Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 166(3): 981-90, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22145625

ABSTRACT

BACKGROUND AND PURPOSE: Previous studies have linked a reduction in pH in airway, caused by either environmental factors, microaspiration of gastric acid or inflammation, with airway smooth muscle (ASM) contraction and increased airway resistance. Neural mechanisms have been shown to mediate airway contraction in response to reductions in airway pH to < 6.5; whether reduced extracellular pH (pHo) has direct effects on ASM is unknown. EXPERIMENTAL APPROACH: Intracellular signalling events stimulated by reduced pHo in human cultured ASM cells were examined by immunoblotting, phosphoinositide hydrolysis and calcium mobilization assays. ASM cell contractile state was examined using magnetic twisting cytometry. The expression of putative proton-sensing GPCRs in ASM was assessed by real-time PCR. The role of ovarian cancer G protein-coupled receptor 1 (OGR1 or GPR68) in acid-induced ASM signalling and contraction was assessed in cultures subjected to siRNA-mediated OGR1 knockdown. KEY RESULTS: ASM cells responded to incremental reductions in pHo (from pH 8.0 to pH 6.8) by activating multiple signalling pathways, involving p42/p44, PKB, PKA and calcium mobilization. Coincidently, ASM cells contracted in response to decreased pHo with similar 'dose'-dependence. Real-time PCR suggested OGR1 was the only proton-sensing GPCR expressed in ASM cells. Both acid-induced signalling (with the exception of PKB activation) and contraction were significantly attenuated by knockdown of OGR1. CONCLUSIONS AND IMPLICATIONS: These studies reveal OGR1 to be a physiologically relevant GPCR in ASM cells, capable of pleiotropic signalling and mediating contraction in response to small reductions in extracellular pH. Accordingly, ASM OGR1 may contribute to asthma pathology and represent a therapeutic target in obstructive lung diseases.


Subject(s)
Extracellular Fluid/chemistry , Muscle Contraction/drug effects , Muscle, Smooth/metabolism , Receptors, G-Protein-Coupled/physiology , Signal Transduction/physiology , Bronchi/cytology , Bronchi/drug effects , Cell Culture Techniques , Cells, Cultured , Cyclic AMP/metabolism , Cyclooxygenase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Humans , Hydrochloric Acid/pharmacology , Hydrogen-Ion Concentration , Indomethacin/pharmacology , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Real-Time Polymerase Chain Reaction , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Trachea/cytology , Trachea/drug effects
2.
Br J Pharmacol ; 163(1): 18-28, 2011 May.
Article in English | MEDLINE | ID: mdl-21175591

ABSTRACT

In the last two decades several significant changes have been proposed in the receptor theory that describes how ligands can interact with G protein-coupled receptors (GPCRs). Here we briefly summarize the evolution of receptor theory and detail recent prominent advances. These include: (i) the existence of spontaneously active GPCRs that are capable of signalling even though they are unoccupied by any ligand; (ii) the discovery of ligands that can inactivate these spontaneously active receptors; (iii) the notion that a ligand may simultaneously activate more than one GPCR signalling pathway; and (iv) the notion that certain ligands may be able to preferentially direct receptor signalling to a specific pathway. Because the data supporting these receptor theory ideas are derived primarily from studies using artificial expression systems, the physiological relevance of these new paradigms remains in question. As a potential example of how these new perspectives in receptor theory relate to drug actions and clinical outcomes, we discuss their relevance to the recent controversy regarding the chronic use of ß(2) -adrenoceptor agonists in the treatment of asthma.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Adrenergic beta-2 Receptor Agonists/therapeutic use , Receptors, Adrenergic, beta-2/metabolism , Animals , Asthma/drug therapy , Asthma/metabolism , Humans , Ligands , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
3.
Kidney Int ; 71(12): 1249-61, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17429344

ABSTRACT

We have shown that A1 adenosine receptors (A1ARs) are cytoprotective against renal tubular necrosis and apoptosis both in vivo and in vitro. To study the role of A1AR numbers on renal epithelial cell survival, we stably overexpressed the human A1 receptor in a porcine renal tubule cell line and utilized primary cultures of proximal tubules obtained from A1AR knockout mice. Receptor-overexpressing cells were protected against peroxide-induced necrosis and tumor necrosis factor-alpha/cycloheximide-induced apoptosis. Conversely, cultured proximal tubule cells from receptor knockout mice showed more necrotic and apoptotic cell loss than corresponding cells from wild-type mice. Overexpression of the receptor resulted in a significantly higher baseline expression of both total and phosphorylated heat-shock protein (HSP)27; the latter due to A1 receptor enhancement of p38 and AP2 mitogen-activated protein kinase activities. The resistance to cell death in the porcine cells was reversed by selective A1 receptor antagonism and by a selective inhibitor of HSP synthesis. Receptor activation in wild-type mice in vivo led to increased total and phosphorylated HSP27, whereas receptor knockout mice showed decreased baseline and adenosine-mediated HSP phosphorylation. These studies show that endogenous A1AR activation produces cytoprotective effects in renal proximal tubules by modulating HSP27 signaling pathways.


