Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Headache Pain ; 24(1): 42, 2023 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-37072694

RESUMO

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.


Assuntos
Transtornos de Enxaqueca , Nitroglicerina , Camundongos , Animais , Nitroglicerina/farmacologia , Elastase de Leucócito , Receptor PAR-2 , Interleucina-6 , Transtornos de Enxaqueca/induzido quimicamente , Dura-Máter , Cefaleia , Modelos Animais de Doenças
2.
Biochem Biophys Res Commun ; 591: 13-19, 2022 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-34990903

RESUMO

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.


Assuntos
Alternaria/fisiologia , Brônquios/patologia , Células Epiteliais/microbiologia , Inflamação/patologia , Receptor PAR-2/metabolismo , Transdução de Sinais , Sequência de Bases , Sistemas CRISPR-Cas/genética , Linhagem Celular , Células Epiteliais/metabolismo , Humanos
3.
Am J Physiol Lung Cell Mol Physiol ; 315(6): L1042-L1057, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30335499

RESUMO

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.


Assuntos
Inflamação/metabolismo , Receptor PAR-2/metabolismo , Serina Proteases/metabolismo , Transdução de Sinais/fisiologia , beta-Arrestina 2/metabolismo , Alérgenos/metabolismo , Animais , Asma/metabolismo , Proteínas de Bactérias/metabolismo , Endopeptidases/metabolismo , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Serina/metabolismo , Serina Endopeptidases/metabolismo
4.
Proc Natl Acad Sci U S A ; 109(7): E442-51, 2012 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-22308427

RESUMO

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.


Assuntos
Fatores de Despolimerização de Actina/fisiologia , Arrestinas/fisiologia , Espinhas Dendríticas/fisiologia , Aprendizagem , Depressão Sináptica de Longo Prazo , N-Metilaspartato/fisiologia , Plasticidade Neuronal/fisiologia , Animais , Calcineurina/metabolismo , Camundongos , Camundongos Knockout , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Receptores de N-Metil-D-Aspartato/metabolismo , beta-Arrestina 2 , beta-Arrestinas
5.
Proc Natl Acad Sci U S A ; 109(41): 16660-5, 2012 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-23012429

RESUMO

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.


Assuntos
Arrestinas/metabolismo , Inflamação/metabolismo , Pulmão/metabolismo , Receptor PAR-2/metabolismo , Animais , Arrestinas/genética , Asma/genética , Asma/metabolismo , Asma/patologia , Hiper-Reatividade Brônquica/genética , Hiper-Reatividade Brônquica/metabolismo , Hiper-Reatividade Brônquica/patologia , Líquido da Lavagem Broncoalveolar/química , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Citocinas/metabolismo , Dinoprostona/metabolismo , Citometria de Fluxo , Inflamação/genética , Inflamação/patologia , Interleucina-13/metabolismo , Leucócitos/metabolismo , Leucócitos/patologia , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Receptor PAR-2/genética , beta-Arrestina 2 , beta-Arrestinas
6.
J Biol Chem ; 288(5): 3265-74, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23235155

RESUMO

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.


Assuntos
Arrestinas/metabolismo , Receptor PAR-2/química , Receptor PAR-2/metabolismo , Receptores da Neurocinina-1/química , Receptores da Neurocinina-1/metabolismo , Transdução de Sinais , Fatores de Despolimerização de Actina/metabolismo , Sequência de Aminoácidos , Animais , Células CHO , Cálcio/metabolismo , Movimento Celular , Cricetinae , Endocitose , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Espaço Intracelular/metabolismo , Cinética , Camundongos , Proteínas Mutantes/metabolismo , Antagonistas dos Receptores de Neurocinina-1 , Fosforilação , Receptor PAR-2/antagonistas & inibidores , Relação Estrutura-Atividade , Frações Subcelulares/metabolismo , beta-Arrestinas
7.
Handb Exp Pharmacol ; 219: 341-59, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24292838

RESUMO

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.


Assuntos
Citoesqueleto de Actina/metabolismo , Arrestinas/metabolismo , Quimiotaxia/fisiologia , Actinas/metabolismo , Animais , Movimento Celular/fisiologia , Humanos , Receptores de Quimiocinas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , beta-Arrestinas
9.
J Pain ; 24(11): 1980-1993, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37315729

RESUMO

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.


Assuntos
Paclitaxel , Doenças do Sistema Nervoso Periférico , Masculino , Feminino , Camundongos , Animais , Paclitaxel/efeitos adversos , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Receptor PAR-2/genética , Receptor PAR-2/uso terapêutico , Gliose/induzido quimicamente , Gliose/complicações , Gliose/patologia , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Dor/complicações , Células Receptoras Sensoriais , Camundongos Knockout , Gânglios Espinais
10.
J Pain ; 24(4): 605-616, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36417966

RESUMO

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.


Assuntos
Peptídeo Hidrolases , Receptor PAR-2 , Camundongos , Animais , Peptídeo Hidrolases/metabolismo , Peptídeo Hidrolases/farmacologia , beta-Arrestinas/metabolismo , beta-Arrestinas/farmacologia , Receptor PAR-2/metabolismo , Dor/tratamento farmacológico , Dor/metabolismo , Transdução de Sinais/fisiologia
11.
Br J Pharmacol ; 180(5): 667-680, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-35735078

RESUMO

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.


Assuntos
Asma , Hiper-Reatividade Brônquica , Hipersensibilidade Respiratória , Camundongos , Humanos , Animais , Alérgenos , Receptor PAR-2 , beta-Arrestinas , Asma/tratamento farmacológico , Hipersensibilidade Respiratória/tratamento farmacológico , beta-Arrestina 1 , Inflamação/tratamento farmacológico , Camundongos Endogâmicos BALB C , Pulmão , Hiper-Reatividade Brônquica/tratamento farmacológico
12.
J Biol Chem ; 286(28): 24638-48, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21576245

RESUMO

Human neutrophil proteinases (elastase, proteinase-3, and cathepsin-G) are released at sites of acute inflammation. We hypothesized that these inflammation-associated proteinases can affect cell signaling by targeting proteinase-activated receptor-2 (PAR(2)). The PAR family of G protein-coupled receptors is triggered by a unique mechanism involving the proteolytic unmasking of an N-terminal self-activating tethered ligand (TL). Proteinases can either activate PAR signaling by unmasking the TL sequence or disarm the receptor for subsequent enzyme activation by cleaving downstream from the TL sequence. We found that none of neutrophil elastase, cathepsin-G, and proteinase-3 can activate G(q)-coupled PAR(2) calcium signaling; but all of these proteinases can disarm PAR(2), releasing the N-terminal TL sequence, thereby preventing G(q)-coupled PAR(2) signaling by trypsin. Interestingly, elastase (but neither cathepsin-G nor proteinase-3) causes a TL-independent PAR(2)-mediated activation of MAPK that, unlike the canonical trypsin activation, does not involve either receptor internalization or recruitment of ß-arrestin. Cleavage of synthetic peptides derived from the extracellular N terminus of PAR(2), downstream of the TL sequence, demonstrated distinct proteolytic sites for all three neutrophil-derived enzymes. We conclude that in inflammation, neutrophil proteinases can modulate PAR(2) signaling by preventing/disarming the G(q)/calcium signal pathway and, via elastase, can selectively activate the p44/42 MAPK pathway. Our data illustrate a new mode of PAR regulation that involves biased PAR(2) signaling by neutrophil elastase and a disarming/silencing effect of cathepsin-G and proteinase-3.


Assuntos
Sinalização do Cálcio/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Receptor PAR-2/agonistas , Receptor PAR-2/metabolismo , Animais , Arrestinas/genética , Arrestinas/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Catepsina G/genética , Catepsina G/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Inflamação/genética , Inflamação/metabolismo , Elastase de Leucócito , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mieloblastina/genética , Mieloblastina/metabolismo , Peptídeos/farmacologia , Estrutura Terciária de Proteína , Ratos , Receptor PAR-2/genética , beta-Arrestinas
13.
Br J Pharmacol ; 179(10): 2208-2222, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34841515

RESUMO

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.


Assuntos
Asma , Receptor PAR-2 , Alérgenos , Alternaria/metabolismo , Animais , Asma/tratamento farmacológico , Asma/metabolismo , Células HEK293 , Humanos , Camundongos
14.
Am J Physiol Cell Physiol ; 300(1): C113-23, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21068362

RESUMO

Studies suggest that there are two distinct pools of proteinase-activated receptor-2 (PAR2) present in intestinal epithelial cells: an apical pool accessible from the lumen, and a basolateral pool accessible from the interstitial space and blood. Although introduction of PAR2 agonists such as 2-furoyl-LIGRL-O-NH2 (2fAP) to the intestinal lumen can activate PAR2, the presence of accessible apical PAR2 has not been definitively shown. Furthermore, some studies have suggested that basolateral PAR2 responses in the intestinal epithelium are mediated indirectly by neuropeptides released from enteric nerve fibers, rather than by intestinal PAR2 itself. Here we identified accessible pools of both apical and basolateral PAR2 in cultured Caco2-BBe monolayers and in mouse ileum. Activation of basolateral PAR2 transiently increased short-circuit current by activating electrogenic Cl⁻ secretion, promoted dephosphorylation of the actin filament-severing protein, cofilin, and activated the transcription factor, AP-1, whereas apical PAR2 did not. In contrast, both pools of PAR2 activated extracellular signal-regulated kinase 1/2 (ERK1/2) via temporally and mechanistically distinct pathways. Apical PAR2 promoted a rapid, biphasic PLCß/Ca²(+)/PKC-dependent ERK1/2 activation, resulting in nuclear localization, whereas basolateral PAR2 promoted delayed ERK1/2 activation which was predominantly restricted to the cytosol, involving both PLCß/Ca²(+) and ß-arrestin-dependent pathways. These results suggest that the outcome of PAR2 activation is dependent on the specific receptor pool that is activated, allowing for fine-tuning of the physiological responses to different agonists.


Assuntos
Mucosa Intestinal/citologia , Receptor PAR-2/metabolismo , Transdução de Sinais/fisiologia , Animais , Células CACO-2 , Cloretos/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica/fisiologia , Humanos , Íleo/fisiologia , Mucosa Intestinal/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor PAR-2/genética
15.
Mol Pharmacol ; 80(5): 760-8, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21836019

RESUMO

First identified as mediators of G-protein-coupled receptor desensitization and internalization and later as signaling platforms, ß-arrestins play a requisite role in chemotaxis and reorganization of the actin cytoskeleton, downstream of multiple receptors. However, the precise molecular mechanisms underlying their involvement have remained elusive. Initial interest in ß-arrestins as facilitators of cell migration and actin reorganization stemmed from the known interplay between receptor endocytosis and actin filament formation, because disruption of the actin cytoskeleton inhibits these ß-arrestin-dependent events. With growing interest in the mechanisms by which cells can sense a gradient of agonist during cell migration, investigators began to hypothesize that ß-arrestins may contribute to directed migration by controlling chemotactic receptor turnover at the plasma membrane. Finally, increasing evidence emerged that ß-arrestins are more than just clathrin adaptor proteins involved in turning off receptor signals; they are actually capable of generating their own signals by scaffolding signaling molecules and controlling the activity of multiple cellular enzymes. This new role of ß-arrestins as signaling scaffolds has led to the hypothesis that they can facilitate cell migration by sequestering actin assembly activities and upstream regulators of actin assembly at the leading edge. This Minireview discusses recent advances in our understanding of how ß-arrestin scaffolds contribute to cell migration, focusing on recently identified ß-arrestin interacting proteins and phosphorylation targets that have known roles in actin reorganization.


Assuntos
Actinas/metabolismo , Arrestinas/fisiologia , Movimento Celular , Citoesqueleto/metabolismo , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Transdução de Sinais , beta-Arrestinas
16.
J Biol Chem ; 285(19): 14318-29, 2010 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-20207744

RESUMO

Protease-activated receptor-2 (PAR-2) mediates pro-inflammatory signals in a number of organs, including enhancing leukocyte recruitment to sites of injury and infection. At the cellular level, PAR-2 promotes activation of the actin filament-severing protein cofilin, which is crucial for the reorganization of the actin cytoskeleton and chemotaxis. These responses require the scaffolding functions of beta-arrestins; however, the mechanism by which beta-arrestins spatially regulate cofilin activity and the role of this pathway in primary cells has not been investigated. Here, using size-exclusion chromatography and co-immunoprecipitation, we demonstrate that PAR-2 promotes the formation of a complex containing beta-arrestins, cofilin, and chronophin (CIN) in primary leukocytes and cultured cells. Both association of cofilin with CIN and cell migration are inhibited in leukocytes from beta-arrestin-2(-/-) mice. We show that, in response to PAR-2 activation, beta-arrestins scaffold cofilin with its upstream activator CIN, to facilitate the localized generation of free actin barbed ends, leading to membrane protrusion. These studies suggest that a major role of beta-arrestins in chemotaxis is to spatially regulate cofilin activity to facilitate the formation of a leading edge, and that this pathway may be important for PAR-2-stimulated immune cell migration.


Assuntos
Citoesqueleto de Actina/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Arrestinas/fisiologia , Movimento Celular , Extensões da Superfície Celular/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Receptor PAR-2/metabolismo , Animais , Membrana Celular/metabolismo , Embrião de Mamíferos/metabolismo , Feminino , Fibroblastos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , beta-Arrestina 2 , beta-Arrestinas
17.
Circ Res ; 104(4): 496-505, 2009 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-19131647

RESUMO

Endothelial nitric oxide synthase (eNOS) plays a central role in maintaining cardiovascular homeostasis by controlling NO bioavailability. The activity of eNOS in vascular endothelial cells (ECs) largely depends on posttranslational modifications, including phosphorylation. Because the activity of AMP-activated protein kinase (AMPK) in ECs can be increased by multiple cardiovascular events, we studied the phosphorylation of eNOS Ser633 by AMPK and examined its functional relevance in the mouse models. Shear stress, atorvastatin, and adiponectin all increased AMPK Thr172 and eNOS Ser633 phosphorylations, which were abolished if AMPK was pharmacologically inhibited or genetically ablated. The constitutively active form of AMPK or an AMPK agonist caused a sustained Ser633 phosphorylation. Expression of gain-/loss-of-function eNOS mutants revealed that Ser633 phosphorylation is important for NO production. The aorta of AMPKalpha2(-/-) mice showed attenuated atorvastatin-induced eNOS phosphorylation. Nano-liquid chromatography/tandem mass spectrometry (LC/MS/MS) confirmed that eNOS Ser633 was able to compete with Ser1177 or acetyl-coenzyme A carboxylase Ser79 for AMPKalpha phosphorylation. Nano-LC/MS/MS confirmed that eNOS purified from AICAR-treated ECs was phosphorylated at both Ser633 and Ser1177. Our results indicate that AMPK phosphorylation of eNOS Ser633 is a functional signaling event for NO bioavailability in ECs.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Células Endoteliais/enzimologia , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/metabolismo , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/deficiência , Proteínas Quinases Ativadas por AMP/genética , Acetil-CoA Carboxilase/metabolismo , Adiponectina/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Atorvastatina , Bovinos , Células Cultivadas , Cromatografia Líquida/métodos , Células Endoteliais/efeitos dos fármacos , Ativação Enzimática , Ácidos Heptanoicos/farmacologia , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Nanotecnologia , Óxido Nítrico Sintase Tipo III/genética , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Pirazóis/farmacologia , Pirimidinas/farmacologia , Pirróis/farmacologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ribonucleotídeos/farmacologia , Serina , Transdução de Sinais/efeitos dos fármacos , Estresse Mecânico , Espectrometria de Massas em Tandem , Fatores de Tempo , Transfecção
18.
Front Pain Res (Lausanne) ; 2: 695962, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35295409

RESUMO

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.

19.
J Neurosci ; 29(25): 8129-42, 2009 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-19553453

RESUMO

Dendritic spines are the postsynaptic sites of most excitatory synapses in the brain and are highly enriched in polymerized F-actin, which drives the formation and maintenance of mature dendritic spines and synapses. We propose that suppressing the activity of the actin-severing protein cofilin plays an important role in the stabilization of mature dendritic spines, and is accomplished through an EphB receptor-focal adhesion kinase (FAK) pathway. Our studies revealed that Cre-mediated knock-out of loxP-flanked fak prompted the reversion of mature dendritic spines to an immature filopodial-like phenotype in primary hippocampal cultures. The effects of FAK depletion on dendritic spine number, length, and morphology were rescued by the overexpression of the constitutively active FAK(Y397E), but not FAK(Y397F), indicating the significance of FAK activation by phosphorylation on tyrosine 397. Our studies demonstrate that FAK acts downstream of EphB receptors in hippocampal neurons and EphB2-FAK signaling controls the stability of mature dendritic spines by promoting cofilin phosphorylation, thereby inhibiting cofilin activity. While constitutively active nonphosphorylatable cofilin(S3A) induced an immature spine profile, phosphomimetic cofilin(S3D) restored mature spine morphology in neurons with disrupted EphB activity or lacking FAK. Further, we found that EphB-mediated regulation of cofilin activity at least partially depends on the activation of Rho-associated kinase (ROCK) and LIMK-1. These findings indicate that EphB2-mediated dendritic spine stabilization relies, in part, on the ability of FAK to activate the RhoA-ROCK-LIMK-1 pathway, which functions to suppress cofilin activity and inhibit cofilin-mediated dendritic spine remodeling.


Assuntos
Fatores de Despolimerização de Actina/metabolismo , Espinhas Dendríticas/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Hipocampo/metabolismo , Vias Neurais/metabolismo , Neurônios/metabolismo , Receptores da Família Eph/metabolismo , Fatores de Despolimerização de Actina/genética , Animais , Células Cultivadas , Imunofluorescência , Proteína-Tirosina Quinases de Adesão Focal/deficiência , Proteína-Tirosina Quinases de Adesão Focal/genética , Vetores Genéticos , Imuno-Histoquímica , Quinases Lim/metabolismo , Camundongos , Fosforilação/fisiologia , Reação em Cadeia da Polimerase , Receptores da Família Eph/genética , Transdução de Sinais/fisiologia , Transfecção , Regulação para Cima/genética , Regulação para Cima/fisiologia , Quinases Associadas a rho/metabolismo
20.
BMC Biochem ; 11: 36, 2010 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-20858278

RESUMO

BACKGROUND: Proteinase-activated-receptor-2 (PAR2) is a seven transmembrane receptor that can activate two separate signaling arms: one through Gαq and Ca2+ mobilization, and a second through recruitment of ß-arrestin scaffolds. In some cases downstream targets of the Gαq/Ca2+ signaling arm are directly inhibited by ß-arrestins, while in other cases the two pathways are synergistic; thus ß-arrestins act as molecular switches capable of modifying the signal generated by the receptor. RESULTS: Here we demonstrate that PAR2 can activate adenosine monophosphate-activated protein kinase (AMPK), a key regulator of cellular energy balance, through Ca2+-dependent Kinase Kinase ß (CAMKKß), while inhibiting AMPK through interaction with ß-arrestins. The ultimate outcome of PAR2 activation depended on the cell type studied; in cultured fibroblasts with low endogenous ß-arrestins, PAR2 activated AMPK; however, in primary fat and liver, PAR2 only activated AMPK in ß-arrestin-2-/- mice. ß-arrestin-2 could be co-immunoprecipitated with AMPK and CAMKKß under baseline conditions from both cultured fibroblasts and primary fat, and its association with both proteins was increased by PAR2 activation. Addition of recombinant ß-arrestin-2 to in vitro kinase assays directly inhibited phosphorylation of AMPK by CAMKKß on Thr172. CONCLUSIONS: Studies have shown that decreased AMPK activity is associated with obesity and Type II Diabetes, while AMPK activity is increased with metabolically favorable conditions and cholesterol lowering drugs. These results suggest a role for ß-arrestin in the inhibition of AMPK signaling, raising the possibility that ß-arrestin-dependent PAR2 signaling may act as a molecular switch turning a positive signal to AMPK into an inhibitory one.


Assuntos
Arrestinas/fisiologia , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Proteínas Quinases/metabolismo , Receptor PAR-2/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Animais , Camundongos , Células NIH 3T3 , Fosforilação , Proteínas Quinases/química , Proteínas Recombinantes/metabolismo , Transdução de Sinais , beta-Arrestina 2 , beta-Arrestinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA