Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Mol Cell ; 81(7): 1384-1396.e6, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33636126

ABSTRACT

G proteins play a central role in signal transduction and pharmacology. Signaling is initiated by cell-surface receptors, which promote guanosine triphosphate (GTP) binding and dissociation of Gα from the Gßγ subunits. Structural studies have revealed the molecular basis of subunit association with receptors, RGS proteins, and downstream effectors. In contrast, the mechanism of subunit dissociation is poorly understood. We use cell signaling assays, molecular dynamics (MD) simulations, and biochemistry and structural analyses to identify a conserved network of amino acids that dictates subunit release. In the presence of the terminal phosphate of GTP, a glycine forms a polar network with an arginine and glutamate, putting torsional strain on the subunit binding interface. This "G-R-E motif" secures GTP and, through an allosteric link, discharges the Gßγ dimer. Replacement of network residues prevents subunit dissociation regardless of agonist or GTP binding. These findings reveal the molecular basis of the final committed step of G protein activation.


Subject(s)
Guanosine Triphosphate , Heterotrimeric GTP-Binding Proteins , Molecular Dynamics Simulation , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/enzymology , Allosteric Regulation , Amino Acid Motifs , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , HEK293 Cells , Heterotrimeric GTP-Binding Proteins/chemistry , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism
2.
J Biol Chem ; 299(5): 104617, 2023 05.
Article in English | MEDLINE | ID: mdl-36933807

ABSTRACT

Gene duplications are common in biology and are likely to be an important source of functional diversification and specialization. The yeast Saccharomyces cerevisiae underwent a whole-genome duplication event early in evolution, and a substantial number of duplicated genes have been retained. We identified more than 3500 instances where only one of two paralogous proteins undergoes posttranslational modification despite having retained the same amino acid residue in both. We also developed a web-based search algorithm (CoSMoS.c.) that scores conservation of amino acid sequences based on 1011 wild and domesticated yeast isolates and used it to compare differentially modified pairs of paralogous proteins. We found that the most common modifications-phosphorylation, ubiquitylation, and acylation but not N-glycosylation-occur in regions of high sequence conservation. Such conservation is evident even for ubiquitylation and succinylation, where there is no established 'consensus site' for modification. Differences in phosphorylation were not associated with predicted secondary structure or solvent accessibility but did mirror known differences in kinase-substrate interactions. Thus, differences in posttranslational modification likely result from differences in adjoining amino acids and their interactions with modifying enzymes. By integrating data from large-scale proteomics and genomics analysis, in a system with such substantial genetic diversity, we obtained a more comprehensive understanding of the functional basis for genetic redundancies that have persisted for 100 million years.


Subject(s)
Evolution, Molecular , Protein Processing, Post-Translational , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Gene Duplication , Genomics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
3.
PLoS Genet ; 17(7): e1009640, 2021 07.
Article in English | MEDLINE | ID: mdl-34214075

ABSTRACT

Heterotrimeric G proteins were originally discovered through efforts to understand the effects of hormones, such as glucagon and epinephrine, on glucose metabolism. On the other hand, many cellular metabolites, including glucose, serve as ligands for G protein-coupled receptors. Here we investigate the consequences of glucose-mediated receptor signaling, and in particular the role of a Gα subunit Gpa2 and a non-canonical Gß subunit, known as Asc1 in yeast and RACK1 in animals. Asc1/RACK1 is of particular interest because it has multiple, seemingly unrelated, functions in the cell. The existence of such "moonlighting" operations has complicated the determination of phenotype from genotype. Through a comparative analysis of individual gene deletion mutants, and by integrating transcriptomics and metabolomics measurements, we have determined the relative contributions of the Gα and Gß protein subunits to glucose-initiated processes in yeast. We determined that Gpa2 is primarily involved in regulating carbohydrate metabolism while Asc1 is primarily involved in amino acid metabolism. Both proteins are involved in regulating purine metabolism. Of the two subunits, Gpa2 regulates a greater number of gene transcripts and was particularly important in determining the amplitude of response to glucose addition. We conclude that the two G protein subunits regulate distinct but complementary processes downstream of the glucose-sensing receptor, as well as processes that lead ultimately to changes in cell growth and metabolism.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , GTP-Binding Proteins/metabolism , Glucose/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Adaptor Proteins, Signal Transducing/genetics , Carbohydrate Metabolism , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Proteins/genetics , Gene Expression Profiling , Metabolomics , Mutation , Purines/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction
4.
Mol Cell ; 55(1): 85-96, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24954905

ABSTRACT

G proteins and their associated receptors process information from a variety of environmental stimuli to induce appropriate cellular responses. Generally speaking, each cell in a population responds within defined limits, despite large variation in the expression of protein signaling components. Therefore, we postulated that noise suppression is encoded within the signaling system. Using the yeast mating pathway as a model, we evaluated the ability of a regulator of G protein signaling (RGS) protein to suppress noise. We found that the RGS protein Sst2 limits variability in transcription and morphogenesis in response to pheromone stimulation. While signal suppression is a result of both the GAP (GTPase accelerating) and receptor binding functions of Sst2, noise suppression requires only the GAP activity. Taken together, our findings reveal a hitherto overlooked role of RGS proteins as noise suppressors and demonstrate an ability to uncouple signal and noise in a prototypical stimulus-response pathway.


Subject(s)
GTP-Binding Proteins/metabolism , GTPase-Activating Proteins/physiology , Saccharomyces cerevisiae Proteins/physiology , Cell Polarity , Pheromones/metabolism , Signal Transduction , Transcription, Genetic , cdc42 GTP-Binding Protein, Saccharomyces cerevisiae/metabolism
5.
J Biol Chem ; 295(41): 14189-14202, 2020 10 09.
Article in English | MEDLINE | ID: mdl-32788210

ABSTRACT

Autophagy is a conserved process that recycles cellular contents to promote survival. Although nitrogen limitation is the canonical inducer of autophagy, recent studies have revealed several other nutrients important to this process. In this study, we used a quantitative, high-throughput assay to identify potassium starvation as a new and potent inducer of autophagy in the yeast Saccharomyces cerevisiae We found that potassium-dependent autophagy requires the core pathway kinases Atg1, Atg5, and Vps34, and other components of the phosphatidylinositol 3-kinase complex. Transmission EM revealed abundant autophagosome formation in response to both stimuli. RNA-Seq indicated distinct transcriptional responses: nitrogen affects transport of ions such as copper, whereas potassium targets the organization of other cellular components. Thus, nitrogen and potassium share the ability to influence molecular supply and demand but do so in different ways. Both inputs promote catabolism through bulk autophagy, but result in distinct mechanisms of cellular remodeling and synthesis.


Subject(s)
Autophagy , Potassium/metabolism , Saccharomyces cerevisiae/metabolism , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Class III Phosphatidylinositol 3-Kinases/genetics , Class III Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
6.
Mol Cell ; 51(4): 531-8, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23954348

ABSTRACT

In response to environmental stress, cells often generate pH signals that serve to protect vital cellular components and reprogram gene expression for survival. A major barrier to our understanding of this process has been the identification of signaling proteins that detect changes in intracellular pH. To identify candidate pH sensors, we developed a computer algorithm that searches proteins for networks of proton-binding sidechains. This analysis indicates that Gα subunits, the principal transducers of G protein-coupled receptor (GPCR) signals, are pH sensors. Our structure-based calculations and biophysical investigations reveal that Gα subunits contain networks of pH-sensing sidechains buried between their Ras and helical domains. Further, we show that proton binding induces changes in conformation that promote Gα phosphorylation and suppress receptor-initiated signaling. Together, our computational, biophysical, and cellular analyses reveal an unexpected function for G proteins as mediators of stress-response signaling.


Subject(s)
Algorithms , GTP-Binding Protein alpha Subunits/chemistry , Protons , Receptors, G-Protein-Coupled/metabolism , Second Messenger Systems , Stress, Physiological , GTP-Binding Protein alpha Subunits/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Hydrogen-Ion Concentration , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Signal Transduction , Temperature
7.
J Biol Chem ; 294(49): 18613-18623, 2019 12 06.
Article in English | MEDLINE | ID: mdl-31645437

ABSTRACT

Many sensory and chemical signal inputs are transmitted by intracellular GTP-binding (G) proteins. G proteins make up two major subfamilies: "large" G proteins comprising three subunits and "small" G proteins, such as the proto-oncogene product RAS, which contains a single subunit. Members of both subfamilies are regulated by post-translational modifications, including lipidation, proteolysis, and carboxyl methylation. Emerging studies have shown that these proteins are also modified by ubiquitination. Much of our current understanding of this post-translational modification comes from investigations of the large G-protein α subunit from yeast (Gpa1) and the three RAS isotypes in humans, NRAS, KRAS, and HRAS. Gα undergoes both mono- and polyubiquitination, and these modifications have distinct consequences for determining the sites and mechanisms of its degradation. Genetic and biochemical reconstitution studies have revealed the enzymes and binding partners required for addition and removal of ubiquitin, as well as the delivery and destruction of both the mono- and polyubiquitinated forms of the G protein. Complementary studies of RAS have identified multiple ubiquitination sites, each having distinct consequences for binding to regulatory proteins, shuttling to and from the plasma membrane, and degradation. Here, we review what is currently known about these two well-studied examples, Gpa1 and the human RAS proteins, that have revealed additional mechanisms of signal regulation and dysregulation relevant to human physiology. We also compare and contrast the effects of G-protein ubiquitination with other post-translational modifications of these proteins.


Subject(s)
GTP-Binding Proteins/metabolism , Monomeric GTP-Binding Proteins/metabolism , Animals , Cysteine Endopeptidases/metabolism , GTP-Binding Proteins/genetics , Humans , Monomeric GTP-Binding Proteins/genetics , Proto-Oncogene Mas , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , ras Proteins/metabolism
8.
J Biol Chem ; 294(40): 14717-14731, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31399514

ABSTRACT

The mating pathway in yeast Saccharomyces cerevisiae has long been used to reveal new mechanisms of signal transduction. The pathway comprises a pheromone receptor, a heterotrimeric G protein, and intracellular effectors of morphogenesis and transcription. Polarized cell growth, in the direction of a potential mating partner, is accomplished by the G-protein ßγ subunits and the small G-protein Cdc42. Transcription induction, needed for cell-cell fusion, is mediated by Gßγ and the mitogen-activated protein kinase (MAPK) scaffold protein Ste5. A potential third pathway is initiated by the G-protein α subunit Gpa1. Gpa1 signaling was shown previously to involve the F-box adaptor protein Dia2 and an endosomal effector protein, the phosphatidylinositol 3-kinase Vps34. Vps34 is also required for proper vacuolar sorting and autophagy. Here, using a panel of reporter assays, we demonstrate that mating pheromone stimulates vacuolar targeting of a cytoplasmic reporter protein and that this process depends on Vps34. Through a systematic analysis of F-box deletion mutants, we show that Dia2 is required to sustain pheromone-induced vacuolar targeting. We also found that other F-box proteins selectively regulate morphogenesis (Ydr306, renamed Pfu1) and transcription (Ucc1). These findings point to the existence of a new and distinct branch of the pheromone-signaling pathway, one that likely leads to vacuolar engulfment of cytoplasmic proteins and recycling of cellular contents in preparation for mating.


Subject(s)
Class III Phosphatidylinositol 3-Kinases/genetics , F-Box Proteins/genetics , Genes, Mating Type, Fungal/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Adaptor Proteins, Signal Transducing/genetics , Cell Cycle/genetics , Endosomes/genetics , F-Box Proteins/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein gamma Subunits/chemistry , GTP-Binding Protein gamma Subunits/genetics , Morphogenesis/genetics , Pheromones/genetics , Pheromones/metabolism , Saccharomyces cerevisiae/physiology , Sequence Deletion/genetics , Signal Transduction , Transcription, Genetic , Vacuoles/genetics , Vacuoles/metabolism , cdc42 GTP-Binding Protein/genetics
9.
PLoS Genet ; 13(5): e1006829, 2017 May.
Article in English | MEDLINE | ID: mdl-28558063

ABSTRACT

All cells respond to osmotic stress by implementing molecular signaling events to protect the organism. Failure to properly adapt can lead to pathologies such as hypertension and ischemia-reperfusion injury. Mitogen-activated protein kinases (MAPKs) are activated in response to osmotic stress, as well as by signals acting through G protein-coupled receptors (GPCRs). For proper adaptation, the action of these kinases must be coordinated. To identify second messengers of stress adaptation, we conducted a mass spectrometry-based global metabolomics profiling analysis, quantifying nearly 300 metabolites in the yeast S. cerevisiae. We show that three branched-chain amino acid (BCAA) metabolites increase in response to osmotic stress and require the MAPK Hog1. Ectopic addition of these BCAA derivatives promotes phosphorylation of the G protein α subunit and dampens G protein-dependent transcription, similar to that seen in response to osmotic stress. Conversely, genetic ablation of Hog1 activity or the BCAA-regulatory enzymes leads to diminished phosphorylation of Gα and increased transcription. Taken together, our results define a new class of candidate second messengers that mediate cross talk between osmotic stress and GPCR signaling pathways.


Subject(s)
Amino Acids/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Osmotic Pressure , Saccharomyces cerevisiae/metabolism , Signal Transduction , GTP-Binding Protein alpha Subunits/genetics , Gene Expression Regulation, Fungal , Metabolome , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
10.
J Biol Chem ; 293(7): 2318-2329, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29284676

ABSTRACT

The yeast Saccharomyces cerevisiae employs multiple pathways to coordinate sugar availability and metabolism. Glucose and other sugars are detected by a G protein-coupled receptor, Gpr1, as well as a pair of transporter-like proteins, Rgt2 and Snf3. When glucose is limiting, however, an ATP-driven proton pump (Pma1) is inactivated, leading to a marked decrease in cytoplasmic pH. Here we determine the relative contribution of the two sugar-sensing pathways to pH regulation. Whereas cytoplasmic pH is strongly dependent on glucose abundance and is regulated by both glucose-sensing pathways, ATP is largely unaffected and therefore cannot account for the changes in Pma1 activity. These data suggest that the pH is a second messenger of the glucose-sensing pathways. We show further that different sugars differ in their ability to control cellular acidification, in the manner of inverse agonists. We conclude that the sugar-sensing pathways act via Pma1 to invoke coordinated changes in cellular pH and metabolism. More broadly, our findings support the emerging view that cellular systems have evolved the use of pH signals as a means of adapting to environmental stresses such as those caused by hypoxia, ischemia, and diabetes.


Subject(s)
Cytoplasm/metabolism , Glucose/metabolism , Saccharomyces cerevisiae/metabolism , Cytoplasm/chemistry , Gene Expression Regulation, Fungal , Hydrogen-Ion Concentration , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Proton-Translocating ATPases/genetics , Proton-Translocating ATPases/metabolism , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
11.
Yeast ; 36(8): 495-518, 2019 08.
Article in English | MEDLINE | ID: mdl-31022772

ABSTRACT

The pheromone response pathway of the yeast Saccharomyces cerevisiae is a well-established model for the study of G proteins and mitogen-activated protein kinase (MAPK) cascades. Our longstanding ability to combine sophisticated genetic approaches with established functional assays has provided a thorough understanding of signalling mechanisms and regulation. In this report, we compare new and established methods used to quantify pheromone-dependent MAPK phosphorylation, transcriptional induction, mating morphogenesis, and gradient tracking. These include both single-cell and population-based assays of activity. We describe several technical advances, provide example data for benchmark mutants, highlight important differences between newer and established methodologies, and compare the advantages and disadvantages of each as applied to the yeast model. Quantitative measurements of pathway activity have been used to develop mathematical models and reveal new regulatory mechanisms in yeast. It is our expectation that experimental and computational approaches developed in yeast may eventually be adapted to human systems biology and pharmacology.


Subject(s)
Pheromones/metabolism , Saccharomyces cerevisiae/metabolism , Signal Transduction , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Morphogenesis , Mutation , Pheromones/genetics , Phosphorylation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Single-Cell Analysis , Systems Biology , Transcription, Genetic
12.
Mol Cell ; 38(5): 746-57, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20542006

ABSTRACT

The yeast pheromone pathway consists of a canonical heterotrimeric G protein and MAP kinase cascade. To identify additional signaling components, we systematically evaluated 870 essential genes using a library of repressible-promoter strains. Quantitative transcription-reporter and MAPK activity assays were used to identify strains that exhibit altered pheromone sensitivity. Of the 92 newly identified essential genes required for proper G protein signaling, those involved with protein degradation were most highly represented. Included in this group are members of the Skp, Cullin, F box (SCF) ubiquitin ligase complex. Further genetic and biochemical analysis reveals that SCF(Cdc4) acts together with the Cdc34 ubiquitin-conjugating enzyme at the level of the G protein; promotes degradation of the G protein alpha subunit, Gpa1, in vivo; and catalyzes Gpa1 ubiquitination in vitro. These insights to the G protein signaling network reveal the essential genome as an untapped resource for identifying new components and regulators of signal transduction pathways.


Subject(s)
GTP-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction/genetics , Anaphase-Promoting Complex-Cyclosome , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cluster Analysis , F-Box Proteins/genetics , F-Box Proteins/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation, Fungal , Genome, Fungal , Humans , Phenotype , Pheromones/genetics , Pheromones/metabolism , Promoter Regions, Genetic , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reproducibility of Results , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Substrate Specificity , Ubiquitin-Conjugating Enzymes , Ubiquitin-Protein Ligase Complexes/genetics , Ubiquitin-Protein Ligase Complexes/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
13.
Proc Natl Acad Sci U S A ; 112(18): 5702-7, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25902551

ABSTRACT

Seven-transmembrane receptors (7TMRs) have evolved in prokaryotes and eukaryotes over hundreds of millions of years. Comparative structural analysis suggests that these receptors may share a remote evolutionary origin, despite their lack of sequence similarity. Here we used structure-based computations to compare 221 7TMRs from all domains of life. Unexpectedly, we discovered that these receptors contain spatially conserved networks of buried ionizable groups. In microbial 7TMRs these networks are used to pump ions across the cell membrane in response to light. In animal 7TMRs, which include light- and ligand-activated G protein-coupled receptors (GPCRs), homologous networks were found to be characteristic of activated receptor conformations. These networks are likely relevant to receptor function because they connect the ligand-binding pocket of the receptor to the nucleotide-binding pocket of the G protein. We propose that agonist and G protein binding facilitate the formation of these electrostatic networks and promote important structural rearrangements such as the displacement of transmembrane helix-6. We anticipate that robust classification of activated GPCR structures will aid the identification of ligands that target activated GPCR structural states.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Algorithms , Archaea/metabolism , Archaeal Proteins/metabolism , Bacteria/metabolism , Bacterial Proteins/metabolism , Bacteriorhodopsins/chemistry , Computational Biology , Databases, Protein , Epinephrine/chemistry , Evolution, Molecular , Hydrogen-Ion Concentration , Ions/chemistry , Ligands , Models, Molecular , Neurotransmitter Agents/chemistry , Norepinephrine/chemistry , Opsins/chemistry , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Rhodopsin/chemistry
14.
J Biol Chem ; 291(15): 7786-7, 2016 Apr 08.
Article in English | MEDLINE | ID: mdl-26907688

ABSTRACT

All cells discriminate environmental signals and generate appropriate intracellular responses. Our understanding of these signal transduction mechanisms has benefitted from studies across the kingdoms of life, from fungi and fish to mice and men. This thematic minireview series examines lessons learned from three of the simplest (and best understood) eukaryotic model organisms. The first article focuses on the mating pheromone pathway in budding yeastSaccharomyces cerevisiae The second describes stress-mediated signaling in the roundwormCaenorhabditis elegans The third outlines some of the signaling pathways that dictate growth and development in the fruit flyDrosophila melanogaster Each system has provided unique insights into hormone and neurotransmitter signaling mechanisms, in particular those mediated by the MAPKs. The advances described in these articles will continue to improve our understanding of human physiology and pharmacology.


Subject(s)
Caenorhabditis elegans/physiology , Drosophila melanogaster/growth & development , Saccharomyces cerevisiae/physiology , Signal Transduction , Animals , Caenorhabditis elegans/cytology , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Humans , Mitogen-Activated Protein Kinases/metabolism , Models, Animal , Pheromones/metabolism , Saccharomyces cerevisiae/cytology
15.
Biochemistry ; 55(3): 534-42, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26701741

ABSTRACT

Protein folding is governed by a variety of molecular forces including hydrophobic and ionic interactions. Less is known about the molecular determinants of protein stability. Here we used a recently developed computer algorithm (pHinder) to investigate the relationship between buried charge and thermostability. Our analysis revealed that charge networks in the protein core are generally smaller in thermophilic organisms as compared to mesophilic organisms. To experimentally test whether core network size influences protein thermostability, we purified 18 paralogous Ras superfamily GTPases from yeast and determined their melting temperatures (Tm, or temperature at which 50% of the protein is unfolded). This analysis revealed a wide range of Tm values (35-63 °C) that correlated significantly (R = 0.87) with core network size. These results suggest that thermostability depends in part on the arrangement of ionizable side chains within a protein core. An improved capacity to predict protein thermostability may be useful for selecting the best candidates for protein crystallography, the development of protein-based therapeutics, as well as for industrial enzyme applications.


Subject(s)
Saccharomyces cerevisiae Proteins/chemistry , ras Proteins/chemistry , Cysteine/chemistry , Enzyme Stability , Hot Temperature , Hydrogen-Ion Concentration , Models, Molecular , Protein Denaturation
16.
J Biol Chem ; 290(32): 19469-70, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26126823

ABSTRACT

Over the past half-century, The Journal of Biological Chemistry has been the venue for many landmark publications on the topic of G protein-coupled receptors (GPCRs, also known as seven-transmembrane receptors). The GPCR superfamily in humans is composed of about 800 members, and is the target of about one-third of all pharmaceuticals. Most of these drugs target a very small subset of GPCRs, and do so by mimicking or competing with endogenous hormones and neurotransmitters. This thematic minireview series examines some emerging trends in GPCR drug discovery. The first article describes efforts to systematically interrogate the human "GPCR-ome," including more than 150 uncharacterized "orphan" receptors. The second article describes recent efforts to target alternative receptor binding sites with drugs that act as allosteric modulators of orthosteric ligands. The third article describes how the recent expansion of GPCR structures is providing new opportunities for computer-guided drug discovery. Collectively, these three articles provide a roadmap for the most important emerging trends in GPCR pharmacology.


Subject(s)
Drug Discovery , Receptors, G-Protein-Coupled/metabolism , Small Molecule Libraries/pharmacology , Allosteric Regulation , Computer-Aided Design , Humans , Ligands , Molecular Mimicry , Protein Binding , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry
17.
J Biol Chem ; 290(11): 6679-80, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25605716

ABSTRACT

This thematic series is on the topic of cell signaling from a cell biology perspective, with a particular focus on G proteins. G protein-coupled receptors (GPCRs, also known as seven-transmembrane receptors) are typically found at the cell surface. Upon agonist binding, these receptors will activate a GTP-binding G protein at the cytoplasmic face of the plasma membrane. Additionally, there is growing evidence that G proteins can also be activated by non-receptor binding partners, and they can signal from non-plasma membrane compartments. The production of second messengers at multiple, spatially distinct locations represents a type of signal encoding that has been largely neglected. The first minireview in the series describes biosensors that are being used to monitor G protein signaling events in live cells. The second describes the implementation of antibody-based biosensors to dissect endosome signaling by G proteins and their receptors. The third describes the function of a non-receptor, cytoplasmic activator of G protein signaling, called GIV (Girdin). Collectively, the advances described in these articles provide a deeper understanding and emerging opportunities for new pharmacology.


Subject(s)
GTP-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Biosensing Techniques/methods , Endosomes/metabolism , Humans , Protein Binding
18.
Mol Cell ; 30(5): 649-56, 2008 Jun 06.
Article in English | MEDLINE | ID: mdl-18538663

ABSTRACT

Cell differentiation requires the ability to detect and respond appropriately to a variety of extracellular signals. Here we investigate a differentiation switch induced by changes in the concentration of a single stimulus. Yeast cells exposed to high doses of mating pheromone undergo cell division arrest. Cells at intermediate doses become elongated and divide in the direction of a pheromone gradient (chemotropic growth). Either of the pheromone-responsive MAP kinases, Fus3 and Kss1, promotes cell elongation, but only Fus3 promotes chemotropic growth. Whereas Kss1 is activated rapidly and with a graded dose-response profile, Fus3 is activated slowly and exhibits a steeper dose-response relationship (ultrasensitivity). Fus3 activity requires the scaffold protein Ste5; when binding to Ste5 is abrogated, Fus3 behaves like Kss1, and the cells no longer respond to a gradient or mate efficiently with distant partners. We propose that scaffold proteins serve to modulate the temporal and dose-response behavior of the MAP kinase.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/enzymology , Cell Differentiation/drug effects , Enzyme Activation , Pheromones/pharmacology , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/growth & development
19.
J Biol Chem ; 289(21): 15052-63, 2014 May 23.
Article in English | MEDLINE | ID: mdl-24722989

ABSTRACT

Heterotrimeric G proteins are well known to transmit signals from cell surface receptors to intracellular effector proteins. There is growing appreciation that G proteins are also present at endomembrane compartments, where they can potentially interact with a distinct set of signaling proteins. Here, we examine the cellular trafficking function of the G protein α subunit in yeast, Gpa1. Gpa1 contains a unique 109-amino acid insert within the α-helical domain that undergoes a variety of posttranslational modifications. Among these is monoubiquitination, catalyzed by the NEDD4 family ubiquitin ligase Rsp5. Using a newly optimized method for G protein purification together with biophysical measures of structure and function, we show that the ubiquitination domain does not influence enzyme activity. By screening a panel of 39 gene deletion mutants, each lacking a different ubiquitin binding domain protein, we identify seven that are necessary to deliver Gpa1 to the vacuole compartment including four proteins (Ede1, Bul1, Ddi1, and Rup1) previously not known to be involved in this process. Finally, we show that proper endocytosis of the G protein is needed for sustained cellular morphogenesis and mating in response to pheromone stimulation. We conclude that a cascade of ubiquitin-binding proteins serves to deliver the G protein to its final destination within the cell. In this instance and in contrast to the previously characterized visual system, endocytosis from the plasma membrane is needed for proper signal transduction rather than for signal desensitization.


Subject(s)
Carrier Proteins/metabolism , Endocytosis , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Ubiquitin/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Carrier Proteins/genetics , Crosses, Genetic , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunoblotting , Microscopy, Confocal , Models, Molecular , Molecular Sequence Data , Morphogenesis , Mutation , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Sequence Homology, Amino Acid , Time-Lapse Imaging , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
20.
PLoS Genet ; 8(1): e1002437, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22242015

ABSTRACT

All eukaryotes have the ability to detect and respond to environmental and hormonal signals. In many cases these signals evoke cellular changes that are incompatible and must therefore be orchestrated by the responding cell. In the yeast Saccharomyces cerevisiae, hyperosmotic stress and mating pheromones initiate signaling cascades that each terminate with a MAP kinase, Hog1 and Fus3, respectively. Despite sharing components, these pathways are initiated by distinct inputs and produce distinct cellular behaviors. To understand how these responses are coordinated, we monitored the pheromone response during hyperosmotic conditions. We show that hyperosmotic stress limits pheromone signaling in at least three ways. First, stress delays the expression of pheromone-induced genes. Second, stress promotes the phosphorylation of a protein kinase, Rck2, and thereby inhibits pheromone-induced protein translation. Third, stress promotes the phosphorylation of a shared pathway component, Ste50, and thereby dampens pheromone-induced MAPK activation. Whereas all three mechanisms are dependent on an increase in osmolarity, only the phosphorylation events require Hog1. These findings reveal how an environmental stress signal is able to postpone responsiveness to a competing differentiation signal, by acting on multiple pathway components, in a coordinated manner.


Subject(s)
MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinases/metabolism , Osmolar Concentration , Pheromones/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Gene Expression Regulation, Fungal , Mitogen-Activated Protein Kinases/genetics , Pheromones/genetics , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae/physiology , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL