Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Clin Exp Allergy ; 45(12): 1844-55, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26312432

ABSTRACT

BACKGROUND: Proteinase-activated receptor 2 (PAR2 ) is a G protein-coupled receptor activated by trypsin-like serine proteinases. PAR2 activation has been associated with inflammation including allergic airway inflammation. We have also shown that PAR2 activation in the airways leads to allergic sensitization. The exact contribution of PAR2 in the development of eosinophilic inflammation and airway hyperresponsiveness (AHR) in sensitized individuals is not clear. OBJECTIVE: To investigate whether functional inhibition of PAR2 during allergen challenge of allergic mice would inhibit allergen-induced AHR and inflammation in mouse models of asthma. METHODS: Mice were sensitized and challenged with ovalbumin (OVA) or cockroach extract (CE). To investigate the role of PAR2 in the development of AHR and airway inflammation, we administered blocking anti-PAR2 antibodies, or a cell permeable peptide inhibitor of PAR2 signalling, pepducin, i.n. before allergen challenges and then assessed AHR and airway inflammation. RESULTS: Administration of anti-PAR2 antibodies significantly inhibited OVA- and CE-induced AHR and airway inflammation. In particular, two anti-PAR2 antibodies, the monoclonal SAM-11 and polyclonal B5, inhibited AHR, airway eosinophilia, the increase of cytokines in the lung tissue and antigen-specific T cell proliferation, but had no effect on antigen-specific IgG and IgE levels. Pepducin was also effective in inhibiting AHR and airway inflammation in an OVA model of allergic airway inflammation. CONCLUSIONS AND CLINICAL RELEVANCE: Functional blockade of PAR2 in the airways during allergen challenge improves allergen-induced AHR and inflammation in mice. Therefore, topical PAR2 blockade in the airways, through anti-PAR2 antibodies or molecules that interrupt PAR2 signalling, has the potential to be used as a therapeutic option in allergic asthma.


Subject(s)
Allergens/immunology , Asthma/immunology , Asthma/metabolism , Receptor, PAR-2/antagonists & inhibitors , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Asthma/genetics , Biomarkers , Cytokines/biosynthesis , Disease Models, Animal , Immunization , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Lung/immunology , Lung/metabolism , Male , Mice , Mice, Knockout , Ovalbumin/immunology , Respiratory Hypersensitivity/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
2.
Br J Pharmacol ; 153 Suppl 1: S298-309, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18037927

ABSTRACT

G-protein-coupled receptors (GPCRs), also known as seven transmembrane receptors (7-TMRs), are the largest protein receptor superfamily in the body. These receptors and their ligands direct a diverse array of physiological responses, and hence have broad relevance to numerous diseases. As a result, they have generated considerable interest in the pharmaceutical industry as drug targets. Recently, GPCRs have been demonstrated to elicit signals through interaction with the scaffolding proteins, beta-arrestins-1 and 2, independent of heterotrimeric G-protein coupling. This review discusses several known G-protein-independent, beta-arrestin-dependent pathways and their potential physiological and pharmacological significance. The emergence of G-protein-independent signalling changes the way in which GPCR signalling is evaluated, from a cell biological to a pharmaceutical perspective and raises the possibility for the development of pathway specific therapeutics.


Subject(s)
Arrestins/physiology , GTP-Binding Proteins/physiology , Signal Transduction/physiology , Actin Depolymerizing Factors/physiology , Animals , Arrestins/chemistry , Enzyme Activation/physiology , GTP-Binding Proteins/chemistry , Humans , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/physiology , Phosphatidylinositol 3-Kinases/physiology , beta-Arrestins
3.
Am J Physiol Cell Physiol ; 293(1): C346-57, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17442737

ABSTRACT

Beta-arrestins-1 and 2 are known to play important roles in desensitization of membrane receptors and facilitation of signal transduction pathways. It has been previously shown that beta-arrestins are required for signal termination, internalization, and ERK1/2 activation downstream of protease-activated-receptor-2 (PAR-2), but it is unclear whether they are functionally redundant or mediate specific events. Here, we demonstrate that in mouse embryonic fibroblasts (MEFs) from beta-arrestin-1/2 knockout mice, G alpha q signaling by PAR-2, as measured by mobilization of intracellular Ca(2+), is prolonged. Only expression of beta-arrestin-1 shortened the signal duration, whereas either beta-arrestin-1 or 2 was able to restore PKC-induced receptor desensitization. Beta-arrestin-1 also mediated early, while beta-arrestin-2 mediated delayed, receptor internalization and membrane-associated ERK1/2 activation. While beta-arrestin-1 colocalized with a lysosomal marker (LAMP-1), beta-arrestin-2 did not, suggesting a specific role for beta-arrestin-1 in lysosomal receptor degradation. Together, these data suggest distinct temporal and functional roles for beta-arrestins in PAR-2 signaling, desensitization, and internalization.


Subject(s)
Arrestins/metabolism , Calcium Signaling , Endocytosis , Fibroblasts/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptor, PAR-2/metabolism , Animals , Arrestins/deficiency , Arrestins/genetics , CHO Cells , Calcium Signaling/drug effects , Cell Line, Tumor , Cricetinae , Cricetulus , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Endocytosis/drug effects , Enzyme Activation , Exocytosis , Fibroblasts/drug effects , Fibroblasts/enzymology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Kinetics , Lysosomal-Associated Membrane Protein 1/metabolism , Lysosomes/metabolism , Mice , Mice, Knockout , Oligopeptides/pharmacology , Protein Isoforms/metabolism , Protein Kinase C/metabolism , Receptor, PAR-2/agonists , Receptor, PAR-2/genetics , Recombinant Fusion Proteins/metabolism , Transfection , Trypsin/metabolism , beta-Arrestin 1 , beta-Arrestin 2 , beta-Arrestins
4.
J Biol Chem ; 276(27): 25427-37, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11306580

ABSTRACT

Understanding the molecular mechanisms of agonist-induced trafficking of G-protein-coupled receptors is important because of the essential role of trafficking in signal transduction. We examined the role of the GTPases dynamin 1 and Rab5a in substance P (SP)-induced trafficking and signaling of the neurokinin 1 receptor (NK1R), an important mediator of pain, depression, and inflammation, by studying transfected cells and enteric neurons that naturally express the NK1R. In unstimulated cells, the NK1R colocalized with dynamin at the plasma membrane, and Rab5a was detected in endosomes. SP induced translocation of the receptor into endosomes containing Rab5a immediately beneath the plasma membrane and then in a perinuclear location. Expression of the dominant negative mutants dynamin 1 K44E and Rab5aS34N inhibited endocytosis of SP by 45 and 32%, respectively. Dynamin K44E caused membrane retention of the NK1R, whereas Rab5aS34N also impeded the translocation of the receptor from superficially located to perinuclear endosomes. Both dynamin K44E and Rab5aS34N strongly inhibited resensitization of SP-induced Ca(2+) mobilization by 60 and 85%, respectively, but had no effect on NK1R desensitization. Dynamin K44E but not Rab5aS34N markedly reduced SP-induced phosphorylation of extracellular signal regulated kinases 1 and 2. Thus, dynamin mediates the formation of endosomes containing the NK1R, and Rab5a mediates both endosomal formation and their translocation from a superficial to a perinuclear location. Dynamin and Rab5a-dependent trafficking is essential for NK1R resensitization but is not necessary for desensitization of signaling. Dynamin-dependent but not Rab5a-dependent trafficking is required for coupling of the NK1R to the mitogen-activated protein kinase cascade. These processes may regulate the nociceptive, depressive, and proinflammatory effects of SP.


Subject(s)
GTP Phosphohydrolases/metabolism , Receptors, Neurokinin-1/metabolism , Signal Transduction , rab5 GTP-Binding Proteins/metabolism , Animals , Calcium/metabolism , Dogs , Dynamin I , Dynamins , Endocytosis , Fluorescent Antibody Technique , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Rats , Transfection , Tumor Cells, Cultured
5.
Am J Physiol Cell Physiol ; 280(5): C1097-106, 2001 May.
Article in English | MEDLINE | ID: mdl-11287322

ABSTRACT

An understanding of the mechanisms that regulate signaling by the substance P (SP) or neurokinin 1 receptor (NK1-R) is of interest because of their role in inflammation and pain. By using activators and inhibitors of protein kinase C (PKC) and NK1-R mutations of potential PKC phosphorylation sites, we determined the role of PKC in desensitization of responses to SP. Activation of PKC abolished SP-induced Ca(2+) mobilization in cells that express wild-type NK1-R. This did not occur in cells expressing a COOH-terminally truncated NK1-R (NK1-Rdelta324), which may correspond to a naturally occurring variant, or a point mutant lacking eight potential PKC phosphorylation sites within the COOH tail (NK1-R Ser-338, Thr-339, Ser-352, Ser-387, Ser-388, Ser-390, Ser-392, Ser-394/Ala, NK1-RKC4). Compared with wild-type NK1-R, the t(1/2) of SP-induced Ca(2+) mobilization was seven- and twofold greater in cells expressing NK1-Rdelta324 and NK1-RKC4, respectively. In cells expressing wild-type NK1-R, inhibition of PKC caused a 35% increase in the t(1/2) of SP-induced Ca(2+) mobilization. Neither inhibition of PKC nor receptor mutation affected desensitization of Ca(2+) mobilization to repeated challenge with SP or SP-induced endocytosis of the NK1-R. Thus PKC regulates SP-induced Ca(2+) mobilization by full-length NK1-R and does not regulate a naturally occurring truncated variant. PKC does not mediate desensitization to repeated stimulation or endocytosis of the NK1-R.


Subject(s)
Calcium Signaling/physiology , Protein Kinase C/metabolism , Receptors, Neurokinin-1/physiology , Substance P/pharmacology , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , Calcium Signaling/drug effects , Cell Line , DNA Primers , Enzyme Activation , Enzyme Inhibitors/pharmacology , Indoles , Kidney , Maleimides , Molecular Sequence Data , Mutagenesis, Site-Directed , Phorbol 12,13-Dibutyrate/pharmacology , Phosphorylation , Phosphoserine/metabolism , Phosphothreonine/metabolism , Protein Conformation , Rats , Receptors, Neurokinin-1/chemistry , Receptors, Neurokinin-1/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Sequence Deletion , Transfection
6.
Proc Natl Acad Sci U S A ; 97(20): 11086-91, 2000 Sep 26.
Article in English | MEDLINE | ID: mdl-10995467

ABSTRACT

A requirement for scaffolding complexes containing internalized G protein-coupled receptors and beta-arrestins in the activation and subcellular localization of extracellular signal-regulated kinases 1 and 2 (ERK1/2) has recently been proposed. However, the composition of these complexes and the importance of this requirement for function of ERK1/2 appear to differ between receptors. Here we report that substance P (SP) activation of neurokinin-1 receptor (NK1R) stimulates the formation of a scaffolding complex comprising internalized receptor, beta-arrestin, src, and ERK1/2 (detected by gel filtration, immunoprecipitation, and immunofluorescence). Inhibition of complex formation, by expression of dominant-negative beta-arrestin or a truncated NK1R that fails to interact with beta-arrestin, inhibits both SP-stimulated endocytosis of the NK1R and activation of ERK1/2, which is required for the proliferative and antiapoptotic effects of SP. Thus, formation of a beta-arrestin-containing complex facilitates the proliferative and antiapoptotic effects of SP, and these effects of SP could be diminished in cells expressing truncated NK1R corresponding to a naturally occurring variant.


Subject(s)
Apoptosis/drug effects , Apoptosis/physiology , Arrestins/physiology , Receptors, Neurokinin-1/physiology , Signal Transduction/drug effects , Substance P/pharmacology , Animals , Cell Division/drug effects , Cell Division/physiology , Cell Line , MAP Kinase Signaling System , Rats , beta-Arrestins
7.
Biochem Soc Trans ; 28(4): 419-26, 2000.
Article in English | MEDLINE | ID: mdl-10961932

