Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Cell ; 185(9): 1506-1520.e17, 2022 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-35385687

RESUMEN

Schistosomes cause morbidity and death throughout the developing world due to the massive numbers of eggs female worms deposit into the blood of their host. Studies dating back to the 1920s show that female schistosomes rely on constant physical contact with a male worm both to become and remain sexually mature; however, the molecular details governing this process remain elusive. Here, we uncover a nonribosomal peptide synthetase that is induced in male worms upon pairing with a female and find that it is essential for the ability of male worms to stimulate female development. We demonstrate that this enzyme generates ß-alanyl-tryptamine that is released by paired male worms. Furthermore, synthetic ß-alanyl-tryptamine can replace male worms to stimulate female sexual development and egg laying. These data reveal that peptide-based pheromone signaling controls female schistosome sexual maturation, suggesting avenues for therapeutic intervention and uncovering a role for nonribosomal peptides as metazoan signaling molecules.


Asunto(s)
Péptidos , Feromonas , Schistosoma/crecimiento & desarrollo , Animales , Femenino , Masculino , Biosíntesis de Péptidos Independientes de Ácidos Nucleicos , Triptaminas
4.
J Biol Chem ; 295(8): 2270-2284, 2020 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-31949046

RESUMEN

Besides being regulated by G-protein-coupled receptors, the activity of heterotrimeric G proteins is modulated by many cytoplasmic proteins. GIV/Girdin and DAPLE (Dvl-associating protein with a high frequency of leucine) are the best-characterized members of a group of cytoplasmic regulators that contain a Gα-binding and -activating (GBA) motif and whose dysregulation underlies human diseases, including cancer and birth defects. GBA motif-containing proteins were originally reported to modulate G proteins by binding Gα subunits of the Gi/o family (Gαi) over other families (such as Gs, Gq/11, or G12/13), and promoting nucleotide exchange in vitro However, some evidence suggests that this is not always the case, as phosphorylation of the GBA motif of GIV promotes its binding to Gαs and inhibits nucleotide exchange. The G-protein specificity of DAPLE and how it might affect nucleotide exchange on G proteins besides Gαi remain to be investigated. Here, we show that DAPLE's GBA motif, in addition to Gαi, binds efficiently to members of the Gs and Gq/11 families (Gαs and Gαq, respectively), but not of the G12/13 family (Gα12) in the absence of post-translational phosphorylation. We pinpointed Met-1669 as the residue in the GBA motif of DAPLE that diverges from that in GIV and enables better binding to Gαs and Gαq Unlike the nucleotide-exchange acceleration observed for Gαi, DAPLE inhibited nucleotide exchange on Gαs and Gαq These findings indicate that GBA motifs have versatility in their G-protein-modulating effect, i.e. they can bind to Gα subunits of different classes and either stimulate or inhibit nucleotide exchange depending on the G-protein subtype.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/metabolismo , Secuencia de Aminoácidos , Animales , Bovinos , Células HEK293 , Humanos , Modelos Biológicos , Proteínas Mutantes/metabolismo , Péptidos/metabolismo , Unión Proteica
5.
Elife ; 82019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30835201

RESUMEN

G protein-coupled receptors (GPCRs) are a family of proteins containing seven transmembrane helices, with the N- and C-terminus of the protein located at the extracellular space and cytosol, respectively. Here, we report that ceramide or related sphingolipids might invert the topology of many GPCRs that contain a GXXXN motif in their first transmembrane helix. The functional significance of this topological regulation is illustrated by the CCR5 chemokine receptor. In the absence of lipopolysaccharide (LPS), CCR5 adopts a topology consistent with that of GPCR, allowing mouse peritoneal macrophages to migrate toward its ligand CCL5. LPS stimulation results in increased production of dihydroceramide, which inverts the topology of CCR5, preventing macrophages from migrating toward CCL5. These results suggest that GPCRs may not always adopt the same topology and can be regulated through topological inversion. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that major issues remain unresolved (see decision letter).


Asunto(s)
Ceramidas/metabolismo , Receptores CCR5/química , Receptores CCR5/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Regulación Alostérica , Animales , Movimiento Celular , Células Cultivadas , Lipopolisacáridos/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/fisiología , Ratones Endogámicos C57BL , Conformación Proteica
6.
Proc Natl Acad Sci U S A ; 115(9): E2085-E2094, 2018 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-29440403

RESUMEN

Regulator of G protein signaling z1 (RGSz1), a member of the RGS family of proteins, is present in several networks expressing mu opioid receptors (MOPRs). By using genetic mouse models for global or brain region-targeted manipulations of RGSz1 expression, we demonstrated that the suppression of RGSz1 function increases the analgesic efficacy of MOPR agonists in male and female mice and delays the development of morphine tolerance while decreasing the sensitivity to rewarding and locomotor activating effects. Using biochemical assays and next-generation RNA sequencing, we identified a key role of RGSz1 in the periaqueductal gray (PAG) in morphine tolerance. Chronic morphine administration promotes RGSz1 activity in the PAG, which in turn modulates transcription mediated by the Wnt/ß-catenin signaling pathway to promote analgesic tolerance to morphine. Conversely, the suppression of RGSz1 function stabilizes Axin2-Gαz complexes near the membrane and promotes ß-catenin activation, thereby delaying the development of analgesic tolerance. These data show that the regulation of RGS complexes, particularly those involving RGSz1-Gαz, represents a promising target for optimizing the analgesic actions of opioids without increasing the risk of dependence or addiction.


Asunto(s)
Analgésicos Opioides/farmacología , Proteínas RGS/antagonistas & inhibidores , Vía de Señalización Wnt , Analgesia , Animales , Condicionamiento Psicológico , Femenino , Proteínas de Unión al GTP/metabolismo , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfina/farmacología , Neuronas/metabolismo , Sustancia Gris Periacueductal/metabolismo , Proteínas RGS/metabolismo , Análisis de Secuencia de ARN , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
7.
J Biol Chem ; 292(40): 16787-16801, 2017 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-28842497

RESUMEN

Upon activation by the Gq family of Gα subunits, Gßγ subunits, and some Rho family GTPases, phospholipase C-ß (PLC-ß) isoforms hydrolyze phosphatidylinositol 4,5-bisphosphate to the second messengers inositol 1,4,5-trisphosphate and diacylglycerol. PLC-ß isoforms also function as GTPase-activating proteins, potentiating Gq deactivation. To elucidate the mechanism of this mutual regulation, we measured the thermodynamics and kinetics of PLC-ß3 binding to Gαq FRET and fluorescence correlation spectroscopy, two physically distinct methods, both yielded Kd values of about 200 nm for PLC-ß3-Gαq binding. This Kd is 50-100 times greater than the EC50 for Gαq-mediated PLC-ß3 activation and for the Gαq GTPase-activating protein activity of PLC-ß. The measured Kd was not altered either by the presence of phospholipid vesicles, phosphatidylinositol 4,5-bisphosphate and Ca2+, or by the identity of the fluorescent labels. FRET-based kinetic measurements were also consistent with a Kd of 200 nm We determined that PLC-ß3 hysteresis, whereby PLC-ß3 remains active for some time following either Gαq-PLC-ß3 dissociation or PLC-ß3-potentiated Gαq deactivation, is not sufficient to explain the observed discrepancy between EC50 and Kd These results indicate that the mechanism by which Gαq and PLC-ß3 mutually regulate each other is far more complex than a simple, two-state allosteric model and instead is probably kinetically determined.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/química , Modelos Químicos , Fosfolipasa C beta/química , Regulación Alostérica/fisiología , Calcio/química , Calcio/metabolismo , Activación Enzimática , Transferencia Resonante de Energía de Fluorescencia , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Unión Proteica
8.
J Biol Chem ; 291(43): 22414-22426, 2016 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-27587390

RESUMEN

The mechanistic target of rapamycin complex 1 (mTORC1) coordinates cell growth with its nutritional, hormonal, energy, and stress status. Amino acids are critical regulators of mTORC1 that permit other inputs to mTORC1 activity. However, the roles of individual amino acids and their interactions in mTORC1 activation are not well understood. Here we demonstrate that activation of mTORC1 by amino acids includes two discrete and separable steps: priming and activation. Sensitizing mTORC1 activation by priming amino acids is a prerequisite for subsequent stimulation of mTORC1 by activating amino acids. Priming is achieved by a group of amino acids that includes l-asparagine, l-glutamine, l-threonine, l-arginine, l-glycine, l-proline, l-serine, l-alanine, and l-glutamic acid. The group of activating amino acids is dominated by l-leucine but also includes l-methionine, l-isoleucine, and l-valine. l-Cysteine predominantly inhibits priming but not the activating step. Priming and activating steps differ in their requirements for amino acid concentration and duration of treatment. Priming and activating amino acids use mechanisms that are distinct both from each other and from growth factor signaling. Neither step requires intact tuberous sclerosis complex of proteins to activate mTORC1. Concerted action of priming and activating amino acids is required to localize mTORC1 to lysosomes and achieve its activation.


Asunto(s)
Aminoácidos/metabolismo , Lisosomas/metabolismo , Complejos Multiproteicos/metabolismo , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Aminoácidos/genética , Animales , Células HeLa , Humanos , Lisosomas/genética , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Noqueados , Complejos Multiproteicos/genética , Serina-Treonina Quinasas TOR/genética
9.
J Biol Chem ; 291(21): 11394-406, 2016 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-27002154

RESUMEN

Mammalian phospholipase C-ß (PLC-ß) isoforms are stimulated by heterotrimeric G protein subunits and members of the Rho GTPase family of small G proteins. Although recent structural studies showed how Gαq and Rac1 bind PLC-ß, there is a lack of consensus regarding the Gßγ binding site in PLC-ß. Using FRET between cerulean fluorescent protein-labeled Gßγ and the Alexa Fluor 594-labeled PLC-ß pleckstrin homology (PH) domain, we demonstrate that the PH domain is the minimal Gßγ binding region in PLC-ß3. We show that the isolated PH domain can compete with full-length PLC-ß3 for binding Gßγ but not Gαq, Using sequence conservation, structural analyses, and mutagenesis, we identify a hydrophobic face of the PLC-ß PH domain as the Gßγ binding interface. This PH domain surface is not solvent-exposed in crystal structures of PLC-ß, necessitating conformational rearrangement to allow Gßγ binding. Blocking PH domain motion in PLC-ß by cross-linking it to the EF hand domain inhibits stimulation by Gßγ without altering basal activity or Gαq response. The fraction of PLC-ß cross-linked is proportional to the fractional loss of Gßγ response. Cross-linked PLC-ß does not bind Gßγ in a FRET-based Gßγ-PLC-ß binding assay. We propose that unliganded PLC-ß exists in equilibrium between a closed conformation observed in crystal structures and an open conformation where the PH domain moves away from the EF hands. Therefore, intrinsic movement of the PH domain in PLC-ß modulates Gßγ access to its binding site.


Asunto(s)
Subunidades beta de la Proteína de Unión al GTP/química , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/química , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Fosfolipasa C beta/química , Fosfolipasa C beta/metabolismo , Sitios de Unión , Proteínas Sanguíneas/química , Transferencia Resonante de Energía de Fluorescencia , Subunidades beta de la Proteína de Unión al GTP/genética , Subunidades gamma de la Proteína de Unión al GTP/genética , Humanos , Modelos Moleculares , Fosfolipasa C beta/genética , Fosfoproteínas/química , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología Estructural de Proteína , Proteína de Unión al GTP rac1/química , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
10.
Mol Endocrinol ; 29(8): 1114-22, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26168033

RESUMEN

The MAPKs ERK1/2 respond to nutrients and other insulin secretagogues in pancreatic ß-cells and mediate nutrient-dependent insulin gene transcription. Nutrients also stimulate the mechanistic target of rapamycin complex 1 (mTORC1) to regulate protein synthesis. We showed previously that activation of both ERK1/2 and mTORC1 in the MIN6 pancreatic ß-cell-derived line by extracellular amino acids (AAs) is at least in part mediated by the heterodimeric T1R1/T1R3, a G protein-coupled receptor. We show here that AAs differentially activate these two signaling pathways in MIN6 cells. Pretreatment with pertussis toxin did not prevent the activation of either ERK1/2 or mTORC1 by AAs, indicating that G(I) is not central to either pathway. Although glucagon-like peptide 1, an agonist for a G(s-)coupled receptor, activated ERK1/2 well and mTORC1 to a small extent, AAs had no effect on cytosolic cAMP accumulation. Ca(2+) entry is required for ERK1/2 activation by AAs but is dispensable for AA activation of mTORC1. Pretreatment with UBO-QIC, a selective G(q) inhibitor, reduced the activation of ERK1/2 but had little effect on the activation of mTORC1 by AAs, suggesting a differential requirement for G(q). Inhibition of G(12/13) by the overexpression of the regulator of G protein signaling domain of p115 ρ-guanine nucleotide exchange factor had no effect on mTORC1 activation by AAs, suggesting that these G proteins are also not involved. We conclude that AAs regulate ERK1/2 and mTORC1 through distinct signaling pathways.


Asunto(s)
Aminoácidos/química , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Complejos Multiproteicos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Animales , Calcio/metabolismo , AMP Cíclico/metabolismo , Endosomas/metabolismo , Regulación de la Expresión Génica , Células HeLa , Humanos , Células Secretoras de Insulina/citología , Lisosomas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Neuronas/metabolismo , Multimerización de Proteína
11.
Cell Logist ; 4: e29391, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25279250

RESUMEN

G protein-coupled receptors and heterotrimeric G proteins can diffuse laterally in the plasma membrane such that one receptor can catalyze the activation (GDP/GTP exchange) of multiple G proteins. In some cases, these processes are fast enough to support molecular signal amplification, where a single receptor maintains the activation of multiple G proteins at steady-state. Amplification in cells is probably highly regulated. It depends upon the identities of the G receptor and G protein - some do and some don't - and upon the activities of GTPase-activating proteins, membrane scaffolds, and other regulatory partners.

12.
Annu Rev Physiol ; 75: 127-54, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23140367

RESUMEN

Phospholipase C (PLC) converts phosphatidylinositol 4,5-bisphosphate (PIP(2)) to inositol 1,4,5-trisphosphate (IP(3)) and diacylglycerol (DAG). DAG and IP(3) each control diverse cellular processes and are also substrates for synthesis of other important signaling molecules. PLC is thus central to many important interlocking regulatory networks. Mammals express six families of PLCs, each with both unique and overlapping controls over expression and subcellular distribution. Each PLC also responds acutely to its own spectrum of activators that includes heterotrimeric G protein subunits, protein tyrosine kinases, small G proteins, Ca(2+), and phospholipids. Mammalian PLCs are autoinhibited by a region in the catalytic TIM barrel domain that is the target of much of their acute regulation. In combination, the PLCs act as a signaling nexus that integrates numerous signaling inputs, critically governs PIP(2) levels, and regulates production of important second messengers to determine cell behavior over the millisecond to hour timescale.


Asunto(s)
Sistemas de Mensajero Secundario/fisiología , Transducción de Señal/fisiología , Fosfolipasas de Tipo C/fisiología , Animales , Diglicéridos/fisiología , Humanos , Inositol 1,4,5-Trifosfato/fisiología , Fosfatidilinositol 4,5-Difosfato/fisiología , Fosfolipasas de Tipo C/química
13.
PLoS One ; 7(9): e45651, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23029161

RESUMEN

We describe the design, construction and validation of a fluorescence sensor to measure activation by agonist of the m1 muscarinic cholinergic receptor, a prototypical class I G(q)-coupled receptor. The sensor uses an established general design in which Förster resonance energy transfer (FRET) from a circularly permuted CFP mutant to FlAsH, a selectively reactive fluorescein, is decreased 15-20% upon binding of a full agonist. Notably, the sensor displays essentially wild-type capacity to catalyze activation of Gα(q), and the purified and reconstituted sensor displays appropriate regulation of affinity for agonists by G(q). We describe the strategies used to increase the agonist-driven change in FRET while simultaneously maintaining regulatory interactions with Gα(q), in the context of the known structures of Class I G protein-coupled receptors. The approach should be generally applicable to other Class I receptors which include numerous important drug targets.


Asunto(s)
Técnicas Biosensibles , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Receptor Muscarínico M1/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Biocatálisis , Calcio/metabolismo , Transferencia Resonante de Energía de Fluorescencia
14.
Curr Biol ; 21(23): 1979-87, 2011 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-22119528

RESUMEN

BACKGROUND: PLC-ß signaling is generally thought to be mediated by allosteric activation by G proteins and Ca(2+). Although availability of the phosphatidylinositol-4,5-biphosphate (PIP(2)) substrate is limiting in some cases, its production has not been shown to be independently regulated as a signaling mechanism. WNK1 protein kinase is known to regulate ion homeostasis and cause hypertension when expression is increased by gene mutations. However, its signaling functions remain largely elusive. RESULTS: Using diacylglycerol-stimulated TRPC6 and inositol trisphosphate-mediated Ca(2+) transients as cellular biosensors, we show that WNK1 stimulates PLC-ß signaling in cells by promoting the synthesis of PIP(2) via stimulation of phosphatidylinositol 4-kinase IIIα. WNK1 kinase activity is not required. Stimulation of PLC-ß by WNK1 and by Gα(q) are synergistic; WNK1 activity is essential for regulation of PLC-ß signaling by G(q)-coupled receptors, and basal input from G(q) is necessary for WNK1 signaling via PLC-ß. WNK1 further amplifies PLC-ß signaling when it is phosphorylated by Akt kinase in response to insulin-like growth factor. CONCLUSIONS: WNK1 is a novel regulator of PLC-ß that acts by controlling substrate availability. WNK1 thereby coordinates signaling between G protein and Akt kinase pathways. Because PIP(2) is itself a signaling molecule, regulation of PIP(2) synthesis by WNK1 also allows the cell to initiate PLC signaling while independently controlling the effects of PIP(2) on other targets. These findings describe a new signaling pathway for Akt-activating growth factors, a mechanism for G protein-growth factor crosstalk, and a means to independently control PLC signaling and PIP(2) availability.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Modelos Biológicos , Fosfatidilinositol 4,5-Difosfato/biosíntesis , Fosfolipasa C beta/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/fisiología , Animales , Calcio/metabolismo , ADN Complementario/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Antígenos de Histocompatibilidad Menor , Oligonucleótidos Antisentido/genética , Técnicas de Placa-Clamp , Proteínas Serina-Treonina Quinasas/genética , ARN Interferente Pequeño/genética , Ratas , Transducción de Señal/genética , Canales Catiónicos TRPC/metabolismo , Canal Catiónico TRPC6 , Proteína Quinasa Deficiente en Lisina WNK 1
15.
Sci Signal ; 4(159): pe5, 2011 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-21304157

RESUMEN

Heterotrimeric G proteins and G protein-coupled receptors represent conserved protein families with origins in the prokaryotes, but the various G protein-regulated effectors are heterogeneous in structure and function. The effectors apparently evolved ways to listen to G proteins late in their evolutionary histories. The structure of a complex between the effector protein phospholipase C-ß3 (PLC-ß3) and its activator, Gα(q), suggests that several effectors independently evolved a structurally similar helix-turn-helix segment for G protein recognition. PLC-ßs are also guanosine triphosphatase (GTPase)-activating proteins (GAPs) for the G(q) that activates them. In a second example of convergent evolution, the GAP activity of these proteins depends on a flexible asparagine-containing loop that resembles the GAP site on RGS proteins, another family of G protein GAPs. Together, these two sites are proposed to cooperate to enable fast binding to activated Gα(q), followed by fast deactivation. This cycle allows rapid sampling of the activation state of G(q)-coupled receptors while providing efficient signal transduction.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/química , Fosfolipasa C beta/química , Estructura Terciaria de Proteína , Transducción de Señal , Animales , Sitios de Unión , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Guanosina Difosfato/química , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/química , Guanosina Trifosfato/metabolismo , Humanos , Hidrólisis , Modelos Biológicos , Modelos Moleculares , Fosfolipasa C beta/metabolismo , Unión Proteica
16.
J Biol Chem ; 286(2): 942-51, 2011 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-21036901

RESUMEN

Cross-talk between Gα(i)- and Gα(q)-linked G-protein-coupled receptors yields synergistic Ca(2+) responses in a variety of cell types. Prior studies have shown that synergistic Ca(2+) responses from macrophage G-protein-coupled receptors are primarily dependent on phospholipase Cß3 (PLCß3), with a possible contribution of PLCß2, whereas signaling through PLCß4 interferes with synergy. We here show that synergy can be induced by the combination of Gßγ and Gα(q) activation of a single PLCß isoform. Synergy was absent in macrophages lacking both PLCß2 and PLCß3, but it was fully reconstituted following transduction with PLCß3 alone. Mechanisms of PLCß-mediated synergy were further explored in NIH-3T3 cells, which express little if any PLCß2. RNAi-mediated knockdown of endogenous PLCßs demonstrated that synergy in these cells was dependent on PLCß3, but PLCß1 and PLCß4 did not contribute, and overexpression of either isoform inhibited Ca(2+) synergy. When synergy was blocked by RNAi of endogenous PLCß3, it could be reconstituted by expression of either human PLCß3 or mouse PLCß2. In contrast, it could not be reconstituted by human PLCß3 with a mutation of the Y box, which disrupted activation by Gßγ, and it was only partially restored by human PLCß3 with a mutation of the C terminus, which partly disrupted activation by Gα(q). Thus, both Gßγ and Gα(q) contribute to activation of PLCß3 in cells for Ca(2+) synergy. We conclude that Ca(2+) synergy between Gα(i)-coupled and Gα(q)-coupled receptors requires the direct action of both Gßγ and Gα(q) on PLCß and is mediated primarily by PLCß3, although PLCß2 is also competent.


Asunto(s)
Señalización del Calcio/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Fosfolipasa C beta/metabolismo , Animales , Complemento C5a/metabolismo , Humanos , Macrófagos/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Mutagénesis , Células 3T3 NIH , Fosfolipasa C beta/genética , ARN Interferente Pequeño , Receptores Purinérgicos P2/metabolismo , Uridina Difosfato/metabolismo
17.
Curr Biol ; 20(15): 1327-35, 2010 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-20579885

RESUMEN

BACKGROUND: Receptors that couple to G(i) and G(q) often interact synergistically in cells to elicit cytosolic Ca(2+) transients that are several-fold higher than the sum of those driven by each receptor alone. Such synergism is commonly assumed to be complex, requiring regulatory interaction between components, multiple pathways, or multiple states of the target protein. RESULTS: We show that cellular G(i)-G(q) synergism derives from direct supra-additive stimulation of phospholipase C-beta3 (PLC-beta3) by G protein subunits Gbetagamma and Galpha(q), the relevant components of the G(i) and G(q) signaling pathways. No additional pathway or proteins are required. Synergism is quantitatively explained by the classical and simple two-state (inactive<-->active) allosteric mechanism. We show generally that synergistic activation of a two-state enzyme reflects enhanced conversion to the active state when both ligands are bound, not merely the enhancement of ligand affinity predicted by positive cooperativity. The two-state mechanism also explains why synergism is unique to PLC-beta3 among the four PLC-beta isoforms and, in general, why one enzyme may respond synergistically to two activators while another does not. Expression of synergism demands that an enzyme display low basal activity in the absence of ligand and becomes significant only when basal activity is

Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Modelos Químicos , Fosfolipasa C beta/metabolismo , Regulación Alostérica , Animales , Activación Enzimática , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Humanos , Isoenzimas/metabolismo , Ligandos , Ratones , Ratas
18.
Curr Biol ; 18(17): R777-R783, 2008 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-18786383

RESUMEN

G-protein-mediated signaling is intrinsically kinetic. Signal output at steady state is a balance of the rates of GTP binding, which causes activation, and of GTP hydrolysis, which terminates activation. This GTPase catalytic cycle is regulated by receptors, which accelerate GTP binding, and GTPase-activating proteins (GAPs), which accelerate hydrolysis. Receptors and GAPs similarly control the rates of signal initiation and termination. To allow independent control of signal amplitude and of the rates of turning the signal on and off, the activities of receptors and GAPs must be coordinated. Here, the principles of such coordination and the mechanisms by which it is achieved are discussed.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Transducción de Señal , Activación Enzimática , Proteínas de Unión al GTP/química , Proteínas Activadoras de GTPasa/fisiología , Cinética , Modelos Biológicos
19.
PLoS Comput Biol ; 4(8): e1000148, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18716678

RESUMEN

Signal output from receptor-G-protein-effector modules is a dynamic function of the nucleotide exchange activity of the receptor, the GTPase-accelerating activity of GTPase-activating proteins (GAPs), and their interactions. GAPs may inhibit steady-state signaling but may also accelerate deactivation upon removal of stimulus without significantly inhibiting output when the receptor is active. Further, some effectors (e.g., phospholipase C-beta) are themselves GAPs, and it is unclear how such effectors can be stimulated by G proteins at the same time as they accelerate G protein deactivation. The multiple combinations of protein-protein associations and interacting regulatory effects that allow such complex behaviors in this system do not permit the usual simplifying assumptions of traditional enzyme kinetics and are uniquely subject to systems-level analysis. We developed a kinetic model for G protein signaling that permits analysis of both interactive and independent G protein binding and regulation by receptor and GAP. We evaluated parameters of the model (all forward and reverse rate constants) by global least-squares fitting to a diverse set of steady-state GTPase measurements in an m1 muscarinic receptor-G(q)-phospholipase C-beta1 module in which GTPase activities were varied by approximately 10(4)-fold. We provide multiple tests to validate the fitted parameter set, which is consistent with results from the few previous pre-steady-state kinetic measurements. Results indicate that (1) GAP potentiates the GDP/GTP exchange activity of the receptor, an activity never before reported; (2) exchange activity of the receptor is biased toward replacement of GDP by GTP; (3) receptor and GAP bind G protein with negative cooperativity when G protein is bound to either GTP or GDP, promoting rapid GAP binding and dissociation; (4) GAP indirectly stabilizes the continuous binding of receptor to G protein during steady-state GTPase hydrolysis, thus further enhancing receptor activity; and (5) receptor accelerates GDP/GTP exchange primarily by opening an otherwise closed nucleotide binding site on the G protein but has minimal effect on affinity (K(assoc) = k(assoc)/k(dissoc)) of G protein for nucleotide. Model-based simulation explains how GAP activity can accelerate deactivation >10-fold upon removal of agonist but still allow high signal output while the receptor is active. Analysis of GTPase flux through distinct reaction pathways and consequent accumulation of specific GTPase cycle intermediates indicate that, in the presence of a GAP, the receptor remains bound to G protein throughout the GTPase cycle and that GAP binds primarily during the GTP-bound phase. The analysis explains these behaviors and relates them to the specific regulatory phenomena described above. The work also demonstrates the applicability of appropriately data-constrained system-level analysis to signaling networks of this scale.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Unión Competitiva , Activación Enzimática/fisiología , Inhibidores Enzimáticos/metabolismo , Proteínas de Unión al GTP/química , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Hidrólisis , Cinética , Análisis de los Mínimos Cuadrados , Modelos Biológicos , Fosfolipasa C beta/metabolismo , Unión Proteica/fisiología , Biología de Sistemas/métodos , Liposomas Unilamelares , Regulación hacia Arriba
20.
J Biol Chem ; 282(14): 10576-84, 2007 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-17283075

RESUMEN

Regulation of intracellular cyclic adenosine 3 ',5 '-monophosphate (cAMP) is integral in mediating cell growth, cell differentiation, and immune responses in hematopoietic cells. To facilitate studies of cAMP regulation we developed a BRET (bioluminescence resonance energy transfer) sensor for cAMP, CAMYEL (cAMP sensor using YFP-Epac-RLuc), which can quantitatively and rapidly monitor intracellular concentrations of cAMP in vivo. This sensor was used to characterize three distinct pathways for modulation of cAMP synthesis stimulated by presumed G(s)-dependent receptors for isoproterenol and prostaglandin E(2). Whereas two ligands, uridine 5 '-diphosphate and complement C5a, appear to use known mechanisms for augmentation of cAMP via G(q)/calcium and G(i), the action of sphingosine 1-phosphate (S1P) is novel. In these cells, S1P, a biologically active lysophospholipid, greatly enhances increases in intracellular cAMP triggered by the ligands for G(s)-coupled receptors while having only a minimal effect by itself. The enhancement of cAMP by S1P is resistant to pertussis toxin and independent of intracellular calcium. Studies with RNAi and chemical perturbations demonstrate that the effect of S1P is mediated by the S1P(2) receptor and the heterotrimeric G(13) protein. Thus in these macrophage cells, all four major classes of G proteins can regulate intracellular cAMP.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Técnicas Biosensibles , Señalización del Calcio/fisiología , AMP Cíclico/metabolismo , Lisofosfolípidos/metabolismo , Macrófagos/metabolismo , Esfingosina/análogos & derivados , Factor de Transcripción Activador 6 , Agonistas Adrenérgicos beta/farmacología , Animales , Señalización del Calcio/efectos de los fármacos , Línea Celular , Complemento C5a/farmacología , Humanos , Factores Inmunológicos/farmacología , Isoproterenol/farmacología , Ratones , Toxina del Pertussis/farmacología , Prostaglandinas E/farmacología , ARN Interferente Pequeño/farmacología , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/metabolismo , Uridina Difosfato/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...