Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters











Publication year range
1.
Oncogene ; 39(7): 1466-1483, 2020 02.
Article in English | MEDLINE | ID: mdl-31659259

ABSTRACT

Over half of cutaneous melanoma tumors have BRAFV600E/K mutations. Acquired resistance to BRAF inhibitors (BRAFi) remains a major hurdle in attaining durable therapeutic responses. In this study we demonstrate that ~50-60% of melanoma cell lines with vemurafenib resistance acquired in vitro show activation of RhoA family GTPases. In BRAFi-resistant melanoma cell lines and tumors, activation of RhoA is correlated with decreased expression of melanocyte lineage genes. Using a machine learning approach, we built gene expression-based models to predict drug sensitivity for 265 common anticancer compounds. We then projected these signatures onto the collection of TCGA cutaneous melanoma and found that poorly differentiated tumors were predicted to have increased sensitivity to multiple Rho kinase (ROCK) inhibitors. Two transcriptional effectors downstream of Rho, MRTF and YAP1, are activated in the RhoHigh BRAFi-resistant cell lines, and resistant cells are more sensitive to inhibition of these transcriptional mechanisms. Taken together, these results support the concept of targeting Rho-regulated gene transcription pathways as a promising therapeutic approach to restore sensitivity to BRAFi-resistant tumors or as a combination therapy to prevent the onset of drug resistance.


Subject(s)
Cell Dedifferentiation/drug effects , Melanoma/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Signal Transduction/drug effects , rho-Associated Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Line, Tumor , Enzyme Activation/drug effects , Humans , Melanocytes/drug effects , Melanocytes/pathology , Transcription Factors/metabolism , Transcription, Genetic/drug effects , YAP-Signaling Proteins
2.
Integr Biol (Camb) ; 7(11): 1378-86, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26374065

ABSTRACT

Many biological processes are rhythmic and proper timing is increasingly appreciated as being critical for development and maintenance of physiological functions. To understand how temporal modulation of an input signal influences downstream responses, we employ microfluidic pulsatile stimulation of a G-protein coupled receptor, the muscarinic M3 receptor, in single cells with simultaneous real-time imaging of both intracellular calcium and NFAT nuclear localization. Interestingly, we find that reduced stimulation with pulses of ligand can give more efficient transcription factor activation, if stimuli are timed appropriately. Our experiments and computational analyses show that M3 receptor-induced calcium oscillations form a low pass filter while calcium-induced NFAT translocation forms a high pass filter. The combination acts as a band-pass filter optimized for intermediate frequencies of stimulation. We demonstrate that receptor desensitization and NFAT translocation rates determine critical features of the band-pass filter and that the band-pass may be shifted for different receptors or NFAT dynamics. As an example, we show that the two NFAT isoforms (NFAT4 and NFAT1) have shifted band-pass windows for the same receptor. While we focus specifically on the M3 muscarinic receptor and NFAT translocation, band-pass processing is expected to be a general theme that applies to multiple signaling pathways.


Subject(s)
NFATC Transcription Factors/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Active Transport, Cell Nucleus , Area Under Curve , Calcium/metabolism , Computer Simulation , HEK293 Cells , Humans , Ligands , Microfluidics , Models, Theoretical
3.
J Pept Res ; 63(2): 141-6, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15009535

ABSTRACT

Regulators of G-protein signaling (RGS) proteins form a multifunctional signaling family. A key role of RGS proteins is binding to the G-protein Galpha-subunit and acting as GTPase-activating proteins (GAPs), thereby rapidly terminating G protein-coupled receptor (GPCR) signaling. Using the published RGS4-Gialpha1 X-ray structure we have designed and synthesized a series of cyclic peptides, modeled on the Gialpha Switch I region, that inhibit RGS4 GAP activity. These compounds should prove useful for elucidating RGS-mediated activity and serve as a starting point for the development of a novel class of therapeutic agent.


Subject(s)
Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , RGS Proteins/antagonists & inhibitors , Animals , CHO Cells , Cricetinae , Cricetulus , GTPase-Activating Proteins/metabolism , Inhibitory Concentration 50 , Peptides, Cyclic/chemical synthesis , RGS Proteins/metabolism , Structure-Activity Relationship
4.
J Pept Res ; 60(6): 312-6, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12464108

ABSTRACT

Many drugs of abuse signal through receptors that couple to G proteins (GPCRs), so the factors that control GPCR signaling are likely to be important to the understanding of drug abuse. Contributions by the recently identified protein family, regulators of G protein signaling (RGS) to the control of GPCR function are just beginning to be understood. RGS proteins can accelerate the deactivation of G proteins by 1000-fold and in cell systems they profoundly inhibit signaling by many receptors, including mu-opioid receptors. Coupled with the known dynamic regulation of RGS protein expression and function, they are of obvious interest in understanding tolerance and dependence mechanisms. Furthermore, drugs that could inhibit their activity could be useful in preventing the development of or in treating drug dependence.


Subject(s)
Central Nervous System Agents/pharmacology , GTP-Binding Proteins/metabolism , RGS Proteins/physiology , Animals , GTPase-Activating Proteins/metabolism , Gene Expression/physiology , Humans , Protein Structure, Tertiary , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Substance-Related Disorders/metabolism
5.
Biochemistry ; 40(35): 10532-41, 2001 Sep 04.
Article in English | MEDLINE | ID: mdl-11523995

ABSTRACT

G protein coupled receptors activate signal transducing guanine nucleotide-binding proteins (G proteins), which consist of an alpha subunit and a betagamma dimer. Whole cell studies have reported that receptors signal through specific betagamma subtypes. Membrane reconstitution studies with the adenosine A(1) and alpha(2A) adrenergic receptors have reached a similar conclusion. We aimed to test the generality of this finding by comparing the gamma subtype specificity for four G(i)-coupled receptors: alpha(2A) adrenergic; A1 adenosine (A(1)-R); 5-hydroxytryptamine(1A) (5-HT(1A)-R); mu opioid. Membranes were reconstituted with Galpha(i)(1) and five gamma subtypes (dimerized to beta1). Using a sensitive alpha-betagamma binding assay, we show that all recombinant betagamma (except beta1gamma1) had comparable affinity for alpha(i)(1). Using high affinity agonist binding as a measure of receptor-G protein coupling, betagamma-containing gamma11 was the most potent for A(1)-R and 5-HT(1A)-R (p < 0.05, one way ANOVA) while gamma7 was most potent for the other two receptors. gamma11 was 3-8-fold more potent for the A(1)-R than were the other gamma subtypes. Also, gamma11 was 2-8-fold more potent for A(1)-R than at the other receptors, suggesting a unique coupling specificity of the A(1)-R for gamma11. In contrast, the discrimination by receptors for the other betagamma subtypes (beta1 and gamma1, gamma2, gamma7, and gamma10) was limited (2-3-fold). Thus the exquisite betagamma specificity of individual receptors reported in whole cell studies may depend on in vivo mechanisms beyond direct receptor recognition of betagamma subtypes.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Receptors, Purinergic P1/metabolism , Receptors, Serotonin/metabolism , Animals , CHO Cells , Cell Line , Cricetinae , Heterotrimeric GTP-Binding Proteins/agonists , Heterotrimeric GTP-Binding Proteins/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Opioid, mu/metabolism , Receptors, Serotonin, 5-HT1 , Swine , Tumor Cells, Cultured
6.
Am J Physiol Cell Physiol ; 281(3): C801-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11502557

ABSTRACT

ANG II type 1 (AT(1)) receptors respond to sustained exposure to ANG II by undergoing downregulation of absolute receptor numbers. It has been assumed previously that downregulation involves endocytosis. The present study hypothesized that AT(1) receptor downregulation occurs independently of receptor endocytosis or G protein coupling. Mutant AT(1) receptors with carboxy-terminal deletions internalized <5% of radioligand compared with 65% for wild-type AT(1) receptors. The truncated AT(1) receptors retained the ability to undergo downregulation. These data suggest the existence of an alternative pathway to AT(1) receptor degradation that does not require endocytosis, per se. Point mutations in either the second transmembrane region or second intracellular loop impaired G protein (G(q)) coupling. These receptors exhibited a biphasic pattern of downregulation. The earliest phase of downregulation (0-2 h) was independent of coupling to G(q), but no additional downregulation was observed after 2 h of ANG II exposure in the receptors with impaired coupling to G(q). These data suggest that coupling to G(q) is required for the later phase (2-24 h) of AT(1) receptor downregulation.


Subject(s)
Down-Regulation/physiology , GTP-Binding Proteins/physiology , Receptors, Angiotensin/physiology , Amino Acid Substitution , Angiotensin II/metabolism , Animals , Binding Sites , COS Cells , Calcium/metabolism , Chlorocebus aethiops , Cloning, Molecular , Endocytosis , Iodine Radioisotopes , Kinetics , Mutagenesis, Site-Directed , Phosphatidylinositols/metabolism , Radioligand Assay , Rats , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/chemistry , Receptors, Angiotensin/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Deletion , Transfection
7.
J Pharmacol Exp Ther ; 297(3): 837-45, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11356902

ABSTRACT

G protein-coupled receptors (GPCRs) play a major role in signal transduction and are targets of many therapeutic drugs. The regulator of G protein signaling (RGS) proteins form a recently identified protein family, and they strongly modulate the activity of G proteins. Their best known function is to inhibit G protein signaling by accelerating GTP hydrolysis [GTPase activating protein (GAP)] thus turning off G protein signals. RGS proteins also possess non-GAP functions, through both their RGS domains and various non-RGS domains and motifs (e.g., GGL, DEP, DH/PH, PDZ domains and a cysteine string motif). They are a highly diverse protein family, have unique tissue distributions, are strongly regulated by signal transduction events, and will likely play diverse functional roles in living cells. Thus they represent intriguing, novel pharmacological/therapeutic targets. Drugs targeting RGS proteins can be divided into five groups: 1) potentiators of endogenous agonist function, 2) potentiators/desensitization blockers of exogenous GPCR agonists, 3) specificity enhancers of exogenous agonists, 4) antagonists of effector signaling by an RGS protein, and 5) RGS agonists. In addition, a novel subsite distinction within the RGS domain has been proposed with significant functional implications and defined herein as "A-site" and "B-site". Therefore, RGS proteins should provide exciting new opportunities for drug development.


Subject(s)
Drug Design , RGS Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Amino Acid Motifs/physiology , Animals , Binding Sites/drug effects , GTP-Binding Proteins/metabolism , GTPase-Activating Proteins/metabolism , Guanosine Triphosphate/metabolism , Humans , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/physiology , RGS Proteins/agonists , RGS Proteins/antagonists & inhibitors , Substrate Specificity/drug effects
8.
Anal Biochem ; 291(1): 109-17, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11262163

ABSTRACT

Three BODIPY GTPgammaS analogs (FL, 515, and TR), BODIPY FL GppNHp and BODIPY FL GTP molecules were synthesized as possible fluorescent probes to study guanine nucleotide binding spectroscopically. Binding to G(alphao) increases baseline analog fluorescence by 6-, 8.5-, 2.8-, 3.5-, and 3.0-fold, respectively. Binding of GTPgammaS and GppNHp analogs to G(alphao) is of high affinity (K(D) 11, 17, 55, and 110 nM, respectively) and reaches a stable plateau while fluorescence of BODIPY FL GTP shows a transient increase which returns to baseline. Furthermore, BODIPY FL GTPgammaS shows varying affinities for alpha(o), alpha(s), alpha(i1), and alpha(i2) (6, 58, 150, and 300 nM). The affinities of BODIPY FL GppNHp for all four G(alpha) subunits are 10-fold lower than for BODIPY FL GTPgammaS. Half-times for the fluorescence increase are consistent with known GDP release rates for those proteins. Enhancement of fluorescence upon binding the G(alpha) subunit is most likely due to a rotation around the gamma-thiol (GTPgammaS) or the 3' ribose-hydroxyl (GppNHp) bond to relieve the quenching of BODIPY fluorescence by the guanine base. Binding to G(alpha) exposes the BODIPY moiety to the external environment, as seen by an increase in sodium iodide quenching. The visible excitation and emission spectra and high fluorescence levels of these probes permit robust real-time detection of nucleotide binding.


Subject(s)
Boron Compounds/chemical synthesis , Fluorescent Dyes/chemistry , GTP-Binding Proteins/chemistry , Guanosine Triphosphate/analogs & derivatives , Guanosine Triphosphate/chemistry , Escherichia coli , GTP-Binding Proteins/metabolism , Guanosine Diphosphate/chemistry , Protein Binding , RGS Proteins/metabolism
9.
Biochem J ; 354(Pt 2): 337-44, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11171112

ABSTRACT

G-protein-coupled receptors activate signal-transducing G-proteins, which consist of an alpha subunit and a betagamma dimer. Membrane extraction with 5-7 M urea has been used to uncouple receptors from endogenous G-proteins to permit reconstitution with purified G-proteins. We show that alpha(i) subunits are inactivated with 5 M urea whereas the betagamma dimer requires at least 7 M urea for its inactivation. There is no significant loss of receptors. Surprisingly, Western-blot analysis indicates that the urea-denatured alpha(i) subunit remains mostly membrane-bound and that beta is only partially removed. After 7 M urea treatment, both alpha(i1) and betagamma subunits are required to restore high-affinity agonist binding and receptor-catalysed guanosine 5'-[gamma-thio]triphosphate binding. We demonstrate the generality of this approach for four G(i)-coupled receptors (alpha(2A)-adrenergic, adenosine A1, 5-hydroxytryptamine(1A) and mu-opioid) expressed in insect cells and two mammalian cell lines. Thus a selectivity of urea for G-protein alpha versus betagamma subunits is established in both concentration and mechanism.


