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.
Artigo em Inglês | MEDLINE | ID: mdl-35385687

RESUMO

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.


Assuntos
Peptídeos , Feromônios , Schistosoma/crescimento & desenvolvimento , Animais , Feminino , Masculino , Biossíntese de Peptídeos Independentes de Ácido Nucleico , Triptaminas
2.
J Biol Chem ; 295(8): 2270-2284, 2020 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-31949046

RESUMO

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.


Assuntos
Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/metabolismo , Sequência de Aminoácidos , Animais , Bovinos , Células HEK293 , Humanos , Modelos Biológicos , Proteínas Mutantes/metabolismo , Peptídeos/metabolismo , Ligação Proteica
3.
Proc Natl Acad Sci U S A ; 115(9): E2085-E2094, 2018 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-29440403

RESUMO

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.


Assuntos
Analgésicos Opioides/farmacologia , Proteínas RGS/antagonistas & inibidores , Via de Sinalização Wnt , Analgesia , Animais , Condicionamento Psicológico , Feminino , Proteínas de Ligação ao GTP/metabolismo , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfina/farmacologia , Neurônios/metabolismo , Substância Cinzenta Periaquedutal/metabolismo , Proteínas RGS/metabolismo , Análise de Sequência de RNA , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
4.
J Biol Chem ; 292(40): 16787-16801, 2017 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-28842497

RESUMO

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.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/química , Modelos Químicos , Fosfolipase C beta/química , Regulação Alostérica/fisiologia , Cálcio/química , Cálcio/metabolismo , Ativação Enzimática , Transferência Ressonante de Energia de Fluorescência , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Fosfolipase C beta/genética , Fosfolipase C beta/metabolismo , Ligação Proteica
5.
J Biol Chem ; 291(21): 11394-406, 2016 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-27002154

RESUMO

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.


Assuntos
Subunidades beta da Proteína de Ligação ao GTP/química , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/química , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Fosfolipase C beta/química , Fosfolipase C beta/metabolismo , Sítios de Ligação , Proteínas Sanguíneas/química , Transferência Ressonante de Energia de Fluorescência , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades gama da Proteína de Ligação ao GTP/genética , Humanos , Modelos Moleculares , Fosfolipase C beta/genética , Fosfoproteínas/química , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia Estrutural de Proteína , Proteínas rac1 de Ligação ao GTP/química , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
6.
J Biol Chem ; 291(43): 22414-22426, 2016 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-27587390

RESUMO

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.


Assuntos
Aminoácidos/metabolismo , Lisossomos/metabolismo , Complexos Multiproteicos/metabolismo , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Aminoácidos/genética , Animais , Células HeLa , Humanos , Lisossomos/genética , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Complexos Multiproteicos/genética , Serina-Treonina Quinases TOR/genética
7.
Annu Rev Physiol ; 75: 127-54, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23140367

RESUMO

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.


Assuntos
Sistemas do Segundo Mensageiro/fisiologia , Transdução de Sinais/fisiologia , Fosfolipases Tipo C/fisiologia , Animais , Diglicerídeos/fisiologia , Humanos , Inositol 1,4,5-Trifosfato/fisiologia , Fosfatidilinositol 4,5-Difosfato/fisiologia , Fosfolipases Tipo C/química
10.
J Biol Chem ; 286(2): 942-51, 2011 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-21036901

RESUMO

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.


Assuntos
Sinalização do Cálcio/fisiologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Fosfolipase C beta/metabolismo , Animais , Complemento C5a/metabolismo , Humanos , Macrófagos/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Mutagênese , Células NIH 3T3 , Fosfolipase C beta/genética , RNA Interferente Pequeno , Receptores Purinérgicos P2/metabolismo , Difosfato de Uridina/metabolismo
11.
Nat Cell Biol ; 7(4): 405-11, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15793568

RESUMO

Signalling by G proteins is controlled by the regulator of G-protein signalling (RGS) proteins that accelerate the GTPase activity of Galpha subunits and act in a G-protein-coupled receptor (GPCR)-specific manner. The conserved RGS domain accelerates the G subunit GTPase activity, whereas the variable amino-terminal domain participates in GPCR recognition. How receptor recognition is achieved is not known. Here, we show that the scaffold protein spinophilin (SPL), which binds the third intracellular loop (3iL) of several GPCRs, binds the N-terminal domain of RGS2. SPL also binds RGS1, RGS4, RGS16 and GAIP. When expressed in Xenopus laevis oocytes, SPL markedly increased inhibition of alpha-adrenergic receptor (alphaAR) Ca2+ signalling by RGS2. Notably, the constitutively active mutant alphaAR(A293E) (the mutation being in the 3iL) did not bind SPL and was relatively resistant to inhibition by RGS2. Use of betaAR-alphaAR chimaeras identified the 288REKKAA293 sequence as essential for the binding of SPL and inhibition of Ca2+ signalling by RGS2. Furthermore, alphaAR-evoked Ca2+ signalling is less sensitive to inhibition by SPL in rgs2-/- cells and less sensitive to inhibition by RGS2 in spl-/- cells. These findings provide a general mechanism by which RGS proteins recognize GPCRs to confer signalling specificity.


