Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 295(10): 3316-3329, 2020 03 06.
Article in English | MEDLINE | ID: mdl-31964716

ABSTRACT

Phosphorylation of specific residues in the activation loops of AGC kinase group (protein kinase A, G, and C families) is required for activity of most of these kinases, including the catalytic subunit of PKA (PKAc). Although many phosphorylated AGC kinases are sensitive to phosphatase-mediated dephosphorylation, the PKAc activation loop uniquely resists dephosphorylation, rendering it "constitutively" phosphorylated in cells. Previous biophysical experiments and structural modeling have suggested that the N-terminal myristoylation signal and the C-terminal FXXF motif in PKAc regulate its thermal stability and catalysis. Here, using site-directed mutagenesis, molecular modeling, and in cell-free and cell-based systems, we demonstrate that substitutions of either the PKAc myristoylation signal or the FXXF motif only modestly reduce phosphorylation and fail to affect PKAc function in cells. However, we observed that these two sites cooperate with an N-terminal FXXW motif to cooperatively establish phosphatase resistance of PKAc while not affecting kinase-dependent phosphorylation of the activation loop. We noted that this tripartite cooperative mechanism of phosphatase resistance is functionally relevant, as demonstrated by changes in morphology, adhesion, and migration of human airway smooth muscle cells transfected with PKAc variants containing amino acid substitutions in these three sites. These findings establish that three allosteric sites located at the PKAc N and C termini coordinately regulate the phosphatase sensitivity of this enzyme. This cooperative mechanism of phosphatase resistance of AGC kinase opens new perspectives toward therapeutic manipulation of kinase signaling in disease.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Amino Acid Motifs , Catalytic Domain , Cell Adhesion , Cell Line , Cell Movement , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/genetics , Cytosol/metabolism , HEK293 Cells , Humans , Mutagenesis, Site-Directed , Phosphorylation , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Signal Transduction
2.
Am J Physiol Lung Cell Mol Physiol ; 310(4): L365-76, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26684251

ABSTRACT

Airway remodeling is a hallmark feature of asthma and chronic obstructive pulmonary disease. Clinical studies and animal models have demonstrated increased airway smooth muscle (ASM) mass, and ASM thickness is correlated with severity of the disease. Current medications control inflammation and reverse airway obstruction effectively but have limited effect on remodeling. Recently we identified the expression of bitter taste receptors (TAS2R) on ASM cells, and activation with known TAS2R agonists resulted in ASM relaxation and bronchodilation. These studies suggest that TAS2R can be used as new therapeutic targets in the treatment of obstructive lung diseases. To further establish their effectiveness, in this study we aimed to determine the effects of TAS2R agonists on ASM growth and promitogenic signaling. Pretreatment of healthy and asthmatic human ASM cells with TAS2R agonists resulted in a dose-dependent inhibition of ASM proliferation. The antimitogenic effect of TAS2R ligands was not dependent on activation of protein kinase A, protein kinase C, or high/intermediate-conductance calcium-activated K(+) channels. Immunoblot analyses revealed that TAS2R agonists inhibit growth factor-activated protein kinase B phosphorylation without affecting the availability of phosphatidylinositol 3,4,5-trisphosphate, suggesting TAS2R agonists block signaling downstream of phosphatidylinositol 3-kinase. Furthermore, the antimitogenic effect of TAS2R agonists involved inhibition of induced transcription factors (activator protein-1, signal transducer and activator of transcription-3, E2 factor, nuclear factor of activated T cells) and inhibition of expression of multiple cell cycle regulatory genes, suggesting a direct inhibition of cell cycle progression. Collectively, these findings establish the antimitogenic effect of TAS2R agonists and identify a novel class of receptors and signaling pathways that can be targeted to reduce or prevent airway remodeling as well as bronchoconstriction in obstructive airway disease.


Subject(s)
Bronchodilator Agents/pharmacology , Muscle, Smooth/drug effects , Myocytes, Smooth Muscle/drug effects , Receptors, G-Protein-Coupled/agonists , Respiratory System/drug effects , Asthma/drug therapy , Asthma/metabolism , Bronchoconstriction/drug effects , Calcium Signaling/drug effects , Humans , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Respiratory System/metabolism , Taste/physiology
3.
Biochem J ; 471(1): 37-51, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26201515

ABSTRACT

The Akt protein kinase, also known as protein kinase B, plays key roles in insulin receptor signalling and regulates cell growth, survival and metabolism. Recently, we described a mechanism to enhance Akt phosphorylation that restricts access of cellular phosphatases to the Akt activation loop (Thr(308) in Akt1 or protein kinase B isoform alpha) in an ATP-dependent manner. In the present paper, we describe a distinct mechanism to control Thr(308) dephosphorylation and thus Akt deactivation that depends on intramolecular interactions of Akt C-terminal sequences with its kinase domain. Modifications of amino acids surrounding the Akt1 C-terminal mTORC2 (mammalian target of rapamycin complex 2) phosphorylation site (Ser(473)) increased phosphatase resistance of the phosphorylated activation loop (pThr(308)) and amplified Akt phosphorylation. Furthermore, the phosphatase-resistant Akt was refractory to ceramide-dependent dephosphorylation and amplified insulin-dependent Thr(308) phosphorylation in a regulated fashion. Collectively, these results suggest that the Akt C-terminal hydrophobic groove is a target for the development of agents that enhance Akt phosphorylation by insulin.


Subject(s)
Insulin/metabolism , Multiprotein Complexes/metabolism , Protein Processing, Post-Translational/physiology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line , Enzyme Activation/physiology , Insulin/genetics , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/genetics , Phosphorylation/physiology , Proto-Oncogene Proteins c-akt/genetics , Rats , TOR Serine-Threonine Kinases/genetics
4.
FASEB J ; 29(10): 4227-35, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26103985

ABSTRACT

Arrestins have been shown to regulate numerous G protein-coupled receptors (GPCRs) in studies employing receptor/arrestin overexpression in artificial cell systems. Which arrestin isoforms regulate which GPCRs in primary cell types is poorly understood. We sought to determine the effect of ß-arrestin-1 or ß-arrestin-2 inhibition or gene ablation on signaling and function of multiple GPCRs endogenously expressed in airway smooth muscle (ASM). In vitro [second messenger (calcium, cAMP generation)], ex vivo (ASM tension generation in suspended airway), and in vivo (invasive airway resistance) analyses were performed on human ASM cells and murine airways/whole animal subject to ß-arrestin-1 or -2 knockdown or knockout (KO). In both human and murine model systems, knockdown or KO of ß-arrestin-2 relative to control missense small interfering RNA or wild-type mice selectively increased (40-60%) ß2-adrenoceptor signaling and function. ß-arrestin-1 knockdown or KO had no effect on signaling and function of ß2-adrenoceptor or numerous procontractile GPCRs, but selectively inhibited M3 muscarinic acetylcholine receptor signaling (∼50%) and function (∼25% ex vivo, >50% in vivo) without affecting EC50 values. Arrestin subtypes differentially regulate ASM GPCRs and ß-arrestin-1 inhibition represents a novel approach to managing bronchospasm in obstructive lung diseases.


Subject(s)
Arrestin/metabolism , Muscle, Smooth/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Arrestin/genetics , Arrestins/genetics , Arrestins/metabolism , Cells, Cultured , Cyclic AMP/metabolism , Humans , Immunoblotting , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle Contraction/genetics , Muscle Contraction/physiology , Muscle, Smooth/cytology , Muscle, Smooth/physiology , RNA Interference , Receptors, Adrenergic, beta/metabolism , Receptors, Muscarinic/metabolism , Respiratory System/metabolism , beta-Arrestin 1 , beta-Arrestin 2 , beta-Arrestins
5.
J Biol Chem ; 289(33): 23065-23074, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24973219

ABSTRACT

Inhaled ß-agonists are effective at reversing bronchoconstriction in asthma, but the mechanism by which they exert this effect is unclear and controversial. PKA is the historically accepted effector, although this assumption is made on the basis of associative and not direct evidence. Recent studies have asserted that exchange protein activated by cAMP (Epac), not PKA, mediates the relaxation of airway smooth muscle (ASM) observed with ß-agonist treatment. This study aims to clarify the role of PKA in the prorelaxant effects of ß-agonists on ASM. Inhibition of PKA activity via expression of the PKI and RevAB peptides results in increased ß-agonist-mediated cAMP release, abolishes the inhibitory effect of isoproterenol on histamine-induced intracellular calcium flux, and significantly attenuates histamine-stimulated MLC-20 phosphorylation. Analyses of ASM cell and tissue contraction demonstrate that PKA inhibition eliminates most, if not all, ß-agonist-mediated relaxation of contracted smooth muscle. Conversely, Epac knockdown had no effect on the regulation of contraction or procontractile signaling by isoproterenol. These findings suggest that PKA, not Epac, is the predominant and physiologically relevant effector through which ß-agonists exert their relaxant effects.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Isoproterenol/pharmacology , Muscle Relaxation/drug effects , Muscle, Smooth/metabolism , Respiratory System/metabolism , Cell Line , Guanine Nucleotide Exchange Factors/metabolism , Humans , Muscle, Smooth/cytology , Respiratory System/cytology
6.
FASEB J ; 28(2): 956-65, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24132623

ABSTRACT

To clarify the potential utility of targeting GRK2/3-mediated desensitization as a means of manipulating airway smooth muscle (ASM) contractile state, we assessed the specificity of GRK2/3 regulation of procontractile and relaxant G-protein-coupled receptors in ASM. Functional domains of GRK2/3 were stably expressed, or siRNA-mediated GRK2/3 knockdown was performed, in human ASM cultures, and agonist-induced signaling was assessed. Regulation of contraction of murine tracheal rings expressing GRK2 C terminus was also assessed. GRK2/3 knockdown or expression of the GRK2 C terminus caused a significant (∼ 30-90%) increase in maximal ß-agonist and histamine [phosphoinositide (PI) hydrolysis] signaling, without affecting the calculated EC50. GRK2 C-terminal expression did not affect signaling by methacholine, thrombin, or LTD4. Expression of the GRK2 N terminus or kinase-dead holo-GRK2 diminished (∼ 30-70%) both PI hydrolysis and Ca(2+) mobilization by every Gq-coupled receptor examined. Under conditions of GRK2 C-terminal expression, ß-agonist inhibition of methacholine-stimulated PI hydrolysis was greater. Finally, transgenic expression of the GRK2 C terminus in murine ASM enabled ∼ 30-50% greater ß-agonist-mediated relaxation of methacholine-induced contraction. Collectively these data demonstrate the relative selectivity of GRKs for the ß2AR in ASM and the ability to exploit GRK2/3 functional domains to render ASM hyporesponsive to contractile agents while increasing responsiveness to bronchodilating ß-agonist.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinase 3/metabolism , Muscle, Smooth/metabolism , Calcium/metabolism , Cyclic AMP/metabolism , G-Protein-Coupled Receptor Kinase 2/chemistry , G-Protein-Coupled Receptor Kinase 3/chemistry , Humans , RNA, Small Interfering , Receptors, G-Protein-Coupled/metabolism , Respiratory System/cytology , Signal Transduction/physiology
7.
J Am Soc Nephrol ; 21(10): 1745-55, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20798260

ABSTRACT

Proton receptors are G protein-coupled receptors that accept protons as ligands and function as pH sensors. One of the proton receptors, GPR4, is relatively abundant in the kidney, but its potential role in acid-base homeostasis is unknown. In this study, we examined the distribution of GPR4 in the kidney, its function in kidney epithelial cells, and the effects of its deletion on acid-base homeostasis. We observed GPR4 expression in the kidney cortex, in the outer and inner medulla, in isolated kidney collecting ducts, and in cultured outer and inner medullary collecting duct cells (mOMCD1 and mIMCD3). Cultured mOMCD1 cells exhibited pH-dependent accumulation of intracellular cAMP, characteristic of GPR4 activation; GPR4 knockdown attenuated this accumulation. In vivo, deletion of GPR4 decreased net acid secretion by the kidney and resulted in a nongap metabolic acidosis, indicating that GPR4 is required to maintain acid-base homeostasis. Collectively, these findings suggest that GPR4 is a pH sensor with an important role in regulating acid secretion in the kidney collecting duct.


Subject(s)
Acid-Base Equilibrium , Kidney Tubules, Collecting/metabolism , Receptors, G-Protein-Coupled/metabolism , Acidosis, Renal Tubular/metabolism , Acids/metabolism , Animals , Cell Line , Cyclic AMP/metabolism , Humans , Hydrogen-Ion Concentration , Mice , Mice, Knockout , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...