis. Together, these results suggest a previously unanticipated role of endocytosis, and the early endocytic pathway, in supporting rapid dopaminergic neurotransmission.


Asunto(s)
Dopamina/metabolismo , Endocitosis/fisiología , Neuronas/fisiología , Transducción de Señal/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Adenilil Ciclasas/farmacología , Animales , Benzazepinas/farmacología , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Células Cultivadas , Cuerpo Estriado/citología , AMP Cíclico/farmacología , Dopamina/farmacología , Agonistas de Dopamina/farmacología , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos , Endocitosis/efectos de los fármacos , Citometría de Flujo/métodos , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Hidrazonas/farmacología , Microscopía Fluorescente/métodos , Neuronas/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Ratas , Receptores de Dopamina D1/genética , Factores de Tiempo , Transfección/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA