Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
2.
J Pain ; 24(11): 1980-1993, 2023 11.
Article in English | MEDLINE | ID: mdl-37315729

ABSTRACT

Chemotherapy-induced peripheral neuropathy (CIPN) is a common, dose-limiting side effect of cancer therapy. Protease-activated receptor 2 (PAR2) is implicated in a variety of pathologies, including CIPN. In this study, we demonstrate the role of PAR2 expressed in sensory neurons in a paclitaxel (PTX)-induced model of CIPN in mice. PAR2 knockout/wildtype (WT) mice and mice with PAR2 ablated in sensory neurons were treated with PTX administered via intraperitoneal injection. In vivo behavioral studies were done in mice using von Frey filaments and the Mouse Grimace Scale. We then examined immunohistochemical staining of dorsal root ganglion (DRG) and hind paw skin samples from CIPN mice to measure satellite cell gliosis and intra-epidermal nerve fiber (IENF) density. The pharmacological reversal of CIPN pain was tested with the PAR2 antagonist C781. Mechanical allodynia caused by PTX treatment was alleviated in PAR2 knockout mice of both sexes. In the PAR2 sensory neuronal conditional knockout (cKO) mice, both mechanical allodynia and facial grimacing were attenuated in mice of both sexes. In the DRG of the PTX-treated PAR2 cKO mice, satellite glial cell activation was reduced compared to control mice. IENF density analysis of the skin showed that the PTX-treated control mice had a reduction in nerve fiber density while the PAR2 cKO mice had a comparable skin innervation as the vehicle-treated animals. Similar results were seen with satellite cell gliosis in the DRG, where gliosis induced by PTX was absent in PAR cKO mice. Finally, C781 was able to transiently reverse established PTX-evoked mechanical allodynia. PERSPECTIVE: Our work demonstrates that PAR2 expressed in sensory neurons plays a key role in PTX-induced mechanical allodynia, spontaneous pain, and signs of neuropathy, suggesting PAR2 as a possible therapeutic target in multiple aspects of PTX CIPN.


Subject(s)
Paclitaxel , Peripheral Nervous System Diseases , Male , Female , Mice , Animals , Paclitaxel/adverse effects , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Receptor, PAR-2/genetics , Receptor, PAR-2/therapeutic use , Gliosis/chemically induced , Gliosis/complications , Gliosis/pathology , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/drug therapy , Pain/complications , Sensory Receptor Cells , Mice, Knockout , Ganglia, Spinal
3.
J Headache Pain ; 24(1): 42, 2023 Apr 19.
Article in English | MEDLINE | ID: mdl-37072694

ABSTRACT

BACKGROUND: Migraine is a severely debilitating disorder that affects millions of people worldwide. Studies have indicated that activation of protease-activated receptor-2 (PAR2) in the dura mater causes headache responses in preclinical models. It is also well known that vasodilators such as nitric oxide (NO) donors can trigger migraine attacks in migraine patients but not controls. In the current study we examined whether activation of PAR2 in the dura causes priming to the NO donor glyceryl trinitrate (GTN). METHODS: A preclinical behavioral model of migraine was used where stimuli (PAR2 agonists: 2at-LIGRL-NH2 (2AT) or neutrophil elastase (NE); and IL-6) were applied to the mouse dura through an injection made at the intersection of the lamdoidal and sagittal sutures on the skull. Following dural injection, periorbital von Frey thresholds and facial grimace responses were measured until their return to baseline. GTN was then given by intraperitoneal injection and periorbital hypersensitivity and facial grimace responses observed until they returned to baseline. RESULTS: We found that application of the selective PAR2 agonist 2at-LIGRL-NH2 (2AT) onto the dura causes headache-related behavioral responses in WT but not PAR2-/- mice with no differences between sexes. Additionally, dural PAR2 activation with 2AT caused priming to GTN (1 mg/kg) at 14 days after primary dural stimulation. PAR2-/- mice showed no priming to GTN. We also tested behavioral responses to the endogenous protease neutrophil elastase, which can cleave and activate PAR2. Dural neutrophil elastase caused both acute responses and priming to GTN in WT but not PAR2-/- mice. Finally, we show that dural IL-6 causes acute responses and priming to GTN that is identical in WT and PAR2-/- mice, indicating that IL-6 does not act through PAR2 in this model. CONCLUSIONS: These results indicate that PAR2 activation in the meninges can cause acute headache behavioral responses and priming to an NO donor, and support further exploration of PAR2 as a novel therapeutic target for migraine.


Subject(s)
Migraine Disorders , Nitroglycerin , Mice , Animals , Nitroglycerin/pharmacology , Leukocyte Elastase , Receptor, PAR-2 , Interleukin-6 , Migraine Disorders/chemically induced , Dura Mater , Headache , Disease Models, Animal
4.
Br J Pharmacol ; 180(5): 667-680, 2023 03.
Article in English | MEDLINE | ID: mdl-35735078

ABSTRACT

BACKGROUND AND PURPOSE: Asthma is a heterogenous disease strongly associated with inflammation that has many different causes and triggers. Current asthma treatments target symptoms such as bronchoconstriction and airway inflammation. Despite recent advances in biological therapies, there remains a need for new classes of therapeutic agents with novel, upstream targets. The proteinase-activated receptor-2 (PAR2) has long been implicated in allergic airway inflammation and asthma and it remains an intriguing target for novel therapies. Here, we describe the actions of C781, a newly developed low MW PAR2 biased antagonist, in vitro and in vivo in the context of acute allergen exposure. EXPERIMENTAL APPROACH: A human bronchial epithelial cell line expressing PAR2 (16HBE14o- cells) was used to evaluate the modulation in vitro, by C781, of physiological responses to PAR2 activation and downstream ß-arrestin/MAPK and Gq/Ca2+ signalling. Acute Alternaria alternata sensitized and challenged mice were used to evaluate C781 as a prophylactically administered modulator of airway hyperresponsiveness, inflammation and mucus overproduction in vivo. KEY RESULTS: C781 reduced in vitro physiological signalling in response to ligand and proteinase activation. C781 effectively antagonized ß-arrestin/MAPK signalling without significant effect on Gq/Ca2+ signalling in vitro. Given prophylactically, C781 modulated airway hyperresponsiveness, airway inflammation and mucus overproduction of the small airways in an acute allergen-challenged mouse model. CONCLUSION AND IMPLICATIONS: Our work demonstrates the first biased PAR2 antagonist for ß-arrestin/MAPK signalling. C781 is efficacious as a prophylactic treatment for allergen-induced airway hyperresponsiveness and inflammation in mice. It exemplifies a key pharmacophore for PAR2 that can be optimized for clinical development.


Subject(s)
Asthma , Bronchial Hyperreactivity , Respiratory Hypersensitivity , Mice , Humans , Animals , Allergens , Receptor, PAR-2 , beta-Arrestins , Asthma/drug therapy , Respiratory Hypersensitivity/drug therapy , beta-Arrestin 1 , Inflammation/drug therapy , Mice, Inbred BALB C , Lung , Bronchial Hyperreactivity/drug therapy
5.
J Pain ; 24(4): 605-616, 2023 04.
Article in English | MEDLINE | ID: mdl-36417966

ABSTRACT

Given the limited options and often harmful side effects of current analgesics and the suffering caused by the opioid crisis, new classes of pain therapeutics are needed. Protease-activated receptors (PARs), particularly PAR2, are implicated in a variety of pathologies, including pain. Since the discovery of the role of PAR2 in pain, development of potent and specific antagonists has been slow. In this study, we describe the in vivo characterization of a novel small molecule/peptidomimetic hybrid compound, C781, as a ß-arrestin-biased PAR2 antagonist. In vivo behavioral studies were done in mice using von Frey filaments and the Mouse Grimace Scale. Pharmacokinetic studies were done to assess pharmacokinetic/pharmacodynamic relationship in vivo. We used both prevention and reversal paradigms with protease treatment to determine whether C781 could attenuate protease-evoked pain. C781 effectively prevented and reversed mechanical and spontaneous nociceptive behaviors in response to small molecule PAR2 agonists, mast cell activators, and neutrophil elastase. The ED50 of C781 (intraperitoneal dosing) for inhibition of PAR2 agonist (20.9 ng 2-AT)-evoked nociception was 6.3 mg/kg. C781 was not efficacious in the carrageenan inflammation model. Pharmacokinetic studies indicated limited long-term systemic bioavailability for C781 suggesting that optimizing pharmacokinetic properties could improve in vivo efficacy. Our work demonstrates in vivo efficacy of a biased PAR2 antagonist that selectively inhibits ß-arrestin/MAPK signaling downstream of PAR2. Given the importance of this signaling pathway in PAR2-evoked nociception, C781 exemplifies a key pharmacophore for PAR2 that can be optimized for clinical development. PERSPECTIVE: Our work provides evidence that PAR2 antagonists that only block certain aspects of signaling by the receptor can be effective for blocking protease-evoked pain in mice. This is important because it creates a rationale for developing safer PAR2-targeting approaches for pain treatment.


Subject(s)
Peptide Hydrolases , Receptor, PAR-2 , Mice , Animals , Peptide Hydrolases/metabolism , Peptide Hydrolases/pharmacology , beta-Arrestins/metabolism , beta-Arrestins/pharmacology , Receptor, PAR-2/metabolism , Pain/drug therapy , Pain/metabolism , Signal Transduction/physiology
6.
Biochem Biophys Res Commun ; 591: 13-19, 2022 02 05.
Article in English | MEDLINE | ID: mdl-34990903

ABSTRACT

Inhalation of the fungus Alternaria alternata is associated with an increased risk of allergic asthma development and exacerbations. Recent work in acute exposure animal models suggests that A. alternata-induced asthma symptoms, which include inflammation, mucus overproduction and airway hyperresponsiveness, are due to A. alternata proteases that act via protease-activated receptor-2 (PAR2). However, because other active components present in A. alternata may be contributing to asthma pathophysiology through alternative signaling, the specific role PAR2 plays in asthma initiation and maintenance remains undefined. Airway epithelial cells provide the first encounter with A. alternata and are thought to play an important role in initiating the physiologic response. To better understand the role for PAR2 airway epithelial signaling we created a PAR2-deficient human bronchial epithelial cell line (16HBEPAR-/-) from a model bronchial parental line (16HBE14o-). Comparison of in vitro physiologic responses in these cell lines demonstrated a complete loss of PAR2 agonist (2at-LIGRL-NH2) response and significantly attenuated protease (trypsin and elastase) and A. alternata responses in the 16HBEPAR-/- line. Apical application of A. alternata to 16HBE14o- and 16HBEPAR2-/- grown at air-liquid interface demonstrated rapid, PAR2-dependent and independent, inflammatory cytokine, chemokine and growth factor basolateral release. In conclusion, the novel human PAR2-deficient cell line allows for direct in vitro examination of the role(s) for PAR2 in allergen challenge with polarized human airway epithelial cells.


Subject(s)
Alternaria/physiology , Bronchi/pathology , Epithelial Cells/microbiology , Inflammation/pathology , Receptor, PAR-2/metabolism , Signal Transduction , Base Sequence , CRISPR-Cas Systems/genetics , Cell Line , Epithelial Cells/metabolism , Humans
7.
Br J Pharmacol ; 179(10): 2208-2222, 2022 05.
Article in English | MEDLINE | ID: mdl-34841515

ABSTRACT

BACKGROUND AND PURPOSE: Despite the availability of a variety of treatment options, many asthma patients have poorly controlled disease with frequent exacerbations. Proteinase-activated receptor-2 (PAR2) has been identified in preclinical animal models as important to asthma initiation and progression following allergen exposure. Proteinase activation of PAR2 raises intracellular Ca2+ , inducing MAPK and ß-arrestin signalling in the airway, leading to inflammatory and protective effects. We have developed C391, a potent PAR2 antagonist effective in blocking peptidomimetic- and trypsin-induced PAR2 signalling in vitro as well as reducing inflammatory PAR2-associated pain in vivo. We hypothesized that PAR2 antagonism by C391 would attenuate allergen-induced acutely expressed asthma indicators in murine models. EXPERIMENTAL APPROACH: We evaluated the ability of C391 to alter Alternaria alternata-induced PAR2 signalling pathways in vitro using a human airway epithelial cell line that naturally expresses PAR2 (16HBE14o-) and a transfected embryonic cell line (HEK 293). We next evaluated the ability for C391 to reduce A. alternata-induced acutely expressed asthma indicators in vivo in two murine strains. KEY RESULTS: C391 blocked A. alternata-induced, PAR2-dependent Ca2+ and MAPK signalling in 16HBE14o- cells, as well as ß-arrestin recruitment in HEK 293 cells. C391 effectively attenuated A. alternata-induced inflammation, mucus production, mucus cell hyperplasia and airway hyperresponsiveness in acute allergen-challenged murine models. CONCLUSIONS AND IMPLICATIONS: To our best knowledge, this is the first demonstration of pharmacological intervention of PAR2 to reduce allergen-induced asthma indicators in vivo. These data support further development of PAR2 antagonists as potential first-in-class allergic asthma drugs.


Subject(s)
Asthma , Receptor, PAR-2 , Allergens , Alternaria/metabolism , Animals , Asthma/drug therapy , Asthma/metabolism , HEK293 Cells , Humans , Mice
8.
Front Pain Res (Lausanne) ; 2: 695962, 2021.
Article in English | MEDLINE | ID: mdl-35295409

ABSTRACT

Mu opioid receptor (MOPr) agonists are well-known and frequently used clinical analgesics but are also rewarding due to their highly addictive and often abusive properties. This may lead to opioid use disorder (OUD) a disorder that effects millions of people worldwide. Therefore, novel compounds are urgently needed to treat OUD. As opioids are effective analgesics and OUD often occurs in conjunction with chronic pain, these novel compounds may be opioids, but they must have a low abuse liability. This could be mediated by diminishing or slowing blood-brain barrier transport, slowing target receptor binding kinetics, and showing a long half-life. NKTR-181 is a PEGylated oxycodol and a MOPr agonist that has slowed blood-brain barrier transport, a long half-life, and diminished likeability in clinical trials. In this study, we examined the signaling and behavioral profile of NKTR-181 in comparison with oxycodone to determine whether further therapeutic development of this compound may be warranted. For this preclinical study, we used a number of in vitro and in vivo assays. The signaling profile of NKTR-181 was determined by the electrophysiological assessment of MOPr-Ca2+ channel inhibition in the nociceptive neurons of rodent dorsal root ganglia. Heterologous cell-based assays were used to assess biased agonism and receptor trafficking. Different rodent behavioral models were used to define the NKTR-181-induced relief of effective and reflexive nociception and drug-seeking behavior as assessed by an intravenous self-administration (IVSA) of NKTR-181. We found that NKTR-181 and oxycodone are partial agonists in G-protein signaling and Ca2+ channel inhibition assays and promote limited MOPr desensitization. However, NKTR-181 inhibits Ca2+ channels by a different mechanism than oxycodone and induces a different pattern of arrestin recruitment. In addition, NKTR-181 has a slower receptor on-rate and a slower rate of Ca2+ channel coupling than oxycodone. This signaling profile is coupled with a slower onset of antinociception and limited drug-seeking behavior in comparison with oxycodone. Together with its known long half-life and slow blood-brain barrier transport, these data suggest that NKTR-181 could be further studied as a pharmacotherapeutic treatment modality for OUD.

9.
Am J Physiol Lung Cell Mol Physiol ; 315(6): L1042-L1057, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30335499

ABSTRACT

Alternaria alternata is a fungal allergen associated with severe asthma and asthma exacerbations. Similarly to other asthma-associated allergens, Alternaria secretes a serine-like trypsin protease(s) that is thought to act through the G protein-coupled receptor protease-activated receptor-2 (PAR2) to induce asthma symptoms. However, specific mechanisms underlying Alternaria-induced PAR2 activation and signaling remain ill-defined. We sought to determine whether Alternaria-induced PAR2 signaling contributed to asthma symptoms via a PAR2/ß-arrestin signaling axis, identify the protease activity responsible for PAR2 signaling, and determine whether protease activity was sufficient for Alternaria-induced asthma symptoms in animal models. We initially used in vitro models to demonstrate Alternaria-induced PAR2/ß-arrestin-2 signaling. Alternaria filtrates were then used to sensitize and challenge wild-type, PAR2-/- and ß-arrestin-2-/- mice in vivo. Intranasal administration of Alternaria filtrate resulted in a protease-dependent increase of airway inflammation and mucin production in wild-type but not PAR2-/- or ß-arrestin-2-/- mice. Protease was isolated from Alternaria preparations, and select in vitro and in vivo experiments were repeated to evaluate sufficiency of the isolated Alternaria protease to induce asthma phenotype. Administration of a single isolated serine protease from Alternaria, Alternaria alkaline serine protease (AASP), was sufficient to fully activate PAR2 signaling and induce ß-arrestin-2-/--dependent eosinophil and lymphocyte recruitment in vivo. In conclusion, Alternaria filtrates induce airway inflammation and mucus hyperplasia largely via AASP using the PAR2/ß-arrestin signaling axis. Thus, ß-arrestin-biased PAR2 antagonists represent novel therapeutic targets for treating aeroallergen-induced asthma.


Subject(s)
Inflammation/metabolism , Receptor, PAR-2/metabolism , Serine Proteases/metabolism , Signal Transduction/physiology , beta-Arrestin 2/metabolism , Allergens/metabolism , Animals , Asthma/metabolism , Bacterial Proteins/metabolism , Endopeptidases/metabolism , Lung/metabolism , Mice , Mice, Inbred C57BL , Serine/metabolism , Serine Endopeptidases/metabolism
10.
11.
J Cell Biol ; 212(7): 861-75, 2016 Mar 28.
Article in English | MEDLINE | ID: mdl-27002170

ABSTRACT

Dynamic changes in membrane protein composition of the primary cilium are central to development and homeostasis, but we know little about mechanisms regulating membrane protein flux. Stimulation of the sonic hedgehog (Shh) pathway in vertebrates results in accumulation and activation of the effector Smoothened within cilia and concomitant disappearance of a negative regulator, the orphan G protein-coupled receptor (GPCR), Gpr161. Here, we describe a two-step process determining removal of Gpr161 from cilia. The first step involves ß-arrestin recruitment by the signaling competent receptor, which is facilitated by the GPCR kinase Grk2. An essential factor here is the ciliary trafficking and activation of Smoothened, which by increasing Gpr161-ß-arrestin binding promotes Gpr161 removal, both during resting conditions and upon Shh pathway activation. The second step involves clathrin-mediated endocytosis, which functions outside of the ciliary compartment in coordinating Gpr161 removal. Mechanisms determining dynamic compartmentalization of Gpr161 in cilia define a new paradigm for down-regulation of GPCRs during developmental signaling from a specialized subcellular compartment.


Subject(s)
Arrestins/metabolism , Endocytosis , Fibroblasts/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Arrestins/genetics , Biosensing Techniques , Cilia/metabolism , Down-Regulation , G-Protein-Coupled Receptor Kinase 2/metabolism , HEK293 Cells , Hedgehog Proteins/metabolism , Humans , Mice , Mutation , NIH 3T3 Cells , Protein Binding , Protein Transport , RNA Interference , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Smoothened Receptor , Time Factors , Transfection , beta-Arrestins
12.
Br J Pharmacol ; 172(18): 4535-4545, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26140338

ABSTRACT

BACKGROUND AND PURPOSE: Proteinase-activated receptor-2 (PAR2) is a GPCR linked to diverse pathologies, including acute and chronic pain. PAR2 is one of the four PARs that are activated by proteolytic cleavage of the extracellular amino terminus, resulting in an exposed, tethered peptide agonist. Several peptide and peptidomimetic agonists, with high potency and efficacy, have been developed to probe the functions of PAR2, in vitro and in vivo. However, few similarly potent and effective antagonists have been described. EXPERIMENTAL APPROACH: We modified the peptidomimetic PAR2 agonist, 2-furoyl-LIGRLO-NH2 , to create a novel PAR2 peptidomimetic ligand, C391. C391 was evaluated for PAR2 agonist/antagonist activity to PAR2 across Gq signalling pathways using the naturally expressing PAR2 cell line 16HBE14o-. For antagonist studies, a highly potent and specific peptidomimetic agonist (2-aminothiazo-4-yl-LIGRL-NH2 ) and proteinase agonist (trypsin) were used to activate PAR2. C391 was also evaluated in vivo for reduction of thermal hyperalgesia, mediated by mast cell degranulation, in mice. KEY RESULTS: C391 is a potent and specific peptidomimetic antagonist, blocking multiple signalling pathways (Gq -dependent Ca2+ , MAPK) induced following peptidomimetic or proteinase activation of human PAR2. In a PAR2-dependent behavioural assay in mice, C391 dose-dependently (75 µg maximum effect) blocked the thermal hyperalgesia, mediated by mast cell degranulation. CONCLUSIONS AND IMPLICATIONS: C391 is the first low MW antagonist to block both PAR2 Ca2+ and MAPK signalling pathways activated by peptidomimetics and/or proteinase activation. C391 represents a new molecular structure for PAR2 antagonism and can serve as a basis for further development for this important therapeutic target.