Subject(s)
Heterotrimeric GTP-Binding Proteins/antagonists & inhibitors , Urea/pharmacology , Animals , Blotting, Western , CHO Cells , Catalysis , Cricetinae , Dimerization , Dose-Response Relationship, Drug , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Protein Subunits , Structure-Activity Relationship , Tumor Cells, Cultured , Urea/administration & dosage
10.
Mol Pharmacol ; 59(3): 532-42, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11179449

ABSTRACT

Constitutive activation of G protein-coupled receptors (GPCRs) is now well recognized and many classical GPCR antagonists have been found to be inverse agonists. For the alpha(2A)-adrenergic receptor (AR) we determine the relative inverse efficacies of a series of antagonists and utilize the extended ternary complex model to estimate the fraction of constitutively active mutant (CAM) receptors in the active state. Stable Chinese hamster ovary cell lines expressing the porcine alpha(2A)-AR in its wild-type (WT) and constitutively activated (CAM-T373K) form were isolated. Activation of both G(i) and G(s) was enhanced for CAM receptors. cAMP production was suppressed in cells with the CAM alpha(2A)-AR and this suppression was reversed by alpha(2)-adrenergic antagonists with an order of inverse efficacy of rauwolscine > yohimbine > RX821002 > MK912, whereas phentolamine and idazoxan were essentially neutral antagonists. This striking difference in inverse efficacy between idazoxan and RX821002 may account for in vivo pharmacological differences between these two alpha(2)-adrenergic antagonists. Agonist binding affinity to the non-G protein-coupled CAM receptor was 3- to 9-fold higher than to WT, whereas binding of the most efficacious inverse agonists, yohimbine and rauwolscine, was 1.7- and 2.1-fold weaker. Analysis of this difference by the extended ternary complex model indicates that approximately 50% of the CAM alpha(2A)-AR is in the active (R*) state although there is no detectable constitutive activity of the WT receptor in the absence of agonist.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists , Adrenergic alpha-Antagonists/pharmacology , Idazoxan/analogs & derivatives , Idazoxan/pharmacology , Alkaloids/pharmacology , Animals , CHO Cells , Cricetinae , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Imidazoline Receptors , Models, Biological , Mutation , Protein Conformation , Radioligand Assay , Receptors, Adrenergic, alpha-2/chemistry , Receptors, Adrenergic, alpha-2/genetics , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Drug/antagonists & inhibitors
11.
J Biol Chem ; 276(10): 7526-33, 2001 Mar 09.
Article in English | MEDLINE | ID: mdl-11099504

ABSTRACT

We generated mutants of the transporter associated with antigen-processing subunits TAP1 and TAP2 that were altered at the conserved lysine residue in the Walker A motifs of the nucleotide binding domains (NBD). In other ATP binding cassette transporters, mutations of the lysine have been shown to reduce or abrogate the ATP hydrolysis activity and in some cases impair nucleotide binding. Mutants TAP1(K544M) and TAP2(K509M) were expressed in insect cells, and the effects of the mutations on nucleotide binding, peptide binding, and peptide translocation were assessed. The mutant TAP1 subunit is significantly impaired for nucleotide binding relative to wild type TAP1. The identical mutation in TAP2 does not significantly impair nucleotide binding relative to wild type TAP2. Using fluorescence quenching assays to measure the binding of fluorescent peptides, we show that both mutants, in combination with their wild type partners, can bind peptides. Since the mutant TAP1 is significantly impaired for nucleotide binding, these results indicate that nucleotide binding to TAP1 is not a requirement for peptide binding to TAP complexes. Peptide translocation is undetectable for TAP1.TAP2(K509M) complexes, but low levels of translocation are detectable with TAP1(K544M).TAP2 complexes. These results suggest an impairment in nucleotide hydrolysis by TAP complexes containing either mutant TAP subunit and indicate that the presence of one intact TAP NBD is insufficient for efficient catalysis of peptide translocation. Taken together, these results also suggest the possibility of distinct functions for TAP1 and TAP2 NBD during a single translocation cycle.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , Mutation , ATP Binding Cassette Transporter, Subfamily B, Member 3 , ATP-Binding Cassette Transporters/genetics , Adenosine Diphosphate/metabolism , Amino Acid Motifs , Animals , Apyrase/metabolism , Cell Line , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Hydrolysis , Insecta , Kinetics , Lysine/chemistry , Microsomes/metabolism , Mutagenesis, Site-Directed , Peptides/metabolism , Precipitin Tests , Protein Binding , Protein Transport , Spectrometry, Fluorescence , Temperature , Time Factors
12.
J Biol Chem ; 276(2): 915-23, 2001 Jan 12.
Article in English | MEDLINE | ID: mdl-11042171

