Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cancer Sci ; 114(5): 2001-2013, 2023 May.
Article in English | MEDLINE | ID: mdl-36718954

ABSTRACT

G-proteins are intracellular partners of G-protein-coupled receptors. As a member of the G-protein family, GNB1 has been shown to play a pro-cancer role in lung cancer and breast cancer. However, the biological function and detailed mechanisms of GNB1 in hepatocellular carcinoma progression are unclear. In this study, we investigated the effects of GNB1 and its possible mechanism of action in hepatocellular carcinoma (HCC). The clinical significance of GNB1 was evaluated in a large cohort of HCC patients, showing that GNB1 was overexpressed in HCC compared to adjacent normal liver tissues, and increased GNB1 expression was associated with poor prognosis. We also demonstrated that GNB1 enhances cell proliferation, colony formation, and cell migration and invasion in vitro and promotes the epithelial-to-mesenchymal transition process in HCC cells. Tumor xenograft model assay confirmed the oncogenic role of GNB1 in tumorigenicity in nude mice. Activation of P38 signaling was found in the GNB1 overexpressed HCC cells. Further intervention of P38 confirmed it as an important signaling pathway for the oncogenic role of GNB1 in HCC. Moreover, co-immunoprecipitation followed by liquid chromatograph-mass spectrometry identified that GNB1 exerted oncogenic functions via the interaction of BAG2 and activated P38 signaling pathway. Together, our results reveal that GNB1 plays a pivotal oncogenic role in HCC by promoting the P38 pathway via cooperating with BAG2. GNB1 may serve as a prognostic biomarker for patients with HCC.


Subject(s)
Carcinoma, Hepatocellular , GTP-Binding Protein beta Subunits , Liver Neoplasms , Animals , Mice , Humans , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Mice, Nude , Cell Line, Tumor , MAP Kinase Signaling System , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Cell Movement/genetics , Prognosis , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein beta Subunits/pharmacology , Molecular Chaperones/metabolism
2.
Proc Natl Acad Sci U S A ; 117(46): 29090-29100, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33122432

ABSTRACT

TRPM3 channels play important roles in the detection of noxious heat and in inflammatory thermal hyperalgesia. The activity of these ion channels in somatosensory neurons is tightly regulated by µ-opioid receptors through the signaling of Gßγ proteins, thereby reducing TRPM3-mediated pain. We show here that Gßγ directly binds to a domain of 10 amino acids in TRPM3 and solve a cocrystal structure of this domain together with Gßγ. Using these data and mutational analysis of full-length proteins, we pinpoint three amino acids in TRPM3 and their interacting partners in Gß1 that are individually necessary for TRPM3 inhibition by Gßγ. The 10-amino-acid Gßγ-interacting domain in TRPM3 is subject to alternative splicing. Its inclusion in or exclusion from TRPM3 channel proteins therefore provides a mechanism for switching on or off the inhibitory action that Gßγ proteins exert on TRPM3 channels.


Subject(s)
GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein beta Subunits/pharmacology , GTP-Binding Protein gamma Subunits/metabolism , GTP-Binding Protein gamma Subunits/pharmacology , TRPM Cation Channels/chemistry , TRPM Cation Channels/drug effects , TRPM Cation Channels/metabolism , Binding Sites , Calcium/metabolism , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein gamma Subunits/chemistry , HEK293 Cells , Humans , Hyperalgesia/metabolism , Models, Molecular , Mutation , Neurons/metabolism , Pain/metabolism , Receptors, Opioid/metabolism , TRPM Cation Channels/genetics
3.
Elife ; 62017 08 15.
Article in English | MEDLINE | ID: mdl-28829742

ABSTRACT

Transient receptor potential melastatin 3 (TRPM3) channels are activated by heat, and chemical ligands such as pregnenolone sulphate (PregS) and CIM0216. Here, we show that activation of receptors coupled to heterotrimeric Gi/o proteins inhibits TRPM3 channels. This inhibition was alleviated by co-expression of proteins that bind the ßγ subunits of heterotrimeric G-proteins (Gßγ). Co-expression of Gßγ, but not constitutively active Gαi or Gαo, inhibited TRPM3 currents. TRPM3 co-immunoprecipitated with Gß, and purified Gßγ proteins applied to excised inside-out patches inhibited TRPM3 currents, indicating a direct effect. Baclofen and somatostatin, agonists of Gi-coupled receptors, inhibited Ca2+ signals induced by PregS and CIM0216 in mouse dorsal root ganglion (DRG) neurons. The GABAB receptor agonist baclofen also inhibited inward currents induced by CIM0216 in DRG neurons, and nocifensive responses elicited by this TRPM3 agonist in mice. Our data uncover a novel signaling mechanism regulating TRPM3 channels.


Subject(s)
GTP-Binding Protein beta Subunits/pharmacology , GTP-Binding Protein gamma Subunits/pharmacology , TRPM Cation Channels/drug effects , Animals , Baclofen/antagonists & inhibitors , Behavior Rating Scale , Calcium/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/pharmacology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , HEK293 Cells , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Neurons/drug effects , Neurons/metabolism , Pregnenolone/pharmacology , Somatostatin/antagonists & inhibitors
4.
Elife ; 62017 08 15.
Article in English | MEDLINE | ID: mdl-28826482

ABSTRACT

Opioids, agonists of µ-opioid receptors (µORs), are the strongest pain killers clinically available. Their action includes a strong central component, which also causes important adverse effects. However, µORs are also found on the peripheral endings of nociceptors and their activation there produces meaningful analgesia. The cellular mechanisms downstream of peripheral µORs are not well understood. Here, we show in neurons of murine dorsal root ganglia that pro-nociceptive TRPM3 channels, present in the peripheral parts of nociceptors, are strongly inhibited by µOR activation, much more than other TRP channels in the same compartment, like TRPV1 and TRPA1. Inhibition of TRPM3 channels occurs via a short signaling cascade involving Gßγ proteins, which form a complex with TRPM3. Accordingly, activation of peripheral µORs in vivo strongly attenuates TRPM3-dependent pain. Our data establish TRPM3 inhibition as important consequence of peripheral µOR activation indicating that pharmacologically antagonizing TRPM3 may be a useful analgesic strategy.


Subject(s)
GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein beta Subunits/pharmacology , GTP-Binding Protein gamma Subunits/metabolism , GTP-Binding Protein gamma Subunits/pharmacology , Receptors, Opioid, mu/metabolism , TRPM Cation Channels/drug effects , Analgesics, Opioid/agonists , Animals , Behavior Rating Scale , Calcium/metabolism , Calcium Signaling/physiology , Ganglia, Spinal/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Nociceptors/physiology , Pain/metabolism , Receptors, Opioid/metabolism , TRPA1 Cation Channel/metabolism , TRPV Cation Channels/metabolism
5.
J Pharmacol Exp Ther ; 352(1): 148-55, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25339760

ABSTRACT

The α1-subunit containing glycine receptors (GlyRs) is potentiated by ethanol, in part, by intracellular Gßγ actions. Previous studies have suggested that molecular requirements in the large intracellular domain are involved; however, the lack of structural data about this region has made it difficult to describe a detailed mechanism. Using circular dichroism and molecular modeling, we generated a full model of the α1-GlyR, which includes the large intracellular domain and provides new information on structural requirements for allosteric modulation by ethanol and Gßγ. The data strongly suggest the existence of an α-helical conformation in the regions near transmembrane (TM)-3 and TM4 of the large intracellular domain. The secondary structure in the N-terminal region of the large intracellular domain near TM3 appeared critical for ethanol action, and this was tested using the homologous domain of the γ2-subunit of the GABAA receptor predicted to have little helical conformation. This region of γ2 was able to bind Gßγ and form a functional channel when combined with α1-GlyR, but it was not sensitive to ethanol. Mutations in the N- and C-terminal regions introduced to replace corresponding amino acids of the α1-GlyR sequence restored the ability to be modulated by ethanol and Gßγ. Recovery of the sensitivity to ethanol was associated with the existence of a helical conformation similar to α1-GlyR, thus being an essential secondary structural requirement for GlyR modulation by ethanol and G protein.


Subject(s)
Ethanol/pharmacology , GTP-Binding Protein beta Subunits/pharmacology , GTP-Binding Protein gamma Subunits/pharmacology , Intracellular Space/metabolism , Receptors, Glycine/chemistry , Receptors, Glycine/metabolism , Amino Acid Sequence , Animals , Dose-Response Relationship, Drug , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , HEK293 Cells , Humans , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary/drug effects , Protein Structure, Tertiary/drug effects , Rats , Receptors, GABA-A/metabolism
6.
Int J Biochem Cell Biol ; 42(6): 1052-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20348012

ABSTRACT

The hematopoietic-specific G(q) subfamily members, Galpha(16) and Galpha(14) proteins have recently been shown to be capable of stimulating the signal transducer and activator of transcription 3 (STAT3) as well as STAT1. In the present study we examined whether this activation was STAT-member specific as well as determining the possible involvement of Gbetagamma dimers. Despite clear stimulation of STAT3, the constitutively active mutants of Galpha(16) (Galpha(16)QL) and Galpha(14) (Galpha(14)QL) failed to induce the phosphorylation of several STAT family members, including STAT2, STAT4 and STAT5 in human embryonic kidney 293 cells. On the other hand, transient expression of specific combinations of Gbetagamma complexes induced STAT3 phosphorylation. Among the 48 combinations tested, 13 permutations of Gbetagamma stimulated STAT3 phosphorylation and all of them contain the neuronal-specific Ggamma(2), Ggamma(4), Ggamma(7) and Ggamma(9). These results suggested that the activation of STAT family members by Galpha(16) or Galpha(14) was selective and that distinct combinations of Gbetagamma complexes can also regulate the STAT signaling pathway.


Subject(s)
GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , STAT3 Transcription Factor/metabolism , Dimerization , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/pharmacology , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/pharmacology , GTP-Binding Protein gamma Subunits/genetics , GTP-Binding Protein gamma Subunits/pharmacology , HeLa Cells , Hematopoiesis , Humans , Mutation/genetics , Neurons/metabolism , Phosphorylation , Protein Binding , Signal Transduction , Transgenes/genetics
7.
J Pharmacol Exp Ther ; 333(2): 393-403, 2010 May.
Article in English | MEDLINE | ID: mdl-20110378

ABSTRACT

Signaling through G protein-coupled receptors (GPCRs) promotes breast cancer metastasis. G proteins convey GPCR signals by dissociating into Galpha and Gbetagamma subunits. The aim of the present study was to determine whether blockade of Gbetagamma signaling suppresses breast cancer cell migration and invasion, which are critical components of metastasis. Conditioned media (CM) of NIH-3T3 fibroblasts are widely used as chemoattractants in in vitro cancer metastasis studies. Expression of a Gbetagamma scavenger peptide attenuated NIH-3T3 CM-induced migration and invasion of both metastatic breast cancer MDA-MB-231 and MDA-MB-436 cells by 40 to 50% without effects on cell viability. Migration and invasion of cells in response to NIH-3T3 CM were also blocked by 8-(4,5,6-trihydroxy-3-oxo-3H-xanthen-9-yl)-1-naph-thalene-carboxylic acid) (M119K), a Gbetagamma inhibitor, with maximum inhibition exceeding 80% and half-maximal inhibitory concentration (IC50) values of 1 to 2 microM. M119K also attenuated Rac-dependent formation of lamellipodia, a key structure required for metastasis. Constitutively active Rac1 rescued Gbetagamma blockade-mediated inhibition of breast cancer cell migration, whereas dominant negative Rac1 inhibited cell migration similar to Gbetagamma blockade. Furthermore, M119K suppressed Gi protein-coupled CXC chemokine receptor 4 (CXCR4)-dependent MDA-MB-231 cell migration by 80% with an IC50 value of 1 microM, whereas tyrosine kinase receptor-dependent cell migration was significantly less inhibited. However, CXCR4-dependent inhibition of adenylyl cyclase, a Gialpha-mediated response in MDA-MB-231 cells, was not blocked by M119K but was blocked by pertussis toxin, which selectively inactivates Gialpha. This report is the first to directly demonstrate the role of Gbetagamma in cancer cell migration and invasion and suggests that targeting Gbetagamma signaling pathways may provide a novel strategy for suppressing breast cancer metastasis.