13.
Handb Exp Pharmacol ; 219: 341-59, 2014.
Article in English | MEDLINE | ID: mdl-24292838

ABSTRACT

ß-Arrestins play a crucial role in cell migration downstream of multiple G-protein-coupled receptors (GPCRs) through multiple mechanisms. There is considerable evidence that ß-arrestin-dependent scaffolding of actin assembly proteins facilitates the formation of a leading edge in response to a chemotactic signal. Conversely, there is substantial support for the hypothesis that ß-arrestins facilitate receptor turnover through their ability to desensitize and internalize GPCRs. This chapter discusses both theories for ß-arrestin-dependent chemotaxis in the context of recent studies, specifically addressing known actin assembly proteins regulated by ß-arrestins, chemokine receptors, and signaling by chemotactic receptors.


Subject(s)
Actin Cytoskeleton/metabolism , Arrestins/metabolism , Chemotaxis/physiology , Actins/metabolism , Animals , Cell Movement/physiology , Humans , Receptors, Chemokine/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , beta-Arrestins
14.
Cell Rep ; 5(4): 1010-21, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24239352

ABSTRACT

G-protein-coupled receptors (GPCRs) are typically present in a basal, inactive state but, when bound to an agonist, activate downstream signaling cascades. In studying arrestin regulation of opioid receptors in dorsal root ganglia (DRG) neurons, we find that agonists of delta opioid receptors (δORs) activate cofilin through Rho-associated coiled-coil-containing protein kinase (ROCK), LIM domain kinase (LIMK), and ß-arrestin 1 (ß-arr1) to regulate actin polymerization. This controls receptor function, as assessed by agonist-induced inhibition of voltage-dependent Ca(2+) channels in DRGs. Agonists of opioid-receptor-like receptors (ORL1) similarly influence the function of this receptor through ROCK, LIMK, and ß-arr1. Functional evidence of this cascade was demonstrated in vivo, where the behavioral effects of δOR or ORL1 agonists were enhanced in the absence of ß-arr1 or prevented by inhibiting ROCK. This pathway allows δOR and ORL1 agonists to rapidly regulate receptor function.


Subject(s)
Arrestins/metabolism , Cofilin 1/metabolism , Lim Kinases/metabolism , Receptors, Opioid, delta/metabolism , Receptors, Opioid/metabolism , rho-Associated Kinases/metabolism , Animals , Benzamides/pharmacology , Calcium Channels , Cells, Cultured , Enzyme Activation , Female , Ganglia, Spinal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pain/drug therapy , Patch-Clamp Techniques , Phosphoprotein Phosphatases/metabolism , Piperazines/pharmacology , Receptors, Opioid/agonists , Receptors, Opioid, delta/agonists , beta-Arrestin 1 , beta-Arrestins , Nociceptin Receptor
15.
Prog Mol Biol Transl Sci ; 118: 205-22, 2013.
Article in English | MEDLINE | ID: mdl-23764055

ABSTRACT

Arrestins have emerged as important regulators of actin reorganization and cell migration. Both in their classical roles as mediators of receptor desensitization and internalization, and in their newer role as signaling scaffolds, ß-arrestins help orchestrate the cellular response to chemotactic signals. However, there is still a considerable amount to be learned about the precise molecular mechanisms underlying these processes. This review discusses how, by regulating receptor internalization and by scaffolding of signaling molecules in discrete cellular locations, arrestins facilitate gradient sensing and cytoskeletal reorganization, ultimately resulting in cell migration. In addition, putative new targets of ß-arrestin regulation that may play important roles in cell migration are discussed, as continued research on these targets may provide important details to fill in the current gaps in our understanding of these processes.


Subject(s)
Actins/metabolism , Arrestins/metabolism , Cell Movement , Animals , Disease , Health , Humans , Receptors, Chemokine/metabolism
16.
Article in English | MEDLINE | ID: mdl-23319470

ABSTRACT

G-protein-coupled receptors (GPCRs) can signal through heterotrimeric G-proteins or through ß-arrestins to elicit responses to a plethora of extracellular stimuli. While the mechanisms underlying G-protein signaling is relatively well understood, the mechanisms by which ß-arrestins regulate the diverse set of proteins with which they associate remain unclear. Multi-protein complexes are a common feature of ß-arrestin-dependent signaling. The first two such complexes discovered were the mitogen-activated kinases modules associated with extracellular regulated kinases (ERK1/2) and Jnk3. Subsequently a number of other kinases have been shown to undergo ß-arrestin-dependent regulation, including Akt, phosphatidylinositol-3kinase (PI3K), Lim-domain-containing kinase (LIMK), calcium calmodulin kinase II (CAMKII), and calcium calmodulin kinase kinase ß (CAMKKß). Some are positively and some negatively regulated by ß-arrestin association. One of the missing links to understanding these pathways is the molecular mechanisms by which the activity of these kinases is regulated. Do ß-arrestins merely serve as scaffolds to bring enzyme and substrate together or do they have a direct effect on the enzymatic activities of target kinases? Recent evidence suggests that both mechanisms are involved and that the mechanisms by which ß-arrestins regulate kinase activity varies with the target kinase. This review discusses recent advances in the field focusing on 5 kinases for which considerable mechanistic detail and specific sites of interaction have been elucidated.


Subject(s)
Arrestins/chemistry , Protein Serine-Threonine Kinases/chemistry , Arrestins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , beta-Arrestins
17.
J Biol Chem ; 288(5): 3265-74, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23235155

ABSTRACT

ß-Arrestins are multifunctional adaptor proteins that, upon recruitment to an activated G-protein-coupled receptor, can promote desensitization of G-protein signaling and receptor internalization while simultaneously eliciting an independent signal. The result of ß-arrestin signaling depends upon the activating receptor. For example, activation of two Gα(q)-coupled receptors, protease-activated receptor-2 (PAR(2)) and neurokinin-1 receptor (NK1R), results in drastically different signaling events. PAR(2) promotes ß-arrestin-dependent membrane-sequestered extracellular signal-regulated kinase (ERK1/2) activation, cofilin activation, and cell migration, whereas NK1R promotes nuclear ERK1/2 activation and proliferation. Using bioluminescence resonance energy transfer to monitor receptor/ß-arrestin interactions in real time, we observe that PAR(2) has a higher apparent affinity for both ß-arrestins than does NK1R, recruits them at a faster rate, and exhibits more rapid desensitization of the G-protein signal. Furthermore, recruitment of ß-arrestins to PAR(2) does not require prior Gα(q) signaling events, whereas inhibition of Gα(q) signaling intermediates inhibits recruitment of ß-arrestins to NK1R. Using chimeric receptors in which the C terminus of PAR(2) is fused to the N terminus of NK1R and vice versa and a critical Ser/Thr mutant of PAR(2), we demonstrate that interactions between ß-arrestins and specific phosphoresidues in the C termini of each receptor are crucial for determining the rate and magnitude of ß-arrestin recruitment as well as the ultimate signaling outcome.


Subject(s)
Arrestins/metabolism , Receptor, PAR-2/chemistry , Receptor, PAR-2/metabolism , Receptors, Neurokinin-1/chemistry , Receptors, Neurokinin-1/metabolism , Signal Transduction , Actin Depolymerizing Factors/metabolism , Amino Acid Sequence , Animals , CHO Cells , Calcium/metabolism , Cell Movement , Cricetinae , Endocytosis , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , HEK293 Cells , Humans , Intracellular Space/metabolism , Kinetics , Mice , Mutant Proteins/metabolism , Neurokinin-1 Receptor Antagonists , Phosphorylation , Receptor, PAR-2/antagonists & inhibitors , Structure-Activity Relationship , Subcellular Fractions/metabolism , beta-Arrestins
18.
Proc Natl Acad Sci U S A ; 109(41): 16660-5, 2012 Oct 09.
Article in English | MEDLINE | ID: mdl-23012429

ABSTRACT

Proteinase-Activated receptor-2 (PAR(2)), a G-protein-coupled Receptor, activated by serine proteinases, is reported to have both protective and proinflammatory effects in the airway. Given these opposing actions, both inhibitors and activators of PAR(2) have been proposed for treating asthma. PAR(2) can signal through two independent pathways: a ß-arrestin-dependent one that promotes leukocyte migration, and a G-protein/Ca(2+) one that is required for prostaglandin E(2) (PGE(2)) production and bronchiolar smooth muscle relaxation. We hypothesized that the proinflammatory responses to PAR(2) activation are mediated by ß-arrestins, whereas the protective effects are not. Using a mouse ovalbumin model for PAR(2)-modulated airway inflammation, we observed decreased leukocyte recruitment, cytokine production, and mucin production in ß-arrestin-2(-/-) mice. In contrast, PAR(2)-mediated PGE(2) production, smooth muscle relaxation, and decreased baseline airway resistance (measures of putative PAR(2) "protective" effects) were independent of ß-arrestin-2. Flow cytometry and cytospins reveal that lung eosinophil and CD4 T-cell infiltration, and production of IL-4, IL-6, IL-13, and TNFα, were enhanced in wild-type but not ß-arrestin-2(-/-) mice. Using the forced oscillation technique to measure airway resistance reveals that PAR(2) activation protects against airway hyperresponsiveness by an unknown mechanism, possibly involving smooth muscle relaxation. Our data suggest that the PAR(2)-enhanced inflammatory process is ß-arrestin-2 dependent, whereas the protective anticonstrictor effect of bronchial epithelial PAR(2) may be ß-arrestin independent.


Subject(s)
Arrestins/metabolism , Inflammation/metabolism , Lung/metabolism , Receptor, PAR-2/metabolism , Animals , Arrestins/genetics , Asthma/genetics , Asthma/metabolism , Asthma/pathology , Bronchial Hyperreactivity/genetics , Bronchial Hyperreactivity/metabolism , Bronchial Hyperreactivity/pathology , Bronchoalveolar Lavage Fluid/chemistry , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Cytokines/metabolism , Dinoprostone/metabolism , Flow Cytometry , Inflammation/genetics , Inflammation/pathology , Interleukin-13/metabolism , Leukocytes/metabolism , Leukocytes/pathology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Receptor, PAR-2/genetics , beta-Arrestin 2 , beta-Arrestins
19.
Proc Natl Acad Sci U S A ; 109(7): E442-51, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22308427

ABSTRACT

Dendritic spines are dynamic, actin-rich structures that form the postsynaptic sites of most excitatory synapses in the brain. The F-actin severing protein cofilin has been implicated in the remodeling of dendritic spines and synapses under normal and pathological conditions, by yet unknown mechanisms. Here we report that ß-arrestin-2 plays an important role in NMDA-induced remodeling of dendritic spines and synapses via translocation of active cofilin to dendritic spines. NMDAR activation triggers cofilin activation through calcineurin and phosphatidylinositol 3-kinase (PI3K)-mediated dephosphorylation and promotes cofilin translocation to dendritic spines that is mediated by ß-arrestin-2. Hippocampal neurons lacking ß-arrestin-2 develop mature spines that fail to remodel in response to NMDA. ß-Arrestin-2-deficient mice exhibit normal hippocampal long-term potentiation, but significantly impaired NMDA-dependent long-term depression and spatial learning deficits. Moreover, ß-arrestin-2-deficient hippocampal neurons are resistant to Aß-induced dendritic spine loss. Our studies demonstrate unique functions of ß-arrestin-2 in NMDAR-mediated dendritic spine and synapse plasticity through spatial control over cofilin activation.


Subject(s)
Actin Depolymerizing Factors/physiology , Arrestins/physiology , Dendritic Spines/physiology , Learning , Long-Term Synaptic Depression , N-Methylaspartate/physiology , Neuronal Plasticity/physiology , Animals , Calcineurin/metabolism , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Receptors, N-Methyl-D-Aspartate/metabolism , beta-Arrestin 2 , beta-Arrestins
20.
Nat Rev Drug Discov ; 11(1): 69-86, 2012 Jan 03.
Article in English | MEDLINE | ID: mdl-22212680

ABSTRACT

Proteinase-activated receptors (PARs), a family of four seven-transmembrane G protein-coupled receptors, act as targets for signalling by various proteolytic enzymes. PARs are characterized by a unique activation mechanism involving the proteolytic unmasking of a tethered ligand that stimulates the receptor. Given the emerging roles of these receptors in cancer as well as in disorders of the cardiovascular, musculoskeletal, gastrointestinal, respiratory and central nervous system, PARs have become attractive targets for the development of novel therapeutics. In this Review we summarize the mechanisms by which PARs modulate cell function and the roles they can have in physiology and diseases. Furthermore, we provide an overview of possible strategies for developing PAR antagonists.


Subject(s)
Drug Delivery Systems/trends , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/metabolism , Receptors, Proteinase-Activated/metabolism , Animals , Drug Delivery Systems/methods , Guanidines/administration & dosage , Guanidines/chemistry , Guanidines/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/metabolism , Oligopeptides/administration & dosage , Oligopeptides/chemistry , Oligopeptides/metabolism , Pharmaceutical Preparations/chemistry , Receptors, Proteinase-Activated/agonists , Receptors, Proteinase-Activated/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...