Subject(s)
Apoptosis , Heat-Shock Proteins/metabolism , Kidney Tubules/pathology , Receptor, Adenosine A1/metabolism , Adenosine A1 Receptor Antagonists , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/genetics , Humans , Intracellular Signaling Peptides and Proteins , Kidney Tubules/metabolism , Mice , Mice, Knockout , Necrosis , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Adenosine A1/genetics , Swine , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Clin Exp Allergy ; 35(1): 8-17, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15649260

ABSTRACT

The linear model of maturation of IFN-gamma-producing cells from a proliferative pool of type 2 cytokine-producing T cells represents a fundamental shift in interpreting how changes in cytokine production by T cell populations are regulated. A major tenet of this model is antigen-independent, bystander proliferation of type 2 T cells and their maturation to IFN-gamma+ cells. Both clinical observations and prevailing theories of immune system development in asthma are consistent with this highly interpretative in vitro model, which allows unambiguous characterization of the modulation of the intrinsic features of T cell proliferation and differentiation by environmental and genetic factors. Hypotheses based on the linear model of T cell maturation are readily testable and should lead to a greater understanding of not only allergen-specific responses, but also the non-specific, bystander effects associated with specific responses to allergens or pathogens. Topics to be discussed in the context of the linear model of T cell maturation in this review include: (1) allergic responses to an inciting allergen that may enhance sensitivity to subsequent yet different allergens; (2) dampening the preferential accumulation of type 2 T cells during a typical immune response against viral and bacterial pathogens; (3) allergen-independent sensitization in asthmatics: (4) the 'hygiene hypothesis' for the reported increased allergy development in industrialized countries; (5) elevated IFN-gamma levels in asthmatics, in addition to the expected high levels of type 2 cytokines; (6) testing the effects of inflammatory mediators, as well as various anti-inflammation therapies on T cell maturation; and (7) testing the influence of gene variation on T cell maturation.


Subject(s)
Asthma/immunology , Hypersensitivity/immunology , T-Lymphocytes/immunology , Bystander Effect , Cell Differentiation/physiology , Cytokines/immunology , Humans , Hypersensitivity/drug therapy , Hypersensitivity/genetics , Interferon-gamma/immunology , Models, Animal , Research Design
5.
Am J Physiol Lung Cell Mol Physiol ; 281(6): L1425-35, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11704539

ABSTRACT

Numerous in vitro and in vivo studies have implicated the cytokines interleukin-1 beta (IL-1 beta) and tumor necrosis factor-alpha (TNF-alpha) as mediators of airway inflammation and therefore potentially important substances in the pathogenesis of asthma. In this study, we examined the mechanisms by which IL-1 beta and TNF-alpha affect inhibition of cell growth, G protein-coupled receptor (GPCR) desensitization, and the recently reported adenylyl cyclase sensitization in human airway smooth muscle (HASM) cultures. Our findings demonstrate that adenylyl cyclase sensitization is independent of cytokine-mediated cyclooxygenase type 2 (COX-2) and prostaglandin E(2) (PGE(2)) induction, whereas COX-2 induction appears to be required for both growth inhibition and GPCR desensitization. However, GPCR desensitization was highly dependent on the presence of EGF during chronic treatment with cytokines, which could be explained by a synergistic effect of EGF on cytokine-mediated COX-2 and PGE(2) induction. Interestingly, various agents (including inhibitors of p42/p44 and p38 mitogen-activated protein kinase signaling) were significantly more effective in inhibiting cytokine-mediated PGE(2) induction, GPCR desensitization, and cell growth inhibition than in inhibiting COX-2 induction. These data demonstrate disparity in the requirement and sufficiency of COX-2 induction in promoting different functional effects of IL-1 beta and TNF-alpha in HASM.


Subject(s)
GTP-Binding Proteins/metabolism , Interleukin-1/pharmacology , JNK Mitogen-Activated Protein Kinases , MAP Kinase Signaling System/drug effects , Muscle, Smooth/metabolism , Trachea/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Adenylyl Cyclases/metabolism , Butadienes/pharmacology , Cell Division/drug effects , Cells, Cultured , Cyclooxygenase 2 , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , Humans , Imidazoles/pharmacology , Isoenzymes/metabolism , MAP Kinase Kinase 4 , Membrane Proteins , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth/cytology , Nitriles/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Pyridines/pharmacology , Receptors, Cell Surface/metabolism , Trachea/cytology , p38 Mitogen-Activated Protein Kinases
6.
J Biol Chem ; 276(35): 32648-56, 2001 Aug 31.
Article in English | MEDLINE | ID: mdl-11418617

ABSTRACT

Despite a widely accepted role of arrestins as "uncouplers" of G protein-coupled receptor (GPCR) signaling, few studies have demonstrated the ability of arrestins to affect second messenger generation by endogenously expressed receptors in intact cells. In this study we demonstrate arrestin specificity for endogenous GPCRs in primary cultures of human airway smooth muscle (HASM). Expression of arrestin-green fluorescent protein (ARR2-GFP or ARR3-GFP) chimeras in HASM significantly attenuated isoproterenol (beta(2)-adrenergic receptor (beta(2)AR)-mediated)- and 5'-(N-ethylcarboxamido)adenosine (A2b adenosine receptor-mediated)-stimulated cAMP production, with fluorescent microscopy demonstrating agonist-promoted redistribution of cellular ARR2-GFP into a punctate formation. Conversely, prostaglandin E(2) (PGE(2))-mediated cAMP production was unaffected by arrestin-GFP, and PGE(2) had little effect on arrestin-GFP distribution. The pharmacological profile of various selective EP receptor ligands suggested a predominantly EP2 receptor population in HASM. Further analysis in COS-1 cells revealed that ARR2-GFP expression increased agonist-promoted internalization of wild type beta(2)AR and EP4 receptors, whereas EP2 receptors remained resistant to internalization. However, expression of an arrestin whose binding to GPCRs is largely independent of receptor phosphorylation (ARR2(R169E)-GFP) enabled substantial agonist-promoted EP2 receptor internalization, increased beta(2)AR internalization to a greater extent than did ARR2-GFP, yet promoted EP4 receptor internalization to the same degree as did ARR2-GFP. Signaling via endogenous EP4 receptors in CHO-K1 cells was attenuated by ARR2-GFP expression, whereas ARR2(R169E)-GFP expression in HASM inhibited EP2 receptor-mediated cAMP production. These findings demonstrate differential effects of arrestins in altering endogenous GPCR signaling in a physiologically relevant cell type and reveal a variable dependence on receptor phosphorylation in dictating arrestin-receptor interaction.


Subject(s)
Arrestins/physiology , Muscle, Smooth/physiology , Phosphoproteins/physiology , Receptors, Adrenergic, beta-2/physiology , Receptors, Prostaglandin E/physiology , Receptors, Purinergic P1/physiology , Signal Transduction/physiology , Trachea/physiology , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Animals , Arrestins/genetics , CHO Cells , COS Cells , Cell Line , Cells, Cultured , Chlorocebus aethiops , Cricetinae , Cyclic AMP/metabolism , Dinoprostone/pharmacology , GTP-Binding Proteins/metabolism , Genes, Reporter , Green Fluorescent Proteins , Humans , Isoproterenol/pharmacology , Kinetics , Luminescent Proteins/genetics , Muscle, Smooth/cytology , Phosphoproteins/genetics , Phosphorylation , Protein Transport , Receptors, Adrenergic, beta-2/drug effects , Receptors, Prostaglandin E/drug effects , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Purinergic P1/drug effects , Recombinant Fusion Proteins/metabolism , Trachea/cytology , Transfection
8.
Am J Respir Cell Mol Biol ; 24(5): 633-9, 2001 May.
Article in English | MEDLINE | ID: mdl-11350835

ABSTRACT

Rhinovirus (RV) is a major cause of wheezing in asthmatics and has been reported to cause beta2 adrenergic receptor hyporesponsiveness in human airway smooth muscle (HASM) via cellular secretion of interleukin (IL)-1beta. We studied the effects of IL-1beta and RV on cyclic adenosine monophosphate (cAMP) production in HASM cells. Chronic incubation with IL-1beta or RV caused a significant increase (approximately 3- and approximately 2-fold, respectively) in forskolin (FSK)-stimulated cAMP production, suggesting a sensitization of adenylyl cyclase (AC). The observed augmentation of FSK-stimulated cAMP formation by IL-1beta was completely abrogated by pretreatment with an IL-1 receptor antagonist or cycloheximide, demonstrating that the effect is mediated via the IL-1 receptor 1 (IL-1R1) and that de novo protein synthesis is required. In contrast, RV-induced AC sensitization was not mediated via the IL-1R1 but was observed to be protein kinase C-dependent. We suggest that the sensitization of AC observed after exposure to IL-1beta or RV infection is a cellular defense mechanism to promote pathways that induce relaxation in the inflamed airway.


Subject(s)
Adenylyl Cyclases/metabolism , Interleukin-1/pharmacology , Muscle, Smooth/enzymology , Rhinovirus/metabolism , Trachea , Adenylate Cyclase Toxin , Adrenergic beta-Agonists/pharmacology , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/metabolism , Cycloheximide/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Feedback , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , Humans , Interleukin 1 Receptor Antagonist Protein , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Muscle, Smooth/virology , Picornaviridae Infections/enzymology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Synthesis Inhibitors/pharmacology , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/metabolism , Receptors, Interleukin-1 Type I , Sialoglycoproteins/pharmacology , Virulence Factors, Bordetella/pharmacology
9.
Am J Respir Cell Mol Biol ; 24(2): 155-63, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11159049

ABSTRACT

Adenosine is a mediator of bronchoconstriction in asthmatics and is believed to mediate its effects through adenosine receptor activation in inflammatory cells. In this study, we identify human airway smooth muscle (ASM) as a direct target of adenosine. Acute exposure of human ASM cultures to adenosine receptor (AR) agonists resulted in rapid accumulation of cyclic adenosine monophosphate (cAMP) with a pharmacologic profile consistent with A(2b)AR activation. Little or no evidence of A1AR or A3AR expression was suggested on acute addition of various AR ligands, although a low level of A1ARs was identified in radioligand binding studies. Treatment with adenosine deaminase suggested that human ASM cultures secrete adenosine that feeds back on A(2b)ARs and regulates basal cAMP levels as well as a small degree of A(2b)AR, beta(2)AR, and prostaglandin E(2) receptor desensitization. When subjected to chronic treatment with AR agonists or agents that enhance accumulation of endogenous, extracellular adenosine, a dual effect of A(2b)AR desensitization and adenylyl cyclase (AC) sensitization was observed. This AC sensitization was eliminated by pertussis toxin and partially reversed by the A1AR antagonist 8-cyclopentyl-1,3-dipropylxanthine, suggesting a contributory role for the A1AR. Overexpression of A1ARs and A(2b)ARs in human ASM cultures resulted in differential effects on basal, agonist-, and AC-mediated cAMP production. These data demonstrate that human ASM is a direct target of exogenous and autocrine adenosine, with effects determined by differential contributions of A(2b) and A1 adenosine receptors that are time-dependent. Accordingly, the relative distribution and activation of AR subtypes in ASM in vivo may influence airway function in diseases such as asthma and warrant consideration in therapeutic strategies that target ARs or alter nucleotide/ nucleoside levels in the airway.


Subject(s)
Adenosine/pharmacology , Adenylyl Cyclases/drug effects , Adenylyl Cyclases/metabolism , GTP-Binding Protein Regulators/pharmacology , Muscle, Smooth/drug effects , Respiratory System/drug effects , Adenosine/metabolism , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Cells, Cultured/drug effects , Cyclic AMP/biosynthesis , DNA Primers/chemistry , Fluorescence , Gene Expression Regulation, Enzymologic , Green Fluorescent Proteins , Humans , Isoenzymes/biosynthesis , Isoenzymes/metabolism , Luminescent Proteins/metabolism , Muscle, Smooth/cytology , Muscle, Smooth/enzymology , Polymerase Chain Reaction , Purinergic P1 Receptor Antagonists , RNA, Messenger/analysis , Receptor, Adenosine A2B , Respiratory System/cytology , Respiratory System/enzymology , Reverse Transcriptase Polymerase Chain Reaction , Transfection
10.
Am J Respir Cell Mol Biol ; 23(6): 794-802, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11104733

ABSTRACT

Although 3':5' cyclic adenosine monophosphate (cAMP) is known to modulate cytokine production in a number of cell types, little information exists regarding cAMP-mediated effects on this synthetic function of human airway smooth-muscle (HASM) cells. We examined the effect of increasing intracellular cAMP concentration ([cAMP](i)) on tumor necrosis factor (TNF)-alpha-induced regulated on activation, normal T cells expressed and secreted (RANTES) and interleukin (IL)-6 secretion from cultured HASM cells. Pretreatment of HASM with prostaglandin (PG) E(2), forskolin, or dibutyryl cAMP inhibited TNF-alpha-induced RANTES secretion but increased TNF-alpha-induced IL-6 secretion. Moreover, stimulation with PGE(2), forskolin, or dibutyryl cAMP alone increased basal IL-6 secretion in a concentration-dependent manner. SB 207499, a specific phosphodiesterase type 4 inhibitor, augmented the inhibitory effects of PGE(2) and forskolin on TNF-alpha-induced RANTES. Collectively, these data demonstrate that increasing [cAMP](i) in HASM effectively increases IL-6 secretion but reduces RANTES secretion promoted by TNF-alpha. Reverse transcriptase/polymerase chain reaction and ribonuclease protection assays suggested that these opposite effects of increased [cAMP](i) on TNF-alpha- induced IL-6 and RANTES secretion may occur at the transcriptional level. Accordingly, we examined the effects of TNF- alpha and cAMP on the regulation of nuclear factor (NF)-kappaB, a transcription factor known to modulate cytokine synthesis in numerous cell types. Stimulation of HASM cells with TNF-alpha increased NF-kappaB DNA-binding activity. However, increased [cAMP](i) in HASM neither activated NF-kappaB nor altered TNF-alpha- induced NF-kappaB DNA-binding activity. These results were confirmed using a NF-kappaB-luciferase reporter assay. Together, our data suggest that TNF-alpha-induced IL-6 and RANTES secretion may be associated with NF-kappaB activation, and that inhibition of TNF-alpha-stimulated RANTES secretion and augmentation of IL-6 secretion by increased [cAMP](i) in HASM cells occurs via an NF-kappaB-independent mechanism.


Subject(s)
Chemokine CCL5/metabolism , Interleukin-6/metabolism , Muscle, Smooth/drug effects , Trachea/drug effects , Tumor Necrosis Factor-alpha/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Bucladesine/pharmacology , Cells, Cultured , Chemokine CCL5/genetics , Colforsin/pharmacology , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4 , Cyclohexanecarboxylic Acids/pharmacology , Dinoprostone/pharmacology , Dose-Response Relationship, Drug , Humans , Interleukin-6/genetics , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , NF-kappa B/drug effects , NF-kappa B/metabolism , Nitriles , Phosphodiesterase Inhibitors/pharmacology , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors , Trachea/cytology , Trachea/metabolism , Transcription, Genetic/drug effects
11.
Am J Respir Cell Mol Biol ; 23(4): 546-54, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11017921