ABSTRACT

A novel regulator of G-protein signaling (RGS) has been isolated from a highly purified population of mouse long-term hematopoietic stem cells, and designated RGS18. It has 234 amino acids consisting of a central RGS box and short divergent NH(2) and COOH termini. The calculated molecular weight of RGS18 is 27,610 and the isoelectric point is 8.63. Mouse RGS18 is expressed from a single gene and shows tissue specific distribution. It is most highly expressed in bone marrow followed by fetal liver, spleen, and then lung. In bone marrow, RGS18 level is highest in long-term and short-term hematopoietic stem cells, and is decreased as they differentiate into more committed multiple progenitors. The human RGS18 ortholog has a tissue-specific expression pattern similar to that of mouse RGS18. Purified RGS18 interacts with the alpha subunit of both G(i) and G(q) subfamilies. The results of in vitro GTPase single-turnover assays using Galpha(i) indicated that RGS18 accelerates the intrinsic GTPase activity of Galpha(i). Transient overexpression of RGS18 attenuated inositol phosphates production via angiotensin receptor and transcriptional activation through cAMP-responsive element via M1 muscarinic receptor. This suggests RGS18 can act on G(q)-mediated signaling pathways in vivo.


Subject(s)
Carrier Proteins/physiology , GTP-Binding Proteins/physiology , Hematopoietic Stem Cells/physiology , Intracellular Signaling Peptides and Proteins , Signal Transduction , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line , Cloning, Molecular , GTP Phosphohydrolases/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Genetic Variation , Humans , Mice , Molecular Sequence Data , Molecular Weight , Organ Specificity , RGS Proteins , Sequence Alignment , Sequence Homology, Amino Acid , Transfection
13.
J Biol Chem ; 275(43): 33497-503, 2000 Oct 27.
Article in English | MEDLINE | ID: mdl-10942773

ABSTRACT

Regulator of G-protein signaling (RGS) proteins accelerate GTP hydrolysis by Galpha subunits speeding deactivation. Galpha deactivation kinetics mediated by RGS are too fast to be directly studied using conventional radiochemical methods. We describe a stopped-flow spectroscopic approach to visualize these rapid kinetics by measuring the intrinsic tryptophan fluorescence decrease of Galpha accompanying GTP hydrolysis and Galpha deactivation on the millisecond time scale. Basal k(cat) values for Galpha(o), Galpha(i1), and Galpha(i2) at 20 degrees C were similar (0.025-0.033 s(-1)). Glutathione S-transferase fusion proteins containing RGS4 and an RGS7 box domain (amino acids 305-453) enhanced the rate of Galpha deactivation in a manner linear with RGS concentration. RGS4-stimulated rates could be measured up to 5 s(-1) at 3 microm, giving a catalytic efficiency of 1.7-2.8 x 10(6) m(-1) s(-1) for all three Galpha subunits. In contrast, RGS7 showed catalytic efficiencies of 0.44, 0.10, and 0.02 x 10(6) m(-1) s(-1) toward Galpha(o), Galpha(i2), and Galpha(i1), respectively. Thus RGS7 is a weaker GTPase activating protein than RGS4 toward all Galpha subunits tested, but it is specific for Galpha(o) over Galpha(i1) or Galpha(i2). Furthermore, the specificity of RGS7 for Galpha(o) does not depend on N- or C-terminal extensions or a Gbeta(5) subunit but resides in the RGS domain itself.


Subject(s)
GTP-Binding Proteins/metabolism , RGS Proteins/physiology , Fluorescence , GTP Phosphohydrolases/metabolism , Guanosine Triphosphate/metabolism , Hydrolysis , Kinetics , Protein Subunits
14.
J Neurochem ; 75(3): 1190-9, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10936202

ABSTRACT

Two constructs encoding the human micro-opioid receptor (hMOR) fused at its C terminus to either one of two Galpha subunits, Galpha(o1) (hMOR-Galpha(o1)) and Galpha(i2) (hMOR-Galpha(i2)), were expressed in Escherichia coli at levels suitable for pharmacological studies (0.4-0.5 pmol/mg). Receptors fused to Galpha(o1) or to Galpha(i2) maintained high-affinity binding of the antagonist diprenorphine. Affinities of the micro-selective agonists morphine, [D-Ala(2),N-Me-Phe(4),Gly(5)-ol]enkephalin (DAMGO), and endomorphins as well as their potencies and intrinsic activities in stimulating guanosine 5'-O-(3-[(35)S]thiotriphosphate) ([(35)S]GTPgammaS) binding were assessed in the presence of added purified Gbetagamma subunits. Both fusion proteins displayed high-affinity agonist binding and agonist-stimulated [(35)S]GTPgammaS binding. In the presence of Gbetagamma dimers, the affinities of DAMGO and endomorphin-1 and -2 were higher at hMOR-Galpha(i2) than at hMOR-Galpha(o1), whereas morphine displayed similar affinities at the two chimeras. Potencies of the four agonists in stimulating [(35)S]GTPgammaS binding at hMOR-Galpha(o1) were similar, whereas at hMOR-Galpha(i2), endomorphin-1 and morphine were more potent than DAMGO and endomorphin-2. The intrinsic activities of the four agonists at the two fusion constructs were similar. The results confirm hMOR coupling to Galpha(o1) and Galpha(i2) and support the hypothesis of the existence of multiple receptor conformational states, depending on the nature of the G protein to which it is coupled.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go , Heterotrimeric GTP-Binding Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Opioid, mu/metabolism , Animals , Brain/metabolism , Cattle , Diprenorphine/pharmacokinetics , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Escherichia coli , GTP-Binding Protein alpha Subunit, Gi2 , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Heterotrimeric GTP-Binding Proteins/genetics , Humans , Kinetics , Morphine/pharmacology , Oligopeptides/pharmacology , Proto-Oncogene Proteins/genetics , Radioligand Assay , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/genetics , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/metabolism , Tritium
15.
J Pharmacol Exp Ther ; 294(2): 539-47, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10900230

ABSTRACT

In this study, we investigated the hypothesis of agonist-directed trafficking of receptor signaling for the alpha(2A)-adrenergic receptor (alpha(2A)-AR). alpha(2A)-ARs couple to both G(s) and G(i) to stimulate or inhibit adenylyl cyclase activity. Chinese hamster ovary-K1 cell lines expressing the porcine alpha(2A)-AR at high (alpha(2A)-H) and low (alpha(2A)-L) levels were used to estimate the relative efficacies (R.e.s) of a series of agonists for the G(s) and G(i) pathways. G(s)-mediated responses were measured after pertussis toxin treatment to inactivate G(i) in alpha(2A)-H, whereas G(i) responses were measured in alpha(2A)-L, where G(s) responses were absent. The full agonist UK-14,304 showed a large receptor reserve for G(i) responses in alpha(2A)-H but little receptor reserve for G(s) responses in alpha(2A)-H or for G(i) responses in alpha(2A)-L. With the exception of l-isoproterenol (ISO), all agonists showed similar R.e.s at the alpha(2A)-AR for G(s) and G(i) responses, with rank orders of R.e.s as follows: l-epinephrine = l-norepinephrine = UK-14,304 > p-aminoclonidine > or = BHT-920 > or = BHT-933 > clonidine = p-iodoclonidine > or = xylazine > or = guanabenz. Interestingly, ISO had the highest efficacy at the alpha(2A)-AR for activating G(s) versus G(i) (9-fold higher); however, it had low potency for both. By several criteria, the ISO response was mediated by the alpha(2A)-AR, supporting the hypothesis of agonist-directed trafficking of receptor signaling or agonist-specific G protein selectivity. In contrast, the apparent G(i) pathway selectivity of oxymetazoline appears to be mediated by an endogenous serotonergic receptor. It is intriguing that a classic beta-AR agonist that activates G(s) through beta(2)-ARs also appears to produce a G(s)-selective conformation of the G(i)-coupled alpha(2A)-AR.


