Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
PLoS One ; 13(8): e0203170, 2018.
Article in English | MEDLINE | ID: mdl-30148884

ABSTRACT

Primary cilia are solitary organelles that emanate from the plasma membrane during growth arrest in almost all mammalian cells. The canonical Hedgehog (HH) pathway requires trafficking of the G protein-coupled receptor SMOOTHENED (SMO) and the GLI transcription factors to the primary cilium upon binding of a HH ligand to PATCHED1. However, it is unknown if activation of the small GTPase RHOA by SMO coupling to heterotrimeric Gi proteins, a form of non-canonical HH signaling, requires localization of SMO in the primary cilium. In this study, we compared RHOA and Gi protein stimulation by activation of SMO or sphingosine 1-phosphate receptor (S1P) receptors in WT and KIF3A-deficient mouse embryonic fibroblasts that lack primary cilia. We found that activation of SMO in response to Sonic HH (SHH) or purmorphamine (PUR), a small molecule agonist of SMO, stimulates Gi proteins and RHOA independently of the presence of primary cilia, similar to the effects of S1P. However, while S1P induced a fast activation of AKT that is sensitive to the Gi inhibitor pertussis toxin, HH pathway activators did not significantly activate AKT, suggesting that RHOA activation is not downstream of AKT. Our findings demonstrate that early events in some forms of non-canonical HH signaling occur in extraciliary membranes, which might be particularly relevant for actively-cycling cells, for some cancers characterized by loss of primary cilia, and in ciliopathies.


Subject(s)
Cell Membrane/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Kinesins/deficiency , Organelles/metabolism , Smoothened Receptor/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Cells, Cultured , Fibroblasts/metabolism , Hedgehog Proteins/metabolism , Kinesins/genetics , Mice, Knockout , Morpholines/administration & dosage , Morpholines/metabolism , Patched-1 Receptor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Purines/administration & dosage , Purines/metabolism , Receptors, Lysosphingolipid/metabolism , Signal Transduction , rhoA GTP-Binding Protein
2.
J Biol Chem ; 293(28): 11022-11032, 2018 07 13.
Article in English | MEDLINE | ID: mdl-29802197

ABSTRACT

SMO (Smoothened), the central transducer of Hedgehog signaling, is coupled to heterotrimeric Gi proteins in many cell types, including cardiomyocytes. In this study, we report that activation of SMO with SHH (Sonic Hedgehog) or a small agonist, purmorphamine, rapidly causes a prolongation of the action potential duration that is sensitive to a SMO inhibitor. In contrast, neither of the SMO agonists prolonged the action potential in cardiomyocytes from transgenic GiCT/TTA mice, in which Gi signaling is impaired, suggesting that the effect of SMO is mediated by Gi proteins. Investigation of the mechanism underlying the change in action potential kinetics revealed that activation of SMO selectively reduces outward voltage-gated K+ repolarizing (Kv) currents in isolated cardiomyocytes and that it induces a down-regulation of membrane levels of Kv4.3 in cardiomyocytes and intact hearts from WT but not from GiCT/TTA mice. Moreover, perfusion of intact hearts with Shh or purmorphamine increased the ventricular repolarization time (QT interval) and induced ventricular arrhythmias. Our data constitute the first report that acute, noncanonical Hh signaling mediated by Gi proteins regulates K+ currents density in cardiomyocytes and sensitizes the heart to the development of ventricular arrhythmias.


Subject(s)
Action Potentials/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Hedgehog Proteins/metabolism , Myocytes, Cardiac/metabolism , Potassium Channels, Voltage-Gated/metabolism , Potassium/metabolism , Smoothened Receptor/metabolism , Animals , Cells, Cultured , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Hedgehog Proteins/genetics , Ion Channel Gating , Mice , Mice, Transgenic , Myocytes, Cardiac/cytology , Smoothened Receptor/genetics
3.
PLoS One ; 13(5): e0197442, 2018.
Article in English | MEDLINE | ID: mdl-29763457

ABSTRACT

The Hedgehog family of morphogens has long been known to utilize, through the 7-transmembrane protein Smoothened (Smo), the heterotrimeric G protein Gi in both canonical and noncanonical forms of signaling. Other G proteins, while not specifically utilized by Smo, may nonetheless provide access to some of the events controlled by it. We reported several years ago that the G protein G13 activates one or more forms of the Gli family of transcription factors. While the Gli transcription factors are well known targets for Smo, the uncertain mechanism of activation by G13 and the identity of the targeted Gli(s) limited predictions as to the extent to which G13 might mimic Smo's actions. We evaluate here the potential for overlap in G13 and Smo signaling using C3H10T1/2 and 3T3-L1 cells as models of osteogenesis and adipogenesis, respectively. We find in C3H10T1/2 cells that a constitutively active form of Gα13 (Gα13QL) increases Gli1 mRNA, as does a constitutively active form of Smo (SmoA1). We find as well that Gα13QL induces alkaline phosphatase activity, a marker of osteogenesis, albeit the induction is far less substantial than that achieved by SmoA1. In 3T3-L1 cells both Gα13QL and SmoA1 markedly suppress adipogenic differentiation as determined by triglyceride accumulation. RNA sequencing reveals that Gα13QL and SmoA1 regulate many of the same genes but that quantitative and qualitative differences exist. Differences also exist, we find, between SmoA1 and purmorphamine, an agonist for Smo. Therefore, while comparisons of constitutively active proteins are informative, extrapolations to the setting of agonists require care.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Signal Transduction/drug effects , 3T3-L1 Cells , Adipogenesis/drug effects , Adipogenesis/genetics , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Mice , Morpholines/pharmacology , Purines/pharmacology , Signal Transduction/genetics
4.
Methods Mol Biol ; 1322: 35-44, 2015.
Article in English | MEDLINE | ID: mdl-26179037

ABSTRACT

The utilization of heterotrimeric G protein, and in particular those of the Gi, family, by Hedgehogs through Smoothened has become increasingly clear. We describe here a method for evaluating the activity of Smoothened toward G proteins in membranes derived from human embryonic kidney-293 (HEK293) cells. The assay relies on receptor-promoted exchange of GDP for [(35)S]GTPγS on the Gα subunit. The assay is best suited for analysis of the constitutive activity of Smoothened, inverse agonism superimposed on this activity, and neutral antagonism superimposed on inverse agonism. The assay would also be suitable for several other applications requiring a proximal, quantifiable readout of Smoothened activity.


Subject(s)
GTP-Binding Proteins/metabolism , Guanosine 5'-O-(3-Thiotriphosphate) , Receptors, G-Protein-Coupled/metabolism , GTP-Binding Proteins/chemistry , Guanosine 5'-O-(3-Thiotriphosphate)/chemistry , HEK293 Cells , Humans , Immunoprecipitation/methods , Protein Binding , Receptors, G-Protein-Coupled/chemistry , Smoothened Receptor , Sulfur Radioisotopes/chemistry
5.
Mol Pharmacol ; 83(3): 691-7, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23292797

ABSTRACT

Smoothened (Smo) is a 7-transmembrane protein essential to the activation of Gli transcription factors (Gli) by hedgehog morphogens. The structure of Smo implies interactions with heterotrimeric G proteins, but the degree to which G proteins participate in the actions of hedgehogs remains controversial. We posit that the G(i) family of G proteins provides to hedgehogs the ability to expand well beyond the bounds of Gli. In this regard, we evaluate here the efficacy of Smo as it relates to the activation of G(i), by comparing Smo with the 5-hydroxytryptamine(1A) (5-HT(1A)) receptor, a quintessential G(i)-coupled receptor. We find that with use of [(35)S]guanosine 5'-(3-O-thio)triphosphate, first, with forms of G(i) endogenous to human embryonic kidney (HEK)-293 cells made to express epitope-tagged receptors and, second, with individual forms of Gα(i) fused to the C terminus of each receptor, Smo is equivalent to the 5-HT(1A) receptor in the assay as it relates to capacity to activate G(i). This finding is true regardless of subtype of G(i) (e.g., G(i2), G(o), and G(z)) tested. We also find that Smo endogenous to HEK-293 cells, ostensibly through inhibition of adenylyl cyclase, decreases intracellular levels of cAMP. The results indicate that Smo is a receptor that can engage not only Gli but also other more immediate effectors.


