Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
Add more filters

Publication year range
1.
Cell ; 183(2): 503-521.e19, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33007266

ABSTRACT

The control over the extent and timing of G protein signaling is provided by the regulator of G protein signaling (RGS) proteins that deactivate G protein α subunits (Gα). Mammalian genomes encode 20 canonical RGS and 16 Gα genes with key roles in physiology and disease. To understand the principles governing the selectivity of Gα regulation by RGS, we examine the catalytic activity of all canonical human RGS proteins and their selectivity for a complete set of Gα substrates using real-time kinetic measurements in living cells. The data reveal rules governing RGS-Gα recognition, the structural basis of its selectivity, and provide principles for engineering RGS proteins with defined selectivity. The study also explores the evolution of RGS-Gα selectivity through ancestral reconstruction and demonstrates how naturally occurring non-synonymous variants in RGS alter signaling. These results provide a blueprint for decoding signaling selectivity and advance our understanding of molecular recognition principles.


Subject(s)
GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein alpha Subunits/physiology , RGS Proteins/genetics , Animals , Female , GTP-Binding Protein Regulators/metabolism , GTP-Binding Protein alpha Subunits/genetics , HEK293 Cells , Humans , Kinetics , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Primary Cell Culture , Protein Binding , RGS Proteins/metabolism , RGS Proteins/physiology , Signal Transduction/genetics
2.
Proc Natl Acad Sci U S A ; 119(20): e2120870119, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35544691

ABSTRACT

Transient receptor potential canonical 4 (TRPC4) is a receptor-operated cation channel codependent on both the Gq/11­phospholipase C signaling pathway and Gi/o proteins for activation. This makes TRPC4 an excellent coincidence sensor of neurotransmission through Gq/11- and Gi/o-coupled receptors. In whole-cell slice recordings of lateral septal neurons, TRPC4 mediates a strong depolarizing plateau that shuts down action potential firing, which may or may not be followed by a hyperpolarization that extends the firing pause to varying durations depending on the strength of Gi/o stimulation. We show that the depolarizing plateau is codependent on Gq/11-coupled group I metabotropic glutamate receptors and on Gi/o-coupled γ-aminobutyric acid type B receptors. The hyperpolarization is mediated by Gi/o activation of G protein­activated inwardly rectifying K+ (GIRK) channels. Moreover, the firing patterns, elicited by either electrical stimulation or receptor agonists, encode information about the relative strengths of Gq/11 and Gi/o inputs in the following fashion. Pure Gq/11 input produces weak depolarization accompanied by firing acceleration, whereas pure Gi/o input causes hyperpolarization that pauses firing. Although coincident Gq/11­Gi/o inputs also pause firing, the pause is preceded by a burst, and both the pause duration and firing recovery patterns reflect the relative strengths of Gq/11 versus Gi/o inputs. Computer simulations demonstrate that different combinations of TRPC4 and GIRK conductances are sufficient to produce the range of firing patterns observed experimentally. Thus, concurrent neurotransmission through the Gq/11 and Gi/o pathways is converted to discernible electrical responses by the joint actions of TRPC4 and GIRK for communication to downstream neurons.


Subject(s)
Action Potentials , G Protein-Coupled Inwardly-Rectifying Potassium Channels , GTP-Binding Protein alpha Subunits, Gi-Go , GTP-Binding Protein alpha Subunits , Neurons , Synaptic Transmission , TRPC Cation Channels , Animals , Cell Communication , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , GTP-Binding Protein alpha Subunits/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Mice , Neurons/physiology , TRPC Cation Channels/physiology
3.
J Biol Chem ; 296: 100472, 2021.
Article in English | MEDLINE | ID: mdl-33639168

ABSTRACT

Heterotrimeric G protein subunits Gαq and Gα11 are inhibited by two cyclic depsipeptides, FR900359 (FR) and YM-254890 (YM), both of which are being used widely to implicate Gq/11 proteins in the regulation of diverse biological processes. An emerging major research question therefore is whether the cellular effects of both inhibitors are on-target, that is, mediated via specific inhibition of Gq/11 proteins, or off-target, that is, the result of nonspecific interactions with other proteins. Here we introduce a versatile experimental strategy to discriminate between these possibilities. We developed a Gαq variant with preserved catalytic activity, but refractory to FR/YM inhibition. A minimum of two amino acid changes were required and sufficient to achieve complete inhibitor resistance. We characterized the novel mutant in HEK293 cells depleted by CRISPR-Cas9 of endogenous Gαq and Gα11 to ensure precise control over the Gα-dependent cellular signaling route. Using a battery of cellular outcomes with known and concealed Gq contribution, we found that FR/YM specifically inhibited cellular signals after Gαq introduction via transient transfection. Conversely, both inhibitors were inert across all assays in cells expressing the drug-resistant variant. These findings eliminate the possibility that inhibition of non-Gq proteins contributes to the cellular effects of the two depsipeptides. We conclude that combined application of FR or YM along with the drug-resistant Gαq variant is a powerful in vitro strategy to discern on-target Gq against off-target non-Gq action. Consequently, it should be of high value for uncovering Gq input to complex biological processes with high accuracy and the requisite specificity.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/physiology , GTP-Binding Protein alpha Subunits/physiology , Signal Transduction/physiology , Depsipeptides/pharmacology , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , HEK293 Cells , Heterotrimeric GTP-Binding Proteins/metabolism , Heterotrimeric GTP-Binding Proteins/physiology , Humans , Peptides, Cyclic/pharmacology , Signal Transduction/drug effects
4.
Nat Immunol ; 10(4): 403-11, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19252491

ABSTRACT

Immature B cells developing in the bone marrow are found in the parenchyma and sinusoids. The mechanisms that control the positioning of B cells in the sinusoids are not understood. Here we show that the integrin alpha(4)beta(1) (VLA-4) and its ligand VCAM-1 were required, whereas the chemokine receptor CXCR4 was dispensable, for sinusoidal retention of B cells. Instead, cannabinoid receptor 2 (CB2), a Galpha(i) protein-coupled receptor upregulated in immature B cells, was required for sinusoidal retention. Using two-photon microscopy, we found immature B cells entering and crawling in sinusoids; these immature B cells were displaced by CB2 antagonism. Moreover, CB2-deficient mice had a lower frequency of immunoglobulin lambda-chain-positive B cells in the peripheral blood and spleen. Our findings identify unique requirements for the retention of B cells in the bone marrow sinusoidal niche and suggest involvement of CB2 in the generation of the B cell repertoire.


Subject(s)
Bone Marrow/physiology , Precursor Cells, B-Lymphoid/physiology , Receptor, Cannabinoid, CB2/physiology , Animals , Bone Marrow/immunology , Cell Movement/physiology , GTP-Binding Protein alpha Subunits/physiology , Immunoglobulin lambda-Chains/blood , Immunoglobulin lambda-Chains/immunology , Integrin alpha4beta1/immunology , Integrin alpha4beta1/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/immunology , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/immunology , Receptors, CXCR4/immunology , Receptors, CXCR4/metabolism , Spleen/immunology , Up-Regulation , Vascular Cell Adhesion Molecule-1/immunology , Vascular Cell Adhesion Molecule-1/physiology
5.
PLoS Comput Biol ; 15(10): e1007382, 2019 10.
Article in English | MEDLINE | ID: mdl-31665146

ABSTRACT

Long-term potentiation and depression of synaptic activity in response to stimuli is a key factor in reinforcement learning. Strengthening of the corticostriatal synapses depends on the second messenger cAMP, whose synthesis is catalysed by the enzyme adenylyl cyclase 5 (AC5), which is itself regulated by the stimulatory Gαolf and inhibitory Gαi proteins. AC isoforms have been suggested to act as coincidence detectors, promoting cellular responses only when convergent regulatory signals occur close in time. However, the mechanism for this is currently unclear, and seems to lie in their diverse regulation patterns. Despite attempts to isolate the ternary complex, it is not known if Gαolf and Gαi can bind to AC5 simultaneously, nor what activity the complex would have. Using protein structure-based molecular dynamics simulations, we show that this complex is stable and inactive. These simulations, along with Brownian dynamics simulations to estimate protein association rates constants, constrain a kinetic model that shows that the presence of this ternary inactive complex is crucial for AC5's ability to detect coincident signals, producing a synergistic increase in cAMP. These results reveal some of the prerequisites for corticostriatal synaptic plasticity, and explain recent experimental data on cAMP concentrations following receptor activation. Moreover, they provide insights into the regulatory mechanisms that control signal processing by different AC isoforms.


Subject(s)
Adenylyl Cyclases/metabolism , GTP-Binding Protein alpha Subunits/physiology , Adenylyl Cyclases/physiology , Animals , Corpus Striatum/physiology , Dogs , Kinetics , Molecular Dynamics Simulation , Neuronal Plasticity , Neurons/physiology , Protein Isoforms/metabolism , Rats , Signal Transduction/physiology
6.
J Neurosci ; 37(50): 12202-12213, 2017 12 13.
Article in English | MEDLINE | ID: mdl-29118104

ABSTRACT

The olfactory system can discriminate a vast number of odorants. This ability derives from the existence of a large family of odorant receptors expressed in the cilia of the olfactory sensory neurons. Odorant receptors signal through the olfactory-specific G-protein subunit, Gαolf. Ric-8b, a guanine nucleotide exchange factor, interacts with Gαolf and can amplify odorant receptor signal transduction in vitro To explore the function of Ric-8b in vivo, we generated a tissue specific knock-out mouse by crossing OMP-Cre transgenic mice to Ric-8b floxed mice. We found that olfactory-specific Ric-8b knock-out mice of mixed sex do not express the Gαolf protein in the olfactory epithelium. We also found that in these mice, the mature olfactory sensory neuron layer is reduced, and that olfactory sensory neurons show increased rate of cell death compared with wild-type mice. Finally, behavioral tests showed that the olfactory-specific Ric-8b knock-out mice show an impaired sense of smell, even though their motivation and mobility behaviors remain normal.SIGNIFICANCE STATEMENT Ric-8b is a guanine nucleotide exchange factor (GEF) expressed in the olfactory epithelium and in the striatum. Ric-8b interacts with the olfactory Gαolf subunit, and can amplify odorant signaling through odorant receptors in vitro However, the functional significance of this GEF in the olfactory neurons in vivo remains unknown. We report that deletion of Ric-8b in olfactory sensory neurons prevents stable expression of Gαolf. In addition, we demonstrate that olfactory neurons lacking Ric-8b (and consequently Gαolf) are more susceptible to cell death. Ric-8b conditional knock-out mice display impaired olfactory guided behavior. Our results reveal that Ric-8b is essential for olfactory function, and suggest that it may also be essential for Gαolf-dependent functions in the brain.


Subject(s)
Appetitive Behavior/physiology , Avoidance Learning/physiology , Guanine Nucleotide Exchange Factors/physiology , Nerve Tissue Proteins/physiology , Olfactory Receptor Neurons/physiology , Animals , Animals, Suckling , Butyric Acid , Cell Count , Cell Death , Crosses, Genetic , Female , Food , GTP-Binding Protein alpha Subunits/deficiency , GTP-Binding Protein alpha Subunits/physiology , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , Male , Mice , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Odorants , Olfactory Mucosa/pathology , Receptors, Odorant/physiology
7.
J Neurosci ; 35(37): 12903-16, 2015 Sep 16.
Article in English | MEDLINE | ID: mdl-26377475

ABSTRACT

The gonadotropin-releasing hormone (GnRH) is the master regulator of fertility and kisspeptin (KP) is a potent trigger of GnRH secretion from GnRH neurons. KP signals via KISS1R, a Gαq/11-coupled receptor, and mice bearing a global deletion of Kiss1r (Kiss1r(-/-)) or a GnRH neuron-specific deletion of Kiss1r (Kiss1r(d/d)) display hypogonadotropic hypogonadism and infertility. KISS1R also signals via ß-arrestin, and in mice lacking ß-arrestin-1 or -2, KP-triggered GnRH secretion is significantly diminished. Based on these findings, we hypothesized that ablation of Gαq/11 in GnRH neurons would diminish but not completely block KP-triggered GnRH secretion and that Gαq/11-independent GnRH secretion would be sufficient to maintain fertility. To test this, Gnaq (encodes Gαq) was selectively inactivated in the GnRH neurons of global Gna11 (encodes Gα11)-null mice by crossing Gnrh-Cre and Gnaq(fl/fl);Gna11(-/-) mice. Experimental Gnaq(fl/fl);Gna11(-/-);Gnrh-Cre (Gnaq(d/d)) and control Gnaq(fl/fl);Gna11(-/-) (Gnaq(fl/fl)) littermate mice were generated and subjected to reproductive profiling. This process revealed that testicular development and spermatogenesis, preputial separation, and anogenital distance in males and day of vaginal opening and of first estrus in females were significantly less affected in Gnaq(d/d) mice than in previously characterized Kiss1r(-/-) or Kiss1r(d/d) mice. Additionally, Gnaq(d/d) males were subfertile, and although Gnaq(d/d) females did not ovulate spontaneously, they responded efficiently to a single dose of gonadotropins. Finally, KP stimulation triggered a significant increase in gonadotropins and testosterone levels in Gnaq(d/d) mice. We therefore conclude that the milder reproductive phenotypes and maintained responsiveness to KP and gonadotropins reflect Gαq/11-independent GnRH secretion and activation of the neuroendocrine-reproductive axis in Gnaq(d/d) mice. SIGNIFICANCE STATEMENT: The gonadotropin-releasing hormone (GnRH) is the master regulator of fertility. Over the last decade, several studies have established that the KISS1 receptor, KISS1R, is a potent trigger of GnRH secretion and inactivation of KISS1R on the GnRH neuron results in infertility. While KISS1R is best understood as a Gαq/11-coupled receptor, we previously demonstrated that it could couple to and signal via non-Gαq/11-coupled pathways. The present study confirms these findings and, more importantly, while it establishes Gαq/11-coupled signaling as a major conduit of GnRH secretion, it also uncovers a significant role for non-Gαq/11-coupled signaling in potentiating reproductive development and function. This study further suggests that by augmenting signaling via these pathways, GnRH secretion can be enhanced to treat some forms of infertility.


Subject(s)
GTP-Binding Protein alpha Subunits/deficiency , Gonadotropin-Releasing Hormone/physiology , Hypogonadism/physiopathology , Infertility, Female/physiopathology , Infertility, Male/physiopathology , Animals , Blastocyst/pathology , Embryonic Development , Female , GTP-Binding Protein alpha Subunits/physiology , Gene Expression Profiling , Genitalia, Female/pathology , Genitalia, Female/physiopathology , Genitalia, Male/pathology , Genitalia, Male/physiopathology , Gonadal Steroid Hormones/metabolism , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Gonadotropins, Pituitary/metabolism , Gonadotropins, Pituitary/pharmacology , Hypogonadism/genetics , Hypogonadism/pathology , Hypothalamo-Hypophyseal System/physiopathology , Hypothalamus/pathology , Infertility, Female/embryology , Infertility, Female/genetics , Infertility, Male/embryology , Infertility, Male/genetics , Kisspeptins/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Oligopeptides/pharmacology , Ovariectomy , Ovulation/drug effects , Peptide Fragments/pharmacology , Peptides/pharmacology , Phenotype , Receptors, G-Protein-Coupled , Receptors, Kisspeptin-1 , Spermatogenesis
8.
Plant J ; 81(3): 388-98, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25430066

ABSTRACT

Heterotrimeric G proteins are crucial for the perception of external signals and subsequent signal transduction in animal and plant cells. In both model systems, the complex comprises one Gα, one Gß, and one Gγ subunit. However, in addition to the canonical Gγ subunits (class A), plants also possess two unusual, plant-specific classes of Gγ subunits (classes B and C) that have not yet been found in animals. These include Gγ subunits lacking the C-terminal CaaX motif (class B), which is important for membrane anchoring of the protein; the presence of such subunits gives rise to a flexible sub-population of Gß/γ heterodimers that are not necessarily restricted to the plasma membrane. Plants also contain class C Gγ subunits, which are twice the size of canonical Gγ subunits, with a predicted transmembrane domain and a large cysteine-rich extracellular C-terminus. However, neither the presence of the transmembrane domain nor the membrane topology have been unequivocally demonstrated. Here, we provide compelling evidence that AGG3, a class C Gγ subunit of Arabidopsis, contains a functional transmembrane domain, which is sufficient but not essential for plasma membrane localization, and that the cysteine-rich C-terminus is extracellular.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis/metabolism , GTP-Binding Protein gamma Subunits/chemistry , Arabidopsis/genetics , Arabidopsis Proteins/analysis , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/physiology , Cell Membrane/metabolism , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein alpha Subunits/physiology , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein beta Subunits/physiology , GTP-Binding Protein gamma Subunits/analysis , GTP-Binding Protein gamma Subunits/genetics , Models, Molecular , Phylogeny , Protein Structure, Tertiary , Sequence Analysis, Protein
9.
Cell Mol Life Sci ; 72(1): 165-79, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24958088

ABSTRACT

Melanopsin expressing photosensitive retinal ganglion cells (pRGCs) represent a third class of ocular photoreceptors and mediate a range of non-image forming responses to light. Melanopsin is a G protein coupled receptor (GPCR) and existing data suggest that it employs a membrane bound signalling cascade involving Gnaq/11 type G proteins. However, to date the precise identity of the Gα subunits involved in melanopsin phototransduction remains poorly defined. Here we show that Gnaq, Gna11 and Gna14 are highly co-expressed in pRGCs of the mouse retina. Furthermore, using RNAi based gene silencing we show that melanopsin can signal via Gnaq, Gna11 or Gna14 in vitro, and demonstrate that multiple members of the Gnaq/11 subfamily, including Gna14 and at least Gnaq or Gna11, can participate in melanopsin phototransduction in vivo and contribute to the pupillary light responses of mice lacking rod and cone photoreceptors. This diversity of G protein interactions suggests additional complexity in the melanopsin phototransduction cascade and may provide a basis for generating the diversity of light responses observed from pRGC subtypes.


Subject(s)
GTP-Binding Protein alpha Subunits/physiology , Pupil/physiology , RNA, Small Interfering/genetics , Retinal Ganglion Cells/metabolism , Rod Opsins/physiology , Animals , Blotting, Western , Calcium/metabolism , Cells, Cultured , Female , GTP-Binding Protein alpha Subunits/antagonists & inhibitors , Immunoenzyme Techniques , Integrases/metabolism , Light , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Photic Stimulation , Pupil/radiation effects , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/radiation effects , Reverse Transcriptase Polymerase Chain Reaction , Rod Opsins/antagonists & inhibitors
10.
Immunol Cell Biol ; 93(7): 616-24, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25732870

ABSTRACT

Gαq, the α-subunit of Gq protein, is ubiquitously expressed in mammalian cells. It initially attracted attention for its physiological significance in cardiovascular system. In recent years, studies have also indicated the important roles of Gαq in regulating immunity, supplying us a new insight into the mechanism of immune regulation. T helper type 17 (Th17) cells are potent inducers of tissue inflammation. Many studies have shown that Th17 cells are major effector cells in the pathogenesis of many experimental autoimmune diseases and human inflammatory conditions such as rheumatoid arthritis (RA). One of our previous studies has shown that Gαq negatively controls the disease activity of RA. However, how Gαq controls the pathogenesis of autoimmune disease is not clear. Whether this effect is via the regulation of Th17 differentiation is still not known. We aimed to find out the role of Gαq in control of Th17 differentiation. We investigated the relationship between Gαq and Th17 in RA patients. We then investigated the mechanism of how Gαq regulated Th17 differentiation by using Gnaq(-/-) mice. We observed that the expression of Gαq was negatively associated with interleukin-17A expression in RA patients, indicating that Gαq negatively controlled the differentiation of Th17 cells. By using Gnaq(-/-) mice, we demonstrated that Gαq inhibited the differentiation of Th17 cell via regulating the activity of extracellular signal-regulated kinase-1/2 to control the expression of STAT3 (signal transducer and activator of transcription 3) and RORα (RAR-related orphan receptor-α). These data suggest the possibility of targeting Gαq to develop a novel therapeutic regimen for autoimmune disease.


Subject(s)
Arthritis, Rheumatoid/pathology , GTP-Binding Protein alpha Subunits/physiology , Th17 Cells/pathology , Animals , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , CD4-Positive T-Lymphocytes/metabolism , Dendritic Cells/metabolism , Flavonoids/pharmacology , GTP-Binding Protein alpha Subunits/biosynthesis , GTP-Binding Protein alpha Subunits/deficiency , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits, Gq-G11 , Gene Expression Regulation , Humans , Interleukin-17/biosynthesis , Interleukin-17/genetics , Interleukin-6/biosynthesis , Interleukin-6/genetics , Lymphopoiesis/genetics , Lymphopoiesis/physiology , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 1/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Radiation Chimera , STAT3 Transcription Factor/physiology , Spondylitis, Ankylosing/genetics , Spondylitis, Ankylosing/metabolism , Th17 Cells/immunology
11.
PLoS Pathog ; 8(2): e1002553, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22383884

ABSTRACT

Heterotrimeric G-proteins are molecular switches integral to a panoply of different physiological responses that many organisms make to environmental cues. The switch from inactive to active Gαßγ heterotrimer relies on nucleotide cycling by the Gα subunit: exchange of GTP for GDP activates Gα, whereas its intrinsic enzymatic activity catalyzes GTP hydrolysis to GDP and inorganic phosphate, thereby reverting Gα to its inactive state. In several genetic studies of filamentous fungi, such as the rice blast fungus Magnaporthe oryzae, a G42R mutation in the phosphate-binding loop of Gα subunits is assumed to be GTPase-deficient and thus constitutively active. Here, we demonstrate that Gα(G42R) mutants are not GTPase deficient, but rather incapable of achieving the activated conformation. Two crystal structure models suggest that Arg-42 prevents a typical switch region conformational change upon Gα(i1)(G42R) binding to GDP·AlF(4)(-) or GTP, but rotameric flexibility at this locus allows for unperturbed GTP hydrolysis. Gα(G42R) mutants do not engage the active state-selective peptide KB-1753 nor RGS domains with high affinity, but instead favor interaction with Gßγ and GoLoco motifs in any nucleotide state. The corresponding Gα(q)(G48R) mutant is not constitutively active in cells and responds poorly to aluminum tetrafluoride activation. Comparative analyses of M. oryzae strains harboring either G42R or GTPase-deficient Q/L mutations in the Gα subunits MagA or MagB illustrate functional differences in environmental cue processing and intracellular signaling outcomes between these two Gα mutants, thus demonstrating the in vivo functional divergence of G42R and activating G-protein mutants.


Subject(s)
GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/physiology , Magnaporthe/pathogenicity , Mycoses/genetics , Point Mutation , Protein Folding , Amino Acid Substitution/physiology , Catalytic Domain/genetics , Crystallography, X-Ray , GTP-Binding Protein alpha Subunits/chemistry , GTP-Binding Protein alpha Subunits/metabolism , Hordeum/microbiology , Magnaporthe/genetics , Magnaporthe/metabolism , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutant Proteins/physiology , Plant Diseases/genetics , Plant Diseases/microbiology , Plant Leaves/microbiology , Point Mutation/physiology , Protein Structure, Tertiary/genetics , Signal Transduction/genetics
12.
Br J Cancer ; 109(2): 493-6, 2013 Jul 23.
Article in English | MEDLINE | ID: mdl-23778528

ABSTRACT

BACKGROUND: Mutations in GNAQ and GNA11, encoding the oncogenic G-protein alpha subunit q and 11, respectively, occur frequently in the majority of uveal melanomas. METHODS: Exons 4 and 5 from GNAQ and GNA11 were amplified and sequenced from 92 ciliary body and choroidal melanomas. The mutation status was correlated with disease-free survival (DFS) and other parameters. RESULTS: None of the tumours harboured a GNAQ exon 4 mutation. A GNAQ mutation in exon 5 codon 209 was found in 46 out of 92 (50.0%) of the tumours. Only 1 out of 92 (1.1%) melanomas showed a mutation in GNA11 exon 4 codon 183, whereas 39 out of 92 (42.4%) harboured a mutation in exon 5 of GNA11 codon 209. Six tumours did not show any mutations in exons 4 and 5 of these genes. Univariate analyses showed no correlation between DFS and the mutation status. CONCLUSION: GNAQ and GNA11 mutations are, in equal matter, not associated with patient outcome.


Subject(s)
GTP-Binding Protein alpha Subunits/genetics , Melanoma/genetics , Melanoma/mortality , Mutation/physiology , Oncogenes , Uveal Neoplasms/genetics , Uveal Neoplasms/mortality , Adult , Aged , Aged, 80 and over , DNA Mutational Analysis , Female , GTP-Binding Protein alpha Subunits/physiology , GTP-Binding Protein alpha Subunits, Gq-G11 , Humans , Male , Middle Aged , Survival Analysis , Young Adult
13.
Proc Natl Acad Sci U S A ; 106(3): 791-6, 2009 Jan 20.
Article in English | MEDLINE | ID: mdl-19129493

ABSTRACT

The Gpg1 protein is a Ggamma subunit mimic implicated in the G-protein glucose-signaling pathway in Saccharomyces cerevisiae, and its function is largely unknown. Here we report that Gpg1 blocks the maintenance of [PSI(+)], an aggregated prion form of the translation termination factor Sup35. Although the GPG1 gene is normally not expressed, over-expression of GPG1 inhibits propagation of not only [PSI(+)] but also [PIN(+)], [URE3] prions, and the toxic polyglutamine aggregate in S. cerevisiae. Over-expression of Gpg1 does not affect expression and activity of Hsp104, a protein-remodeling factor required for prion propagation, showing that Gpg1 does not target Hsp104 directly. Nevertheless, prion elimination by Gpg1 is weakened by over-expression of Hsp104. Importantly, Gpg1 protein is prone to self-aggregate and transiently colocalized with Sup35NM-prion aggregates when expressed in [PSI(+)] cells. Genetic selection and characterization of loss-of-activity gpg1 mutations revealed that multiple mutations on the hydrophobic one-side surface of predicted alpha-helices of the Gpg1 protein hampered the activity. Prion elimination by Gpg1 is unaffected in the gpa2Delta and gpb1Delta strains lacking the supposed physiological G-protein partners of Gpg1. These findings suggest a general inhibitory interaction of the Gpg1 protein with other transmissible and nontransmissible amyloids, resulting in prion elimination. Assuming the ability of Gpg1 to form G-protein heterotrimeric complexes, Gpg1 is likely to play a versatile function of reversing the prion state and modulating the G-protein signaling pathway.


Subject(s)
GTP-Binding Protein gamma Subunits/physiology , Prions/antagonists & inhibitors , Saccharomyces cerevisiae/metabolism , GTP-Binding Protein alpha Subunits/physiology , GTP-Binding Protein beta Subunits/physiology , Heat-Shock Proteins/physiology , Mutation , Saccharomyces cerevisiae Proteins/physiology
14.
J Neurosci ; 30(13): 4562-72, 2010 Mar 31.
Article in English | MEDLINE | ID: mdl-20357107

ABSTRACT

A central question in insect chemoreception is whether signaling occurs via G-proteins. Two families of seven-transmembrane-domain chemoreceptors, the odor (Or) and gustatory receptor (Gr) families, have been identified in Drosophila (Clyne et al., 1999, 2000; Vosshall et al., 1999). Ors mediate odor responses, whereas two Grs, Gr21a and Gr63a, mediate CO2 response (Hallem et al., 2004; Jones et al., 2007; Kwon et al., 2007). Using single-sensillum recordings, we systematically investigate the role of Galpha proteins in vivo, initially with RNA interference constructs, competitive peptides, and constitutively active Galpha proteins. The results do not support a role for Galpha proteins in odor sensitivity. In parallel experiments, manipulations of Galpha(q), but not other Galpha proteins, affected CO2 response. Transient, conditional, and ectopic expression analyses consistently supported a role for Galpha(q) in the response of CO2-sensing neurons, but not odor-sensing neurons. Genetic mosaic analysis confirmed that odor responses are normal in the absence of Galpha(q). Ggamma30A is also required for normal CO2 response. The simplest interpretation of these results is that Galpha(q) and Ggamma30A play a role in the response of CO2-sensing neurons, but are not required for Or-mediated odor signaling.


Subject(s)
Chemoreceptor Cells/physiology , Drosophila/physiology , GTP-Binding Protein alpha Subunits/physiology , GTP-Binding Protein gamma Subunits/physiology , Animals , Carbon Dioxide/physiology , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , GTP-Binding Protein alpha Subunits, Gs/genetics , GTP-Binding Protein alpha Subunits, Gs/physiology , GTP-Binding Protein gamma Subunits/genetics , Mutation , Odorants , Olfactory Pathways , Smell
15.
Mol Pharmacol ; 79(3): 488-98, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21088225

ABSTRACT

The chemokine receptor CCR2, which has been implicated in a variety of inflammatory, autoimmune, and cardiovascular conditions, binds several natural chemokine ligands. Here, we assessed the recruitment of ß-arrestin to CCR2 in response to these ligands using bioluminescence resonance energy transfer technology. Compared with CCL2, which was considered as a full agonist, other CCR2 ligands were partial agonists with reduced efficacy and potency. Agonist potencies were not a function of their affinity for CCR2. Efficacy of arrestin recruitment matched that of agonist-induced CCR2 internalization. Although the potency and efficacy rank orders of the ligands in arrestin recruitment were similar to those observed for Gα(i1) activation, arrestin recruitment was at least in part resistant to Gα(i/o)-inactivating pertussis toxin, suggesting partial independence from Gα(i/o). The degree of pertussis toxin resistance of arrestin recruitment was different between the chemokines. Moreover, qualitative differences between the arrestin responses to the different ligands were identified in the stability of the response: although CCL7-induced arrestin recruitment had a half-life of less than 15 min, CCL8 and CCL13 induced stable CCR2-arrestin interactions. Finally, the ligands stabilized different conformations of the CCR2 homodimer. Our results support the validity of models for receptor-ligand interactions in which different ligands stabilize different receptor conformations also for endogenous receptor ligands, with corresponding implications for drug development targeting CCR2.


Subject(s)
Arrestins/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Receptors, CCR2/metabolism , Signal Transduction , Adenylyl Cyclases/metabolism , Dose-Response Relationship, Drug , Endocytosis/drug effects , Flow Cytometry , GTP-Binding Protein alpha Subunits/drug effects , GTP-Binding Protein alpha Subunits/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , HEK293 Cells , Humans , Ligands , Pertussis Toxin/pharmacology , Protein Binding , Radioligand Assay , Receptors, CCR2/drug effects , Signal Transduction/drug effects , beta-Arrestins
16.
Plant Mol Biol ; 77(1-2): 145-58, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21725861

ABSTRACT

The canonical Gα subunit of the heterotrimeric G protein complex from wheat (Triticum aestivum), GA3, and the calcium-binding protein, Clo3, were revealed to interact both in vivo and in vitro and Clo3 was shown to enhance the GTPase activity of GA3. Clo3 is a member of the caleosin gene family in wheat with a single EF-hand domain and is induced during cold acclimation. Bimolecular Fluorescent Complementation (BiFC) was used to localize the interaction between Clo3 and GA3 to the plasma membrane (PM). Even though heterotrimeric G-protein signaling and Ca²âº signaling have both been shown to play a role in the response to environmental stresses in plants, little is known about the interaction between calcium-binding proteins and Gα. The GAP activity of Clo3 towards GA3 suggests it may play a role in the inactivation of GA3 as part of the stress response in plants. GA3 was also shown to interact with the phosphoinositide-specific phospholipase C, PI-PLC1, not only in the PM but also in the endoplasmic reticulum (ER). Surprisingly, Clo3 was also shown to interact with PI-PLC1 in the PM and ER. In vitro analysis of the protein-protein interaction showed that the interaction of Clo3 with GA3 and PI-PLC1 is enhanced by high Ca²âº levels. Three-way affinity characterizations with GA3, Clo3 and PI-PLC1 showed the interaction with Clo3 to be competitive, which suggests that Clo3 may play a role in the Ca²âº-triggered feedback regulation of both GA3 and PI-PLC1. This hypothesis was further supported by the demonstration that Clo3 has GAP activity with GA3.


Subject(s)
Calcium-Binding Proteins/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Phosphoinositide Phospholipase C/metabolism , Plant Proteins/metabolism , Triticum/enzymology , Amino Acid Sequence , Binding, Competitive , Calcium-Binding Proteins/physiology , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , GTP-Binding Protein alpha Subunits/physiology , Molecular Sequence Data , Plant Proteins/physiology , Sequence Alignment , Signal Transduction , Substrate Specificity , Triticum/genetics , Triticum/metabolism
17.
Plant Physiol ; 152(4): 2067-77, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20200073

ABSTRACT

Land plants must balance CO2 assimilation with transpiration in order to minimize drought stress and maximize their reproductive success. The ratio of assimilation to transpiration is called transpiration efficiency (TE). TE is under genetic control, although only one specific gene, ERECTA, has been shown to regulate TE. We have found that the alpha-subunit of the heterotrimeric G protein in Arabidopsis (Arabidopsis thaliana), GPA1, is a regulator of TE. gpa1 mutants, despite having guard cells that are hyposensitive to abscisic acid-induced inhibition of stomatal opening, have increased TE under ample water and drought stress conditions and when treated with exogenous abscisic acid. Leaf-level gas-exchange analysis shows that gpa1 mutants have wild-type assimilation versus internal CO2 concentration responses but exhibit reduced stomatal conductance compared with ecotype Columbia at ambient and below-ambient internal CO2 concentrations. The increased TE and reduced whole leaf stomatal conductance of gpa1 can be primarily attributed to stomatal density, which is reduced in gpa1 mutants. GPA1 regulates stomatal density via the control of epidermal cell size and stomata formation. GPA1 promoter::beta-glucuronidase lines indicate that the GPA1 promoter is active in the stomatal cell lineage, further supporting a function for GPA1 in stomatal development in true leaves.


Subject(s)
Arabidopsis Proteins/physiology , Arabidopsis/physiology , GTP-Binding Protein alpha Subunits/physiology , Plant Transpiration/physiology , Abscisic Acid/pharmacology , Arabidopsis/metabolism , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Biomass , Carbon Isotopes/metabolism , GTP-Binding Protein alpha Subunits/chemistry , GTP-Binding Protein alpha Subunits/genetics , Mutation , Plant Leaves/physiology
18.
Clin Sci (Lond) ; 121(12): 545-54, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21736560

ABSTRACT

Gastro-oesophageal variceal haemorrhage is one of the most dreadful complications of portal hypertension and can be controlled with vasoconstrictors. Nevertheless, sympathetic tone abnormality and vascular hyporesponsiveness in portal hypertension may impede the haemostatic effects of vasoconstrictors. Propranolol, a ß-blocker binding the G-protein-coupled adrenoceptor, is a portal hypotensive agent. However, whether propranolol influences the collateral vasoresponse is unknown. Portal hypertension was induced by PVL (portal vein ligation) in Sprague-Dawley rats. In an acute study with an in situ perfusion model, the collateral responsiveness to AVP (arginine vasopressin) was evaluated with vehicle, propranolol (10 µmol/l), propranolol plus suramin (100 µmol/l, a G(α) inhibitor) or suramin pre-incubation. G(α) mRNA expression in the splenorenal shunt, the most prominent intra-abdominal collateral vessel, was measured. In the chronic study, rats received DW (distilled water) or propranolol (10 mg x kg(-1) of body weight x day(-1)) for 9 days. Then the concentration-response relationship of AVP and G(α) mRNA expression were assessed. Propranolol pre-incubation elevated the perfusion pressure changes of collaterals in response to AVP, which was inhibited by suramin. The splenorenal shunt G(αq) and G(α11) mRNA expression were enhanced by propranolol. The group treated with propranolol plus suramin had a down-regulation of G(α11) as compared with the propranolol group. Chronic propranolol treatment reduced mean arterial pressure, PP (portal pressure) and the perfusion pressure changes of collaterals to AVP. G(αs) expression was up-regulated. In conclusion, propranolol pre-incubation enhanced the portal-systemic collateral AVP responsiveness in portal hypertensive rats, which was related to G(αq) and G(α11) up-regulation. In contrast, the attenuated AVP responsiveness by chronic propranolol treatment was related to G(αs) up-regulation. The G(α) signalling pathway may be a therapeutic target to control variceal bleeding and PP in portal hypertension.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Arginine Vasopressin/pharmacology , Collateral Circulation/drug effects , Hypertension, Portal/physiopathology , Propranolol/pharmacology , Vasoconstrictor Agents/pharmacology , Adrenergic beta-Antagonists/administration & dosage , Animals , Antihypertensive Agents/pharmacology , Arginine Vasopressin/antagonists & inhibitors , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Interactions , GTP-Binding Protein alpha Subunits/antagonists & inhibitors , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/physiology , Gene Expression Regulation/drug effects , Hemodynamics/drug effects , Liver Circulation/drug effects , Liver Circulation/physiology , Male , Propranolol/administration & dosage , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Suramin/pharmacology
19.
Proc Natl Acad Sci U S A ; 105(24): 8476-81, 2008 Jun 17.
Article in English | MEDLINE | ID: mdl-18541915

ABSTRACT

In mammals, basal currents through G protein-coupled inwardly rectifying K(+) (GIRK) channels are repressed by Galpha(i/o)GDP, and the channels are activated by direct binding of free Gbetagamma subunits released upon stimulation of Galpha(i/o)-coupled receptors. However, essentially all information on G protein regulation of GIRK electrophysiology has been gained on the basis of coexpression studies in heterologous systems. A major advantage of the model organism, Arabidopsis thaliana, is the ease with which knockout mutants can be obtained. We evaluated plants harboring mutations in the sole Arabidopsis Galpha (AtGPA1), Gbeta (AGB1), and Regulator of G protein Signaling (AtRGS1) genes for impacts on ion channel regulation. In guard cells, where K(+) fluxes are integral to cellular regulation of stomatal apertures, inhibition of inward K(+) (K(in)) currents and stomatal opening by the phytohormone abscisic acid (ABA) was equally impaired in Atgpa1 and agb1 single mutants and the Atgpa1 agb1 double mutant. AGB1 overexpressing lines maintained a wild-type phenotype. The Atrgs1 mutation did not affect K(in) current magnitude or ABA sensitivity, but K(in) voltage-activation kinetics were altered. Thus, Arabidopsis cells differ from mammalian cells in that they uniquely use the Galpha subunit or regulation of the heterotrimer to mediate K(in) channel modulation after ligand perception. In contrast, outwardly rectifying (K(out)) currents were unaltered in the mutants, and ABA activation of slow anion currents was conditionally disrupted in conjunction with cytosolic pH clamp. Our studies highlight unique aspects of ion channel regulation by heterotrimeric G proteins and relate these aspects to stomatal aperture control, a key determinant of plant biomass acquisition and drought tolerance.


Subject(s)
Abscisic Acid/physiology , Arabidopsis Proteins/physiology , Arabidopsis/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/agonists , GTP-Binding Protein beta Subunits/physiology , RGS Proteins/physiology , Abscisic Acid/pharmacology , Arabidopsis/drug effects , Arabidopsis/genetics , Arabidopsis Proteins/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/physiology , GTP-Binding Protein beta Subunits/genetics , Mutation , RGS Proteins/genetics
20.
Funct Plant Biol ; 48(10): 1005-1016, 2021 09.
Article in English | MEDLINE | ID: mdl-34167638

ABSTRACT

Oridonin is an important diterpenoid, which plays an important role in plant growth and development. PLDα1 and GPA1 are involved in many biotic or abiotic stresses. In this study, using the seedlings of Arabidopsis thaliana L. wild type (WT), PLDα1 defective mutant (pldα1), GPA1 defective mutant (gpa1) and pldα1/gpa1 double mutant as materials, the effect of stomatal apertures responding to Oridonin and the functions of PLDα1 and GPA1 in this response were investigated. The results showed that 60 µmol·L-1 of Oridonin induced stomatal closure and significantly increased the relative expression levels of GPA1 and PLDα1. Oridonin increased H2O2 accumulation in guard cells by inhibiting the antioxidant enzymes. The increase of H2O2 caused the expression of OST1, which is a positive regulatory gene for stomatal closure. Both PLDα1 and GPA1 were involved in Oridonin-induced stomatal closure and PLDα1 acted downstream of GPA1. The results suggested that Oridonin caused stomatal closure by affecting GPA1 and promoting PLDα1 to produce PA, and further accumulating H2O2 to upregulate gene OST1.


Subject(s)
Arabidopsis Proteins/physiology , Arabidopsis , GTP-Binding Protein alpha Subunits/physiology , Phospholipase D/physiology , Plant Stomata/physiology , Arabidopsis/genetics , Arabidopsis/physiology , Diterpenes, Kaurane , Hydrogen Peroxide/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL