Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Nat Commun ; 11(1): 4798, 2020 09 23.
Article in English | MEDLINE | ID: mdl-32968066

ABSTRACT

Myeloid cells are known mediators of hypertension, but their role in initiating renin-induced hypertension has not been studied. Vitamin D deficiency causes pro-inflammatory macrophage infiltration in metabolic tissues and is linked to renin-mediated hypertension. We tested the hypothesis that impaired vitamin D signaling in macrophages causes hypertension using conditional knockout of the myeloid vitamin D receptor in mice (KODMAC). These mice develop renin-dependent hypertension due to macrophage infiltration of the vasculature and direct activation of renal juxtaglomerular (JG) cell renin production. Induction of endoplasmic reticulum stress in knockout macrophages increases miR-106b-5p secretion, which stimulates JG cell renin production via repression of transcription factors E2f1 and Pde3b. Moreover, in wild-type recipient mice of KODMAC/miR106b-/- bone marrow, knockout of miR-106b-5p prevents the hypertension and JG cell renin production induced by KODMAC macrophages, suggesting myeloid-specific, miR-106b-5p-dependent effects. These findings confirm macrophage miR-106b-5p secretion from impaired vitamin D receptor signaling causes inflammation-induced hypertension.


Subject(s)
Hypertension, Renal/metabolism , Hypertension/metabolism , Macrophages/metabolism , MicroRNAs/metabolism , Nephritis/metabolism , Renin/metabolism , Animals , Bone Marrow , Bone Marrow Transplantation , Disease Models, Animal , E2F1 Transcription Factor/metabolism , Endoplasmic Reticulum Stress , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells , Receptors, Calcitriol , Vitamin D
2.
Neuroscience ; 151(4): 969-82, 2008 Feb 19.
Article in English | MEDLINE | ID: mdl-18248908

ABSTRACT

Members of the regulator of G protein signaling 7 (RGS7) (R7) family and Gbeta5 form obligate heterodimers that are expressed predominantly in the nervous system. R7-Gbeta5 heterodimers are GTPase-activating proteins (GAPs) specific for Gi/o-class Galpha subunits, which mediate phototransduction in retina and the action of many modulatory G protein-coupled receptors (GPCRs) in brain. Here we have focused on the R7-family binding protein (R7BP), a recently identified palmitoylated protein that can bind R7-Gbeta5 complexes and is hypothesized to control the intracellular localization and function of the resultant heterotrimeric complexes. We show that: 1) R7-Gbeta5 complexes are obligate binding partners for R7BP in brain because they co-immunoprecipitate and exhibit similar expression patterns. Furthermore, R7BP and R7 protein accumulation in vivo requires Gbeta5. 2) Expression of R7BP in Neuro2A cells at levels approximating those in brain recruits endogenous RGS7-Gbeta5 complexes to the plasma membrane. 3) R7BP immunoreactivity in brain concentrates in neuronal soma, dendrites, spines or unmyelinated axons, and is absent or low in glia, myelinated axons, or axon terminals. 4) RGS7-Gbeta5-R7BP complexes in brain extracts associate inefficiently with detergent-resistant lipid raft fractions with or without G protein activation. 5) R7BP and Gbeta5 protein levels are upregulated strikingly during the first 2-3 weeks of postnatal brain development. Accordingly, we suggest that R7-Gbeta5-R7BP complexes in the mouse or rat could regulate signaling by modulatory Gi/o-coupled GPCRs in the developing and adult nervous systems.


Subject(s)
Brain/growth & development , Brain/metabolism , GTP-Binding Protein beta Subunits/physiology , Gene Expression Regulation, Developmental/physiology , RGS Proteins/metabolism , Animals , Animals, Newborn , Brain/ultrastructure , Cells, Cultured , GTP-Binding Protein beta Subunits/deficiency , Immunoprecipitation/methods , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Immunoelectron/methods , Neurons/metabolism , Neurons/ultrastructure , Protein Binding/physiology , Rats , Rats, Sprague-Dawley
3.
J Biol Chem ; 276(17): 14195-203, 2001 Apr 27.
Article in English | MEDLINE | ID: mdl-11278586

ABSTRACT

RGS proteins negatively regulate heterotrimeric G proteins at the plasma membrane. RGS2-GFP localizes to the nucleus, plasma membrane, and cytoplasm of HEK293 cells. Expression of activated G(q) increased RGS2 association with the plasma membrane and decreased accumulation in the nucleus, suggesting that signal-induced redistribution may regulate RGS2 function. Thus, we identified and characterized a conserved N-terminal domain in RGS2 that is necessary and sufficient for plasma membrane localization. Mutational and biophysical analyses indicated that this domain is an amphipathic alpha-helix that binds vesicles containing acidic phospholipids. However, the plasma membrane targeting function of the amphipathic helical domain did not appear to be essential for RGS2 to attenuate signaling by activated G(q). Nevertheless, truncation mutants indicated that the N terminus is essential, potentially serving as a scaffold that binds receptors, signaling proteins, or nuclear components. Indeed, the RGS2 N terminus directs nuclear accumulation of GFP. Although RGS2 possesses a nuclear targeting motif, it lacks a nuclear import signal and enters the nucleus by passive diffusion. Nuclear accumulation of RGS2 does not limit its ability to attenuate G(q) signaling, because excluding RGS2 from the nucleus was without effect. RGS2 may nonetheless regulate signaling or other processes in the nucleus.


Subject(s)
RGS Proteins/biosynthesis , RGS Proteins/physiology , Amino Acid Sequence , Animals , Cell Line , Cell Membrane/metabolism , Cell Nucleus/metabolism , Circular Dichroism , Conserved Sequence , Cytoplasm/metabolism , DNA Mutational Analysis , DNA, Complementary/metabolism , Gene Deletion , Green Fluorescent Proteins , Humans , Hydrolysis , Liposomes/metabolism , Luminescent Proteins/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Molecular Sequence Data , Peptide Biosynthesis , Point Mutation , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Sequence Homology, Amino Acid , Signal Transduction
4.
J Mol Cell Cardiol ; 33(2): 209-18, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11162127

ABSTRACT

The intrinsic GTPase activity of Galpha q is low, and RGS proteins which activate GTPase are expressed in the heart; however, their functional relevance in vivo is unknown. Transgenic mice with cardiac-specific overexpression of Galpha q in myocardium exhibit cardiac hypertrophy, enhanced PKC xi membrane translocation, embryonic gene expression, and depressed cardiac contractility. We recently reported that transgenic mice with cardiac-specific expression of RGS4, a Galpha q and Galpha i GTPase activator, exhibit decreased left ventricular hypertrophy and ANF induction in response to pressure overload. To test the hypothesis that RGS4 can act as a Galpha q-specific GTPase activating protein (GAP) in the in vivo heart, dual transgenic Galpha q-40xRGS4 mice were generated to determine if RGS4 co-expression would ameliorate the Galpha q-40 phenotype. At age 4 weeks, percent fractional shortening was normalized in dual transgenic mice as was left ventricular internal dimension and posterior and septal wall thicknesses. PKC xi membrane translocation and ANF and alpha -skeletal actin mRNA levels were also normalized. Compound transgenic mice eventually developed depressed cardiac contractility that was evident by 9 weeks of age. These studies establish for the first time a role for RGS4 as a GAP for Galpha q in the in vivo heart, and demonstrate that its regulated expression can have pathophysiologic consequences.


Subject(s)
Cardiomegaly/genetics , Myocardial Contraction/physiology , RGS Proteins/metabolism , RGS Proteins/physiology , Actins/metabolism , Animals , Atrial Natriuretic Factor/metabolism , Blotting, Northern , Blotting, Western , Cell Nucleus/metabolism , Echocardiography , GTPase-Activating Proteins/metabolism , Isoenzymes/metabolism , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Phenotype , Protein Kinase C/metabolism , Protein Kinase C-epsilon , Protein Transport , RNA, Messenger/metabolism , Time Factors
5.
Development ; 127(13): 2773-84, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10851124

ABSTRACT

RGS family members are GTPase activating proteins (GAPs) that antagonize signaling by heterotrimeric G proteins. Injection of Xenopus embryos with RNA encoding rat RGS4 (rRGS4), a GAP for G(i) and G(q), resulted in shortened trunks and decreased skeletal muscle. This phenotype is nearly identical to the effect of injection of either frzb or dominant negative Xwnt-8. Injection of human RGS2, which selectively deactivates G(q), had similar effects. rRGS4 inhibited the ability of early Xwnt-8 but not Xdsh misexpression to cause axis duplication. This effect is distinct from axin family members that contain RGS-like domains but act downstream of Xdsh. We identified two Xenopus RGS4 homologs, one of which, Xrgs4a, was expressed as a Spemann organizer component. Injection of Xenopus embryos with Xrgs4a also resulted in shortened trunks and decreased skeletal muscle. These results suggest that RGS proteins modulate Xwnt-8 signaling by attenuating the function of a G protein.


Subject(s)
Glycoproteins , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , RGS Proteins/biosynthesis , RGS Proteins/physiology , Signal Transduction , Xenopus/embryology , Zebrafish Proteins , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Cloning, Molecular , Dishevelled Proteins , Embryo, Nonmammalian/metabolism , GTP-Binding Proteins/metabolism , Genes, Dominant , Humans , Immunohistochemistry , In Situ Hybridization , Intracellular Signaling Peptides and Proteins , Mice , Microinjections , Molecular Sequence Data , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , Phenotype , Phosphoproteins/metabolism , Plasmids , Protein Binding , Proteins/metabolism , RGS Proteins/genetics , RGS Proteins/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Time Factors , Wnt Proteins , Xenopus Proteins
6.
Curr Biol ; 10(6): 341-4, 2000 Mar 23.
Article in English | MEDLINE | ID: mdl-10744981

ABSTRACT

Hormones, sensory stimuli, neurotransmitters and chemokines signal by activating G-protein-coupled receptors (GPCRs) [1]. Although GPCRs are thought to function as monomers, they can form SDS-resistant dimers, and coexpression of two non-functional or related GPCRs can result in rescue of activity or modification of function [2-10]. Furthermore, dimerization of peptides corresponding to the third cytoplasmic loops of GPCRs increases their potency as activators of G proteins in vitro [11], and peptide inhibitors of dimerization diminish beta(2)-adrenergic receptor signaling [3]. Nevertheless, it is not known whether GPCRs exist as monomers or oligomers in intact cells and membranes, whether agonist binding regulates monomer-oligomer equilibrium, or whether oligomerization governs GPCR function. Here, we report that the alpha-factor receptor, a GPCR that is the product of the STE2 gene in the yeast Saccharomyces cerevisiae, is oligomeric in intact cells and membranes. Coexpression of receptors tagged with the cyan or yellow fluorescent proteins (CFP or YFP) resulted in efficient fluorescence resonance energy transfer (FRET) due to stable association rather than collisional interaction. Monomer-oligomer equilibrium was unaffected by binding of agonist, antagonist, or G protein heterotrimers. Oligomerization was further demonstrated by rescuing endocytosis-defective receptors with coexpressed wild-type receptors. Dominant-interfering receptor mutants inhibited signaling by interacting with wild-type receptors rather than by sequestering G protein heterotrimers. We suggest that oligomerization is likely to govern GPCR signaling and regulation.


Subject(s)
Fungal Proteins/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Oligopeptides/metabolism , Receptors, Peptide/metabolism , Signal Transduction , Transcription Factors , Endocytosis/physiology , Fungal Proteins/genetics , Oligopeptides/genetics , Receptors, Mating Factor , Receptors, Peptide/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Spectrometry, Fluorescence/methods
7.
Mol Biol Cell ; 11(3): 957-68, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10712512

ABSTRACT

To establish the biological function of thioacylation (palmitoylation), we have studied the heterotrimeric guanine nucleotide-binding protein (G protein) subunits of the pheromone response pathway of Saccharomyces cerevisiae. The yeast G protein gamma subunit (Ste18p) is unusual among G(gamma) subunits because it is farnesylated at cysteine 107 and has the potential to be thioacylated at cysteine 106. Substitution of either cysteine results in a strong signaling defect. In this study, we found that Ste18p is thioacylated at cysteine 106, which depended on prenylation of cysteine 107. Ste18p was targeted to the plasma membrane even in the absence of prenylation or thioacylation. However, G protein activation released prenylation- or thioacylation-defective Ste18p into the cytoplasm. Hence, lipid modifications of the G(gamma) subunit are dispensable for G protein activation by receptor, but they are required to maintain the plasma membrane association of G(betagamma) after receptor-stimulated release from G(alpha). The G protein alpha subunit (Gpa1p) is tandemly modified at its N terminus with amide- and thioester-linked fatty acids. Here we show that Gpa1p was thioacylated in vivo with a mixture of radioactive myristate and palmitate. Mutation of the thioacylation site in Gpa1p resulted in yeast cells that displayed partial activation of the pathway in the absence of pheromone. Thus, dual lipidation motifs on Gpa1p and Ste18p are required for a fully functional pheromone response pathway.


Subject(s)
GTP-Binding Protein alpha Subunits , GTP-Binding Protein gamma Subunits , Heterotrimeric GTP-Binding Proteins/metabolism , Palmitic Acid/metabolism , Pheromones/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/metabolism , Amino Acid Motifs , Animals , Cell Membrane/metabolism , Cells, Cultured , Cysteine/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11 , Green Fluorescent Proteins , Heterotrimeric GTP-Binding Proteins/genetics , Insecta , Luminescent Proteins , Pheromones/chemistry , Saccharomyces cerevisiae/chemistry
8.
J Cell Biol ; 147(4): 845-56, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10562285

ABSTRACT

During the cell cycle of the yeast Saccharomyces cerevisiae, the actin cytoskeleton and cell surface growth are polarized, mediating bud emergence, bud growth, and cytokinesis. We have determined whether p21-activated kinase (PAK)-family kinases regulate cell and actin polarization at one or several points during the yeast cell cycle. Inactivation of the PAK homologues Ste20 and Cla4 at various points in the cell cycle resulted in loss of cell and actin cytoskeletal polarity, but not in depolymerization of F-actin. Loss of PAK function in G1 depolarized the cortical actin cytoskeleton and blocked bud emergence, but allowed isotropic growth and led to defects in septin assembly, indicating that PAKs are effectors of the Rho-guanosine triphosphatase Cdc42. PAK inactivation in S/G2 resulted in depolarized growth of the mother and bud and a loss of actin polarity. Loss of PAK function in mitosis caused a defect in cytokinesis and a failure to polarize the cortical actin cytoskeleton to the mother-bud neck. Cla4-green fluorescent protein localized to sites where the cortical actin cytoskeleton and cell surface growth are polarized, independently of an intact actin cytoskeleton. Thus, PAK family kinases are primary regulators of cell and actin cytoskeletal polarity throughout most or all of the yeast cell cycle. PAK-family kinases in higher organisms may have similar functions.


Subject(s)
Actins/metabolism , Cell Cycle/physiology , Cell Polarity/physiology , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/physiology , Fungal Proteins/metabolism , Genotype , Intracellular Signaling Peptides and Proteins , Kinetics , MAP Kinase Kinase Kinases , Microscopy, Video , Mitosis , Protein Serine-Threonine Kinases/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , cdc42 GTP-Binding Protein/metabolism
9.
J Biol Chem ; 274(48): 34253-9, 1999 Nov 26.
Article in English | MEDLINE | ID: mdl-10567399

ABSTRACT

RGS (regulator of G protein signaling) proteins are GTPase-activating proteins that attenuate signaling by heterotrimeric G proteins. Whether the biological functions of RGS proteins are governed by quantitative differences in GTPase-activating protein activity toward various classes of Galpha subunits and how G protein selectivity is achieved by differences in RGS protein structure are largely unknown. Here we provide evidence indicating that the function of RGS2 is determined in part by differences in potency toward G(q) versus G(i) family members. RGS2 was 5-fold more potent than RGS4 as an inhibitor of G(q)-stimulated phosphoinositide hydrolysis in vivo. In contrast, RGS4 was 8-fold more potent than RGS2 as an inhibitor of G(i)-mediated signaling. RGS2 mutants were identified that display increased potency toward G(i) family members without affecting potency toward G(q). These mutations and the structure of RGS4-G(i)alpha(1) complexes suggest that RGS2-G(i)alpha interaction is unfavorable in part because of the geometry of the switch I binding pocket of RGS2 and a potential interaction between the alpha8-alpha9 loop of RGS2 and alphaA of G(i) class alpha subunits. The results suggest that the function of RGS2 relative to other RGS family members is governed in part by quantitative differences in activity toward different classes of Galpha subunits.


Subject(s)
GTP-Binding Proteins/metabolism , RGS Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Binding Sites/genetics , Binding, Competitive , Carbachol/pharmacology , Cell Line , Humans , Models, Molecular , Molecular Sequence Data , Point Mutation , Protein Structure, Tertiary , RGS Proteins/chemistry , RGS Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction/drug effects
10.
Biochem J ; 343 Pt 1: 77-85, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10493914

ABSTRACT

Chinese hamster ovary (CHO) cells stably expressing alpha(2) adrenergic receptor (alpha(2)AR) were pretreated with cholera toxin (CTX) and then treated with or without PMA. The alpha(2A)AR-mediated inhibition of forskolin-stimulated cAMP accumulation was completely ablated by CTX pretreatment only after additional treatment with PMA. Although the addition of cycloheximide (protein synthesis inhibitor) and H-89 (cAMP dependent protein kinase inhibitor) did not completely counteract the negative regulation, the elevation of cAMP was a primary factor for negative regulation by treatment with CTX and PMA. In contrast with the cAMP response, the inhibition of membrane adenylate cyclase activity and the agonist competition curve were not influenced by treatment with CTX or PMA, suggesting that a cytosolic factor was involved in this negative regulation. The m2-muscarinic-acetylcholine-receptor-mediated inhibition of the forskolin-stimulated accumulation of cAMP was also attenuated by treatment with CTX and PMA. The ablation of alpha(2A)AR-mediated inhibition was not observed when alpha(2A)AR was expressed in Rat2 fibroblast cells, suggesting that this negative regulation is not dependent on the receptor type but is instead a phenomenon common to G(i)-coupled receptors in CHO cells. Reverse-transcriptase-mediated PCR and Northern blot analysis showed that the expression of GOS8/RGS2 mRNA, which is a member of the regulator of G-protein signalling (RGS) group of proteins, was considerably increased by pretreatment with CTX. These results indicate a novel regulatory pathway, whereby a cytosolic factor induced by the elevation of cellular cAMP levels negatively regulates G(i) signalling in a protein-kinase-C-dependent manner.


Subject(s)
GTP-Binding Proteins/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Signal Transduction , Animals , Base Sequence , CHO Cells , Cholera Toxin/pharmacology , Cricetinae , DNA Primers , Enzyme Activation , Protein Binding , Protein Kinase C/metabolism , Rats , Tetradecanoylphorbol Acetate/pharmacology
11.
J Clin Invest ; 104(5): 567-76, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10487771

ABSTRACT

RGS family members are GTPase-activating proteins (GAPs) for heterotrimeric G proteins. There is evidence that altered RGS gene expression may contribute to the pathogenesis of cardiac hypertrophy and failure. We investigated the ability of RGS4 to modulate cardiac physiology using a transgenic mouse model. Overexpression of RGS4 in postnatal ventricular tissue did not affect cardiac morphology or basal cardiac function, but markedly compromised the ability of the heart to adapt to transverse aortic constriction (TAC). In contrast to wild-type mice, the transgenic animals developed significantly reduced ventricular hypertrophy in response to pressure overload and also did not exhibit induction of the cardiac "fetal" gene program. TAC of the transgenic mice caused a rapid decompensation in most animals characterized by left ventricular dilatation, depressed systolic function, and increased postoperative mortality when compared with nontransgenic littermates. These results implicate RGS proteins as a crucial component of the signaling pathway involved in both the cardiac response to acute ventricular pressure overload and the cardiac hypertrophic program.


Subject(s)
Hypertrophy, Left Ventricular/etiology , Proteins/physiology , Ventricular Dysfunction, Left/etiology , Adaptation, Physiological/genetics , Adrenergic alpha-Agonists/pharmacology , Animals , Aorta, Thoracic , Apoptosis , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Constriction , GTPase-Activating Proteins , Gene Expression Regulation , Heart Rate , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/physiopathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Contraction/drug effects , Myocardium/pathology , Myosin Heavy Chains/genetics , Phenylephrine/pharmacology , Pressure , Promoter Regions, Genetic , Proteins/genetics , Signal Transduction , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/physiopathology
12.
Circulation ; 99(3): 441-7, 1999 Jan 26.
Article in English | MEDLINE | ID: mdl-9918533

ABSTRACT

BACKGROUND: RGS family members are GTPase-activating proteins for heterotrimeric Gq and Gi proteins. RGS genes are expressed in heart tissue and in cultured cardiomyocytes. There is evidence that altered RGS gene expression may contribute to the pathogenesis of cardiac hypertrophy and failure. METHODS AND RESULTS: We investigated the ability of RGS proteins to block G-protein signaling in vivo by using a cultured cardiomyocyte transfection system. Endothelin-1, angiotensin II, and phenylephrine signal through Gq or Gi family members and promote the hypertrophy of cardiomyocytes. We found that phenylephrine-mediated and endothelin-1-mediated induction of the atrial natriuretic factor and myosin light chain-2 genes was inhibited in cells that were transfected with RGS4. Phenylephrine-mediated gene induction was not inhibited in cells that were transfected with N128A-RGS4, a point mutant form that lacks GTPase-activating protein activity. Phenylephrine-mediated myofilament organization and cell growth were also blocked in cells by RGS4. CONCLUSIONS: These results demonstrate that RGS protein can inhibit G-protein-mediated signaling in vivo and suggest that increased expression of RGS protein may be a counterregulatory mechanism to inhibit G protein signaling.


Subject(s)
GTP-Binding Proteins/metabolism , Muscle Fibers, Skeletal/enzymology , Proteins/genetics , Proteins/metabolism , RGS Proteins , Signal Transduction/physiology , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/enzymology , Animals , Atrial Natriuretic Factor/pharmacology , Cardiomegaly/metabolism , Cell Size/physiology , Cells, Cultured , Endothelin-1/pharmacology , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation, Enzymologic/physiology , Genes, Reporter , Luciferases/genetics , Muscle Fibers, Skeletal/chemistry , Muscle Fibers, Skeletal/cytology , Myocardium/cytology , Phenylephrine/pharmacology , Point Mutation , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Subcellular Fractions/chemistry , Subcellular Fractions/enzymology , Sympathomimetics/pharmacology , Transcriptional Activation , Transfection
13.
J Biol Chem ; 274(3): 1835-41, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-9880567

ABSTRACT

G protein-coupled receptors that transduce signals for many hormones, neurotransmitters, and inflammatory mediators are internalized and subsequently recycled to the plasma membrane, or down-regulated by targeting to lysosomes for degradation. Here we have characterized yeast alpha-factor receptors tagged with green fluorescent protein (Ste2-GFP) and used them to obtain mutants defective in receptor down-regulation. In wild type cells, Ste2-GFP was functional and localized to the plasma membrane and endocytic compartments. Although GFP was fused to the cytoplasmic tail of the receptor, GFP also accumulated in the lumen of the vacuole, suggesting that the receptor's extracellular and cytoplasmic domains are degraded within the vacuole lumen. Transposon mutagenesis and a visual screen were used to identify mutants displaying aberrant localization of Ste2-GFP. Mutants that accumulated Ste2-GFP in numerous intracellular vesicles carried disruptions of the VAM3/PTH1 gene, which encodes a syntaxin homolog (t-SNARE) required for homotypic vacuole membrane fusion, autophagy and fusion of biosynthetic transport vesicles with the vacuole. We provide evidence that Vam3 is required for the delivery of alpha-factor receptor-ligand complexes to the vacuole. Vam3 homologs in mammalian cells may mediate late steps in the down-regulation and lysosomal degradation pathways of various G protein-coupled receptors.


Subject(s)
Down-Regulation , Fungal Proteins/pharmacology , Receptors, Peptide/genetics , Saccharomyces cerevisiae Proteins , Sequence Homology, Amino Acid , Transcription Factors/genetics , Agglutinins/genetics , Agglutinins/metabolism , Biological Transport , Cell Membrane/metabolism , Endocytosis , Green Fluorescent Proteins , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mating Factor , Membrane Proteins/chemistry , Peptides/genetics , Peptides/metabolism , Pheromones/genetics , Pheromones/metabolism , Qa-SNARE Proteins , Receptors, Mating Factor , Receptors, Peptide/metabolism , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , Vacuoles/metabolism
14.
Curr Biol ; 8(17): 967-70, 1998 Aug 27.
Article in English | MEDLINE | ID: mdl-9742399

ABSTRACT

Cdc42, Rac1 and other Rho-type GTPases regulate gene expression, cell proliferation and cytoskeletal architecture [1,2]. A challenge is to identify the effectors of Cdc42 and Rac1 that mediate these biological responses. Protein kinases of the p21-activated kinase (PAK) family bind activated Rac1 and Cdc42, and switch on mitogen-activated protein (MAP) kinase pathways; however, their roles in regulating actin cytoskeleton organization have not been clearly established [3-5]. Here, we show that mutants of the budding yeast Saccharomyces cerevisiae lacking the PAK homologs Ste20 and Cla4 exhibit actin cytoskeletal defects, in vivo and in vitro, that resemble those of cdc42-1 mutants. Moreover, STE20 overexpression suppresses cdc42-1 growth defects and cytoskeletal defects in vivo, and Ste20 kinase corrects the actin-assembly defects of permeabilized cdc42-1 cells in vitro. Thus, PAKs are effectors of Cdc42 in pathways that regulate the organization of the cortical actin cytoskeleton.


Subject(s)
Actins/metabolism , Cell Cycle Proteins/physiology , Cytoskeleton/metabolism , GTP-Binding Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/cytology , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cell Cycle Proteins/genetics , Cell Polarity , Cytoskeleton/enzymology , GTP-Binding Proteins/genetics , Intracellular Signaling Peptides and Proteins , MAP Kinase Kinase Kinases , Mutation , Protein Serine-Threonine Kinases/genetics , Recombinant Proteins , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Signal Transduction , Temperature , cdc42 GTP-Binding Protein, Saccharomyces cerevisiae
15.
J Biol Chem ; 273(29): 18405-10, 1998 Jul 17.
Article in English | MEDLINE | ID: mdl-9660808

ABSTRACT

The members of a recently identified protein family termed regulators of G-protein signaling (RGS) act as GTPase-activating proteins for certain Galpha subunits in vitro, but their physiological effects in cells are uncertain in the face of similar biochemical activity and overlapping patterns of tissue expression. Consistent with its activity in in vitro GTPase-activating protein assays, RGS4 interacts efficiently with endogenous proteins of the Gi and Gq subclasses of Galpha subunits but not with G12alpha or Gsalpha. Unlike other RGS proteins such as RGS9, RGS-GAIP, and Sst2p, which have been reported to be largely membrane-associated, a majority of cellular RGS4 is found as a soluble protein in the cytoplasm. However, the expression of a GTPase-deficient Gialpha subunit (Gialpha2-Q204L) resulted in the translocation of both wild type RGS4 and a non-Gialpha-binding mutant (L159F) to the plasma membrane. These data suggest that RGS4 may be recruited to the plasma membrane indirectly by G-protein activation and that multiple RGS proteins within a given cell might be differentially localized to determine a physiologic response to a G-protein-linked stimulus.


Subject(s)
Cell Membrane/metabolism , GTP Phosphohydrolases/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go , GTP-Binding Proteins/biosynthesis , Proteins/metabolism , Proto-Oncogene Proteins/biosynthesis , RGS Proteins , Animals , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cytoplasm/metabolism , GTP-Binding Protein alpha Subunit, Gi2 , GTP-Binding Proteins/metabolism , Neurons/metabolism , PC12 Cells , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Rats , Transfection
16.
Genetics ; 149(2): 879-92, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9611199

ABSTRACT

In the yeast Saccharomyces cerevisiae, mating pheromone response is initiated by activation of a G protein- and mitogen-activated protein (MAP) kinase-dependent signaling pathway and attenuated by several mechanisms that promote adaptation or desensitization. To identify genes whose products negatively regulate pheromone signaling, we screened for mutations that suppress the hyperadaptive phenotype of wild-type cells overexpressing signaling-defective G protein beta subunits. This identified recessive mutations in MOT3, which encodes a nuclear protein with two Cys2-His2 Zn fingers. MOT3 was found to be a dosage-dependent inhibitor of pheromone response and pheromone-induced gene expression and to require an intact signaling pathway to exert its effects. Several results suggested that Mot3 attenuates expression of pheromone-responsive genes by mechanisms distinct from those used by the negative transcriptional regulators Cdc36, Cdc39, and Mot2. First, a Mot3-lexA fusion functions as a transcriptional activator. Second, Mot3 is a dose-dependent activator of several genes unrelated to pheromone response, including CYC1, SUC2, and LEU2. Third, insertion of consensus Mot3 binding sites (C/A/T)AGG(T/C)A activates a promoter in a MOT3-dependent manner. These findings, and the fact that consensus binding sites are found in the 5' flanking regions of many yeast genes, suggest that Mot3 is a globally acting transcriptional regulator. We hypothesize that Mot3 regulates expression of factors that attenuate signaling by the pheromone response pathway.


Subject(s)
Gene Expression Regulation, Fungal , Pheromones/physiology , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Signal Transduction/genetics , Transcription Factors/physiology , Zinc Fingers/physiology , Amino Acid Sequence , Binding Sites/genetics , Cloning, Molecular , Consensus Sequence , DNA-Binding Proteins/genetics , Gene Expression Regulation, Fungal/drug effects , Mating Factor , Molecular Sequence Data , Peptides/genetics , Trans-Activators/physiology
17.
J Mol Cell Cardiol ; 30(2): 269-76, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9515003

ABSTRACT

RGS family members are regulatory molecules that act as GTPase activating proteins (GAPs) for G alpha subunits of heterotrimeric G proteins. RGS proteins are able to deactivate G protein subunits of the Gi alpha, Go alpha and Gq alpha subtypes when tested in vitro and in vivo. Although the function of RGS proteins in cardiac physiology is unknown, their ability to deactivate Galpha subunits suggests that they may inhibit the action of muscarinic, alpha-adrenergic, endothelin, and other agonists. To evaluate the role of RGS family members in the regulation of cardiac physiology, we investigated the expression pattern of two RGS genes in normal and diseased rat heart tissue. RGS3 and RGS4 mRNAs and proteins were detected in adult myocardium. RGS3 and RGS4 gene expression was markedly enhanced in two model systems of cardiac hypertrophy: growth factor-stimulated cultured neonatal rat cardiomyocytes and pulmonary artery-banded (PAB) mice. RGS3 and RGS4 mRNA levels were reduced in failing myocardium obtained from SHHF/Mcc-fa(cp) (SHHF) rats. These findings support the hypothesis that RGS gene expression is highly regulated in myocardium and imply that RGS family members play an important role in the regulation of cardiac function.


Subject(s)
GTP Phosphohydrolases/metabolism , GTPase-Activating Proteins , Myocardium/metabolism , Proteins/metabolism , RGS Proteins , Repressor Proteins , Amino Acid Sequence , Animals , Cardiomegaly/etiology , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cells, Cultured , Disease Models, Animal , Enzyme Activation , GTP-Binding Proteins/metabolism , Gene Expression , Heart Failure/genetics , Heart Failure/metabolism , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Mutant Strains
18.
Proc Natl Acad Sci U S A ; 95(10): 5584-9, 1998 May 12.
Article in English | MEDLINE | ID: mdl-9576926

ABSTRACT

RGS4, a mammalian GTPase activating protein for G protein alpha subunits, was identified by its ability to inhibit the pheromone response pathway in Saccharomyces cerevisiae. To define regions of RGS4 necessary for its function in vivo, we assayed mutants for activity in this system. Deletion of the N-terminal 33 aa of RGS4 (Delta1-33) yielded a nonfunctional protein and loss of plasma membrane localization. These functions were restored by addition of a C-terminal membrane-targeting sequence to RGS4 (Delta1-33). Thus, plasma membrane localization is tightly coupled with the ability of RGS4 to inhibit signaling. Fusion of the N-terminal 33 aa of RGS4 to green fluorescent protein was sufficient to localize an otherwise soluble protein to the plasma membrane, defining this N-terminal region as a plasma membrane anchorage domain. RGS4 is palmitoylated, with Cys-2 and Cys-12 the likely sites of palmitoylation. Surprisingly, mutation of the cysteine residues within the N-terminal domain of RGS4 did not affect plasma membrane localization in yeast or the ability to inhibit signaling. Features of the N-terminal domain other than palmitoylation are responsible for the plasma membrane association of RGS4 and its ability to inhibit pheromone response in yeast.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cell Membrane/metabolism , Proteins/physiology , RGS Proteins , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Cysteine/metabolism , Fungal Proteins/physiology , Green Fluorescent Proteins , Luminescent Proteins/metabolism , Molecular Sequence Data , Palmitic Acid/metabolism , Pheromones/physiology , Saccharomyces cerevisiae/enzymology , Signal Transduction , Structure-Activity Relationship
19.
Mol Biol Cell ; 9(4): 885-99, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9529386

ABSTRACT

We have addressed the mechanisms governing the activation and trafficking of G protein-coupled receptors (GPCRs) by analyzing constitutively active mating pheromone receptors (Ste2p and Ste3p) of the yeast Saccharomyces cerevisiae. Substitution of the highly conserved proline residue in transmembrane segment VI of these receptors causes constitutive signaling. This proline residue may facilitate folding of GPCRs into native, inactive conformations, and/or mediate agonist-induced structural changes leading to G protein activation. Constitutive signaling by mutant receptors is suppressed upon coexpression with wild-type, but not G protein coupling-defective, receptors. Wild-type receptors may therefore sequester a limiting pool of G proteins; this apparent "precoupling" of receptors and G proteins could facilitate signal production at sites where cell surface projections form during mating partner discrimination. Finally, rather than being expressed mainly at the cell surface, constitutively active pheromone receptors accumulate in post-endoplasmic reticulum compartments. This is in contrast to other defective membrane proteins, which apparently are targeted by default to the vacuole. We suggest that the quality-control mechanism that retains receptors in post-endoplasmic reticulum compartments may normally allow wild-type receptors to fold into their native, fully inactive conformations before reaching the cell surface. This may ensure that receptors do not trigger a response in the absence of agonist.


Subject(s)
Fungal Proteins/metabolism , GTP-Binding Proteins/metabolism , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled , Receptors, Peptide/genetics , Receptors, Pheromone , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/metabolism , Transcription Factors , Amino Acid Sequence , Binding Sites , Conserved Sequence , Gene Expression Regulation, Fungal , Molecular Sequence Data , Mutation , Pheromones/pharmacology , Proline , Receptors, Cell Surface/metabolism , Receptors, Mating Factor , Receptors, Peptide/metabolism , Saccharomyces cerevisiae/drug effects , Signal Transduction
20.
J Biol Chem ; 273(3): 1529-33, 1998 Jan 16.
Article in English | MEDLINE | ID: mdl-9430692

ABSTRACT

GTP hydrolysis by guanine nucleotide-binding proteins, an essential step in many biological processes, is stimulated by GTPase-activating proteins (GAPs). The mechanisms whereby GAPs stimulate GTP hydrolysis are unknown. We have used mutational, biochemical, and structural data to investigate how RGS4, a GAP for heterotrimeric G protein alpha subunits, stimulates GTP hydrolysis. Many of the residues of RGS4 that interact with Gi alpha 1 are important for GAP activity. Furthermore, optimal GAP activity appears to require the additive effects of interactions along the RGS4-G alpha interface. GAP-defective RGS4 mutants invariably were defective in binding G alpha subunits in their transition state; furthermore, the apparent strengths of GAP and binding defects were correlated. Thus, none of these residues of RGS4, including asparagine 128, the only residue positioned at the active site of Gi alpha 1, is required exclusively for catalyzing GTP hydrolysis. These results and structural data (Tesmer, J. G. G., Berman, D. M., Gilman, A. G., and Sprang, S. R. (1997) Cell 89, 251-261) indicate that RGS4 stimulates GTP hydrolysis primarily by stabilizing the transition state conformation of the switch regions of the G protein, favoring the transition state of the reactants. Therefore, although monomeric and heterotrimeric G proteins are related, their GAPs have evolved distinct mechanisms of action.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Proteins/metabolism , RGS Proteins , Amino Acid Sequence , Amino Acid Substitution , Animals , Asparagine/metabolism , GTP Phosphohydrolases/metabolism , Guanosine Triphosphate/metabolism , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Point Mutation , Protein Binding , Proteins/genetics , Rats , Saccharomyces cerevisiae , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...