Assuntos
Cálcio/metabolismo , Proteínas dos Microfilamentos/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Proteínas RGS/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Animais , Linhagem Celular , Humanos , Camundongos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Oócitos/química , Ligação Proteica/fisiologia , Proteínas RGS/metabolismo , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Xenopus laevis
12.
Curr Biol ; 18(17): R777-R783, 2008 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-18786383

RESUMO

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.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Transdução de Sinais , Ativação Enzimática , Proteínas de Ligação ao GTP/química , Proteínas Ativadoras de GTPase/fisiologia , Cinética , Modelos Biológicos
13.
J Cell Biol ; 162(2): 293-303, 2003 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-12860966

RESUMO

Homers are scaffolding proteins that bind G protein-coupled receptors (GPCRs), inositol 1,4,5-triphosphate (IP3) receptors (IP3Rs), ryanodine receptors, and TRP channels. However, their role in Ca2+ signaling in vivo is not known. Characterization of Ca2+ signaling in pancreatic acinar cells from Homer2-/- and Homer3-/- mice showed that Homer 3 has no discernible role in Ca2+ signaling in these cells. In contrast, we found that Homer 2 tunes intensity of Ca2+ signaling by GPCRs to regulate the frequency of [Ca2+]i oscillations. Thus, deletion of Homer 2 increased stimulus intensity by increasing the potency for agonists acting on various GPCRs to activate PLCbeta and evoke Ca2+ release and oscillations. This was not due to aberrant localization of IP3Rs in cellular microdomains or IP3R channel activity. Rather, deletion of Homer 2 reduced the effectiveness of exogenous regulators of G proteins signaling proteins (RGS) to inhibit Ca2+ signaling in vivo. Moreover, Homer 2 preferentially bound to PLCbeta in pancreatic acini and brain extracts and stimulated GAP activity of RGS4 and of PLCbeta in an in vitro reconstitution system, with minimal effect on PLCbeta-mediated PIP2 hydrolysis. These findings describe a novel, unexpected function of Homer proteins, demonstrate that RGS proteins and PLCbeta GAP activities are regulated functions, and provide a molecular mechanism for tuning signal intensity generated by GPCRs and, thus, the characteristics of [Ca2+]i oscillations.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Isoenzimas/metabolismo , Neuropeptídeos/metabolismo , Proteínas RGS/metabolismo , Fosfolipases Tipo C/metabolismo , Animais , Bombesina/farmacologia , Cálcio/metabolismo , Cálcio/farmacocinética , Sinalização do Cálcio , ATPases Transportadoras de Cálcio/antagonistas & inibidores , ATPases Transportadoras de Cálcio/metabolismo , Carbacol/farmacologia , Proteínas de Transporte/química , Proteínas de Transporte/genética , Colecistocinina/farmacologia , Agonistas Colinérgicos/farmacologia , Inibidores Enzimáticos/farmacologia , Proteínas de Ligação ao GTP/agonistas , Deleção de Genes , Proteínas de Arcabouço Homer , Indóis/farmacologia , Camundongos , Camundongos Knockout , Neuropeptídeos/química , Neuropeptídeos/genética , Pâncreas/citologia , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfolipase C beta , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático
14.
PLoS Comput Biol ; 4(8): e1000148, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18716678

RESUMO

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.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Ligação Competitiva , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/metabolismo , Proteínas de Ligação ao GTP/química , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Hidrólise , Cinética , Análise dos Mínimos Quadrados , Modelos Biológicos , Fosfolipase C beta/metabolismo , Ligação Proteica/fisiologia , Biologia de Sistemas/métodos , Lipossomas Unilamelares , Regulação para Cima
15.
Elife ; 82019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30835201

RESUMO

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).


Assuntos
Ceramidas/metabolismo , Receptores CCR5/química , Receptores CCR5/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Regulação Alostérica , Animais , Movimento Celular , Células Cultivadas , Lipopolissacarídeos/metabolismo , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/fisiologia , Camundongos Endogâmicos C57BL , Conformação Proteica
16.
Mol Endocrinol ; 29(8): 1114-22, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26168033

RESUMO

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.


Assuntos
Aminoácidos/química , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Complexos Multiproteicos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Animais , Cálcio/metabolismo , AMP Cíclico/metabolismo , Endossomos/metabolismo , Regulação da Expressão Gênica , Células HeLa , Humanos , Células Secretoras de Insulina/citologia , Lisossomos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Neurônios/metabolismo , Multimerização Proteica
17.
Cell Logist ; 4: e29391, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25279250

RESUMO

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.

18.
PLoS One ; 7(9): e45651, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23029161

RESUMO

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.


Assuntos
Técnicas Biossensoriais , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Receptor Muscarínico M1/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Biocatálise , Cálcio/metabolismo , Transferência Ressonante de Energia de Fluorescência
19.
Sci Signal ; 4(159): pe5, 2011 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-21304157

RESUMO

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.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/química , Fosfolipase C beta/química , Estrutura Terciária de Proteína , Transdução de Sinais , Animais , Sítios de Ligação , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Guanosina Difosfato/química , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/química , Guanosina Trifosfato/metabolismo , Humanos , Hidrólise , Modelos Biológicos , Modelos Moleculares , Fosfolipase C beta/metabolismo , Ligação Proteica
20.
Curr Biol ; 21(23): 1979-87, 2011 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-22119528

RESUMO

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.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Modelos Biológicos , Fosfatidilinositol 4,5-Difosfato/biossíntese , Fosfolipase C beta/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/fisiologia , Animais , Cálcio/metabolismo , DNA Complementar/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Técnicas de Silenciamento de Genes , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Antígenos de Histocompatibilidade Menor , Oligonucleotídeos Antissenso/genética , Técnicas de Patch-Clamp , Proteínas Serina-Treonina Quinases/genética , RNA Interferente Pequeno/genética , Ratos , Transdução de Sinais/genética , Canais de Cátion TRPC/metabolismo , Canal de Cátion TRPC6 , Proteína Quinase 1 Deficiente de Lisina WNK
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA