Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Oncogene ; 34(24): 3095-106, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25109332

ABSTRACT

Protein geranylgeranylation (GGylation) is an important biochemical process for many cellular signaling molecules. Previous studies have shown that GGylation is essential for cell survival in many types of cancer. However, the molecular mechanism mediating the cell survival effect remains elusive. In this report, we show that the Hippo pathway mediates GGylation-dependent cell proliferation and migration in breast cancer cells. Blockade of GGylation enhanced phosphorylation of Mst1/2 and Lats1, and inhibited YAP and TAZ activity and the Hippo-YAP/TAZ pathway-dependent transcription. The effect of GGylation blockade on inhibition of breast cancer cell proliferation and migration is dependent on the Hippo-YAP/TAZ signaling, in which YAP appears to regulate cell proliferation and TAZ to regulate cell migration. Furthermore, GGylation-dependent cell proliferation is correlated with the activity of YAP/TAZ in breast cancer cells. Finally, Gγ and RhoA are the GGylated proteins that may transduce GGylation signals to the Hippo-YAP/TAZ pathway. Taken together, our studies have demonstrated that the Hippo-YAP/TAZ pathway is essential for GGylation-dependent cancer cell proliferation and migration.


Subject(s)
Breast Neoplasms/pathology , Cell Movement , Cell Proliferation , Prenylation/physiology , Protein Serine-Threonine Kinases/metabolism , Atorvastatin , Benzamides/pharmacology , Breast Neoplasms/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , HEK293 Cells , Heptanoic Acids/pharmacology , Hippo Signaling Pathway , Humans , MCF-7 Cells , Protein Processing, Post-Translational/drug effects , Pyrroles/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Tumor Cells, Cultured
2.
J Biol Chem ; 276(42): 39386-93, 2001 Oct 19.
Article in English | MEDLINE | ID: mdl-11500503

ABSTRACT

The D(1) dopamine receptor, G protein gamma(7) subunit, and adenylylcyclase are selectively expressed in the striatum, suggesting their potential interaction in a common signaling pathway. To evaluate this possibility, a ribozyme strategy was used to suppress the expression of the G protein gamma(7) subunit in HEK 293 cells stably expressing the human D(1) dopamine receptor. Prior in vitro analysis revealed that the gamma(7) ribozyme possessed cleavage activity directed exclusively toward the gamma(7) RNA transcript (Wang, Q., Mullah, B., Hansen, C., Asundi, J., and Robishaw, J. D. (1997) J. Biol. Chem. 272, 26040-26048). In vivo analysis of cells transfected with the gamma(7) ribozyme showed a specific reduction in the expression of the gamma(7) protein. Coincident with the loss of the gamma(7) protein, there was a noticeable reduction in the expression of the beta(1) protein, confirming their interaction in these cells. Finally, functional analysis of ribozyme-mediated suppression of the beta(1) and gamma(7) proteins revealed a significant attenuation of SKF81297-stimulated adenylylcyclase activity in D(1) dopamine receptor-expressing cells. By contrast, ribozyme-mediated suppression of the beta(1) and gamma(7) proteins showed no reduction of SKF81297-stimulated adenylylcyclase activity in D(5) dopamine receptor-expressing cells. Taken together, these data indicate that the structurally related D(1) and D(5) dopamine receptor subtypes utilize G proteins composed of distinct betagamma subunits to stimulate adenylylcyclase in HEK 293 cells. Underscoring the physiological relevance of these findings, single cell reverse transcriptase-polymerase chain reaction analysis revealed that the D(1) dopamine receptor and the G protein gamma(7) subunit are coordinately expressed in substance P containing neurons in rat striatum, suggesting that the G protein gamma(7) subunit may be a new target for drugs to selectively alter dopaminergic signaling within the brain.


Subject(s)
Adenylyl Cyclases/metabolism , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/metabolism , Receptors, Dopamine D1/metabolism , Base Sequence , Cell Line , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Humans , Kinetics , Molecular Sequence Data , Neurons/metabolism , Polymerase Chain Reaction , Protein Binding , RNA, Catalytic/metabolism , Receptors, Dopamine D5 , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Structure-Activity Relationship , Transfection
3.
Oncogene ; 20(13): 1653-60, 2001 Mar 26.
Article in English | MEDLINE | ID: mdl-11313913

ABSTRACT

Heterotrimeric G-proteins are components of the signal transduction pathways for the soluble and cell-contact signals that regulate normal growth and differentiation. There is now a greater appreciation of the role of the Gbetagamma-dimer in the regulation of a variety of intracellular effectors, including ion channels, adenylyl cyclase, and phospholipase Cbeta. In many cases, Gbetagamma-dimers are required for the activation of mitogen activated protein kinase (MAPK) pathways that promote cellular proliferation, although the underlying mechanisms have yet to be fully elucidated. Activation of phosphotidylinositol-3-kinase (PI3K) is a critical step in the intracellular transduction of survival signals. Gbetagamma-dimers directly activate PI3Kgamma as well as the more widely distributed PI3Kbeta. The activation of PI3Kgamma by Gbetagamma-dimers likely involves direct binding of specific Gbetagamma-dimers to both subunits of PI3Kgamma. Thus, Gbetagamma-dimers transmit signals from numerous receptors to a variety of intracellular effectors in distinct cellular contexts. Five distinct Gbeta-subunits and 12 distinct Ggamma-subunits have been identified. New experimental approaches are needed to elucidate the specific roles of individual Gbetagamma-dimers in the pathways that transduce signals for proliferation and survival.