ABSTRACT

Despite recent studies depicting the capacity of G protein-coupled receptors (GPCRs) to activate mitogenic signaling pathways more commonly associated with receptor tyrosine kinases (RTKs), little is known regarding the interactive effects of GPCR and RTK activation on cell growth and signal transduction. Such interactions likely mediate the physiologic growth in most cells in vivo as well as the aberrant, non-neoplastic growth that occurs in diseases such as asthma, where disruptions of the local hormonal or inflammatory state can contribute to significant GPCR activation. In this study, we show that numerous inflammatory or contractile agents, including thrombin, histamine, and carbachol, potentiate epidermal growth factor (EGF)-stimulated proliferation of human airway smooth muscle (ASM), thus demonstrating a clear synergy between RTK and GPCR activation. Alterations in promitogenic nuclear signaling were evidenced by additive or synergistic increases in Elk-1 and activator protein-1 activation, and by increases in cyclin D1 expression. Interestingly, GPCR activation did not cause EGF receptor tyrosine phosphorylation nor did it increase EGF-stimulated autophosphorylation. In the presence of EGF, histamine or carbachol did not alter the time-dependent phosphorylation of p42/p44, whereas thrombin was capable of increasing phospho-p42/p44 levels at selected time points in some, but not all, cultures. In contrast to their relative inability to alter EGF receptor-linked p42/p44 activation, thrombin, histamine, and carbachol consistently increased the late phase (> 1 h) activity of p70 S6 kinase. Collectively, these findings suggest that inflammatory and contractile agents that activate GPCRs can significantly modulate RTK-mediated ASM growth through a p70 S6 kinase-dependent, p42/p44-independent mechanism.


Subject(s)
GTP-Binding Proteins/metabolism , Muscle, Smooth/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Trachea/drug effects , Carbachol/pharmacology , Cells, Cultured , Cyclin D1/metabolism , DNA Replication/drug effects , Enzyme Activation , Epidermal Growth Factor/pharmacology , Histamine/pharmacology , Humans , Muscle, Smooth/enzymology , Phosphorylation , Thrombin/pharmacology , Trachea/enzymology , Trachea/metabolism
12.
J Cell Sci ; 113 ( Pt 13): 2463-70, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10852825

ABSTRACT

Non-visual arrestins (arrestin-2 and arrestin-3) play critical roles in the desensitization and internalization of many G protein-coupled receptors. In vitro experiments have shown that both non-visual arrestins bind with high and approximately comparable affinities to activated, phosphorylated forms of receptors. They also exhibit high affinity binding, again of comparable magnitude, to clathrin. Further, agonist-promoted internalization of many receptors has been found to be stimulated by exogenous over-expression of either arrestin2 or arrestin3. The existence of multiple arrestins raises the question whether stimulated receptors are selective for a specific endogenous arrestin under more physiological conditions. Here we address this question in RBL-2H3 cells, a cell line that expresses comparable levels of endogenous arrestin-2 and arrestin-3. When (beta)(2)-adrenergic receptors are stably expressed in these cells the receptors internalize efficiently following agonist stimulation. However, by immunofluorescence microscopy we determine that only arrestin-3, but not arrestin-2, is rapidly recruited to clathrin coated pits upon receptor stimulation. Similarly, in RBL-2H3 cells that stably express physiological levels of m1AChR, the addition of carbachol selectively induces the localization of arrestin-3, but not arrestin-2, to coated pits. Thus, this work demonstrates coupling of G protein-coupled receptors to a specific non-visual arrestin in an in vivo setting.


Subject(s)
Arrestins/metabolism , Clathrin/metabolism , Coated Pits, Cell-Membrane/metabolism , GTP-Binding Proteins/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Phosphoproteins/metabolism , Receptor, Adenosine A3 , Receptor, Muscarinic M1 , Receptors, Adrenergic, beta-2/metabolism , Receptors, Cell Surface/drug effects , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/metabolism , Receptors, Purinergic P1/drug effects , Receptors, Purinergic P1/metabolism , Tumor Cells, Cultured
13.
Circulation ; 101(17): 2083-9, 2000 May 02.
Article in English | MEDLINE | ID: mdl-10790351

ABSTRACT

BACKGROUND: Desensitization of G protein-coupled receptors (GPCR) is emerging as an important feature of several cardiovascular diseases. G protein-coupled receptor kinase 2 (GRK2) plays a key role in the regulation of a variety of these receptors, and its cardiac expression levels are altered in pathological situations such as chronic heart failure. However, very little is known about the signals and mechanisms that modulate GRK2 expression in cardiovascular cells. METHODS AND RESULTS: We have studied the transcriptional activity of the 1.6-kb-long proximal genomic region of the human GRK2 gene. In an aortic smooth muscle cell line, agents that lead to physiological vasoconstriction and hypertrophy, such as phorbol esters, increased GRK2 promoter activity. Activation of signaling pathways by cotransfected G(alphaq) subunits or alpha(1)-adrenergic receptors also markedly enhanced the expression of the GRK2 promoter constructs. Conversely, proinflammatory cytokines, such as interleukin-1beta, tumor necrosis factor-alpha, or interferon-gamma, led to the opposite effect, decreasing the activity of the GRK2 promoter. CONCLUSIONS: Our results suggest that the expression of GRK2 in vascular cells is tightly controlled at the transcriptional level by the interplay between several extracellular messengers, which may trigger alterations of normal GRK2 levels in some physiopathological circumstances, thus promoting changes in the efficacy of the GPCR signal transduction.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/genetics , Muscle, Smooth, Vascular/metabolism , Promoter Regions, Genetic/drug effects , Animals , Aorta/metabolism , Cell Line , Cytokines/pharmacology , G-Protein-Coupled Receptor Kinase 2 , Humans , Phorbol Esters/pharmacology , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Tumor Cells, Cultured/metabolism , beta-Adrenergic Receptor Kinases
14.
Circ Res ; 86(1): 43-50, 2000.
Article in English | MEDLINE | ID: mdl-10625304

ABSTRACT

G protein-coupled receptor kinases (GRKs) phosphorylate activated G protein-coupled receptors, including alpha(1B)-adrenergic receptors (ARs), resulting in desensitization. In vivo analysis of GRK substrate selectivity has been limited. Therefore, we generated hybrid transgenic mice with myocardium-targeted overexpression of 1 of 3 GRKs expressed in the heart (GRK2 [commonly known as the beta-AR kinase 1], GRK3, or GRK5) with concomitant cardiac expression of a constitutively activated mutant (CAM) or wild-type alpha(1B)AR. Transgenic mice with cardiac CAMalpha(1B)AR overexpression had enhanced myocardial alpha(1)AR signaling and elevated heart-to-body weight ratios with ventricular atrial natriuretic factor expression denoting myocardial hypertrophy. Transgenic mouse hearts overexpressing only GRK2, GRK3, or GRK5 had no hypertrophy. In hybrid transgenic mice, enhanced in vivo signaling through CAMalpha(1B)ARs, as measured by myocardial diacylglycerol content, was attenuated by concomitant overexpression of GRK3 but not GRK2 or GRK5. CAMalpha(1B)AR-induced hypertrophy and ventricular atrial natriuretic factor expression were significantly attenuated with either concurrent GRK3 or GRK5 overexpression. Similar GRK selectivity was seen in hybrid transgenic mice with wild-type alpha(1B)AR overexpression concurrently with a GRK. GRK2 overexpression was without effect on any in vivo CAM or wild-type alpha(1B)AR cardiac phenotype, which is in contrast to previously reported in vitro findings. Furthermore, endogenous myocardial alpha(1)AR mitogen-activated protein kinase signaling in single-GRK transgenic mice also exhibited selectivity, as GRK3 and GRK5 desensitized in vivo alpha(1)AR mitogen-activated protein kinase responses that were unaffected by GRK2 overexpression. Thus, these results demonstrate that GRKs differentially interact with alpha(1B)ARs in vivo such that GRK3 desensitizes all alpha(1B)AR signaling, whereas GRK5 has partial effects and, most interestingly, GRK2 has no effect on in vivo alpha(1B)AR signaling in the heart.


Subject(s)
Cardiomegaly/prevention & control , Cyclic AMP-Dependent Protein Kinases/metabolism , Myocardium/enzymology , Protein Serine-Threonine Kinases/metabolism , Animals , Atrial Natriuretic Factor/genetics , Cell Line , Diglycerides/metabolism , G-Protein-Coupled Receptor Kinase 3 , G-Protein-Coupled Receptor Kinase 5 , Gene Expression/physiology , Hybridization, Genetic , JNK Mitogen-Activated Protein Kinases , Mice , Mice, Transgenic/genetics , Mitogen-Activated Protein Kinases/metabolism , Mutation/physiology , RNA, Messenger/metabolism , Receptors, Adrenergic, alpha/genetics , Receptors, Adrenergic, alpha/metabolism , Transgenes/genetics , beta-Adrenergic Receptor Kinases
15.
Trends Cardiovasc Med ; 10(2): 81-9, 2000 Feb.
Article in English | MEDLINE | ID: mdl-11150735

ABSTRACT

G protein-coupled receptor kinases (GRKs) specifically interact with the agonist-activated form of G protein-coupled receptors (GPCRs) to effect receptor phosphorylation and desensitization. Recent studies demonstrate that GRK function is a highly regulated process, and it is perhaps in this manner that a handful of GRKs (7 have been identified to date) are able to regulate the responsiveness of numerous GPCRs in a given cell type in a coordinated manner. The mechanisms by which GRK activity is regulated can be divided into 3 categories: 1) subcellular localization; 2) alterations in intrinsic kinase activity; and 3) alterations in GRK expression levels. This review will summarize our current understanding of each of these regulatory processes, and offer explanations as to how such mechanisms influence GPCR regulation under various physiologic conditions.


Subject(s)
GTP-Binding Proteins/physiology , Phosphotransferases/physiology , Receptors, Cell Surface/physiology , Animals , Binding Sites/physiology , Catalysis , Heart Diseases/metabolism , Humans , Hypertension/metabolism , Protein Binding/physiology , Structure-Activity Relationship
16.
Am J Respir Cell Mol Biol ; 21(5): 597-606, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10536119

ABSTRACT

Beta-agonists, through activation of the beta(2)-adrenergic receptor (beta(2)AR)-G(s)-adenylyl cyclase (AC) pathway, promote bronchodilation via functional antagonism of airway smooth muscle (ASM) spasmogens associated with the asthmatic state. Although previous studies have demonstrated that beta(2)AR signaling in ASM is subject to homologous (beta-agonist-induced) beta(2)AR desensitization, the potential for inflammatory and contractile agents to impact beta(2)AR signaling in ASM through heterologous mechanisms has not been defined. Here we report that chronic exposure of human ASM (HASM) to carbachol, serotonin, the thromboxane analogue U46619, or histamine induced little change or a small increase in isoproterenol-stimulated cyclic adenosine monophosphate (cAMP) formation, but significantly increased cAMP formation elicited by stimulation with forskolin. This latter increase in intrinsic AC activity was largely reversed by pertussis toxin pretreatment, and was unaffected by protein kinase C inhibition. Analysis of both AC function and isoform expression supports a dominant role of AC VI in HASM, and points to important differences in ASM AC isoform expression among species. Additional studies identify AC as the limiting component in beta(2)AR-G(s)-AC signaling in HASM, and thus a potentially important target of therapeutic strategies designed to influence airway contractile state.


Subject(s)
Adenylyl Cyclases/metabolism , Cholinergic Agonists/pharmacology , Free Radical Scavengers/pharmacology , Muscle, Smooth/drug effects , Respiratory System/drug effects , Vasoconstrictor Agents/pharmacology , Adenylyl Cyclases/biosynthesis , Adrenergic beta-2 Receptor Agonists , Adrenergic beta-Agonists/pharmacology , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , GTP-Binding Protein alpha Subunits, Gs/metabolism , Histamine/pharmacology , Humans , Isoenzymes/biosynthesis , Isoenzymes/metabolism , Muscle, Smooth/cytology , Muscle, Smooth/enzymology , Receptors, Adrenergic, beta-2/metabolism , Respiratory System/cytology , Respiratory System/enzymology , Signal Transduction/drug effects
17.
Am J Physiol ; 277(3): L479-88, 1999 09.
Article in English | MEDLINE | ID: mdl-10484455

ABSTRACT

Asthma is frequently associated with abnormal airway smooth muscle (ASM) growth that may contribute to airway narrowing and hyperresponsiveness to contractile agents. Although numerous hormones and cytokines have been shown to induce human ASM (HASM) proliferation, the cellular and molecular mechanisms underlying HASM hyperplasia are largely unknown. Here we characterize the roles of the mitogen-activated protein kinase (MAPK) superfamily [p42/p44 MAPK, c-Jun amino-terminal kinase/stress-activated protein kinase (JNK/SAPK), and p38] in mediating hormone- and cytokine-induced HASM proliferation. Significant enhancement of [(3)H]thymidine incorporation in HASM cultures was observed only by treatment with agents (epidermal growth factor, platelet-derived growth factor, thrombin, and phorbol 12-myristate 13-acetate) that promoted a strong and sustained activation of p42/p44 MAPK. Significant activation of the JNK/SAPK and p38 pathways was only observed on stimulation with interleukin (IL)-1beta and tumor necrosis factor-alpha, agents that did not appreciably stimulate HASM proliferation. Two different inhibitors of MAPK/extracellular signal-regulated kinase kinase (MEK), PD-98059 and U-0126, inhibited mitogen-induced [3H]thymidine incorporation in a manner consistent with their ability to inhibit p42/p44 activation. Elk-1 and activator protein-1 reporter activation by mitogens was similarly inhibited by inhibition of MEK, suggesting a linkage between p42/p44 activation, transcription factor activation, and HASM proliferation. These findings establish a fundamental role for p42/p44 activation in regulating HASM proliferation and provide insight into species-specific differences observed among studies in ASM mitogenesis.


Subject(s)
DNA-Binding Proteins , Gene Expression Regulation , Mitogen-Activated Protein Kinases/genetics , Multigene Family/genetics , Muscle, Smooth/physiology , Protein Serine-Threonine Kinases , Trachea/physiology , Transcription Factors , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Activation/physiology , Humans , MAP Kinase Kinase 1 , MAP Kinase Kinase 2 , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/physiology , Mitosis/physiology , Muscle, Smooth/cytology , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Trachea/cytology , Transcription Factor AP-1/genetics , Transcription, Genetic , ets-Domain Protein Elk-1
18.
Am J Physiol ; 277(1): L65-78, 1999 07.
Article in English | MEDLINE | ID: mdl-10409232

ABSTRACT

Hypertrophy and hyperplasia of airway smooth muscle (ASM) are important pathological features that contribute to airflow obstruction in chronic severe asthma. Despite considerable research effort, the cellular mechanisms that modulate ASM growth remain unknown. Recent evidence suggests that mitogen-induced activation of phosphoinositide (PI)-specific phospholipase C (PLC) and PI-dependent calcium mobilization are neither sufficient nor necessary to stimulate human ASM proliferation. In this study, we identify phosphatidylinositol (PtdIns) 3-kinase as a key regulator of human ASM proliferation. Pretreatment of human ASM with the PtdIns 3-kinase inhibitors wortmannin and LY-294002 significantly reduced thrombin- and epidermal growth factor (EGF)-induced DNA synthesis (IC(50) approximately 10 nM and approximately 3 microM, respectively). In separate experiments, wortmannin and LY-294002 markedly inhibited PtdIns 3-kinase and 70-kDa S6 protein kinase (pp70(S6k)) activation induced by stimulation of human ASM cells with EGF and thrombin but had no effect on EGF- and thrombin-induced p42/p44 mitogen-activated protein kinase (MAPK) activation. The specificity of wortmannin and LY-294002 was further suggested by the demonstrated inability of these compounds to alter thrombin-induced calcium transients, total PI hydrolysis, or basal cAMP levels. Transient expression of constitutively active PtdIns 3-kinase (p110*) activated pp70(S6k), whereas a dominant-negative PtdIns 3-kinase (Deltap85) blocked EGF- and thrombin-stimulated pp70(S6k) activity. Collectively, these data suggest that activation of PtdIns 3-kinase is required for the mitogenic effect of EGF and thrombin in human ASM cells. Further investigation of the role of PtdIns 3-kinase may offer new therapeutic approaches in the treatment of diseases characterized by smooth muscle cell hyperplasia such as asthma and chronic bronchitis.