Subject(s)
Heterotrimeric GTP-Binding Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Adenylyl Cyclases/metabolism , Cell Line , Cyclic AMP/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , HEK293 Cells , Hedgehog Proteins/metabolism , Humans , Receptor, Serotonin, 5-HT1A/metabolism , Serotonin/metabolism , Smoothened Receptor , Transcription Factors/metabolism
6.
Sci Signal ; 4(200): pt7, 2011 Nov 22.
Article in English | MEDLINE | ID: mdl-22114142

ABSTRACT

The vertebrate Hedgehog (Hh) pathway has essential functions during development and tissue homeostasis in normal physiology, and its dysregulation is a common theme in cancer. The Hh ligands (Sonic Hh, Indian Hh, and Desert Hh) bind to the receptors Patched1 and Patched2, resulting in inhibition of their repressive effect on Smoothened (Smo). Smo is a seven-transmembrane protein, which was only recently shown to function as a G protein-coupled receptor (GPCR) with specificity toward the heterotrimeric guanine nucleotide-binding protein G(i). In addition to activating G(i), Smo signals through its C-terminal tail to inhibit Suppressor of Fused, resulting in stabilization and activation of the Gli family of transcription factors, which execute a transcriptional response to so-called "canonical Hh signaling." In this Presentation, we illustrate two outcomes of Hh signaling that are independent of Gli transcriptional activity and, thus, are defined as "noncanonical." One outcome is dependent on Smo coupling to G(i) proteins and exerts changes to the actin cytoskeleton through stimulation of the small guanosine triphosphatases (GTPases) RhoA and Rac1. These cytoskeletal changes promote migration in fibroblasts and tubulogenesis in endothelial cells. Signaling through the other noncanonical Hh pathway is independent of Smo and inhibits Patched1-induced cell death.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Hedgehog Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Transcription Factors/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cell Death/physiology , Cell Movement/physiology , Cytoskeleton/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Enzyme Activation/physiology , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Patched Receptors , Patched-1 Receptor , Patched-2 Receptor , Protein Structure, Tertiary , Receptors, Cell Surface/metabolism , Repressor Proteins/metabolism , Signal Transduction/physiology , Smoothened Receptor , Zinc Finger Protein GLI1
7.
J Biol Chem ; 286(35): 30714-30722, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21757753

ABSTRACT

Smoothened (Smo) is a seven-transmembrane (7-TM) receptor that is essential to most actions of the Hedgehog family of morphogens. We found previously that Smo couples to members of the G(i) family of heterotrimeric G proteins, which in some cases are integral although alone insufficient in the activation of Gli transcription factors through Hedgehog signaling. In response to a report that the G(12/13) family is relevant to Hedgehog signaling as well, we re-evaluated the coupling of Smo to one member of this family, G(13), and investigated the capacity of this and other G proteins to activate one or more of forms of Gli. We found no evidence that Smo couples directly to G(13). We found nonetheless that Gα(13) and to some extent Gα(q) and Gα(12) are able to effect activation of Gli(s). This capacity is realized in some cells, e.g. C3H10T1/2, MC3T3, and pancreatic cancer cells, but not all cells. The mechanism employed is distinct from that achieved through canonical Hedgehog signaling, as the activation does not involve autocrine signaling or in any other way require active Smo and does not necessarily involve enhanced transcription of Gli1. The activation by Gα(13) can be replicated through a G(q)/G(12/13)-coupled receptor, CCK(A), and is attenuated by inhibitors of p38 mitogen-activated protein kinase and Tec tyrosine kinases. We posit that G proteins, and perhaps G(13) in particular, provide access to Gli that is independent of Smo and that they thus establish a basis for control of at least some forms of Gli-mediated transcription apart from Hedgehogs.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Receptors, G-Protein-Coupled/metabolism , Transcription Factors/metabolism , 3T3 Cells , Animals , Cell Line, Tumor , HEK293 Cells , Hedgehog Proteins/metabolism , Humans , Mice , Mice, Inbred C3H , Models, Biological , Plasmids/metabolism , Protein Structure, Tertiary , Smoothened Receptor , Zinc Finger Protein GLI1
8.
J Biol Chem ; 286(22): 19589-96, 2011 Jun 03.
Article in English | MEDLINE | ID: mdl-21474452

ABSTRACT

Evidence supporting the functionality of Smoothened (SMO), an essential transducer in most pathways engaged by Hedgehog (Hh), as a G(i)-coupled receptor contrasts with the lack of an apparently consistent requirement for G(i) in Hh signal transduction. In the present study, we sought to evaluate the role of SMO-G(i) coupling in fibroblast migration induced by Sonic Hedgehog (Shh). Our results demonstrate an absolute requirement for G(i) in Shh-induced fibroblast migration. We found that Shh acutely stimulates the small Rho GTPases Rac1 and RhoA via SMO through a G(i) protein- and PI3K-dependent mechanism, and that these are required for cell migration. These responses were independent of transcription by Gli and of the C-terminal domain of SMO, as we show using a combination of molecular and genetic tools. Our findings provide a mechanistic model for fibroblast migration in response to Shh and underscore the role of SMO-G(i) coupling in non-canonical Hh signaling.


Subject(s)
Cell Movement/physiology , Fibroblasts/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Hedgehog Proteins/metabolism , Neuropeptides/metabolism , Signal Transduction/physiology , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Fibroblasts/cytology , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Hedgehog Proteins/genetics , Mice , Mice, Knockout , NIH 3T3 Cells , Neuropeptides/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Smoothened Receptor , rac GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein , rho GTP-Binding Proteins/genetics , rhoA GTP-Binding Protein
9.
Mol Pharmacol ; 78(4): 767-77, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20664004

ABSTRACT

Regions of basic amino acids in proteins can promote membrane localization through electrostatic interactions with negatively charged membrane lipid head groups. Previous work showed that the heterotrimeric G protein subunit α(q) contains a polybasic region in its N terminus that contributes to plasma membrane localization. Here, the role of the N-terminal polybasic region of α(q) in signaling was addressed. For α(q) mutants, loss of plasma membrane localization correlated with loss of signaling function, as measured by the ability to couple activated G protein-coupled receptors (GPCRs) to stimulation of inositol phosphate production. However, recovery of plasma membrane localization of α(q) polybasic mutants by introduction of a site for myristoylation or by coexpression of ßγ failed to recover signaling, suggesting a role for N-terminal basic amino acids of α(q) beyond simple plasma membrane localization. It is noteworthy that an α(q)4Q mutant, containing glutamine substitutions at arginines 27, 30, 31, and 34, was identified that failed to mediate signaling yet retained plasma membrane localization. Although α(q)4Q failed to couple activated receptors to inositol phosphate production, it was able to bind ßγ, bind RGS4 in an activation-dependent manner, stimulate inositol phosphate production in a receptor-independent manner, and productively interact with a GPCR in isolated membranes. It is noteworthy that α(q)4Q showed a differing localization to plasma membrane nanodomains compared with wild-type α(q). Thus, basic amino acids in the N terminus of α(q) can affect its lateral segregation on plasma membranes, and changes in such lateral segregation may be responsible for the observed signaling defects of α(q)4Q.


Subject(s)
Amino Acids, Basic/physiology , Cell Membrane/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Nanostructures/chemistry , Peptide Fragments/chemistry , Peptide Fragments/physiology , Signal Transduction/physiology , Amino Acid Motifs/genetics , Amino Acid Sequence , Amino Acids, Basic/genetics , Animals , Cell Line , Cell Membrane/chemistry , Cell Membrane/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Humans , Molecular Sequence Data , Peptide Fragments/genetics , Protein Binding/genetics , Protein Structure, Tertiary/genetics , Spodoptera/cytology , Spodoptera/genetics
10.
Mol Pharmacol ; 75(1): 235-41, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18952767

ABSTRACT

Receptors coupled to the G(q) and G(12) families of heterotrimeric G proteins have surfaced rarely in the context of functional selectivity and always indirectly. We explore here the differential engagement of G(q) and G(13) (of the G(12) family) by the thromboxane A(2) receptor alpha (TPalpha), via agonist-effected [(35)S]-guanosine 5'-O-(3-thio)triphosphate binding when the G proteins themselves are used as reporters. We find for TPalpha introduced into human embryonic kidney 293 cells and for the receptor expressed normally in human platelets an agonist-selective engagement of G(q) versus G(13). Pinane thromboxane A(2) (PTA(2)) activates G(q) in preference to G(13), whereas 8-iso-prostaglandin F(2alpha) activates G(13) in preference to G(q). 9,11-Dideoxy-9alpha,11alpha-methanoepoxy-prosta-5Z,13E-dien-1-oic acid (U46619), in contrast, exhibits no preference. Reserve of receptor in relation to G protein and of G protein in relation to downstream events is apparent in some instances but does not have a bearing on selectivity. Activation of G proteins by PTA(2) is right-shifted from binding of the ligand to receptor, a manifestation of which is a bimodal action: PTA(2) is an antagonist at low concentrations and an agonist at higher concentrations. We posit two populations of TPalpha, or two intrinsic sites of ligand binding, with selectivity evident not only in terms of the G proteins activated but properties of antagonism versus agonism.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Bicyclic Monoterpenes , Bridged Bicyclo Compounds, Heterocyclic , Calcium/analysis , Calcium/metabolism , Cell Line , Cell Shape , Cytosol/metabolism , Dinoprost/analogs & derivatives , Dinoprost/metabolism , Fatty Acids, Unsaturated , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Hydrazines/pharmacology , Inhibitory Concentration 50 , Kidney/cytology , Ligands , Molecular Conformation , Platelet Aggregation , Radioligand Assay , Receptors, Thromboxane A2, Prostaglandin H2/agonists , Receptors, Thromboxane A2, Prostaglandin H2/antagonists & inhibitors , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Thromboxane A2/analogs & derivatives , Thromboxane A2/metabolism
11.
Biochem J ; 403(3): 369-79, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17419683

ABSTRACT

Signalling by Hh (Hedgehog) proteins is among the most actively studied receptor-mediated phenomena relevant to development and post-embryonic homoeostatic events. The impact of signalling by the Hh proteins is profound, and work pertaining to the presentation of these proteins and the pathways engaged by them continues to yield unique insights into basic aspects of morphogenic signalling. We review here the mechanisms of signalling relevant to the actions of Hh proteins in vertebrates. We emphasize findings within the past several years on the recognition of, in particular, Sonic hedgehog by target cells, pathways of transduction employed by the seven-pass transmembrane protein Smoothened and end points of action, as manifest in the regulation of the Gli transcription factors. Topics of extended interest are those regarding the employment of heterotrimeric G-proteins and G-protein-coupled receptor kinases by Smoothened. We also address the pathways, insofar as known, linking Smoothened to the expression and stability of Gli1, Gli2 and Gli3. The mechanisms by which Hh proteins signal have few, if any, parallels. It is becoming clear in vertebrates, however, that several facets of signalling are shared in common with other venues of signalling. The challenge in understanding both the actions of Hh proteins and the overlapping forms of regulation will be in understanding, in molecular terms, both common and divergent signalling events.


Subject(s)
Hedgehog Proteins/physiology , Signal Transduction/physiology , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Kruppel-Like Transcription Factors/physiology , Models, Biological , Patched Receptors , Receptors, Cell Surface/physiology , Receptors, G-Protein-Coupled/physiology , Smoothened Receptor , Transcription, Genetic/drug effects , Zinc Finger Protein GLI1 , Zinc Finger Protein Gli2
12.
Proc Natl Acad Sci U S A ; 103(33): 12607-12, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16885213

ABSTRACT

The mechanisms by which the activation of Smoothened (Smo), a protein essential to the actions of the Hedgehog family of secreted proteins, is translated into signals that converge on the Gli transcription factors are not fully understood. The seven-transmembrane structure of Smo has long implied the utilization of heterotrimeric GTP-binding regulatory proteins (G proteins); however, evidence in this regard has been indirect and contradictory. In the current study we evaluated the capacity of mammalian Smo to couple to G proteins directly. We found that Smo, by virtue of what appears to be constitutive activity, activates all members of the G(i) family but does not activate members of the G(s), G(q), and G(12) families. The activation is suppressed by cyclopamine and other inhibitors of Hedgehog signaling and is enhanced by the Smo agonist purmorphamine. Activation of G(i) by Smo is essential in the activation of Gli in fibroblasts, because disruption of coupling to G(i) with pertussis toxin inhibits the activation of Gli by Sonic hedgehog and a constitutively active form of Smo (SmoM2). However, G(i) does not provide a sufficient signal because a truncated form of Smo, although capable of activating G(i), does not effect activation of Gli. Rescue of pertussis toxin-inhibited activation of Gli by Sonic hedgehog can be achieved with a constitutively active Galpha(i)-subunit. The data suggest that Smo is in fact the source of two signals relevant to the activation of Gli: one involving G(i) and the other involving events at Smo's C-tail independent of G(i).


Subject(s)
Heterotrimeric GTP-Binding Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Animals , Enzyme Activation , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Mice , NIH 3T3 Cells , Oncogene Proteins/metabolism , Pertussis Toxin/metabolism , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Smoothened Receptor , Trans-Activators/metabolism , Veratrum Alkaloids/metabolism , Zinc Finger Protein GLI1
13.
Mol Pharmacol ; 69(4): 1433-40, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16418336

ABSTRACT

Despite advances in the understanding of pathways regulated by the G12 family of heterotrimeric G proteins, much regarding the engagement of this family by receptors remains unclear. We explore here, using the thromboxane A2 receptor TPalpha, the ability of G12 and G13 to report differences in the potency and efficacy of receptor ligands. We were interested especially in the potential of the isoprostane 8-iso-prostaglandin F (8-iso-PGF2alpha), among other ligands examined, to activate G12 and G13 through TPalpha explicitly. We were also interested in the functionality of TPalpha-Galpha fusion proteins germane to G12 and G13. Using fusion proteins in Spodoptera frugiperda (Sf9) cells and independently expressed proteins in human embryonic kidney 293 cells, and using guanosine 5'-O-(3-[35S]thio)triphosphate binding to evaluate Galpha activation directly, we found for Galpha that no ligand tested, including 8-iso-prostaglandin F (8-iso-PGF2alpha and a purported antagonist (pinane thromboxane A2), was silent. The activity of agonists was especially pronounced when evaluated for TPalpha-Galpha13 and in the context of receptor reserve. Agonist activity for 8-iso-PGF2 was diminished and that for pinane thromboxane A nonexistent when Galpha12 was the reporter. These data establish that G12 and G13 can report differentially potency and efficacy and underscore the relevance of receptor and G protein context.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/physiology , Animals , Bridged Bicyclo Compounds, Heterocyclic , Cell Line , Dinoprost/analogs & derivatives , Dinoprost/metabolism , Electrophoresis, Polyacrylamide Gel , Fatty Acids, Unsaturated , Humans , Hydrazines/metabolism , Radioligand Assay , Spodoptera
14.
Trends Pharmacol Sci ; 26(3): 146-54, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15749160

ABSTRACT

Much regarding the engagement of the G(12) family of heterotrimeric G proteins (G(12) and G(13)) by agonist-activated receptors remains unclear. For example, the identity of receptors that couple unequivocally to G(12) and G(13) and how signals are allocated among these and other G proteins remain open questions. Part of the problem in understanding signaling through G(12) and G(13) is that the activation of these G proteins is rarely demonstrated directly and is instead presumed usually from far removed downstream events. Furthermore, receptors that couple to G(12) and G(13) invariably couple to additional G proteins, and thus few events can be linked unambiguously to one G protein or another. In this article, we document receptors that reportedly couple to G(12), G(13) or both G(12) and G(13), evaluate the methodology used to understand the coupling of these receptors, and discuss the ability of these receptors to couple also to G(q).


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Humans , Receptors, G-Protein-Coupled/genetics
15.
Sci STKE ; 2003(196): pe35, 2003 Aug 19.
Article in English | MEDLINE | ID: mdl-12928525

