Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Biochem Pharmacol ; 216: 115795, 2023 10.
Article in English | MEDLINE | ID: mdl-37690571

ABSTRACT

Prolonged vasoconstrictor signalling found in hypertension, increases arterial contraction, and alters vessel architecture by stimulating arterial smooth muscle cell (ASMC) growth, underpinning the development of re-stenosis lesions and vascular remodelling. Vasoconstrictors interact with their cognate G protein coupled receptors activating a variety of signalling pathways to promote smooth muscle proliferation. Here, angiotensin II (AngII) and endothelin 1 (ET1), but not UTP stimulates ASMC proliferation. Moreover, siRNA-mediated depletion of endogenous GRK2 expression, or GRK2 inhibitors, compound 101 or paroxetine, prevented AngII and ET1-promoted ASMC growth. Depletion of GRK2 expression or inhibition of GRK2 activity ablated the prolonged phase of AngII and ET-stimulated ERK signalling, while enhancing and prolonging UTP-stimulated ERK signalling. Increased GRK2 expression enhanced and prolonged AngII and ET1-stimulated ERK signalling, but suppressed UTP-stimulated ERK signalling. In ASMC prepared from 6-week-old WKY and SHR, AngII and ET1-stimulated proliferation rates were similar, however, in cultures prepared from 12-week-old rats AngII and ET1-stimulated growth was enhanced in SHR-derived ASMC, which was reversed following depletion of GRK2 expression. Furthermore, in ASMC cultures isolated from 6-week-old WKY and SHR rats, AngII and ET1-stimulated ERK signals were similar, while in cultures from 12-week-old rats ERK signals were both enhanced and prolonged in SHR-derived ASMC, and were reversed to those seen in age-matched WKY-derived ASMC following pre-treatment of SHR-derived ASMC with compound 101. These data indicate that the presence of GRK2 and its catalytic activity are essential to enable pro-proliferative vasoconstrictors to promote growth via recruitment and activation of the ERK signalling pathway in ASMC.


Subject(s)
G-Protein-Coupled Receptor Kinase 2 , Hypertension , Vasoconstrictor Agents , Animals , Rats , Angiotensin II/pharmacology , Cell Proliferation , Cells, Cultured , Hypertension/metabolism , Muscle, Smooth, Vascular/metabolism , Rats, Inbred SHR , Rats, Inbred WKY , Uridine Triphosphate/pharmacology , Vasoconstrictor Agents/pharmacology , G-Protein-Coupled Receptor Kinase 2/metabolism
2.
Cell Signal ; 51: 86-98, 2018 11.
Article in English | MEDLINE | ID: mdl-30075183

ABSTRACT

Generation of cAMP through Gs-coupled G protein-coupled receptor (GPCR) [e.g. ß2-adrenoceptor (ß2AR), adenosine A2B receptor (A2BR)] activation, induces arterial smooth muscle relaxation, counteracting the actions of vasoconstrictors. Gs-coupled GPCR signalling is regulated by G protein-coupled receptor kinases (GRK) and arrestin proteins, and dysregulation of Gs/GPCR signalling is thought play a role in the development of hypertension, which may be a consequence of enhanced GRK2 and/or arrestin expression. However, despite numerous studies indicating that ß2AR and A2BR can be substrates for GRK/arrestin proteins, currently little is known regarding GRK/arrestin regulation of these endogenous receptors in arterial smooth muscle. Here, endogenous GRK isoenzymes and arrestin proteins were selectively depleted using RNA-interference in rat arterial smooth muscle cells (RASM) and the consequences of this for ß2AR- and A2BR-mediated adenylyl cyclase (AC) signalling were determined by assessing cAMP accumulation. GRK2 or GRK5 depletion enhanced and prolonged ß2AR/AC signalling, while combined deletion of GRK2/5 has an additive effect. Conversely, activation of AC by A2BR was regulated by GRK5, but not GRK2. ß2AR desensitization was attenuated following combined GRK2/GRK5 knockdown, but not by depletion of individual GRKs, arrestins, or by inhibiting PKA. Arrestin3 (but not arrestin2) depletion enhanced A2BR-AC signalling and attenuated A2BR desensitization, while ß2AR-AC signalling was regulated by both arrestin isoforms. This study provides a first demonstration of how different complements of GRK and arrestin proteins contribute to the regulation of signalling and desensitization of these important receptors mediating vasodilator responses in arterial smooth muscle.


Subject(s)
Aorta/metabolism , G-Protein-Coupled Receptor Kinase 2/physiology , G-Protein-Coupled Receptor Kinase 5/physiology , G-Protein-Coupled Receptor Kinases/physiology , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism , Receptor, Adenosine A2B/metabolism , Receptors, Adrenergic, beta-2/metabolism , beta-Arrestin 2/physiology , Adenylyl Cyclases/metabolism , Animals , Aorta/cytology , Arrestins/genetics , Arrestins/physiology , Cells, Cultured , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 5/genetics , Muscle, Smooth/cytology , Myocytes, Smooth Muscle/cytology , Rats , Rats, Wistar , Signal Transduction , beta-Arrestin 2/genetics
3.
Mol Pharmacol ; 94(3): 1079-1091, 2018 09.
Article in English | MEDLINE | ID: mdl-29980659

ABSTRACT

Vasoconstrictor-driven G protein-coupled receptor (GPCR)/phospholipase C (PLC) signaling increases intracellular Ca2+ concentration to mediate arterial contraction. To counteract vasoconstrictor-induced contraction, GPCR/PLC signaling can be desensitized by G protein-coupled receptor kinases (GRKs), with GRK2 playing a predominant role in isolated arterial smooth muscle cells. In this study, we use an array of GRK2 inhibitors to assess their effects on the desensitization of UTP and angiotensin II (AngII)-mediated arterial contractions. The effects of GRK2 inhibitors on the desensitization of UTP- or AngII-stimulated mesenteric third-order arterial contractions, and PLC activity in isolated mesenteric smooth muscle cells (MSMC), were determined using wire myography and Ca2+ imaging, respectively. Applying a stimulation protocol to cause receptor desensitization resulted in reductions in UTP- and AngII-stimulated arterial contractions. Preincubation with the GRK2 inhibitor paroxetine almost completely prevented desensitization of UTP- and attenuated desensitization of AngII-stimulated arterial contractions. In contrast, fluoxetine was ineffective. Preincubation with alternative GRK2 inhibitors (Takeda compound 101 or CCG224063) also attenuated the desensitization of UTP-mediated arterial contractile responses. In isolated MSMC, paroxetine, Takeda compound 101, and CCG224063 also attenuated the desensitization of UTP- and AngII-stimulated increases in Ca2+, whereas fluoxetine did not. In human uterine smooth muscle cells, paroxetine reversed GRK2-mediated histamine H1 receptor desensitization, but not GRK6-mediated oxytocin receptor desensitization. Utilizing various small-molecule GRK2 inhibitors, we confirm that GRK2 plays a central role in regulating vasoconstrictor-mediated arterial tone, highlighting a potentially novel strategy for blood pressure regulation through targeting GRK2 function.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , G-Protein-Coupled Receptor Kinase 2/physiology , Muscle, Smooth, Vascular/physiology , Protein Kinase Inhibitors/pharmacology , Vasoconstriction/physiology , Vasoconstrictor Agents/pharmacology , Animals , Cell Line, Transformed , Dose-Response Relationship, Drug , Humans , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiology , Muscle, Smooth, Vascular/drug effects , Rats , Rats, Wistar , Vasoconstriction/drug effects
4.
Mol Pharmacol ; 94(1): 674-688, 2018 07.
Article in English | MEDLINE | ID: mdl-29724789

ABSTRACT

The structurally related, but distinct neuropeptides, neuromedin U (NmU) and neuromedin S (NmS) are ligands of two G protein-coupled NmU receptors (NMU1 and NMU2). Hypothalamic NMU2 regulates feeding behavior and energy expenditure and has therapeutic potential as an anti-obesity target, making an understanding of its signaling and regulation of particular interest. NMU2 binds both NmU and NmS with high affinity, resulting in receptor-ligand co-internalization. We have investigated whether receptor trafficking events post-internalization are biased by the ligand bound and can therefore influence signaling function. Using recombinant cell lines expressing human NMU2, we demonstrate that acute Ca2+ signaling responses to NmU or NmS are indistinguishable and that restoration of responsiveness (resensitization) requires receptor internalization and endosomal acidification. The rate of NMU2 resensitization is faster following NmU compared with NmS exposure, but is similar if endothelin-converting enzyme-1 activity is inhibited or knocked down. Although acute activation of extracellular signal-regulated kinase (ERK) is also similar, activation by NMU2 is longer lasting if NmS is the ligand. Furthermore, when cells are briefly challenged before removal of free, but not receptor-bound ligand, activation of ERK and p38 mitogen-activated protein kinase by NmS is more sustained. However, only NmU responses are potentiated and extended by endothelin-converting enzyme-1 inhibition. These data indicate that differential intracellular ligand processing produces different signaling and receptor resensitization profiles and add to the findings of other studies demonstrating that intracellular ligand processing can shape receptor behavior and signal transduction.


Subject(s)
Receptors, Neurotransmitter/metabolism , Signal Transduction/physiology , Calcium/metabolism , Calcium Signaling/physiology , Cell Line , Endothelin-Converting Enzymes/metabolism , Energy Metabolism , HEK293 Cells , Humans , Ligands , MAP Kinase Signaling System/physiology , Neuropeptides/metabolism , Obesity/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
5.
PLoS Biol ; 16(4): e2003611, 2018 04.
Article in English | MEDLINE | ID: mdl-29630591

ABSTRACT

Nitric oxide (NO) regulates neuronal function and thus is critical for tuning neuronal communication. Mechanisms by which NO modulates protein function and interaction include posttranslational modifications (PTMs) such as S-nitrosylation. Importantly, cross signaling between S-nitrosylation and prenylation can have major regulatory potential. However, the exact protein targets and resulting changes in function remain elusive. Here, we interrogated the role of NO-dependent PTMs and farnesylation in synaptic transmission. We found that NO compromises synaptic function at the Drosophila neuromuscular junction (NMJ) in a cGMP-independent manner. NO suppressed release and reduced the size of available vesicle pools, which was reversed by glutathione (GSH) and occluded by genetic up-regulation of GSH-generating and de-nitrosylating glutamate-cysteine-ligase and S-nitroso-glutathione reductase activities. Enhanced nitrergic activity led to S-nitrosylation of the fusion-clamp protein complexin (cpx) and altered its membrane association and interactions with active zone (AZ) and soluble N-ethyl-maleimide-sensitive fusion protein Attachment Protein Receptor (SNARE) proteins. Furthermore, genetic and pharmacological suppression of farnesylation and a nitrosylation mimetic mutant of cpx induced identical physiological and localization phenotypes as caused by NO. Together, our data provide evidence for a novel physiological nitrergic molecular switch involving S-nitrosylation, which reversibly suppresses farnesylation and thereby enhances the net-clamping function of cpx. These data illustrate a new mechanistic signaling pathway by which regulation of farnesylation can fine-tune synaptic release.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Nerve Tissue Proteins/metabolism , Neurotransmitter Agents/metabolism , Nitric Oxide/metabolism , Protein Processing, Post-Translational , Adaptor Proteins, Vesicular Transport/genetics , Aldehyde Oxidoreductases/genetics , Aldehyde Oxidoreductases/metabolism , Animals , Brain/metabolism , Cyclic GMP/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Glutamate-Cysteine Ligase/genetics , Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Larva/genetics , Larva/metabolism , Nerve Tissue Proteins/genetics , Neuromuscular Junction/cytology , Neuromuscular Junction/metabolism , Phenotype , Prenylation , SNARE Proteins/genetics , SNARE Proteins/metabolism , Synaptic Transmission , Synaptic Vesicles/metabolism
7.
J Biol Chem ; 291(17): 8862-75, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-26826123

ABSTRACT

Establishing the in vivo activation status of G protein-coupled receptors would not only indicate physiological roles of G protein-coupled receptors but would also aid drug discovery by establishing drug/receptor engagement. Here, we develop a phospho-specific antibody-based biosensor to detect activation of the M1 muscarinic acetylcholine receptor (M1 mAChR) in vitro and in vivo Mass spectrometry phosphoproteomics identified 14 sites of phosphorylation on the M1 mAChR. Phospho-specific antibodies to four of these sites established that serine at position 228 (Ser(228)) on the M1 mAChR showed extremely low levels of basal phosphorylation that were significantly up-regulated by orthosteric agonist stimulation. In addition, the M1 mAChR-positive allosteric modulator, 1-(4-methoxybenzyl)-4-oxo-1,4-dihydroquinoline-3-carboxylic acid, enhanced acetylcholine-mediated phosphorylation at Ser(228) These data supported the hypothesis that phosphorylation at Ser(228) was an indicator of M1 mAChR activation. This was further supported in vivo by the identification of phosphorylated Ser(228) on the M1 mAChR in the hippocampus of mice following administration of the muscarinic ligands xanomeline and 1-(4-methoxybenzyl)-4-oxo-1,4-dihydroquinoline-3-carboxylic acid. Finally, Ser(228) phosphorylation was seen to increase in the CA1 region of the hippocampus following memory acquisition, a response that correlated closely with up-regulation of CA1 neuronal activity. Thus, determining the phosphorylation status of the M1 mAChR at Ser(228) not only provides a means of establishing receptor activation following drug treatment both in vitro and in vivo but also allows for the mapping of the activation status of the M1 mAChR in the hippocampus following memory acquisition thereby establishing a link between M1 mAChR activation and hippocampus-based memory and learning.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/chemistry , Biosensing Techniques/methods , CA1 Region, Hippocampal/metabolism , Learning/physiology , Memory/physiology , Phosphoproteins/metabolism , Receptor, Muscarinic M1/metabolism , Animals , CA1 Region, Hippocampal/cytology , CHO Cells , Cricetinae , Cricetulus , Mice , Phosphoproteins/genetics , Phosphorylation/physiology , Receptor, Muscarinic M1/genetics
8.
Br J Pharmacol ; 173(5): 870-87, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26660275

ABSTRACT

BACKGROUND AND PURPOSE: We investigated the hypothesis that elevated glucose increases contractile responses in vascular smooth muscle and that this enhanced constriction occurs due to the glucose-induced PKC-dependent inhibition of voltage-gated potassium channels. EXPERIMENTAL APPROACH: Patch-clamp electrophysiology in rat isolated mesenteric arterial myocytes was performed to investigate the glucose-induced inhibition of voltage-gated potassium (Kv ) current. To determine the effects of glucose in whole vessel, wire myography was performed in rat mesenteric, porcine coronary and human internal mammary arteries. KEY RESULTS: Glucose-induced inhibition of Kv was PKC-dependent and could be pharmacologically dissected using PKC isoenzyme-specific inhibitors to reveal a PKCß-dependent component of Kv inhibition dominating between 0 and 10 mM glucose with an additional PKCα-dependent component becoming evident at concentrations greater than 10 mM. These findings were supported using wire myography in all artery types used, where contractile responses to vessel depolarization and vasoconstrictors were enhanced by increasing bathing glucose concentration, again with evidence for distinct and complementary PKCα/PKCß-mediated components. CONCLUSIONS AND IMPLICATIONS: Our results provide compelling evidence that glucose-induced PKCα/PKCß-mediated inhibition of Kv current in vascular smooth muscle causes an enhanced constrictor response. Inhibition of Kv current causes a significant depolarization of vascular myocytes leading to marked vasoconstriction. The PKC dependence of this enhanced constrictor response may present a potential therapeutic target for improving microvascular perfusion following percutaneous coronary intervention after myocardial infarction in hyperglycaemic patients.


Subject(s)
Coronary Vessels/drug effects , Glucose/pharmacology , Mammary Arteries/drug effects , Mesenteric Arteries/drug effects , Protein Kinase C beta/physiology , Protein Kinase C-alpha/physiology , Animals , Coronary Vessels/physiology , Humans , In Vitro Techniques , Isoenzymes/antagonists & inhibitors , Isoenzymes/physiology , Male , Mammary Arteries/physiology , Mesenteric Arteries/physiology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Potassium Channels, Voltage-Gated/physiology , Protein Kinase C beta/antagonists & inhibitors , Protein Kinase C-alpha/antagonists & inhibitors , Rats, Wistar , Swine , Vasoconstriction/drug effects
10.
Am J Physiol Cell Physiol ; 309(3): C179-89, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25972452

ABSTRACT

Prolonged vasoconstrictor-stimulated phospholipase C activity can induce arterial constriction, hypertension, and smooth muscle hypertrophy/hyperplasia. Arrestin proteins are recruited by agonist-occupied G protein-coupled receptors to terminate signaling and counteract changes in vascular tone. Here we determine whether the development of hypertension affects arrestin expression in resistance arteries and how such changes alter arterial contractile signaling and function. Arrestin2/3 expression was increased in mesenteric arteries of 12-wk-old spontaneously hypertensive rats (SHR) compared with normotensive Wistar-Kyoto (WKY) controls, while no differences in arrestin expression were observed between 6-wk-old SHR and WKY animals. In mesenteric artery myography experiments, high extracellular K(+)-stimulated contractions were increased in both 6- and 12-wk-old SHR animals. Concentration-response experiments for uridine 5'-triphosphate (UTP) acting through P2Y receptors displayed a leftward shift in 12-wk, but not 6-wk-old animals. Desensitization of UTP-stimulated vessel contractions was increased in 12-wk-old (but not 6-wk-old) SHR animals. Dual IP3/Ca(2+) imaging in mesenteric arterial cells showed that desensitization of UTP and endothelin-1 (ET1) responses was enhanced in 12-wk-old (but not 6-wk-old) SHR compared with WKY rats. siRNA-mediated depletion of arrestin2 for UTP and arrestin3 for ET1, reversed the desensitization of PLC signaling. In conclusion, arrestin2 and 3 expression is elevated in resistance arteries during the emergence of the early hypertensive phenotype, which underlies an enhanced ability to desensitize vasoconstrictor signaling and vessel contraction. Such regulatory changes may act to compensate for increased vasoconstrictor-induced vessel contraction.


Subject(s)
Arrestins/physiology , Hypertension/metabolism , Vasoconstriction/physiology , Animals , Dose-Response Relationship, Drug , Hypertension/pathology , Male , Mesenteric Arteries/metabolism , Mesenteric Arteries/pathology , Organ Culture Techniques , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , beta-Arrestins
11.
PLoS One ; 10(3): e0121285, 2015.
Article in English | MEDLINE | ID: mdl-25793374

ABSTRACT

Voltage-gated potassium channels (Kv) are important regulators of membrane potential in vascular smooth muscle cells, which is integral to controlling intracellular Ca2+ concentration and regulating vascular tone. Previous work indicates that Kv channels can be modulated by receptor-driven alterations of cyclic AMP-dependent protein kinase (PKA) activity. Here, we demonstrate that Kv channel activity is maintained by tonic activity of PKA. Whole-cell recording was used to assess the effect of manipulating PKA signalling on Kv and ATP-dependent K+ channels of rat mesenteric artery smooth muscle cells. Application of PKA inhibitors, KT5720 or H89, caused a significant inhibition of Kv currents. Tonic PKA-mediated activation of Kv appears maximal as application of isoprenaline (a ß-adrenoceptor agonist) or dibutyryl-cAMP failed to enhance Kv currents. We also show that this modulation of Kv by PKA can be reversed by protein phosphatase 2B/calcineurin (PP2B). PKA-dependent inhibition of Kv by KT5720 can be abrogated by pre-treatment with the PP2B inhibitor cyclosporin A, or inclusion of a PP2B auto-inhibitory peptide in the pipette solution. Finally, we demonstrate that tonic PKA-mediated modulation of Kv requires intact caveolae. Pre-treatment of the cells with methyl-ß-cyclodextrin to deplete cellular cholesterol, or adding caveolin-scaffolding domain peptide to the pipette solution to disrupt caveolae-dependent signalling each attenuated PKA-mediated modulation of the Kv current. These findings highlight a novel, caveolae-dependent, tonic modulatory role of PKA on Kv channels providing new insight into mechanisms and the potential for pharmacological manipulation of vascular tone.


Subject(s)
Calcineurin/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Ion Channel Gating , Mesenteric Arteries/cytology , Myocytes, Smooth Muscle/metabolism , Potassium Channels, Voltage-Gated/metabolism , A Kinase Anchor Proteins/metabolism , Adenylyl Cyclases/metabolism , Animals , Carbazoles/pharmacology , Caveolae/drug effects , Caveolae/metabolism , Dideoxyadenosine/pharmacology , Ion Channel Gating/drug effects , Isoproterenol/pharmacology , Male , Myocytes, Smooth Muscle/drug effects , Pyrroles/pharmacology , Rats, Wistar
12.
Respirology ; 19(5): 763-6, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24850215

ABSTRACT

The cause of airway smooth muscle (ASM) hypercontractility in asthma is not fully understood. The relationship of spontaneous intracellular calcium oscillation frequency in ASM to asthma severity was investigated. Oscillations were increased in subjects with impaired lung function abolished by extracellular calcium removal, attenuated by caffeine and unaffected by verapamil or nitrendipine. Whether modulation of increased spontaneous intracellular calcium oscillations in ASM from patients with impaired lung function represents a therapeutic target warrants further investigation.


Subject(s)
Asthma/physiopathology , Calcium Signaling/physiology , Muscle, Smooth/physiopathology , Pulmonary Disease, Chronic Obstructive/physiopathology , Respiratory Muscles/physiopathology , Severity of Illness Index , Adult , Aged , Biopsy , Caffeine/pharmacology , Calcium Signaling/drug effects , Case-Control Studies , Female , Forced Expiratory Volume/physiology , Humans , Male , Middle Aged , Muscle, Smooth/drug effects , Muscle, Smooth/pathology , Nitrendipine/pharmacology , Respiratory Muscles/drug effects , Respiratory Muscles/pathology , Verapamil/pharmacology , Vital Capacity/physiology
13.
Mol Pharmacol ; 86(2): 211-21, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24830940

ABSTRACT

Both human ether-à-go-go-related gene (hERG1) and the closely related human ether-à-go-go (hEAG1) channel are aberrantly expressed in a large proportion of human cancers. In the present study, we demonstrate that transfection of hERG1 into mouse fibroblasts is sufficient to induce many features characteristic of malignant transformation. An important finding of this work is that this transformation could be reversed by chronic incubation (for 2-3 weeks) with the hERG channel blocker dofetilide (100 nM), whereas more acute applications (for 1-2 days) were ineffective. The hERG1 expression resulted in a profound loss of cell contact inhibition, multiple layers of overgrowing cells, and high saturation densities. Cells also changed from fibroblast-like to a more spindle-shaped morphology, which was associated with a smaller cell size, a dramatic increase in cell polarization, a reduction in the number of actin stress fibers, and less punctate labeling of focal adhesions. Analysis of single-cell migration and scratch-wound closure clearly demonstrated that hERG1-expressing cells migrated more rapidly than vector-transfected control cells. In contrast to previous studies on hEAG1, there were no increases in rates of proliferation, or loss of growth factor dependency; however, hERG1-expressing cells were capable of substrate-independent growth. Allogeneic transplantation of hERG1-expressing cells into nude mice resulted in an increased incidence of tumors. In contrast to hEAG1, the mechanism of cellular transformation is dependent on ion conduction. Trafficking-deficient and conduction-deficient hERG1 mutants also prevented cellular transformation. These results provide evidence that hERG1 expression is sufficient to induce cellular transformation by a mechanism distinct from hEAG1. The most important conclusion of this study is that selective hERG1 channel blockers have therapeutic potential in the treatment of hERG1-expressing cancers.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/metabolism , Potassium Channel Blockers/pharmacology , Actins/metabolism , Animals , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/metabolism , Fibroblasts/drug effects , Focal Adhesions/metabolism , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Stress Fibers/metabolism , Transfection
14.
J Biol Chem ; 288(48): 34777-90, 2013 Nov 29.
Article in English | MEDLINE | ID: mdl-24133207

ABSTRACT

Class A G protein-coupled receptors (GPCRs) are able to form homodimers and/or oligomeric arrays. We recently proposed, based on bioluminescence resonance energy transfer studies with the M3 muscarinic receptor (M3R), a prototypic class A GPCR, that the M3R is able to form multiple, structurally distinct dimers that are probably transient in nature (McMillin, S. M., Heusel, M., Liu, T., Costanzi, S., and Wess, J. (2011) J. Biol. Chem. 286, 28584-28598). To provide more direct experimental support for this concept, we employed a disulfide cross-linking strategy to trap various M3R dimeric species present in a native lipid environment (transfected COS-7 cells). Disulfide cross-linking studies were carried out with many mutant M3Rs containing single cysteine (Cys) substitutions within two distinct cytoplasmic M3R regions, the C-terminal portion of the second intracellular loop (i2) and helix H8 (H8). The pattern of cross-links that we obtained, in combination with molecular modeling studies, was consistent with the existence of two structurally distinct M3R dimer interfaces, one involving i2/i2 contacts (TM4-TM5-i2 interface) and the other one characterized by H8-H8 interactions (TM1-TM2-H8 interface). Specific H8-H8 disulfide cross-links led to significant impairments in M3R-mediated G protein activation, suggesting that changes in the structural orientation or mobility of H8 are critical for efficient receptor-G protein coupling. Our findings provide novel structural and functional insights into the mechanisms involved in M3R dimerization (oligomerization). Because the M3R shows a high degree of sequence similarity with many other class A GPCRs, our findings should be of considerable general interest.


Subject(s)
Protein Conformation , Protein Multimerization , Receptor, Muscarinic M3/chemistry , Receptor, Muscarinic M3/metabolism , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Cysteine/chemistry , Mutagenesis, Site-Directed , Phosphatidylinositols/chemistry , Phosphatidylinositols/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Receptor, Muscarinic M3/genetics
15.
J Neurosci ; 33(13): 5626-37, 2013 Mar 27.
Article in English | MEDLINE | ID: mdl-23536077

ABSTRACT

Nitric oxide (NO) is an important signaling molecule with a variety of functions in the CNS, including a potential role in modulating neuronal growth and synapse formation. In the present study, we used tractable, identified neurons in the CNS of the pond snail Lymnaea stagnalis to study the role of endogenous NO signaling in neuronal growth and synaptic remodeling after nerve injury. Axonal damage of L. stagnalis neurons B1 and B2 induces extensive central growth of neurites that is accompanied by changes in existing electrical connections, the transient formation of novel electrical connections, and the formation of a novel excitatory chemical synapse from B2 to B1 neurons. Partial chronic inhibition of endogenous NO synthesis reduces neurite growth in NO-synthase-expressing B2, but has only minor effects on NOS-negative B1 neurons. Chronic application of an NO donor while inhibiting endogenous NO synthesis rescues neurite extension in B2 neurons and boosts growth of B1 neurons. Blocking soluble guanylate cyclase activity completely suppresses neurite extension and synaptic remodeling after nerve crush, demonstrating the importance of cGMP in these processes. Interestingly, inhibition of cGMP-dependent protein kinase only suppresses chemical synapse formation without effects on neuronal growth and electrical synapse remodeling. We conclude that NO signaling via cGMP is an important modulator of both neurite growth and synaptic remodeling after nerve crush. However, differential effects of cGMP-dependent protein kinase inhibition on neurite growth and synaptic remodeling suggest that these effects are mediated by separate signaling pathways.


Subject(s)
Axotomy , Central Nervous System/cytology , Cyclic GMP/metabolism , Neurites/physiology , Neurons/pathology , Nitric Oxide/metabolism , Synapses/physiology , Action Potentials/drug effects , Action Potentials/physiology , Analysis of Variance , Animals , Biophysics , Biotin/analogs & derivatives , Cells, Cultured , Cyclic GMP-Dependent Protein Kinases/metabolism , Electric Stimulation , Electrophysiology , Enzyme Inhibitors/pharmacology , Hydrazines , Iontophoresis , Neurites/drug effects , Neurons/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Organ Culture Techniques , Snails , Synapses/pathology
16.
Biochem Pharmacol ; 84(3): 249-59, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22531220

ABSTRACT

Work published over the past 10-15 years has caused the neuroscience community to engage in a process of constant re-evaluation of the roles of glial cells in the mammalian central nervous system. Recent emerging evidence suggests that, in addition to carrying out various homeostatic functions within the CNS, astrocytes can also engage in a two-way dialogue with neurons. Astrocytes possess many of the receptors, and some of the ion channels, present in neurons endowing them with an ability to sense and respond to an array of neuronal signals. In addition, an expanding number of small molecules and proteins have been shown to be released by astrocytes in both health and disease. In this commentary we will highlight advances in our understanding of G protein-coupled receptor signalling in astrocytes, with a particular emphasis on metabotropic glutamate (mGlu) receptors. Discussion will focus on the major mGlu receptors expressed in astrocytes, mGlu3 and mGlu5, how these receptors can influence different aspects of astrocyte physiology, and how signalling by these G protein-coupled receptors might change under pathophysiological circumstances.


Subject(s)
Astrocytes/physiology , Health Status , Receptors, G-Protein-Coupled/physiology , Receptors, Metabotropic Glutamate/physiology , Signal Transduction/physiology , Animals , Astrocytes/metabolism , Humans , Receptors, G-Protein-Coupled/metabolism , Receptors, Metabotropic Glutamate/metabolism
17.
PLoS One ; 7(1): e29946, 2012.
Article in English | MEDLINE | ID: mdl-22272263

ABSTRACT

BACKGROUND AND OBJECTIVE: Muscarinic acetylcholine receptors (mAChRs) are 7-transmembrane, G protein-coupled receptors that regulate a variety of physiological processes and represent potentially important targets for therapeutic intervention. mAChRs can be stimulated by full and partial orthosteric and allosteric agonists, however the relative abilities of such ligands to induce conformational changes in the receptor remain unclear. To gain further insight into the actions of mAChR agonists, we have developed a fluorescently tagged M(1) mAChR that reports ligand-induced conformational changes in real-time by changes in Förster resonance energy transfer (FRET). METHODS: Variants of CFP and YFP were inserted into the third intracellular loop and at the end of the C-terminus of the mouse M(1) mAChR, respectively. The optimized FRET receptor construct (M(1)-cam5) was expressed stably in HEK293 cells. RESULTS: The variant CFP/YFP-receptor chimera expressed predominantly at the plasma membrane of HEK293 cells and displayed ligand-binding affinities comparable with those of the wild-type receptor. It also retained an ability to interact with Gα(q/11) proteins and to stimulate phosphoinositide turnover, ERK1/2 phosphorylation and undergo agonist-dependent internalization. Addition of the full agonist methacholine caused a reversible decrease in M(1) FRET (F(EYFP)/F(ECFP)) that was prevented by atropine pre-addition and showed concentration-dependent amplitude and kinetics. Partial orthosteric agonists, arecoline and pilocarpine, as well as allosteric agonists, AC-42 and 77-LH-28-1, also caused atropine-sensitive decreases in the FRET signal, which were smaller in amplitude and significantly slower in onset compared to those evoked by methacholine. CONCLUSION: The M(1) FRET-based receptor chimera reports that allosteric and orthosteric agonists induce similar conformational changes in the third intracellular loop and/or C-terminus, and should prove to be a valuable molecular reagent for pharmacological and structural investigations of M(1) mAChR activation.


Subject(s)
Cell Membrane/metabolism , Fluorescence Resonance Energy Transfer/methods , Muscarinic Agonists/metabolism , Receptor, Muscarinic M1/metabolism , Animals , Arecoline/metabolism , Arecoline/pharmacology , Atropine/metabolism , Atropine/pharmacology , Binding, Competitive/drug effects , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Kinetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Methacholine Chloride/metabolism , Methacholine Chloride/pharmacology , Mice , Microscopy, Confocal , Muscarinic Agonists/pharmacology , Piperidines/metabolism , Piperidines/pharmacology , Receptor, Muscarinic M1/agonists , Receptor, Muscarinic M1/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection
18.
Am J Physiol Cell Physiol ; 302(5): C723-34, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22159081

ABSTRACT

Overstimulation of endothelin type A (ET(A)) and nucleotide (P2Y) Gα(q)-coupled receptors in vascular smooth muscle causes vasoconstriction, hypertension, and, eventually, hypertrophy and vascular occlusion. G protein-coupled receptor kinases (GRKs) and arrestin proteins are sequentially recruited by agonist-occupied Gα(q)-coupled receptors to terminate phospholipase C signaling, preventing prolonged/inappropriate contractile signaling. However, these proteins also play roles in the regulation of several mitogen-activated protein kinase (MAPK) signaling cascades known to be essential for vascular remodeling. Here we investigated whether different arrestin isoforms regulate endothelin and nucleotide receptor MAPK signaling in rat aortic smooth muscle cells (ASMCs). When intracellular Ca(2+) levels were assessed in isolated ASMCs loaded with Ca(2+)-sensitive dyes, P2Y(2) and ET(A) receptor desensitization was attenuated by selective small-interfering (si)RNA-mediated depletion of G protein-coupled receptor kinase 2 (GRK2). Using similar siRNA techniques, knockdown of arrestin2 prevented P2Y(2) receptor desensitization and enhanced and prolonged p38 and ERK MAPK signals, while arrestin3 depletion was ineffective. Conversely, arrestin3 knockdown prevented ET(A) receptor desensitization and attenuated ET1-stimulated p38 and ERK signals, while arrestin2 depletion had no effect. Using Transwell assays to assess agonist-stimulated ASMC migration, we found that UTP-stimulated migration was markedly attenuated following arrestin2 depletion, while ET1-stimulated migration was attenuated following knockdown of either arrestin. These data highlight a differential arrestin-dependent regulation of ET(A) and P2Y(2) receptor-stimulated MAPK signaling. GRK2 and arrestin expression are essential for agonist-stimulated ASMC migration, which, as a key process in vascular remodeling, highlights the potential roles of GRK2 and arrestin proteins in the progression of vascular disease.


Subject(s)
Arrestins/metabolism , Hypertension/metabolism , Muscle, Smooth, Vascular/metabolism , Receptor, Endothelin A/metabolism , Receptors, Purinergic P2Y2/metabolism , Animals , Arrestins/antagonists & inhibitors , Arrestins/genetics , Arteries/metabolism , Calcium/analysis , Cell Movement/physiology , Fura-2/analogs & derivatives , Fura-2/analysis , G-Protein-Coupled Receptor Kinase 2/metabolism , Gene Knockdown Techniques , Male , Mitogen-Activated Protein Kinases/metabolism , Myocytes, Smooth Muscle/chemistry , Myocytes, Smooth Muscle/metabolism , Phosphorylation , RNA, Small Interfering/metabolism , Rats , Rats, Wistar , Signal Transduction , Type C Phospholipases/metabolism
19.
Am J Respir Crit Care Med ; 185(3): 267-74, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22108207

ABSTRACT

RATIONALE: Asthma is characterized by disordered airway physiology as a consequence of increased airway smooth muscle contractility. The underlying cause of this hypercontractility is poorly understood. OBJECTIVES: We sought to investigate whether the burden of oxidative stress in airway smooth muscle in asthma is heightened and mediated by an intrinsic abnormality promoting hypercontractility. METHODS: We examined the oxidative stress burden of airway smooth muscle in bronchial biopsies and primary cells from subjects with asthma and healthy controls. We determined the expression of targets implicated in the control of oxidative stress in airway smooth muscle and their role in contractility. MEASUREMENTS AND MAIN RESULTS: We found that the oxidative stress burden in the airway smooth muscle in individuals with asthma is heightened and related to the degree of airflow obstruction and airway hyperresponsiveness. This was independent of the asthmatic environment as in vitro primary airway smooth muscle from individuals with asthma compared with healthy controls demonstrated increased oxidative stress-induced DNA damage together with an increased production of reactive oxygen species. Genome-wide microarray of primary airway smooth muscle identified increased messenger RNA expression in asthma of NADPH oxidase (NOX) subtype 4. This NOX4 overexpression in asthma was supported by quantitative polymerase chain reaction, confirmed at the protein level. Airway smooth muscle from individuals with asthma exhibited increased agonist-induced contraction. This was abrogated by NOX4 small interfering RNA knockdown and the pharmacological inhibitors diphenyleneiodonium and apocynin. CONCLUSIONS: Our findings support a critical role for NOX4 overexpression in asthma in the promotion of oxidative stress and consequent airway smooth muscle hypercontractility. This implicates NOX4 as a potential novel target for asthma therapy.


Subject(s)
Asthma/enzymology , Bronchi/physiopathology , Muscle Contraction/physiology , Muscle, Smooth/physiopathology , NADPH Oxidases/metabolism , Adult , Biomarkers/metabolism , Blotting, Western , Bronchi/enzymology , Case-Control Studies , DNA Damage , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Male , Middle Aged , Muscle, Smooth/enzymology , NADPH Oxidase 4 , Oligonucleotide Array Sequence Analysis , Oxidative Stress , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/metabolism
20.
FASEB J ; 26(2): 604-16, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22031716

ABSTRACT

To explore the structural mechanisms underlying the assembly and activation of family A GPCR dimers, we used the rat M(3) muscarinic acetylcholine receptor (M3R) as a model system. Studies with Cys-substituted mutant M3Rs expressed in COS-7 cells led to the identification of several mutant M3Rs that exclusively existed as cross-linked dimers under oxidizing conditions. The cross-linked residues were located at the bottom of transmembrane domain 5 (TM5) and within the N-terminal portion of the third intracellular loop (i3 loop). Studies with urea-stripped membranes demonstrated that M3R disulfide cross-linking did not require the presence of heterotrimeric G proteins. Molecular modeling studies indicated that the cross-linking data were in excellent agreement with the existence of a low-energy M3R dimer characterized by a TM5-TM5 interface. [(35)S]GTPγS binding/Gα(q/11) immunoprecipitation assays revealed that an M3R dimer that was cross-linked within the N-terminal portion of the i3 loop (264C) was functionally severely impaired (∼50% reduction in receptor-G-protein coupling, as compared to control M3R). These data support the novel concept that agonist-induced activation of M3R dimers requires a conformational change of the N-terminal segment of the i3 loop. Given the high degree of structural homology among family A GPCRs, these findings should be of broad significance.


Subject(s)
Receptor, Muscarinic M3/chemistry , Receptor, Muscarinic M3/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites/genetics , COS Cells , Chlorocebus aethiops , Cross-Linking Reagents , Dimerization , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Structure, Quaternary , Rats , Receptor, Muscarinic M3/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...