Subject(s)
Cell Transformation, Neoplastic , Signal Transduction , Dimerization , Heterotrimeric GTP-Binding Proteins , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Processing, Post-Translational , Protein Subunits , Receptors, Cell Surface/metabolism
4.
Am J Hum Genet ; 67(5): 1314-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11022010

ABSTRACT

Myoclonus-dystonia (M-D) is an autosomal dominant disorder characterized by myoclonic and dystonic muscle contractions that are often responsive to alcohol. The dopamine D2 receptor gene (DRD2) on chromosome 11q has been implicated in one family with this syndrome, and linkage to a 28-cM region on 7q has been reported in another. We performed genetic studies, using eight additional families with M-D, to assess these two loci. No evidence for linkage was found for 11q markers. However, all eight of these families showed linkage to chromosome 7 markers, with a combined multipoint LOD score of 11.71. Recombination events in the families define the disease gene within a 14-cM interval flanked by D7S2212 and D7S821. These data provide evidence for a major locus for M-D on chromosome 7q21.


Subject(s)
Chromosomes, Human, Pair 7/genetics , Dystonia/genetics , Genetic Linkage/genetics , Myoclonus/genetics , Chromosome Mapping , Chromosomes, Human, Pair 11/genetics , Exons/genetics , Female , Genetic Markers/genetics , Humans , Lod Score , Male , Pedigree , Receptors, Dopamine D2/genetics , Recombination, Genetic/genetics , Software
5.
Exp Cell Res ; 257(2): 310-9, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10837145

ABSTRACT

The mammalian gamma subunit family consists of a minimum of 12 members. Analysis of the amino acid sequence conservation suggests that the gamma subunit family can be divided into three distinct subclasses. The division of the gamma subunit family into these classes is based not only on amino acid homology, but also to some extent on functional similarities. In the present study, two new members of the gamma subunit family, the gamma(11) and gamma(14) subunits, are identified and characterized in terms of their expression and function. The gamma(11) and gamma(14) subunits are most closely related to the gamma(1) subunit and share similar biochemical properties, suggesting their inclusion in class I. However, despite their close phylogenetic relationship and similar biochemical properties, the gamma(1), gamma(11), and gamma(14) subunits exhibit very distinct expression patterns, suggesting that class I should be further subdivided and that the signaling functions of each subgroup are distinct. In this regard, the gamma(11) and gamma(14) subunits represent a new subgroup of farnesylated gamma subunits that are expressed outside the retina and have functions other than phototransduction.


Subject(s)
GTP-Binding Proteins/classification , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Cattle , Cell Line , Cloning, Molecular , DNA, Complementary , GTP-Binding Proteins/genetics , GTP-Binding Proteins/physiology , Gene Expression , Humans , Molecular Sequence Data , RNA, Messenger
6.
J Biol Chem ; 275(3): 2087-97, 2000 Jan 21.
Article in English | MEDLINE | ID: mdl-10636913

ABSTRACT

Activation of alpha(1)-adrenergic receptors influences both the contractile activity and the growth potential of cardiac myocytes. However, the signaling pathways linking activation of specific alpha(1)-adrenergic receptor (AR) subtypes to these physiological responses remain controversial. In the present study, a molecular approach was used to identify conclusively the signaling pathways activated in response to the individual alpha(1A)- and alpha(1B)-AR subtypes in cardiac myocytes. For this purpose, a mutant alpha(1a)-AR subtype (alpha(1a)-S(290/293)-AR) was constructed based on analogy to the previously described constitutively active mutant alpha(1b)-AR subtype (alpha(1b)-S(288-294)-AR). The mutant alpha(1a)-S(290/293)-AR subtype displayed constitutive activity based on four criteria. To introduce the constitutively active alpha(1)-AR subtypes into cardiac myocytes, recombinant Sindbis viruses encoding either the alpha(1a)-S(290/293)-AR or alpha(1b)-S(288-294)-AR subtype were used to infect the whole cell population with >90% efficiency, thereby allowing the biochemical activities of the various signaling pathways to be measured. When expressed at comparable levels, the alpha(1a)-S(290/293)-AR subtype exhibited a significantly elevated basal level as well as agonist-stimulated level of inositol phosphate accumulation, coincident with activation of atrial natriuretic factor-luciferase gene expression. By contrast, the alpha(1b)-S(288-294)-AR subtype displayed a markedly increased serum response element-luciferase gene expression but no activation of atrial natriuretic factor-luciferase gene expression. Taken together, this study provides the first molecular evidence for coupling of the alpha(1a)-AR and the alpha(1b)-AR subtypes to different signaling pathways in cardiac myocytes.


Subject(s)
Myocardium/metabolism , Receptors, Adrenergic, alpha-1/genetics , Receptors, Adrenergic, alpha-1/metabolism , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Animals , Binding, Competitive , COS Cells , Cattle , Cells, Cultured , Enzyme Activation , Genes, Reporter , Hydrolysis , Luciferases/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutagenesis, Site-Directed , Phenylephrine/pharmacology , Phosphatidylinositols/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Transfection
7.
Mol Cell Biochem ; 214(1-2): 111-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11195782

ABSTRACT

Activation of alpha-1 adrenergic receptors in the heart has been shown to result in increased contractile activity, cardiac fetal gene re-expression, and myocyte hypertrophy. Three alpha-1 adrenergic receptors have been identified through molecular cloning. Due to the limited selectivities of the currently available alpha-1 adrenergic receptor antagonists, the signaling pathways activated by specific subtypes in the heart remain unresolved. To resolve this dilemma, we have used a molecular approach to identify the signaling pathways and downstream genes that are engaged in response to activation of individual alpha-1 adrenergic subtypes in cardiac cells. We have transfected constitutively active alpha-1 adrenergic receptors (alpha1a-S290/293-AR [1] or the alpha1b-S288/294-AR [2]) subtypes into the cardiac murine myocyte cell line (HL-1) and studied the signal transduction pathway(s) and cardiac gene(s) activated by them. In this study, we demonstrate that the alpha1a-S290/293 -AR [1] subtype preferentially couples to cardiac-specific atrial natriuretic factor (ANF) gene expression, while the alpha1b-S288/294-AR preferentially couples to activation of mitogen-activated protein kinase (MAPK), Ets-like transcription factor-1 (Elk1) and serum response element (SRE) signaling pathways. Endogenous alpha-1 adrenergic receptors are expressed, and stimulate phosphatidylinositol-hydrolysis upon activation with the alpha-1 agonist, phenylephrine.


Subject(s)
Myocardium/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Animals , Atrial Natriuretic Factor/genetics , Cell Line , GTP-Binding Proteins/metabolism , Gene Expression , Heart Atria/cytology , Heart Atria/metabolism , Luciferases/genetics , Mice , Mitogen-Activated Protein Kinases/metabolism , Myocardium/cytology , Phosphatidylinositols/metabolism , Receptors, Adrenergic, alpha-1/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Transfection
8.
J Biol Chem ; 274(24): 17365-71, 1999 Jun 11.
Article in English | MEDLINE | ID: mdl-10358098

ABSTRACT

The complex role that the heterotrimeric G proteins play in signaling pathways has become increasingly apparent with the cloning of countless numbers of receptors, G proteins, and effectors. However, in most cases, the specific combinations of alpha and betagamma subunits comprising the G proteins that participate in the most common signaling pathways, such as beta-adrenergic regulation of adenylyl cyclase activity, are not known. The extent of this problem is evident in the fact that the identities of the betagamma subunits that combine with the alpha subunit of Gs are only now being elucidated almost 20 years after its initial purification. In a previous study, we described the first use of a ribozyme strategy to suppress specifically the expression of the gamma7 subunit of the G proteins, thereby identifying a specific role of this protein in coupling the beta-adrenergic receptor to stimulation of adenylyl cyclase activity in HEK 293 cells. In the present study, we explored the potential utility of a ribozyme approach directed against the gamma7 subunit to identify functional associations with a particular beta and alphas subunit of the G protein in this signaling pathway. Accordingly, HEK 293 cells were transfected with a ribozyme directed against the gamma7 subunit, and the effects of this manipulation on levels of the beta and alphas subunits were determined by immunoblot analysis. Among the five beta alphas subunits detected in these cells, only the beta1 subunit was coordinately reduced following treatment with the ribozyme directed against the gamma7 subunit, thereby demonstrating a functional association between the beta1 and gamma7 subunits. The mechanism for coordinate suppression of the beta1 subunit was due to a striking change in the half-life of the beta1 monomer versus the beta1 heterodimer complexed with the gamma7 subunit. Neither the 52- nor 45-kDa subunits were suppressed following treatment with the ribozyme directed against the gamma7 subunit, thereby providing insights into the assembly of the Gs heterotrimer. Taken together, these data show the utility of a ribozyme approach to identify the role of not only the gamma subunits but also the beta subunits of the G proteins in signaling pathways.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/metabolism , GTP-Binding Protein beta Subunits , GTP-Binding Protein gamma Subunits , Heterotrimeric GTP-Binding Proteins , Receptors, Adrenergic, beta/metabolism , Cell Fractionation , GTP-Binding Protein alpha Subunits, Gs/genetics , GTP-Binding Proteins/metabolism , Half-Life , Humans , Kinetics , Precipitin Tests , Protein Binding , Protein Conformation , RNA/metabolism , RNA, Catalytic/genetics , RNA, Catalytic/metabolism , RNA, Messenger/metabolism , Signal Transduction
9.
J Biol Chem ; 274(19): 13525-33, 1999 May 07.
Article in English | MEDLINE | ID: mdl-10224121

ABSTRACT

In view of the expanding roles of the betagamma subunits of the G proteins in signaling, the possibility was raised that the rich diversity of betagamma subunit combinations might contribute to the specificity of signaling at the level of the receptor. To test this possibility, Sf9 cell membranes expressing the recombinant alpha2A-adrenergic receptor were used to assess the contribution of the betagamma subunit composition. Reconstituted coupling between the receptor and heterotrimeric Gi protein was assayed by high affinity, guanine nucleotide-sensitive binding of the alpha2-adrenergic agonist, [3H]UK-14,304. Supporting this hypothesis, the present study showed clear differences in the abilities of the various betagamma dimers, including those containing the beta3 subtype and the newly described gamma4, gamma10, and gamma11 subtypes, to promote interaction of the same alphai subunit with the alpha2A-adrenergic receptor.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Adrenergic alpha-2 Receptor Agonists , Adrenergic alpha-Agonists/pharmacology , Amino Acid Sequence , Animals , Biopolymers , Brimonidine Tartrate , Cell Membrane/metabolism , Molecular Sequence Data , Protein Binding , Quinoxalines/pharmacology , Spodoptera
10.
J Gen Physiol ; 112(2): 199-210, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9689027

ABSTRACT

The membrane-delimited activation of muscarinic K+ channels by G protein beta gamma subunits plays a prominent role in the inhibitory synaptic transmission in the heart. These channels are thought to be heterotetramers comprised of two homologous subunits, GIRK1 and CIR, both members of the family of inwardly rectifying K+ channels. Here, we demonstrate that muscarinic K+ channels in neonatal rat atrial myocytes exhibit four distinct gating modes. In intact myocytes, after muscarinic receptor activation, the different gating modes were distinguished by differences in both the frequency of channel opening and the mean open time of the channel, which accounted for a 76-fold increase in channel open probability from mode 1 to mode 4. Because of the tetrameric architecture of the channel, the hypothesis that each of the four gating modes reflects binding of a different number of Gbeta gamma subunits to the channel was tested, using recombinant Gbeta1 gamma5. Gbeta1 gamma5 was able to control the equilibrium between the four gating modes of the channel in a manner consistent with binding of Gbeta gamma to four equivalent and independent sites in the protein complex. Surprisingly, however, Gbeta1 gamma5 lacked the ability to stabilize the long open state of the channel that is responsible for the augmentation of the mean open time in modes 3 and 4 after muscarinic receptor stimulation. The modal regulation of muscarinic K+ channel gating by Gbeta gamma provides the atrial cells with at least two major advantages: the ability to filter out small inputs from multiple membrane receptors and yet the ability to create the gradients of information necessary to control the heart rate with great precision.


Subject(s)
Ion Channel Gating/physiology , Myocardium/chemistry , Potassium Channels/physiology , Receptors, Muscarinic/physiology , Adenosine/pharmacology , Animals , Animals, Newborn , Cells, Cultured , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/physiology , Guanosine Triphosphate/pharmacology , Heart Atria/chemistry , Heart Atria/cytology , Ion Channel Gating/drug effects , Muscle Fibers, Skeletal/chemistry , Myocardium/cytology , Patch-Clamp Techniques , Potassium Channels/chemistry , Rats , Rats, Sprague-Dawley , Receptors, Muscarinic/chemistry , Signal Transduction/drug effects , Signal Transduction/physiology , Spodoptera
11.
J Biol Chem ; 272(41): 26040-8, 1997 Oct 10.
Article in English | MEDLINE | ID: mdl-9325341

ABSTRACT

Human HEK 293 cells present a simple and tractable system to directly test the hypothesis that the G protein gamma subunits contribute to the specificity of receptor signaling pathways in vivo. To begin to elucidate the functions of the individual gamma subunits in these cells, a ribozyme strategy was used to specifically inactivate the mRNA encoding the gamma7 subunit. A phosphorothioated DNA-RNA chimeric hammerhead ribozyme was constructed and analyzed for specificity toward the targeted gamma7 subunit. In vitro cleavage analysis of this ribozyme revealed a highly efficient cleavage activity directed exclusively toward the gamma7 RNA transcript. In particular, this ribozyme did not result in cleavage of the gamma12 RNA transcript, which is 75% identical to the gamma7 RNA transcript. Using a transient transfection assay, in vivo analysis of this ribozyme showed a specific reduction in both the mRNA and protein expression of the gamma7 subunit in HEK 293 cells. Coincident with this loss in gamma7 subunit, there was a specific reduction in the protein expression of the beta1 subunit, suggesting that the beta1 and gamma7 subunits may functionally interact to form a betagamma dimer in vivo. Functional analysis of the consequences of ribozyme-mediated suppression of the gamma7 subunit expression indicated that it was associated with significant attenuation of isoproterenol-, but not prostaglandin E1-, stimulated adenylylcyclase activity. Suppression of the gamma7 subunit expression had no effect on carbachol- and ATP-mediated stimulation of phosphatidylinositol turnover. Taken together, these results not only indicate the feasibility of using the ribozyme technology to determine the roles of individual gamma subunits in receptor-G protein-effector pathways in vivo, but they point to a specific role of the gamma7 subunit in the regulation of adenylylcyclase activity in response to isoproterenol.


Subject(s)
Adenylyl Cyclases/metabolism , GTP-Binding Proteins/metabolism , Hormones/metabolism , RNA, Catalytic/metabolism , Adenosine Triphosphate/pharmacology , Adrenergic beta-Agonists/pharmacology , Alprostadil/pharmacology , Base Sequence , Binding Sites , Carbachol/pharmacology , Catalysis , Cell Line , Feasibility Studies , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/genetics , Humans , Isoproterenol/pharmacology , Molecular Sequence Data , Molecular Weight , Phosphatidylinositols/metabolism , Protein Conformation , RNA, Messenger/metabolism , Signal Transduction/drug effects
12.
Biochemistry ; 36(31): 9523-31, 1997 Aug 05.
Article in English | MEDLINE | ID: mdl-9235998

ABSTRACT

Mechanisms contributing to altered heterotrimeric G-protein expression and subsequent signaling events during cholesterol accretion have been unexplored. The influence of cholesterol enrichment on G-protein expression was examined in cultured smooth muscle cells that resemble human atherosclerotic cells by exposure to cationized LDL (cLDL). cLDL, which increases cellular free and esterified cholesterol 2-fold and 10-fold, respectively, reduced the cell membrane content of Galphai-1, Galphai-2, Galphai-3, Gq/11, and Galphas. The following evidence supports the premise that the mechanism by which this occurs is due to reduced isoprenylation of the Ggamma-subunit. First, the inhibitory effect of cholesterol enrichment on the membrane content of Galphai subunits was found to be post-transcriptional, since the mRNA steady-state levels of Galphai(1-3) were unchanged following cholesterol enrichment. Second, the membrane expression of alpha and beta subunits was mimicked by cholesterol and 17-ketocholesterol, both of which inhibit HMG-CoA reductase. Third, inhibition of Galphai and Gbeta expression in cholesterol-enriched cells was overcome by mevalonate, the immediate product of HMG-CoA reductase. Fourth, pulse-chase experiments revealed that cholesterol enrichment did not reduce the degradation rate of membrane-associated Galphai subunits. Fifth, cholesterol enrichment also reduced membrane expression of Ggamma-5, Ggamma-7upper; these gamma subunits are responsible for trafficking of the heterotrimeric G-protein complex to the cell membrane as a result of HMG-CoA reductase-dependent post-translational lipid modification (geranylgeranylation) and subsequent membrane association. Cholesterol enrichment did not alter expression of G-gamma-5 mRNA, as assessed by reverse transcriptase polymerase chain reaction, supporting a post-transcriptional defect in Ggamma subunit expression. Fifth, cholesterol enrichment also reduced the membrane content of p21ras (a low molecular weight G-protein requiring farnesylation for membrane targeting) but did not alter the membrane content of the two proteins that do not require isoprenylation for membrane association&sbd;PDGF-receptor or p60-src. Reduced G-protein content in cholesterol-laden cells was reflected by reduced G-protein-mediated signaling events, including ATP-induced GTPase activity, thrombin-induced inhibition of cyclic AMP accumulation, and MAP kinase activity. Collectively, these results demonstrate that cholesterol enrichment reduces G-protein expression and signaling by inhibiting isoprenylation and subsequent membrane targeting. These results provide a molecular basis for altered G-protein-mediated cell signaling processes in cholesterol-enriched cells.


Subject(s)
Cholesterol/metabolism , GTP-Binding Proteins/metabolism , Mitogen-Activated Protein Kinases , Muscle, Smooth, Vascular/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Membrane/metabolism , Cells, Cultured , Humans , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Oncogene Protein pp60(v-src)/metabolism , Oxygen/metabolism , Protein Prenylation , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/metabolism , Rabbits , Rats , Receptors, Platelet-Derived Growth Factor/metabolism , Steroids/metabolism
13.
Proc Natl Acad Sci U S A ; 94(6): 2180-5, 1997 Mar 18.
Article in English | MEDLINE | ID: mdl-9122168

ABSTRACT

The G protein-coupled receptor (GPCR) kinases (GRKs) phosphorylate and desensitize agonist-occupied GPCRs. GRK2-mediated receptor phosphorylation is preceded by the agonist-dependent membrane association of this enzyme. Previous in vitro studies with purified proteins have suggested that this translocation may be mediated by the recruitment of GRK2 to the plasma membrane by its interaction with the free betagamma subunits of heterotrimeric G proteins (G betagamma). Here we demonstrate that this mechanism operates in intact cells and that specificity is imparted by the selective interaction of discrete pools of G betagamma with receptors and GRKs. Treatment of Cos-7 cells transiently overexpressing GRK2 with a beta-receptor agonist promotes a 3-fold increase in plasma membrane-associated GRK2. This translocation of GRK2 is inhibited by the carboxyl terminus of GRK2, a known G betagamma sequestrant. Furthermore, in cells overexpressing both GRK2 and G beta1 gamma2, activation of lysophosphatidic acid receptors leads to the rapid and transient formation of a GRK/G betagamma complex. That G betagamma specificity exists at the level of the GPCR and the GRK is indicated by the observation that a GRK2/G betagamma complex is formed after agonist occupancy of the lysophosphatidic acid and beta-adrenergic but not thrombin receptors. In contrast to GRK2, GRK3 forms a G betagamma complex after stimulation of all three GPCRs. This G betagamma binding specificity of the GRKs is also reflected at the level of the purified proteins. Thus the GRK2 carboxyl terminus binds G beta1 and G beta2 but not G beta3, while the GRK3 fusion protein binds all three G beta isoforms. This study provides a direct demonstration of a role for G betagamma in mediating the agonist-stimulated translocation of GRK2 and GRK3 in an intact cellular system and demonstrates isoform specificity in the interaction of these components.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , GTP-Binding Proteins/metabolism , Protein Serine-Threonine Kinases , Receptor Protein-Tyrosine Kinases/metabolism , Animals , COS Cells , Cattle , Cell Membrane/drug effects , Cell Membrane/metabolism , Cyclic AMP-Dependent Protein Kinases/drug effects , Cyclic AMP-Dependent Protein Kinases/isolation & purification , G-Protein-Coupled Receptor Kinase 3 , G-Protein-Coupled Receptor Kinase 5 , GTP-Binding Proteins/isolation & purification , Glutathione Transferase , Isoproterenol/pharmacology , Kinetics , Lysophospholipids/pharmacology , Peptide Fragments/pharmacology , Protein Binding , Receptor Protein-Tyrosine Kinases/drug effects , Receptor Protein-Tyrosine Kinases/isolation & purification , Receptors, Thrombin/drug effects , Receptors, Thrombin/metabolism , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Transfection , beta-Adrenergic Receptor Kinases
14.
Eur J Pharmacol ; 339(1): 77-86, 1997 Nov 19.
Article in English | MEDLINE | ID: mdl-9450619

ABSTRACT

Activation of cardiac alpha1-adrenoreceptors has a number of physiological effects. Ascribing these effects to a specific alpha1-adrenoreceptor subtype first requires the elucidation of the subtypes that are present in the tissue of interest. In the present study, mRNA transcripts for the alpha1A, alpha1B and alpha1D-adrenoreceptor subtypes were detected in cultured neonatal rat cardiac myocytes, using reverse transcriptase-polymerase chain reaction analysis. However, binding sites for only the alpha1A and alpha1B-adrenoreceptor subtypes were detected in cultured neonatal rat cardiac myocytes, using competition binding analysis with a variety of alpha1 selective receptor antagonists. Phenylephrine-stimulated phosphatidylinositol hydrolysis was inhibited by alpha1 selective receptor antagonists with affinities consistent with the alpha1A-adrenoreceptor subtype, whereas phenylephrine-induced activation of the mitogen activated protein kinase cascade was inhibited by these same antagonists with affinities more closely resembling the alpha1B-adrenoreceptor subtype. In the case of both signaling pathways, the alpha1D selective receptor antagonist, BMY 7378, exhibited affinities suggestive of the relative absence of a alpha1D-adrenoreceptor subtype. Thus, despite the presence of mRNA transcripts for all three alpha1-adrenoreceptor subtypes, only the alpha1A and alpha1B-adrenoreceptor subtypes were expressed and functionally coupled at detectable levels in neonatal rat cardiac myocytes. Of particular interest, phenylephrine-induced activation of the mitogen activated protein kinase cascade appears to be mediated by a subtype resembling most closely the pharmacological profile of the alpha1B-adrenoreceptor subtype.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Myocardium/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Type C Phospholipases/metabolism , Animals , Animals, Newborn , Binding, Competitive , Cells, Cultured , Hydrolysis , Myocardium/cytology , Phosphatidylinositols/metabolism , Radioligand Assay , Rats , Rats, Sprague-Dawley
15.
J Clin Invest ; 98(8): 1728-36, 1996 Oct 15.
Article in English | MEDLINE | ID: mdl-8878422

