Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Cell Proteomics ; 14(5): 1385-99, 2015 May.
Article in English | MEDLINE | ID: mdl-25759509

ABSTRACT

Several cytoplasmic proteins that are involved in G protein-coupled receptor signaling cascades are known to translocate to the plasma membrane upon receptor activation, such as beta-arrestin2. Based on this example and in order to identify new cytoplasmic proteins implicated in the ON-and-OFF cycle of G protein-coupled receptor, a live-imaging screen of fluorescently labeled cytoplasmic proteins was performed using translocation criteria. The screening of 193 fluorescently tagged human proteins identified eight proteins that responded to activation of the tachykinin NK2 receptor by a change in their intracellular localization. Previously we have presented the functional characterization of one of these proteins, REDD1, that translocates to the plasma membrane. Here we report the results of the entire screening. The process of cell activation was recorded on videos at different time points and all the videos can be visualized on a dedicated website. The proteins BAIAP3 and BIN1, partially translocated to the plasma membrane upon activation of NK2 receptors. Proteins ARHGAP12 and PKM2 translocated toward membrane blebs. Three proteins that associate with the cytoskeleton were of particular interest : PLEKHH2 rearranged from individual dots located near the cell-substrate adhesion surface into lines of dots. The speriolin-like protein, SPATC1L, redistributed to cell-cell junctions. The Chloride intracellular Channel protein, CLIC2, translocated from actin-enriched plasma membrane bundles to cell-cell junctions upon activation of NK2 receptors. CLIC2, and one of its close paralogs, CLIC4, were further shown to respond with the same translocation pattern to muscarinic M3 and lysophosphatidic LPA receptors. This screen allowed us to identify potential actors in signaling pathways downstream of G protein-coupled receptors and could be scaled-up for high-content screening.


Subject(s)
Biological Assay , Molecular Imaging/methods , Receptors, Neurokinin-2/genetics , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chloride Channels/genetics , Chloride Channels/metabolism , Cytoskeletal Proteins , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Transport , Receptors, Neurokinin-2/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thyroid Hormones/genetics , Thyroid Hormones/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Thyroid Hormone-Binding Proteins
2.
J Cell Sci ; 127(Pt 4): 773-87, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24338366

ABSTRACT

The mTORC1 kinase promotes cell growth in response to growth factors by activation of receptor tyrosine kinase. It is regulated by the cellular energy level and the availability of nutrients. mTORC1 activity is also inhibited by cellular stresses through overexpression of REDD1 (regulated in development and DNA damage responses). We report the identification of REDD1 in a fluorescent live-imaging screen aimed at discovering new proteins implicated in G-protein-coupled receptor signaling, based on translocation criteria. Using a sensitive and quantitative plasma membrane localization assay based on bioluminescent resonance energy transfer, we further show that a panel of endogenously expressed GPCRs, through a Ca(2+)/calmodulin pathway, triggers plasma membrane translocation of REDD1 but not of its homolog REDD2. REDD1 and REDD2 share a conserved mTORC1-inhibitory motif characterized at the functional and structural level and differ most in their N-termini. We show that the N-terminus of REDD1 and its mTORC1-inhibitory motif participate in the GPCR-evoked dynamic interaction of REDD1 with the plasma membrane. We further identify REDD1 as a novel effector in GPCR signaling. We show that fast activation of mTORC1 by GPCRs correlates with fast and maximal translocation of REDD1 to the plasma membrane. Overexpression of functional REDD1 leads to a reduction of mTORC1 activation by GPCRs. By contrast, depletion of endogenous REDD1 protein unleashes mTORC1 activity. Thus, translocation to the plasma membrane appears to be an inactivation mechanism of REDD1 by GPCRs, which probably act by sequestering its functional mTORC1-inhibitory motif that is necessary for plasma membrane targeting.


Subject(s)
Cell Membrane/metabolism , Multiprotein Complexes/metabolism , Receptors, Neurokinin-2/metabolism , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Calcium Signaling , Calmodulin/metabolism , Enzyme Activation , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Molecular Sequence Data , Protein Interaction Domains and Motifs , Protein Sorting Signals , Protein Transport , Proteins/metabolism , Transcription Factors/chemistry
3.
Traffic ; 9(3): 305-24, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18088318

ABSTRACT

Activated human neuropeptide Y Y(1) receptors rapidly desensitize and internalize through clathrin-coated pits and recycle from early and recycling endosomes, unlike Y(2) receptors that neither internalize nor desensitize. To identify motifs implicated in Y(1) receptor desensitization and trafficking, mutants with varying C-terminal truncations or a substituted Y(2) C-terminus were constructed. Point mutations of key putative residues were made in a C-terminal conserved motif [phi-H-(S/T)-(E/D)-V-(S/T)-X-T] that we have identified and in the second intracellular i2 loop. Receptors were analyzed by functional assays, spectrofluorimetric measurements on living cells, flow cytometry, confocal imaging and bioluminescence resonance energy transfer assays for beta-arrestin activation and adaptor protein (AP-2) complex recruitment. Inhibitory GTP-binding protein-dependent signaling of Y(1) receptors to adenylyl cyclase and desensitization was unaffected by C-terminal truncations or mutations, while C-terminal deletion mutants of 42 and 61 amino acids no longer internalized. Substitutions of Thr357, Asp358, Ser360 and Thr362 by Ala in the C-terminus abolished both internalization and beta-arrestin activation but not desensitization. A Pro145 substitution by His in an i2 consensus motif reported to mediate phosphorylation-independent recruitment of beta-arrestins affected neither desensitization, internalization or recycling kinetics of activated Y(1) receptors nor beta-arrestin activation. Interestingly, combining Pro145 substitution by His and C-terminal substitutions significantly attenuates Y(1) desensitization. In the Y(2) receptor, replacement of His155 with Pro at this position in the i2 loop motif promotes agonist-mediated desensitization, beta-arrestin activation, internalization and recycling. Overall, our results indicate that beta-arrestin-mediated desensitization and internalization of Y(1) and Y(2) receptors are differentially regulated by the C-terminal motif and the i2 loop consensus motif.


Subject(s)
Receptors, Neuropeptide Y/chemistry , Receptors, Neuropeptide Y/metabolism , Adenylyl Cyclases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Arrestins/metabolism , Biological Transport, Active , Cell Line , Cyclic AMP/metabolism , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Kinetics , Microscopy, Confocal , Molecular Sequence Data , Mutagenesis, Site-Directed , Receptors, Neuropeptide Y/agonists , Receptors, Neuropeptide Y/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Transfection , beta-Arrestins
4.
Microsc Res Tech ; 69(12): 941-56, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17080432

ABSTRACT

Fluorescence resonance energy transfer (FRET) between an adequate pair of fluorophores is an indication of closer proximity than colocalization and is used by biologists to study fluorescently modified protein interactions inside cells. We present a method for visualization of FRET images acquired by confocal sensitized emission, involving excitation of the donor fluorophore and detection of the energy transfer as an emission from the acceptor fluorophore into the FRET channel. Authentic FRET signal measurements require the correction from the FRET channel of the undesired bleed-through signals (BT) resulting from both the leak-through of the donor emission and the direct acceptor emission. Our method reduces the interference of the user to a minimum by analyzing the entire image, pixel by pixel. It proposes imaging treatments and the display of control images to validate the BT calculation and the image corrections. It displays FRET images as a function of the colocalization of the two fluorescent partners. Finally, it proposes an alternative to normalization of the FRET intensities to compare FRET signal variations between samples. This method called "FRET and Colocalization Analyzer" has been implemented in a Plug-in of the freely available ImageJ software. It is particularly adapted when transient expression of the fluorescent proteins is used thereby giving very variable expression levels or when the colocalization of the two partners is varying in proportion, in amount, and in size, as a function of time. The method and program are validated using the analysis of the spatio-temporal interactions between a G-protein coupled receptor, the tachykinin NK2 receptor, and the beta-arrestin 2 as an example.


Subject(s)
Fluorescence Resonance Energy Transfer , Image Processing, Computer-Assisted , Microscopy, Confocal/methods , Arrestins/metabolism , Fluorescent Dyes , Humans , Receptors, G-Protein-Coupled/metabolism , Software , beta-Arrestin 2 , beta-Arrestins
5.
J Biol Chem ; 279(43): 45057-67, 2004 Oct 22.
Article in English | MEDLINE | ID: mdl-15294896