Subject(s)
Mitogens/pharmacology , Muscle, Smooth/cytology , Phosphatidylinositol 3-Kinases/physiology , Trachea/cytology , Androstadienes/pharmacology , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Chromones/pharmacology , Cyclic AMP/metabolism , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , Humans , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositols/metabolism , Phosphoinositide-3 Kinase Inhibitors , Ribosomal Protein S6 Kinases/metabolism , Thrombin/pharmacology , Wortmannin
19.
J Pharmacol Exp Ther ; 288(2): 428-37, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9918542

ABSTRACT

The use of pharmacological inhibitors of protein kinases represents a potentially powerful tool in dissecting the regulatory features of intracellular signaling pathways. However, although the in vitro potency, selectivity, and efficacy of numerous kinase inhibitors have been characterized, little is known regarding the usefulness of these compounds as inhibitors in intact cells. In attempting to characterize the role of protein kinase A (PKA) in regulating the beta-2 adrenergic receptor (AR) in human airway cells, we observed a seemingly profound capacity of the isoquinoline H-89, a potent and widely used PKA inhibitor, to attenuate agonist-mediated desensitization of the beta-2 AR. Although additional experiments identified H-89 as an effective inhibitor of intracellular PKA, extended analysis of the compound determined the principal effect of H-89 was via its action as a beta-2 AR antagonist. Pretreatment with or the acute addition of H-89 significantly attenuated isoproterenol-stimulated cAMP accumulation. In cells pretreated with H-89 and then washed extensively, the subsequent dose-dependent response to isoproterenol suggested beta-2 AR antagonism by retained H-89. Competition binding of [125I]iodopindolol established Ki values of approximately 180 nM and 350 nM for H-89 antagonism of beta-2 AR and beta-1 AR, respectively. Additional receptor binding studies suggest selectivity of H-89 for the beta-2 AR and beta-1 AR, although a weak antagonism (Ki values of approximately 10 microM or greater) of other G protein-coupled receptors was observed. Results from additional pharmacological and biochemical analyses of various protein kinase inhibitors further established the need for careful characterization of pharmacological inhibitors when used in intact cell models.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Enzyme Inhibitors/pharmacology , Isoquinolines/pharmacology , Protein Kinase Inhibitors , Receptors, Adrenergic, beta/drug effects , Receptors, Adrenergic, beta/metabolism , Sulfonamides , Adrenergic beta-Antagonists/metabolism , Animals , COS Cells , Cells, Cultured , Humans , Kidney/cytology , Kinetics , Ligands , Protein Binding/drug effects , Receptors, Adrenergic, beta-1/drug effects , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/drug effects , Receptors, Adrenergic, beta-2/metabolism , Trachea/cytology
20.
Am J Respir Cell Mol Biol ; 19(2): 338-48, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9698608

ABSTRACT

beta2-Adrenergic receptors (beta2ARs) are important regulators of airway smooth muscle tone, and beta-sympathomimetic drugs are the most widely used agents in asthma therapy and are universally recognized as the treatment of choice for acute asthma attacks. Despite the clinical importance of beta-agonists and a good understanding of their mechanism of action in airway smooth muscle relaxation, surprisingly little is known about the manner in which the beta2AR signaling pathway is regulated in human airway smooth muscle (HASM). In this communication, we characterize mechanisms underlying rapid desensitization of the HASM beta2AR-adenylyl cyclase (AC) pathway. Acute homologous desensitization of beta2AR-mediated cyclic adenosine monophosphate (cAMP) production was characterized by an approximately 60% loss of maximal responsiveness to isoproterenol (ISO) when cells were pretreated for 30 min with 1 microM ISO. Acute heterologous beta2AR desensitization was characterized by an approximately 20% and 30% loss of maximal responsiveness to ISO challenge when cells were pretreated with forskolin and prostaglandin E2 (PGE2), respectively. Each form of desensitization was also characterized by an increase in the EC50 for ISO. beta2AR sequestration was associated with but not required for homologous desensitization. However, sequestration was required for rapid resensitization. Minimal alterations in inherent AC activity were observed with both modes of desensitization, suggesting that the beta2AR is the principal locus of regulation. Protein kinase inhibition by staurosporine largely reversed heterologous beta2AR desensitization and had a small but significant effect on homologous desensitization. In contrast, bisindolylmaleimide IX, a specific PKC-inhibitor, had no effect on heterologous or homologous beta2AR desensitization, suggesting that staurosporine effects were mediated by PKA inhibition. Overexpression of the G protein-coupled receptor kinase GRK2 in HASM cultures enhanced homologous desensitization. These data suggest that HASM beta2ARs are highly susceptible to rapid desensitization by multiple agents, and identify both GRKs and PKA as important mediators of acute beta2AR desensitization.


Subject(s)
Adenylyl Cyclases/metabolism , Muscle, Smooth/metabolism , Receptors, Adrenergic, beta-2/metabolism , Trachea/metabolism , Adrenergic beta-2 Receptor Agonists , Adrenergic beta-Agonists/pharmacology , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Muscle, Smooth/cytology , Muscle, Smooth/enzymology , Protein Kinase C/metabolism , Signal Transduction , Trachea/cytology , Trachea/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...