Subject(s)
Breast Neoplasms/physiopathology , Cell Movement/physiology , GTP-Binding Protein beta Subunits/pharmacology , GTP-Binding Protein gamma Subunits/pharmacology , Neoplasm Invasiveness/physiopathology , Adenylyl Cyclases/drug effects , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/physiology , Cyclohexanes/pharmacology , Female , Humans , Microscopy, Fluorescence , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/physiopathology , Peptide Fragments/physiology , Pseudopodia/drug effects , Receptors, CXCR4/physiology , Recombinant Proteins , Signal Transduction/drug effects , Signal Transduction/physiology , Xanthenes/pharmacology , rac GTP-Binding Proteins/drug effects , rac GTP-Binding Proteins/physiology
8.
Cell Signal ; 18(1): 62-8, 2006 Jan.
Article in English | MEDLINE | ID: mdl-15925485

ABSTRACT

The Ca2+-activated adenylyl cyclase type VIII (AC-VIII) has been implicated in several forms of neural plasticity, including drug addiction and learning and memory. It has not been clear whether Gi/o proteins and G-protein coupled receptors regulate the activity of AC-VIII. Here we show in intact mammalian cell system that AC-VIII is inhibited by mu-opioid receptor activation and that this inhibition is pertussis toxin sensitive. Moreover, we show that G(betagamma) subunits inhibit AC-VIII activity, while constitutively active alphai/o subunits do not. Different Gbeta isoforms varied in their efficacies, with Gbeta1gamma2 or Gbeta2gamma2 being more efficient than Gbeta3gamma2 and Gbeta4gamma2, while Gbeta5 (transfected with gamma2) had no effect. As for the Ggamma subunits, Gbeta1 inhibited AC-VIII activity in the presence of all gamma subunits tested except for gamma5 that had only a marginal activity. Moreover, cotransfection with proteins known to serve as scavengers of Gbetagamma dimers, or to reduce Gbetagamma plasma membrane anchorage, markedly attenuated the mu-opioid receptor-induced inhibition of AC-VIII. These results demonstrate that Gbetagamma (originating from agonist activation of these receptors) and probably not Galphai/o subunits are involved in the agonist inhibition of AC-VIII.


Subject(s)
Adenylyl Cyclases/metabolism , GTP-Binding Protein beta Subunits/pharmacology , GTP-Binding Protein gamma Subunits/pharmacology , Adenylyl Cyclase Inhibitors , Animals , COS Cells , Chlorocebus aethiops , Colforsin/pharmacology , Dimerization , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Ionomycin/pharmacology , Morphine/pharmacology , Pertussis Toxin/pharmacology , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/metabolism
9.
Nat Neurosci ; 8(9): 1160-8, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16116443

ABSTRACT

Exocytosis proceeds by either full fusion or 'kiss-and-run' between vesicle and plasma membrane. Switching between these two modes permits the cell to regulate the kinetics and amount of secretion. Here we show that ATP receptor activation reduces secretion downstream from cytosolic Ca2+ elevation in rat adrenal chromaffin cells. This reduction is mediated by activation of a pertussis toxin-sensitive G(i/o) protein, leading to activation of G(betagamma) subunits, which promote the 'kiss-and-run' mode by reducing the total open time of the fusion pore during a vesicle fusion event. Furthermore, parallel activation of the muscarinic acetylcholine receptor removes the inhibitory effects of ATP on secretion. This is mediated by a G(q) pathway through protein kinase C activation. The inhibitory effects of ATP and its reversal by protein kinase C activation are also shared by opioids and somatostatin. Thus, a variety of G protein pathways exist to modulate Ca2+-evoked secretion at specific steps in fusion pore formation.


Subject(s)
Chromaffin Cells/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Protein Kinase C/metabolism , Receptors, G-Protein-Coupled/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Action Potentials/radiation effects , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Adrenal Medulla/cytology , Analgesics, Opioid/pharmacology , Animals , Calcium/metabolism , Cells, Cultured , Chromaffin Cells/drug effects , Dose-Response Relationship, Radiation , Drug Interactions , Dynamins/pharmacology , Electric Stimulation/methods , Electrochemistry/methods , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , GTP-Binding Protein beta Subunits/pharmacology , Ionomycin/pharmacology , Ionophores/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/radiation effects , Muscarine/pharmacology , Neural Inhibition/drug effects , Neural Inhibition/physiology , Patch-Clamp Techniques/methods , Pertussis Toxin/pharmacology , Potassium Chloride/pharmacology , Protein Kinases/pharmacology , Rats , Recombinant Fusion Proteins/pharmacology , Somatostatin/pharmacology , Thionucleotides/pharmacology
10.
J Biol Chem ; 280(25): 23945-59, 2005 Jun 24.
Article in English | MEDLINE | ID: mdl-15824105

ABSTRACT

Voltage-gated Ca2+ channels of the N-, P/Q-, and R-type and G protein inwardly rectifying K+ channels (GIRK) are modulated via direct binding of G proteins. The modulation is mediated by G protein betagamma subunits. By using electrophysiological recordings and fluorescence resonance energy transfer, we characterized the modulatory domains of the G protein beta subunit on the recombinant P/Q-type channel and GIRK channel expressed in HEK293 cells and on native non-L-type Ca2+ currents of cultured hippocampal neurons. We found that Gbeta2 subunit-derived deletion constructs and synthesized peptides can either induce or inhibit G protein modulation of the examined ion channels. In particular, the 25-amino acid peptide derived from the Gbeta2 N terminus inhibits G protein modulation, whereas a 35-amino acid peptide derived from the Gbeta2 C terminus induced modulation of voltage-gated Ca2+ channels and GIRK channels. Fluorescence resonance energy transfer (FRET) analysis of the live action of these peptides revealed that the 25-amino acid peptide diminished the FRET signal between G protein beta2gamma3 subunits, indicating a reorientation between G protein beta2gamma3 subunits in the presence of the peptide. In contrast, the 35-amino acid peptide increased the FRET signal between GIRK1,2 channel subunits, similarly to the Gbetagamma-mediated FRET increase observed for this GIRK subunit combination. Circular dichroism spectra of the synthesized peptides suggest that the 25-amino acid peptide is structured. These results indicate that individual G protein beta subunit domains can act as independent, separate modulatory domains to either induce or inhibit G protein modulation for several effector proteins.


Subject(s)
Calcium Channels/metabolism , GTP-Binding Protein beta Subunits/pharmacology , GTP-Binding Proteins/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Amino Acid Sequence , Cell Line , Cells, Cultured , Fluorescence Resonance Energy Transfer , G Protein-Coupled Inwardly-Rectifying Potassium Channels , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/biosynthesis , Humans , Ion Channel Gating , Molecular Sequence Data , Sequence Deletion
11.
Nat Neurosci ; 8(4): 421-5, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15778713

ABSTRACT

The activation of G protein-coupled receptors (GPCRs) can result in an inhibition of Ca(2+)-dependent hormone and neurotransmitter secretion. This has been attributed in part to G protein inhibition of Ca(2+) influx. However, a frequently dominant inhibitory effect, of unknown mechanism, also occurs distal to Ca(2+) entry. Here we characterize direct inhibitory actions of G protein betagamma (Gbetagamma) on Ca(2+)-triggered vesicle exocytosis in permeable PC12 cells. Gbetagamma inhibition was rapid (<1 s) and was attenuated by cleavage of synaptosome-associated protein of 25 kD (SNAP25). Gbetagamma bound soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complexes, and binding was reduced to SNARE complexes containing cleaved SNAP25 or by Ca(2+)-dependent synaptotagmin binding. Here we show inhibitory coupling between GPCRs and vesicle exocytosis mediated directly by Gbetagamma interactions with the Ca(2+)-dependent fusion machinery.