ABSTRACT

Erythropoietin induces a dose-dependent increase in cytosolic calcium in human erythroblasts that is mediated by a voltage-independent Ca2+ channel. Inhibition of this response to erythropoietin by pertussis toxin suggests involvement of guanine nucleotide-binding regulatory proteins (G-proteins). The role of G-proteins in regulation of the erythropoietin-modulated Ca2+ channel was delineated here by microinjection of G-protein modulators or subunits into human erythroid precursors. This is the first report on the use of microinjection to study erythropoietin signal transduction in normal precursor cells. Fura-2 loaded day-10 burst-forming units-erythroid-derived erythroblasts were used for microinjection and free intracellular calcium concentration ([Ca(i)]) was measured with digital video imaging. BCECF (1,2',7'-bis(2-carboxyethyl)-5-(and -6-)-carboxyfluorescein) was included in microinjectate, and an increase in BCECF fluorescence was evidence of successful microinjection. Cells were microinjected with nonhydrolyzable analogues of GTP, GTPgammaS or GDPbetaS, which maintain the alpha subunit in an activated or inactivated state, respectively. [Ca(i)] increased significantly in a dose-dependent manner after microinjection of GTPgammaS. However, injection of GDPbetaS blocked the erythropoietin-induced calcium increase, providing direct evidence that activation of a G-protein is required. To delineate which G-protein subunits are involved, alpha or betagamma transducin subunits were purified and microinjected as a sink for betagamma or alpha subunits in the erythroblast, respectively. Transducin betagamma, but not alpha, subunits eliminated the calcium response to erythropoietin, demonstrating the primary role of the alpha subunit. Microinjected antibodies to Gi(alpha)2, but not Gi(alpha)1 or Gi(alpha)3, blocked the erythropoietin-stimulated [Ca(i)] rise, identifying Gi(alpha)2 as the subunit involved. This was confirmed by the ability of microinjected recombinant myristoylated Gi(alpha)2, but not Gi(alpha)1 or Gi(alpha)3 subunits, to reconstitute the response of pertussis toxin-treated erythroblasts to erythropoietin. These data directly demonstrate a physiologic function of G-proteins in hematopoietic cells and show that Gi(alpha)2 is required in erythropoietin modulation of [Ca(i)] via influx through calcium channels.


Subject(s)
Erythroid Precursor Cells/metabolism , Erythropoietin/pharmacology , GTP-Binding Proteins/physiology , Signal Transduction/drug effects , Calcium/metabolism , Calcium Channels/physiology , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Humans , Microinjections , Pertussis Toxin , Virulence Factors, Bordetella/pharmacology
16.
Trends Cardiovasc Med ; 6(4): 115-21, 1996 May.
Article in English | MEDLINE | ID: mdl-21232284

ABSTRACT

A family of G proteins, composed of α, ß, and γ subunits, plays a central role in coupling receptors to a variety of enzymes and ion channels. In the cardiovascular system, G proteins are involved in coupling receptors for epinephrine, norepinephrine, acetylcholine, adenosine, angiotensin II, and endothelin to regulation of adenylyl cyclases, phospholipases, and ion channels. For many years, the classic view has been that G protein α subunits provide the requisite specificity for receptor and effector interactions. Recent advances, however, have revealed that the ß and γ subunits also play prominent roles in transducing information from receptors to the appropriate effectors. With the identification of multiple subtypes of ß and γ subunits in the heart, questions are raised regarding their respective roles in signal transduction processes regulating cardiac function.

17.
J Biol Chem ; 270(37): 21765-71, 1995 Sep 15.
Article in English | MEDLINE | ID: mdl-7665596

ABSTRACT

With the growing awareness that the G protein beta and gamma subunits directly regulate the activities of various enzymes and ion channels, the importance of identifying and characterizing these subunits is underscored. In this paper, we report the isolation of cDNA clones encoding eight different human gamma subunits, including three novel forms designated gamma 4, gamma 10, and gamma 11. The predicted protein sequence of gamma 4 shares the most identity (60-77%) with gamma 2, gamma 3, and gamma 7 and the least identity (38%) with gamma 1. The gamma 4 is modified by a geranylgeranyl group and is capable of interacting with both beta 1 and beta 2 but not with beta 3. The predicted protein sequence of gamma 10 shows only modest to low identity (35-53%) with the other known gamma subunits, with most of the differences concentrated in the N-terminal region, suggesting gamma 10 may interact with a unique subclass of alpha. The gamma 10 is modified by a geranylgeranyl group and is capable of interacting with beta 1 and beta 2 but not with beta 3. Finally, the predicted protein sequence of gamma 11 shows the most identity to gamma 1 (76% identity) and the least identity to the other known gamma (33-44%). Unlike most of the other known gamma subunits, gamma 11 is modified by a farnesyl group and is not capable of interacting with beta 2. The close resemblance of gamma 11 to gamma 1 raises intriguing questions regarding its function since the mRNA for gamma 11 is abundantly expressed in all tissues tested except for brain, whereas the mRNA for gamma 1 is expressed only in the retina where the protein functions in phototransduction.


Subject(s)
GTP-Binding Proteins/biosynthesis , GTP-Binding Proteins/chemistry , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Cattle , Cloning, Molecular/methods , DNA, Complementary/isolation & purification , DNA, Complementary/metabolism , Electrophoresis, Polyacrylamide Gel , GTP-Binding Proteins/isolation & purification , Humans , Macromolecular Substances , Molecular Sequence Data , Plasmids , Protein Biosynthesis , Protein Prenylation , RNA, Messenger/isolation & purification , RNA, Messenger/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sequence Homology, Amino Acid , Transcription, Genetic
18.
J Biol Chem ; 270(24): 14835-41, 1995 Jun 16.
Article in English | MEDLINE | ID: mdl-7782351