ABSTRACT

A functional fluorescent neurokinin NK2 receptor, EGFP-NK2, was previously used to follow, by fluorescence resonance energy transfer measurements in living cells, the binding of its fluorescently labeled agonist, bodipy-neurokinin A (NKA). Local agonist application suggested that the activation and desensitization of the NK2 receptors were compartmentalized at the level of the plasma membrane. In this study, fluorescence recovery after photobleaching experiments are carried out at variable observation radius (vrFRAP) to probe EGFP-NK2 receptor mobility and confinement. Experiments are carried out at 20 degrees C to maintain the number of receptors constant at the cell surface during recordings. In the absence of agonist, 35% EGFP-NK2 receptors diffuse within domains of 420 +/- 80 nm in radius with the remaining 65% of receptors able to diffuse with a long range lateral diffusion coefficient between the domains. When cells are incubated with a saturating concentration of NKA, 30% EGFP-NK2 receptors become immobilized in small domains characterized by a radius equal to 170 +/- 50 nm. Biochemical experiments show that the confinement of EGFP-NK2 receptor is not due to its association with rafts at any given time. Colocalization of the receptor with beta-arrestin and transferrin supports that the small domains, containing 30% of activated EGFP-NK2, correspond to clathrin-coated pre-pits. The similar amount of confined EGFP-NK2 receptors found before and after activation (30-35%) is discussed in term of putative transient interactions of the receptors with preexisting scaffolds of signaling molecules.


Subject(s)
Cell Membrane/metabolism , Receptors, Neurokinin-2/chemistry , Arrestins/metabolism , Boron Compounds/pharmacology , Cell Line , Clathrin/chemistry , Diffusion , Dose-Response Relationship, Drug , Fluorescence Recovery After Photobleaching , Fluorescent Dyes/pharmacology , Genetic Vectors , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/metabolism , Humans , Immunoblotting , Lipids/chemistry , Microscopy, Confocal , Microscopy, Fluorescence , Models, Chemical , Neurokinin A/chemistry , Octoxynol/pharmacology , Protein Structure, Tertiary , Receptors, Neurokinin-2/metabolism , Temperature , Time Factors , Transfection , Transferrin/metabolism , Xanthenes/pharmacology , beta-Arrestins
6.
Neurosci Lett ; 356(3): 220-4, 2004 Feb 19.
Article in English | MEDLINE | ID: mdl-15036634

ABSTRACT

Given the existence of functional interactions between opioidergic and dopaminergic systems, we have analyzed by quantitative autoradiography the possible long-term adaptive changes in the expression of D(1)- and D(2)-like dopamine receptors in the brains of mice lacking the micro-opioid receptor gene. An overall significant increase in D(1) and D(2) receptors (7.4 and 12.6%, respectively) across all cerebral regions examined was obtained in mutant mice relative to wild-type mice. However, region by region comparisons failed to reach significance in any individual brain area. These results indicate that only moderate changes in D(1)- and D(2)-like dopamine receptors densities occur in the brains of micro-opioid receptor knockout mice.


Subject(s)
Brain/metabolism , Receptors, Dopamine/analysis , Receptors, Opioid, mu/deficiency , Analysis of Variance , Animals , Autoradiography/methods , Benzazepines/pharmacokinetics , Brain/anatomy & histology , Dopamine Antagonists/pharmacokinetics , Mice , Mice, Knockout , Raclopride/pharmacokinetics , Receptors, Dopamine/classification , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Tritium/pharmacokinetics
7.
Eur J Pharmacol ; 446(1-3): 103-9, 2002 Jun 20.
Article in English | MEDLINE | ID: mdl-12098591

ABSTRACT

The micro-opioid receptor is the main substrate mediating opiate reward. Multiple micro-opioid receptor subtypes have been postulated to underlie opiate actions. Animals treated with antisense oligonucleotides targeting specific micro-opioid receptor exons show differential sensitivity to morphine versus heroin. The present work examined the rewarding and locomotor activating effects of heroin in mutant mice with a disrupted exon 2 of the micro-opioid receptor. Heroin (1-3 mg/kg) produced significant place preferences and stimulated locomotor activity in wild-type mice, whereas it had no effect in micro-opioid receptor-deficient mice. In contrast, treatment with cocaine (10-30 mg/kg) produced comparable place preferences and locomotor activation in both wild-type and micro-opioid receptor-deficient mice, thus providing evidence that the mutant mice are able to show drug-induced effects in the two behavioral paradigms used here. These results support an essential role for the micro-opioid receptor in the rewarding and locomotor activating effects of heroin.


Subject(s)
Hallucinogens/pharmacology , Heroin/pharmacology , Motor Activity/drug effects , Receptors, Opioid, mu/genetics , Reward , Animals , Cocaine/pharmacology , Conditioning, Psychological/drug effects , Female , Male , Mice , Mice, Knockout
8.
Brain Res ; 943(1): 68-79, 2002 Jul 05.
Article in English | MEDLINE | ID: mdl-12088840

ABSTRACT

There is a large body of evidence indicating important interactions between the adenosine and opioid systems in regulating pain at both the spinal and supraspinal level. Mice lacking the mu-opioid receptor (MOR) gene have been successfully developed and the animals show complete loss of analgesic responses to morphine as well as differences in pain sensitivity. To investigate if there are any compensatory alterations in adenosine systems in mutant animals, we have carried out quantitative autoradiographic mapping of A(1) and A(2A) adenosine receptors and nitrobenzylthioinosine (NBTI) sensitive adenosine transporters in the brains and spinal cords of wild type, heterozygous and homozygous mu-opioid receptor knockout mice. Adjacent coronal sections were cut from the brains and spinal cords of +/+, +/- and -/- mice for the determination of binding of [3H]DPCPX, [3H]CGS21680 or [3H]NBTI to A(1) and A(2A) adenosine receptors and NBTI-sensitive adenosine transporters, respectively. A small but significant reduction in [3H]DPCPX and [3H]NBTI binding was detected in mutant mice brains but not in spinal cords. No significant change in A(2A) binding was detected in mu-opioid receptor knockout brains. The results suggest there may be functional interactions between mu-receptors and A(1) adenosine receptors as well as NBTI-sensitive adenosine transporters in the brain but not in the spinal cord.


Subject(s)
Brain/metabolism , Carrier Proteins/analysis , Membrane Transport Proteins , Receptors, Opioid, mu/deficiency , Receptors, Opioid, mu/genetics , Receptors, Purinergic P1/analysis , Spinal Cord/metabolism , Thioinosine/analogs & derivatives , Thioinosine/metabolism , Animals , Autoradiography , Carrier Proteins/metabolism , Female , Male , Mice , Mice, Knockout , Nucleoside Transport Proteins , Receptor, Adenosine A2A , Receptors, Opioid, mu/biosynthesis , Receptors, Purinergic P1/metabolism , Spinal Cord/chemistry , Thioinosine/pharmacology
9.
J Neurosci ; 22(3): 1146-54, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11826143

ABSTRACT

Repeated THC administration produces motivational and somatic adaptive changes leading to dependence in rodents. To investigate the molecular basis for cannabinoid dependence and its possible relationship with the endogenous opioid system, we explored delta9-tetrahydrocannabinol (THC) activity in mice lacking mu-, delta- or kappa-opioid receptor genes. Acute THC-induced hypothermia, antinociception, and hypolocomotion remained unaffected in these mice, whereas THC tolerance and withdrawal were minimally modified in mutant animals. In contrast, profound phenotypic changes are observed in several place conditioning protocols that reveal both THC rewarding and aversive properties. Absence of microreceptors abolishes THC place preference. Deletion of kappa receptors ablates THC place aversion and furthermore unmasks THC place preference. Thus, an opposing activity of mu- and kappa-opioid receptors in modulating reward pathways forms the basis for the dual euphoric-dysphoric activity of THC.


Subject(s)
Cannabinoids/pharmacology , Marijuana Abuse/physiopathology , Motivation , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Analysis of Variance , Animals , Behavior, Animal/drug effects , Cannabinoids/antagonists & inhibitors , Crosses, Genetic , Dronabinol/antagonists & inhibitors , Dronabinol/pharmacology , Drug Tolerance/genetics , Hypothermia/chemically induced , Mice , Mice, Inbred Strains , Mice, Knockout , Motor Activity/drug effects , Piperidines/pharmacology , Psychotropic Drugs/antagonists & inhibitors , Psychotropic Drugs/pharmacology , Pyrazoles/pharmacology , Receptors, Opioid, delta/deficiency , Receptors, Opioid, delta/genetics , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/deficiency , Receptors, Opioid, kappa/genetics , Receptors, Opioid, mu/deficiency , Receptors, Opioid, mu/genetics , Reward , Rimonabant , Spatial Behavior/drug effects , Substance Withdrawal Syndrome/physiopathology
10.
ALTEX ; 13(3): 130-135, 1996.
Article in English | MEDLINE | ID: mdl-11178453

ABSTRACT

Laboratory breeding of lice, fleas and other bloodsucking arthropods is necessary for developing and testing the efficacy of new drugs and alternative arthropod control methods. They are necessary to investigate their role as vectors of pathogens and to develop methods for diagnosis and therapy of arthropod caused or transmitted diseases. To date, it is not possible to replace rabbits as natural blood sources for breeding lice colonies (Pediculus spp.). But on the base of the knowledge about lice feeding and about in vitro-breeding of other arthropods it would be possible to develop an in vitro-feeding method for lice. For about three years an equipment is available in the USA for in vitro-breeding of cat fleas (Ctenocephalides felis). Using this so called "Artificial dog" it would be possible to replace numerous of cats used as hosts of laboratory flea colonies in pharmaceutical companies and other research laboratories

SELECTION OF CITATIONS
SEARCH DETAIL
...