ABSTRACT

Examples of the activation of heterotrimeric G proteins in vivo by any means other than through activated cell surface receptors have been limited to pathophysiological phenomena. With the discovery of proteins apart from receptors that facilitate guanine nucleotide exchange and affect G protein subunit dissociation directly, however, the notion of receptor-independent modes of activation in normal circumstances has become a subject of great interest. Three recent publications, each focusing on G protein regulators (GPRs) in asymmetric positioning of the mitotic spindle in the early Caenorhabditis elegans embryo, provide substantial support for the likelihood of such a form of activation. The C. elegans proteins GPR-1 and GPR-2 each contain a G protein regulatory motif, which supports interaction with Galpha(i)-like subunits. Inactivation of the genes encoding GPR-1 and GPR-2 prevents the correct positioning of the mitotic spindle in the one- and two-cell embryo. This phenotype is identical to that achieved by inactivation of genes encoding the Galpha subunits GOA-1 and GPA-16. Because signaling in the one- and two-cell embryos is "intrinsic," the data suggest a GPR-dependent, receptor-independent mode of G protein activation. The GPRs interact preferentially with the guanosine diphosphate (GDP)-bound form of Galpha subunits, and the GPR motif per se exhibits GDP dissociation inhibitor activity. The actions of the GPRs imply that GDP.Galpha.GPR is a key intermediate or effector in force generation relevant to mitotic spindle positioning.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Heterotrimeric GTP-Binding Proteins/physiology , Receptors, Cell Surface/physiology , Spindle Apparatus/physiology , Animals , Caenorhabditis elegans/cytology , Cell Division/physiology
16.
J Biol Chem ; 277(48): 46035-42, 2002 Nov 29.
Article in English | MEDLINE | ID: mdl-12297509

ABSTRACT

Platelet responses at sites of vascular injury are regulated by intracellular cAMP levels, which rise rapidly when prostacyclin (PGI(2)) is released from endothelial cells. Platelet agonists such as ADP and epinephrine suppress PGI(2)-stimulated cAMP formation by activating receptors coupled to G(i) family members, four of which are present in platelets. To address questions about the specificity of receptor:G protein coupling, the regulation of cAMP formation in vivo and the contribution of G(i)-mediated pathways that do not involve adenylyl cyclase, we studied platelets from mice that lacked the alpha subunits of one or more of the three most abundantly expressed G(i) family members and compared the results with platelets from mice that lacked the PGI(2) receptor, IP. As reported previously, loss of G(i2)alpha or G(z)alpha inhibited aggregation in response to ADP and epinephrine, respectively, producing defects that could not be reversed by adding an adenylyl cyclase inhibitor. Platelets that lacked both G(i2)alpha and G(z)alpha showed impaired responses to both agonists, but the impairment was no greater than in the individual knockouts. Loss of G(i3)alpha had no effect either alone or in combination with G(z)alpha. Loss of either G(z)alpha or G(i2)alpha impaired the ability of ADP and epinephrine to inhibit PGI(2)-stimulated adenylyl cyclase activity and caused a 40%-50% rise in basal cAMP levels, whereas loss of G(i3)alpha did not. Conversely, deletion of IP abolished responses to PGI(2) and caused cAMP levels to fall by 30%, effects that did not translate into enhanced responsiveness to agonists ex vivo. From these results we conclude that 1) cAMP levels in circulating platelets reflect ongoing signaling through G(i2), G(z), and IP, but not G(i3); 2) platelet epinephrine (alpha(2A)-adrenergic) and ADP (P2Y12) receptors display strong preferences among G(i) family members with little evidence of redundancy; and 3) these receptor preferences do not extend to G(i3). Finally, the failure of ADP and epinephrine to inhibit basal, as opposed to PGI(2)-stimulated, cAMP formation highlights the need during platelet activation for G(i) signaling pathways that involve effectors other than adenylyl cyclase.


Subject(s)
Adenylyl Cyclases/metabolism , Blood Platelets/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Signal Transduction , Animals , Blood Platelets/enzymology , Cyclic AMP/metabolism , Epoprostenol/metabolism , Mice
17.
J Pharmacol Exp Ther ; 302(3): 1220-7, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12183683

ABSTRACT

5-(4-[4-(5-Cyano-3-indolyl)-butyl)-butyl]-1-piperazinyl)-benzofuran-2-carboxamide (EMD 68843; vilazodone) is a novel compound with combined high affinity and selectivity for the 5-hydroxytryptamine (5-HT) transporter and 5-HT(1A) receptors. EMD 68843 was tested as a prototype compound, which benefits from dual pharmacological effects that could increase extracellular 5-HT to levels higher than those produced by conventional selective serotonin reuptake inhibitors (SSRIs). In Sf9 cells, EMD 68843 increased guanosine 5'-O-(3-[(35)S]thiotriphosphate) binding to 69% of the magnitude of the full 5-HT(1A) receptor agonist R-(1)-trans-8-hydroxy-2-[N-n-propyl-N-(39-iodo-29-propenyl)] aminotetralin (8-OH-PIPAT), indicating that it is a partial agonist at 5-HT(1A) receptors. Acute, systemic administration of EMD 68843 produced a larger maximal increase of extracellular 5-HT than the SSRI fluoxetine in both the ventral hippocampus (HPv) (558 versus 274%) and the frontal cortex (FC) (527 versus 165%). Regional differences in the response to the two drugs were also observed. These effects may be attributed to the differential regulation of 5-HT release in the HPv and FC by 5-HT(1A) autoreceptors. When challenged with the 5-HT(1A) receptor agonist 8-hydroxy-2-(di-n-propylamino) tetralin (8-OH-DPAT), EMD 68843-induced increases in extracellular 5-HT were greatly reduced in the HPv but to a lesser extent in the FC. In behavioral studies, EMD 68843 produced antidepressant-like effects in the forced swimming test in both rats and mice but only within a narrow dosage range. Like fluoxetine, EMD 68843 did not produce the symptoms of the 5-HT behavioral syndrome in rats but, unlike fluoxetine, pretreatment with EMD 68843 blocked expression of the 5-HT behavioral syndrome induced by 8-OH-DPAT. Taken together, the results show that EMD 68843 augments extracellular 5-HT levels in forebrain regions to a greater extent than fluoxetine. At higher doses, however, weak efficacy of EMD 68843 at postsynaptic 5-HT(1A) receptors may inhibit the expression of rodent antidepressant-like behaviors.


Subject(s)
Behavior, Animal/drug effects , Benzofurans/pharmacology , Brain Chemistry/drug effects , Indoles/pharmacology , Receptors, Serotonin/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin Receptor Agonists/pharmacology , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Antidepressive Agents, Second-Generation/pharmacology , Cell Line , Extracellular Space/drug effects , Extracellular Space/metabolism , Female , Fluoxetine/pharmacology , GTP-Binding Proteins/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Microdialysis , Piperazines , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Serotonin, 5-HT1 , Swimming/psychology , Vilazodone Hydrochloride
19.
Biochim Biophys Acta ; 1582(1-3): 94-9, 2002 May 23.
Article in English | MEDLINE | ID: mdl-12069815

ABSTRACT

Pathways of transduction employed by receptors for sphingosine 1-phosphate (S1P) are identified by the nature of second messengers and/or downstream targets regulated and, more formally, by direct assays of heterotrimeric G protein activation. The different methods generally agree. S1P1 couples to members of the Gi family, apparently selectively, although reported pertussis toxin (PTX)-insensitive actions make categorical statements regarding exclusivity difficult. S1P2 and S1P3 couple to members of the Gi, Gq, and G12/13 families. S1P4 couples to Gi and possibly G12/13, while S1P5 couples to Gi and G12/13 but not to Gq. In virtually all circumstances, coupling of S1P receptors to Gi is reflected in PTX-sensitive inhibition of adenylyl cyclase, activation of extracellular-regulated kinases (ERKs), and, depending on the cell, activation of phospholipase C (PLC). Coupling to Gq is reflected in PTX-insensitive activation of phospholipase C. Coupling to G12/13 is reflected in activation of Rho and subsequent activation of serum response factor (SRF). Specific linkages have been verified in almost all instances by receptor-promoted [35S]GTPgammaS/GDP exchange on identified G proteins.


Subject(s)
GTP-Binding Proteins/physiology , I-kappa B Proteins , Lysophospholipids , Receptors, Cell Surface/physiology , Receptors, G-Protein-Coupled , Signal Transduction/physiology , Sphingosine/analogs & derivatives , Sphingosine/physiology , Animals , DNA-Binding Proteins/physiology , Immediate-Early Proteins/physiology , NF-KappaB Inhibitor alpha , Receptors, Lysophospholipid
SELECTION OF CITATIONS
SEARCH DETAIL
...