ABSTRACT

A family of GTP-binding regulatory proteins (G proteins) transduces signals across the plasma membrane from a large number of receptors to a smaller number of effectors. Recent studies indicate that a series of post-translational modifications are required for their association with the plasma membrane and for their function. In the case of the G protein gamma subunits, the post-translational modifications include the prenylation of a cysteine residue within a carboxyl-terminal CAAX motif. Although prenylation has been shown to involve the addition of either a C15 farnesyl or a C20 geranylgeranyl group to proteins, the structural requirements and functional consequences of adding different types of prenyl groups to various members of the gamma subunit family have not been examined. In the present study, we have employed the baculovirus expression system to study the structural requirements for attaching different types of prenyl groups to various members of the gamma subunit family. We show that the gamma 2 subunit is modified by a C20 geranylgeranyl group, consistent with the presence of a geranylgeranylation target sequence in this protein. However, we found that the gamma 1 and mutant gamma 2(Ser-71) subunits are modified by both C15 farnesyl and C20 geranylgeranyl groups, despite the presence of an accepted farnesylation target sequence in both of these proteins. Using chimeras of the gamma 1 and gamma 2 subunits, we provide evidence indicating that structural elements upstream of the carboxyl-terminal CAAX motif play a role in the recognition of members of the gamma subunit family by the appropriate insect and mammalian prenyltransferases.


Subject(s)
Alkyl and Aryl Transferases , GTP-Binding Proteins/metabolism , Animals , Baculoviridae/genetics , Binding Sites , Cell Line , Cloning, Molecular , Dimethylallyltranstransferase/metabolism , Farnesyltranstransferase , GTP-Binding Proteins/genetics , Protein Prenylation , Spodoptera , Transferases/antagonists & inhibitors , Transferases/genetics , Transferases/metabolism
19.
J Biol Chem ; 270(9): 4189-92, 1995 Mar 03.
Article in English | MEDLINE | ID: mdl-7876173

ABSTRACT

The gamma subunit composition of the major bovine brain Go and Gi proteins (GOA, GOB, GOC, Gi1, and Gi2) was characterized using antibodies against specific gamma isoforms. Each of the purified G protein heterotrimers contained a heterogeneous population of gamma subunits, and the profiles of the gamma subunits found with Gi1, Gi2, and GOA were similar. In contrast, each GO isoform had a distinct pattern of associated gamma subunits. These differences were surprising given that all three alpha O isoforms are thought to share a common amino-terminal sequence important for the binding of beta gamma dimers and that the alpha OA and alpha OC proteins may come from the same alpha O1 mRNA. The free alpha OA and alpha OC subunits had unique elution behaviors during MonoQ chromatography, compatible with differences in their post-translational processing. These results indicate that both the alpha and gamma subunit compositions of heterotrimers define the structure of an intact G protein. Furthermore, the exact subunit composition of G protein heterotrimers may depend upon regulated expression of different subunit isoforms or upon cellular processing of alpha subunits.


Subject(s)
Brain Chemistry , GTP-Binding Proteins/chemistry , Amino Acid Sequence , Animals , Cattle , GTP-Binding Proteins/immunology , Molecular Sequence Data , Protein Processing, Post-Translational
20.
J Biol Chem ; 270(11): 5901-8, 1995 Mar 17.
Article in English | MEDLINE | ID: mdl-7890721

ABSTRACT

Differentiated HL-60 cells acquire responsiveness to fMet-Leu-Phe (fMLP), which activates phospholipase C and O2- generation in a pertussis toxin-sensitive manner. Addition of retinoic acid (RA) for the last 24 h during dimethyl sulfoxide (Me2SO)-induced differentiation enhanced fMLP-dependent signals and interaction between fMLP receptor and G(i). RA modifies both the function and subunit composition of G(i)2, the predominant G(i) of HL-60 membranes, as shown by comparing purified G(i)2 from membranes of Me2SO-treated cells (D-G(i)2) to G(i)2 from membranes of cells treated with both Me2SO and RA (DR-G(i)2). As compared to D-G(i)2, DR-G(i)2 induced more fMLP binding when added to membranes of pertussis toxin-treated HL-60 cells and, in the presence of GTP gamma S, stimulated beta gamma-sensitive phospholipase C in extracts of HL-60 cells to a much greater extent at a lower concentrations. Immunoblasts revealed that RA induced expression of the gamma 2 subunit, which was otherwise undetectable in G(i)2 purified from HL-60 cells or in HL-60 membranes. Possibly by inducing expression of gamma 2, RA alters two functions of the G(i) beta gamma subunit, modulation of fMLP receptor-G(i)2 coupling and activation of the effector, Phospholipase C.


Subject(s)
GTP-Binding Proteins/metabolism , Tretinoin/pharmacology , Type C Phospholipases/metabolism , Cell Differentiation , Cell Line , Cell Membrane/metabolism , Chromatography, Affinity , Dimethyl Sulfoxide/pharmacology , Drug Synergism , Enzyme Activation , GTP-Binding Proteins/isolation & purification , Gene Expression , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Guanosine Triphosphate/metabolism , Humans , Immunoblotting , Kinetics , Leukemia, Promyelocytic, Acute , Macromolecular Substances , N-Formylmethionine Leucyl-Phenylalanine/metabolism , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Tumor Cells, Cultured , Type C Phospholipases/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...