ABSTRACT

Biological responses to neuropeptides are rapidly attenuated by overlapping mechanisms that include peptide degradation by cell-surface proteases, receptor uncoupling from heterotrimeric G-proteins and receptor endocytosis. We have investigated the mechanisms that terminate the proinflammatory effects of the neuropeptide substance P (SP), which are mediated by the neurokinin 1 receptor (NK1R). Neutral endopeptidase degrades SP in the extracellular fluid and is one of the first mechanisms to terminate signalling. G-protein receptor kinases and second-messenger kinases phosphorylate the NK1R to permit interaction with beta-arrestins, which uncouple the receptor from G-proteins to terminate the signal. SP-induces NK1R endocytosis by a beta-arrestin-dependent mechanism, which also involves the GTPases dynamin and Rab5a. Endocytosis contributes to desensitization by depleting receptors from the cell surface. Disruption of these mechanisms results in uncontrolled stimulation and disease. Thus the deletion of neutral endopeptidase in mice exacerbates inflammation of many tissues. There are similarities and distinct differences in the mechanisms that regulate signalling by neuropeptide receptors and other G-protein-coupled receptors, in particular those that are activated irreversibly by proteolysis.


Subject(s)
Endopeptidases/metabolism , Receptors, Neuropeptide/metabolism , Signal Transduction , Substance P/metabolism , Animals , Arrestins/metabolism , Cell Membrane/metabolism , Dynamins , Endocytosis , GTP Phosphohydrolases/metabolism , Humans , Mast Cells/metabolism , Mice , Models, Biological , Neprilysin/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Receptor, PAR-2 , Receptors, Neurokinin-1/metabolism , Receptors, Thrombin/metabolism , Serine Endopeptidases/metabolism , Time Factors , Tryptases , beta-Arrestins , rab5 GTP-Binding Proteins/metabolism
8.
J Cell Biol ; 148(6): 1267-81, 2000 Mar 20.
Article in English | MEDLINE | ID: mdl-10725339

ABSTRACT

Recently, a requirement for beta-arrestin-mediated endocytosis in the activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) by several G protein-coupled receptors (GPCRs) has been proposed. However, the importance of this requirement for function of ERK1/2 is unknown. We report that agonists of Galphaq-coupled proteinase-activated receptor 2 (PAR2) stimulate formation of a multiprotein signaling complex, as detected by gel filtration, immunoprecipitation and immunofluorescence. The complex, which contains internalized receptor, beta-arrestin, raf-1, and activated ERK, is required for ERK1/2 activation. However, ERK1/2 activity is retained in the cytosol and neither translocates to the nucleus nor causes proliferation. In contrast, a mutant PAR2 (PAR2deltaST363/6A), which is unable to interact with beta-arrestin and, thus, does not desensitize or internalize, activates ERK1/2 by a distinct pathway, and fails to promote both complex formation and cytosolic retention of the activated ERK1/2. Whereas wild-type PAR2 activates ERK1/2 by a PKC-dependent and probably a ras-independent pathway, PAR2(deltaST363/6A) appears to activate ERK1/2 by a ras-dependent pathway, resulting in increased cell proliferation. Thus, formation of a signaling complex comprising PAR2, beta-arrestin, raf-1, and activated ERK1/2 might ensure appropriate subcellular localization of PAR2-mediated ERK activity, and thereby determine the mitogenic potential of receptor agonists.


Subject(s)
Arrestins/physiology , Endocytosis , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Receptors, Thrombin/physiology , Animals , Calcium/metabolism , Cell Division , Cell Line , Cell Nucleus/physiology , Cell Nucleus/ultrastructure , Cytosol/physiology , Cytosol/ultrastructure , Enzyme Activation , Humans , Kinetics , Microscopy, Confocal , Mitogen-Activated Protein Kinase 3 , Models, Biological , Mutagenesis , Rats , Receptor, PAR-2 , Receptors, Thrombin/genetics , Recombinant Proteins/metabolism , Transfection , beta-Arrestins
9.
J Biol Chem ; 274(14): 9351-6, 1999 Apr 02.
Article in English | MEDLINE | ID: mdl-10092613

ABSTRACT

Serine/threonine phosphorylation of insulin receptor substrate 1 (IRS-1) has been implicated as a negative regulator of insulin signaling. Prior studies have indicated that this negative regulation by protein kinase C involves the mitogen-activated protein kinase and phosphorylation of serine 612 in IRS-1. In the present studies, the negative regulation by platelet-derived growth factor (PDGF) was compared with that induced by endothelin-1, an activator of protein kinase C. In contrast to endothelin-1, the inhibitory effects of PDGF did not require mitogen-activated protein kinase or the phosphorylation of serine 612. Instead, three other serines in the phosphorylation domain of IRS-1 (serines 632, 662, and 731) were required for the negative regulation by PDGF. In addition, the PDGF-activated serine/threonine kinase called Akt was found to inhibit insulin signaling. Moreover, this inhibition required the same IRS-1 serine residues as the inhibition by PDGF. Finally, the negative regulatory effects of PDGF and Akt were inhibited by rapamycin, an inhibitor of the mammalian target of rapamycin (mTOR), one of the downstream targets of Akt. These studies implicate the phosphatidylinositol 3-kinase/Akt kinase cascade as an additional negative regulatory pathway for the insulin signaling cascade.


Subject(s)
Endothelin-1/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins , Tyrosine/metabolism , 3T3 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Flavonoids/pharmacology , Insulin/pharmacology , Insulin Receptor Substrate Proteins , Mice , Phosphorylation , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins c-akt , Sirolimus/pharmacology
10.
Science ; 279(5352): 827-34, 1998 Feb 06.
Article in English | MEDLINE | ID: mdl-9452375

ABSTRACT

At the endoplasmic reticulum membrane, the prion protein (PrP) can be synthesized in several topological forms. The role of these different forms was explored with transgenic mice expressing PrP mutations that alter the relative ratios of the topological forms. Expression of a particular transmembrane form (termed CtmPrP) produced neurodegenerative changes in mice similar to those of some genetic prion diseases. Brains from these mice contained CtmPrP but not PrPSc, the PrP isoform responsible for transmission of prion diseases. Furthermore, in one heritable prion disease of humans, brain tissue contained CtmPrP but not PrPSc. Thus, aberrant regulation of protein biogenesis and topology at the endoplasmic reticulum can result in neurodegeneration.


Subject(s)
Endoplasmic Reticulum/metabolism , Neurodegenerative Diseases/etiology , PrPC Proteins/chemistry , PrPC Proteins/metabolism , Prions/chemistry , Prions/metabolism , Amino Acid Sequence , Animals , Brain/metabolism , Brain/pathology , Cricetinae , Endopeptidases/metabolism , Endoplasmic Reticulum/chemistry , Gerstmann-Straussler-Scheinker Disease/metabolism , Humans , Intracellular Membranes/chemistry , Mesocricetus , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , PrPC Proteins/biosynthesis , PrPC Proteins/genetics , PrPSc Proteins/chemistry , PrPSc Proteins/metabolism , Prion Diseases/etiology , Prion Diseases/metabolism , Prion Diseases/pathology , Prions/biosynthesis , Prions/genetics , Protein Conformation
11.
Anal Biochem ; 174(2): 393-8, 1988 Nov 01.
Article in English | MEDLINE | ID: mdl-3239744

ABSTRACT

A microscopic method of measuring the orientation of nucleic acids in the agarose gels is described. A nucleic acid undergoing electrophoresis is stained with the dye ethidium bromide and is viewed under high magnification with a polarization microscope. A high-numerical-aperture microscope objective is used to illuminate and to collect the fluorescence signal, and therefore the orientation of the minute quantities of nucleic-acid can be measured: in a typical experiment we can detect the orientation of one-tenth of a picogram (10(13)g) of DNA. Polarization properties of the fluorescent light emitted by the separate bands corresponding to different molecular weights of the DNA are examined. A linear dichroism equation relates the measured fluorescence to the mean orientation of the absorption dipole of the ethidium bromide (and therefore DNA) and to the extent to which it is disorganized. As an example, we measured the orientation of phi X174 DNA RF/HaeIII fragments undergoing electrophoresis in a field of 10 V/cm. Ethidium bromide bound to the fragments with an angle of the absorption dipole largely perpendicular to the direction of the electrophoretic current. The dichroism declined as the molecular weight of the fragments decreased which is interpreted as an increase in the degree of disorder for shorter DNA.


Subject(s)
DNA , Electrophoresis , Ethidium , Gels , Microchemistry , Microscopy, Fluorescence , Sepharose
SELECTION OF CITATIONS
SEARCH DETAIL
...