Subject(s)
Exocytosis/physiology , GTP-Binding Protein beta Subunits/physiology , GTP-Binding Protein gamma Subunits/physiology , Secretory Vesicles/metabolism , Vesicular Transport Proteins/metabolism , Animals , Blotting, Western/methods , Calcium/pharmacology , Calcium-Binding Proteins/metabolism , Cattle , Dose-Response Relationship, Drug , Drug Interactions , Exocytosis/drug effects , Eye Proteins/pharmacology , GTP-Binding Protein Regulators , GTP-Binding Protein beta Subunits/pharmacology , GTP-Binding Protein gamma Subunits/pharmacology , Membrane Glycoproteins/metabolism , Membrane Proteins/pharmacology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/pharmacology , Neural Inhibition/drug effects , Norepinephrine/metabolism , PC12 Cells , Phosphoproteins/pharmacology , Protein Binding/drug effects , Rats , Reaction Time/drug effects , Recombinant Fusion Proteins/metabolism , SNARE Proteins , Secretory Vesicles/drug effects , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins , Synaptosomal-Associated Protein 25 , Synaptotagmins , Time Factors
12.
FEBS Lett ; 556(1-3): 187-92, 2004 Jan 02.
Article in English | MEDLINE | ID: mdl-14706848

ABSTRACT

The p21-activated kinase (PAK) family is homologous to the yeast sterile 20 (Ste20) and regulates a wide variety of cellular responses, including cell morphology, proliferation, and survival. In this study we examined the activation of PAK1 by Gbetagamma subunits. Co-transfection of COS7 cells with Gbeta1gamma2 or Gbeta1gamma5 was sufficient to induce agonist-independent activation of PAK1. Expression of dominant/negative Rac, Cdc42, or Ras did not inhibit this Gbetagamma-dependent activation. Wortmannin, which inhibits phosphoinositide 3-kinase (PI3-kinase) activity, and expression of a dominant/negative form of Akt were sufficient to abrogate the activation of PAK1 that was induced by Gbetagamma. These results reveal that stimulation of PAK1 by Gbetagamma can occur via a PI3-kinase and Akt pathway that does not require Rac1 or Cdc42.


Subject(s)
GTP-Binding Protein beta Subunits/pharmacology , GTP-Binding Protein gamma Subunits/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , Androstadienes/pharmacology , Animals , Blotting, Western , COS Cells , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/genetics , GTP-Binding Protein gamma Subunits/metabolism , Lysophospholipids/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Precipitin Tests , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/agonists , Proto-Oncogene Proteins c-akt , Transfection , Wortmannin , p21-Activated Kinases
13.
Circ Res ; 93(9): 848-56, 2003 Oct 31.
Article in English | MEDLINE | ID: mdl-14512443

ABSTRACT

Rho GTPases integrate the intracellular signaling in a wide range of cellular processes. Activation of these G proteins is tightly controlled by a number of guanine nucleotide exchange factors (GEFs). In this study, we addressed the functional role of the recently identified p114RhoGEF in in vivo experiments. Activation of endogenous G protein-coupled receptors with lysophosphatidic acid resulted in activation of a transcription factor, serum response element (SRE), that was enhanced by p114RhoGEF. This stimulation was inhibited by the functional scavenger of Gbetagamma subunits, transducin. We have determined that Gbetagamma subunits but not Galpha subunits of heterotrimeric G proteins stimulated p114RhoGEF-dependent SRE activity. Using coimmunoprecipitation assay, we have determined that Gbetagamma subunits interacted with full-length and DH/PH domain of p114RhoGEF. Similarly, Gbetagamma subunits stimulated SRE activity induced by full-length and DH/PH domain of p114RhoGEF. Using in vivo pull-down assays and dominant-negative mutants of Rho GTPases, we have determined that p114RhoGEF activated RhoA and Rac1 but not Cdc42 proteins. Functional significance of RhoA activation was established by the ability of p114RhoGEF to induce actin stress fibers and cell rounding. Functional significance of Rac1 activation was established by the ability of p114RhoGEF to induce production of reactive oxygen species (ROS) followed by activation of NADPH oxidase enzyme complex. In summary, our data showed that the novel guanine nucleotide exchange factor p114RhoGEF regulates the activity of RhoA and Rac1, and that Gbetagamma subunits of heterotrimeric G proteins are activators of p114RhoGEF under physiological conditions. The findings help to explain the integrated effects of LPA and other G-protein receptor-coupled agonists on actin stress fiber formation, cell shape change, and ROS production.


Subject(s)
GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Guanine Nucleotide Exchange Factors/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Actins/metabolism , Alternative Splicing , Animals , Cell Line , Cell Size/drug effects , Cell Size/physiology , GTP-Binding Protein beta Subunits/pharmacology , GTP-Binding Protein gamma Subunits/pharmacology , Genes, Reporter , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/drug effects , Guanine Nucleotide Exchange Factors/genetics , Humans , Lysophospholipids/pharmacology , Mice , NADPH Oxidases/metabolism , NIH 3T3 Cells , Organ Specificity , Phosphoproteins/metabolism , Protein Structure, Tertiary/physiology , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Reactive Oxygen Species/metabolism , Rho Guanine Nucleotide Exchange Factors , Serum Response Element/genetics , Stress Fibers/drug effects , Stress Fibers/metabolism , Thrombin/pharmacology , cdc42 GTP-Binding Protein/metabolism
14.
J Gen Physiol ; 121(6): 495-510, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12771191

ABSTRACT

Inhibition of N- (Cav2.2) and P/Q-type (Cav2.1) calcium channels by G-proteins contribute importantly to presynaptic inhibition as well as to the effects of opiates and cannabinoids. Accordingly, elucidating the molecular mechanisms underlying G-protein inhibition of voltage-gated calcium channels has been a major research focus. So far, inhibition is thought to result from the interaction of multiple proposed sites with the Gbetagamma complex (Gbetagamma). Far less is known about the important interaction sites on Gbetagamma itself. Here, we developed a novel electrophysiological paradigm, "compound-state willing-reluctant analysis," to describe Gbetagamma interaction with N- and P/Q-type channels, and to provide a sensitive and efficient screen for changes in modulatory behavior over a broad range of potentials. The analysis confirmed that the apparent (un)binding kinetics of Gbetagamma with N-type are twofold slower than with P/Q-type at the voltage extremes, and emphasized that the kinetic discrepancy increases up to ten-fold in the mid-voltage range. To further investigate apparent differences in modulatory behavior, we screened both channels for the effects of single point alanine mutations within four regions of Gbeta1, at residues known to interact with Galpha. These residues might thereby be expected to interact with channel effectors. Of eight mutations studied, six affected G-protein modulation of both N- and P/Q-type channels to varying degrees, and one had no appreciable effect on either channel. The remaining mutation was remarkable for selective attenuation of effects on P/Q-, but not N-type channels. Surprisingly, this mutation decreased the (un)binding rates without affecting its overall affinity. The latter mutation suggests that the binding surface on Gbetagamma for N- and P/Q-type channels are different. Also, the manner in which this last mutation affected P/Q-type channels suggests that some residues may be important for "steering" or guiding the protein into the binding pocket, whereas others are important for simply binding to the channel.


Subject(s)
Calcium Channels, N-Type/physiology , GTP-Binding Protein beta Subunits/pharmacology , Models, Theoretical , Alanine , Animals , Calcium Channels, N-Type/genetics , Electrophysiology , Humans , Kidney/cytology , Kidney/embryology , Point Mutation , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...