Subject(s)
Adrenergic alpha-2 Receptor Agonists , Adrenergic alpha-Agonists/pharmacology , Adrenergic beta-Agonists/pharmacology , GTP-Binding Protein alpha Subunits, Gs/physiology , Isoproterenol/pharmacology , Signal Transduction/physiology , Adrenergic alpha-Antagonists/pharmacology , Animals , Binding Sites , Brimonidine Tartrate , CHO Cells , Cricetinae , Drug Interactions , GTP-Binding Protein alpha Subunits, Gi-Go/agonists , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , GTP-Binding Protein alpha Subunits, Gs/agonists , Kinetics , Oxymetazoline/antagonists & inhibitors , Oxymetazoline/pharmacology , Pindolol/analogs & derivatives , Pindolol/pharmacology , Protein Binding , Quinoxalines/pharmacology , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Adrenergic, alpha-2/physiology , Serotonin Antagonists/pharmacology , Signal Transduction/drug effects , Swine , Yohimbine/pharmacology
16.
J Bacteriol ; 182(12): 3460-6, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10852878

ABSTRACT

Era is an essential Escherichia coli guanine nucleotide binding protein that appears to play a number of cellular roles. Although the kinetics of Era guanine nucleotide binding and hydrolysis have been described, guanine nucleotide exchange rates have never been reported. Here we describe a kinetic analysis of guanine nucleotide binding, exchange, and hydrolysis by Era using the fluorescent mant (N-methyl-3'-O-anthraniloyl) guanine nucleotide analogs. The equilibrium binding constants (K(D)) for mGDP and mGTP (0.61 +/- 0. 12 microgM and 3.6 +/- 0.80 microM, respectively) are similar to those of the unmodified nucleotides. The single turnover rates for mGTP hydrolysis by Era were 3.1 +/- 0.2 mmol of mGTP hydrolyzed/min/mol in the presence of 5 mM MgCl(2) and 5.6 +/- 0.3 mmol of mGTP hydrolyzed/min/mol in the presence of 0.2 mM MgCl(2). Moreover, Era associates with and exchanges guanine nucleotide rapidly (on the order of seconds) in both the presence and absence of Mg(2+). We suggest that models of Era function should reflect the rapid exchange of nucleotides in addition to the GTPase activity inherent to Era.


Subject(s)
Escherichia coli Proteins , Escherichia coli/metabolism , GTP-Binding Proteins/metabolism , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , RNA-Binding Proteins , Escherichia coli/genetics , GTP-Binding Proteins/genetics , GTP-Binding Proteins/isolation & purification , Guanosine Diphosphate/analogs & derivatives , Guanosine Triphosphate/analogs & derivatives , Histidine/chemistry , Hydrolysis , Kinetics , Time Factors , ortho-Aminobenzoates/metabolism
17.
Life Sci ; 68(6): 647-58, 2000 Dec 29.
Article in English | MEDLINE | ID: mdl-11205879

ABSTRACT

The binding of a drug to a G-protein coupled receptor initiates a complex series of dynamic events that ultimately leads to a cellular response. In addition to the concentrations of receptor, drug and G-protein, important determinants of the cellular response are the rates at which these species interact. However, most models for G-protein coupled receptor signaling are equilibrium models that neglect the role of reaction kinetics. A kinetic ternary-complex model of signaling through G-protein coupled receptors is presented. We demonstrate that this kinetic model can make significantly different predictions than an equilibrium ternary complex model, which provides a different perspective on multiple aspects of the signal transduction cascade, such as agonist efficacy, the effect of precoupled receptors, and the role of RGS proteins. Incorporation of the reaction kinetics is critical for a complete understanding of signal transduction and will ultimately impact the fields of drug discovery and drug design.


Subject(s)
GTP-Binding Proteins/metabolism , Biotransformation , Kinetics , Ligands , Models, Biological
18.
Biochemistry ; 38(42): 13795-800, 1999 Oct 19.
Article in English | MEDLINE | ID: mdl-10529224

ABSTRACT

The role of interdomain contact sites in basal GDP release from heterotrimeric G proteins is unknown. G(alpha)(o) and G(alpha)(i1) display a 5-fold difference in the rate of GDP dissociation with half-times of 2.3 +/- 0.2 and 10.4 +/- 1.3 min, respectively. To identify molecular determinants of the GDP release rate, we evaluated the rate of binding of the fluorescent guanine nucleotide 2'(3')-O-(N-methyl-3'-anthraniloyl)guanosine 5'-O-(3-thiotriphosphate) (mGTPgammaS) to chimers of G(alpha)(o) and G(alpha)(i1). Although no one region of the G protein determined the GDP dissociation rate, when the C-terminal 123 amino acids in G(alpha)(i1) were replaced with those of G(alpha)(o), the GDP release rate increased 3.3-fold compared to that of wild-type G(alpha)(i1). Within the C-terminal portion, modification of four amino acids in a coil between beta4 and the alpha3 helix resulted in GDP release kinetics identical to those of wild-type G(alpha)(o). Based on the G(alpha)(i1)-GDP crystal structure of this region, Leu(232) appeared to form a hydrophobic contact with Arg(144) of the helical domain. The role of this interaction was confirmed by G(alpha)(i1) L232Q and G(alpha)(i1) R144A which displayed 2-5-fold faster GDP release rates compared to wild-type G(alpha)(i1) (t(1/2) 4.7 and 1.5 min, respectively), suggesting that interdomain bridging contacts partially determine the basal rate of GDP release from heterotrimeric G proteins.


Subject(s)
Guanosine Diphosphate/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , Fluorescent Dyes/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/analogs & derivatives , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Guanosine Diphosphate/chemistry , Heterotrimeric GTP-Binding Proteins/chemistry , Heterotrimeric GTP-Binding Proteins/genetics , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spectrometry, Fluorescence , ortho-Aminobenzoates/metabolism
19.
Mol Pharmacol ; 56(5): 1005-13, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10531407

ABSTRACT

The structural determinants of G protein coupling versus activation by G protein-coupled receptors are not well understood. We examine the role of two distinct basic regions in the carboxyl terminal portion of the third intracellular loop of the alpha(2A)-adrenergic receptor to dissect these aspects of function. Changing three arginines to alanines by mutagenesis and stable expression in Chinese hamster ovary-K1 cells impaired the alpha(2)-adrenergic receptor G(s)-mediated stimulation of cyclic AMP (cAMP) accumulation, whereas G(i)-mediated inhibition was normal. When two (B2) or three (B3) basic residues closer to transmembrane span 6 were mutated to alanine, normal ligand binding was observed, but G(i)-mediated inhibition of cAMP accumulation showed 20-fold and 50-fold decreases in agonist potency for the B2 and B3 mutants, respectively. Surprisingly, a normal G(s) response was seen for the B2 mutant, and the B3 mutant showed only a 6-fold decrease in agonist potency. Mutation of both the three alanines and B3 residues to alanines showed a 200-fold decrease in agonist potency for G(i)-mediated inhibition of cAMP accumulation, whereas the G(s) response was nearly completely eliminated. The three basic residues (which include the BB of the BBXXF motif) play a role as G(i) activators rather than in receptor-G protein coupling, because high-affinity agonist binding is intact. Thus, we have identified three basic residues required for activation of G(i) but not required for receptor-G protein coupling. Also, distinct basic residues are required for optimal G(i) and G(s) responses, defining a microspecificity determinant within the carboxyl terminal portion of the third intracellular loop of the alpha(2a) adrenergic receptor.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Amino Acid Sequence , Animals , Arginine/metabolism , CHO Cells , Cricetinae , Membrane Proteins/metabolism , Molecular Sequence Data , Protein Conformation
20.
J Biol Chem ; 273(21): 12794-7, 1998 May 22.
Article in English | MEDLINE | ID: mdl-9582306

ABSTRACT

Regulator of G protein-signaling (RGS) proteins accelerate GTP hydrolysis by Galpha subunits and are thought to be responsible for rapid deactivation of enzymes and ion channels controlled by G proteins. We wanted to identify and characterize Gi-family alpha subunits that were insensitive to RGS action. Based on a glycine to serine mutation in the yeast Galpha subunit Gpa1(sst) that prevents deactivation by Sst2 (DiBello, P. R., Garrison, T. R., Apanovitch, D. M., Hoffman, G., Shuey, D. J., Mason, K., Cockett, M. I., and Dohlman, H. G. (1998) J. Biol. Chem. 273, 5780-5784), site-directed mutagenesis of alphao and alphai1 was done. G184S alphao and G183S alphai1 show kinetics of GDP release and GTP hydrolysis similar to wild type. In contrast, GTP hydrolysis by the G --> S mutant proteins is not stimulated by RGS4 or by a truncated RGS7. Quantitative flow cytometry binding studies show IC50 values of 30 and 96 nM, respectively, for aluminum fluoride-activated wild type alphao and alphai1 to compete with fluorescein isothiocyanate-alphao binding to glutathione S-transferase-RGS4. The G --> S mutant proteins showed a greater than 30-100-fold lower affinity for RGS4. Thus, we have defined the mechanism of a point mutation in alphao and alphai1 that prevents RGS binding and GTPase activating activity. These mutant subunits should be useful in biochemical or expression studies to evaluate the role of endogenous RGS proteins in Gi function.


Subject(s)
GTP-Binding Proteins/metabolism , Mutagenesis, Site-Directed , Signal Transduction , Enzyme Activation , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/genetics , Glycine/genetics , Glycine/metabolism , Guanosine Triphosphate/metabolism , Hydrolysis , Serine/genetics , Serine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL