Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Neurochem Res ; 35(12): 1859-66, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20857334

ABSTRACT

Our laboratory embarked on research to discover proteins the interaction of which with the mu opioid receptor (MOPr) is required for its function and regulation. We performed yeast two-hybrid screens, using the carboxy tail of the human MOPr as bait and a human brain library. This yielded a number of proteins that seemed to bind to the MOPr C-tail. The one we chose to study in detail was filamin A (FLNA). Evidence was obtained that there was indeed protein-protein binding between the C-tail of MOPr and FLNA. A human melanoma cell line (M2) lacking the gene for FLNA and a control cell line (A7) which differed from M2 only in having been transfected with the gene for FLNA and expressing the FLNA protein were made available to us. We transfected these cell lines with the gene for MOPr and used them in our studies. The absence of FLNA strongly reduced MOPr downregulation as well as desensitization of adenylyl cyclase inhibition and G protein activation. A recent finding, published here for the first time, is that FLNA is required for the activation by mu opioid agonists of the MAP kinase p38. Deletion studies indicated that the MOPr binding site on FLNA is in the 24th repeat, close to its C-terminal. It was further found that FLNA lacking the N-terminal actin binding domain is as capable as full length FLNA to restore cells to control status, suggesting that actin binding is not required. A surprising finding was that upregulation of MOPr by morphine and some agonist analogs occurs in M2 cells lacking FLNA, whereas normal receptor downregulation takes place in A7 cells.


Subject(s)
Contractile Proteins/metabolism , Microfilament Proteins/metabolism , Receptors, Opioid, mu/metabolism , Cell Line, Tumor , Contractile Proteins/chemistry , Contractile Proteins/genetics , Filamins , Humans , Melanoma/genetics , Melanoma/pathology , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Morphine/administration & dosage , Morphine/pharmacology , Protein Binding , Sequence Deletion , Two-Hybrid System Techniques , Up-Regulation/drug effects
2.
Neurochem Res ; 33(10): 2054-61, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18404377

ABSTRACT

We have previously reported that the protein filamin A (FLA) binds to the carboxyl tail of the mu opioid receptor (MOPr). Using human melanoma cells, which do not express filamin A, we showed that receptor down-regulation, functional desensitization and trafficking are deficient in the absence of FLA (Onoprishvili et al. Mol Pharmacol 64:1092-1100, 2003). Since FLA has a binding domain for actin and is a member of the family of actin cytoskeleton proteins, it is usually assumed that FLA functions via the actin cytoskeleton. We decided to test this hypothesis by preparing cDNA coding for mutant FLA lacking the actin binding domain (FLA-ABD) and expressing FLA-ABD in the human melanoma cell line M2 (M2-ABD cell line). We report here that this mutant is capable of restoring almost as well as full length FLA the down-regulation of the human MOPr. It is similarly very effective in restoring functional desensitization of MOPr, as assessed by the decrease in G-protein activation after chronic exposure of M2-ABD cells to the mu agonist DAMGO. We also found that A7 cells, expressing wild type FLA, exhibit rapid activation of the MAP kinases, ERK 1 and 2, by DAMGO, as shown by a rise in the level of phospho-ERK 1 and 2. This is followed by rapid dephosphorylation (inactivation), which reaches basal level between 30 and 60 min after DAMGO treatment. M2 cells show normal activation of ERK 1 and 2 in the presence of DAMGO, but very slow inactivation. The rapid rate of MAPK inactivation is partially restored by FLA-ABD. We conclude that some functions of FLA do not act via the actin cytoskeleton. It is likely that other functions, not studied here, may require functional binding of the MOPr-FLA complex to actin.


Subject(s)
Contractile Proteins/genetics , Contractile Proteins/physiology , Melanoma/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/physiology , Receptors, Opioid, mu/metabolism , Actins/metabolism , Cell Line, Tumor , Down-Regulation , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Enzyme Activation , Filamins , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Protein Binding , Protein Structure, Tertiary
3.
Brain Res ; 1177: 9-18, 2007 Oct 26.
Article in English | MEDLINE | ID: mdl-17897634

ABSTRACT

We investigated the effects of morphine and other agonists on the human mu opioid receptor (MOP) expressed in M2 melanoma cells, lacking the actin cytoskeleton protein filamin A and in A7, a subclone of the M2 melanoma cells, stably transfected with filamin A cDNA. The results of binding experiments showed that after chronic morphine treatment (24 h) of A7 cells, MOP-binding sites were down-regulated to 63% of control, whereas, unexpectedly, in M2 cells, MOP binding was up-regulated to 188% of control naive cells. Similar up-regulation was observed with the agonists methadone and levorphanol. The presence of antagonists (naloxone or CTAP) during chronic morphine treatment inhibited MOP down-regulation in A7 cells. In contrast, morphine-induced up-regulation of MOP in M2 cells was further increased by these antagonists. Chronic morphine desensitized MOP in A7 cells, i.e., it decreased DAMGO-induced stimulation of GTPgammaS binding. In M2 cells DAMGO stimulation of GTPgammaS binding was significantly greater than in A7 cells and was not desensitized by chronic morphine. Pertussis toxin treatment abolished morphine-induced receptor up-regulation in M2 cells, whereas it had no effect on morphine-induced down-regulation in A7 cells. These results indicate that, in the absence of filamin A, chronic treatment with morphine, methadone or levorphanol leads to up-regulation of MOP, to our knowledge, the first instance of opioid receptor up-regulation by agonists in cell culture.


Subject(s)
Contractile Proteins/physiology , Microfilament Proteins/physiology , Morphine/pharmacology , Narcotics/pharmacology , Receptors, Opioid, mu/drug effects , Blotting, Western , Cell Line , Cell Line, Tumor , Contractile Proteins/deficiency , Data Interpretation, Statistical , Diprenorphine/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Filamins , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Ligands , Melanoma/genetics , Melanoma/pathology , Microfilament Proteins/deficiency , Narcotic Antagonists/metabolism , Narcotic Antagonists/pharmacology , Pertussis Toxin/pharmacology , Radioligand Assay , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Tubulin/pharmacology , Up-Regulation/drug effects
4.
Mol Pharmacol ; 64(5): 1092-100, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14573758

ABSTRACT

The carboxyl tail of the human mu opioid receptor was shown to bind the carboxyl terminal region of human filamin A, a protein known to couple membrane proteins to actin. Results from yeast two-hybrid screening were confirmed by direct protein-protein binding and by coimmunoprecipitation of filamin and mu opioid receptor from cell lysates. To investigate the role of filamin A in opioid receptor function and regulation, we used the melanoma cell line M2, which does not express filamin A, and its subclone A7, transfected with human filamin A cDNA. Both cell lines were stably transfected with cDNA encoding myc-tagged human mu opioid receptor. Fluorescent studies, using confocal microscopy, provided evidence that filamin and mu opioid receptors were extensively colocalized on the membranes of filamin-expressing melanoma cells. The immunostaining of mu opioid receptors indicated that the lack of filamin had no detectable effect on membrane localization of the receptors. Moreover, mu opioid receptors function normally in the absence of filamin A, as evidenced by studies of opioid binding and DAMGO inhibition of forskolin-stimulated adenylyl cyclase. However, agonist-induced receptor down-regulation and functional desensitization were virtually abolished in cells lacking filamin A. The level of internalized mu-opioid receptors, after 30-min exposure to agonist, was greatly reduced, suggesting a role for filamin in mu opioid receptor trafficking. During these studies, we observed that forskolin activation of adenylyl cyclase was greatly reduced in filamin-lacking cells. An even more unexpected finding was the ability of long-term treatment with [d-Ala2,N-Me-Phe4,Gly5-ol]-enkephalin of M2 cells, containing mu opioid receptors, to restore normal forskolin activation. The mechanism of this effect is currently unknown. It is postulated that the observed effects on mu opioid receptor regulation by filamin A and, by implication, of the actin cytoskeleton may be the result of its role in mu opioid receptor trafficking.


Subject(s)
Contractile Proteins/metabolism , Microfilament Proteins/metabolism , Receptors, Opioid, mu/metabolism , Actins/metabolism , Adenylyl Cyclases/metabolism , Binding Sites , Biological Transport , Cells, Cultured , Filamins , Fluorescent Antibody Technique , Humans , Melanoma/enzymology , Melanoma/pathology , Precipitin Tests , Protein Structure, Tertiary , Transfection , Two-Hybrid System Techniques
5.
BMC Pharmacol ; 2: 5, 2002.
Article in English | MEDLINE | ID: mdl-11897012

ABSTRACT

BACKGROUND: In this study, we investigated the mechanism(s) by which delta opioids induce their potent activation of extracellular signal-regulated protein kinases (ERKs) in different cell lines expressing the cloned delta-opioid receptor (delta-OR). While it has been known for some time that OR stimulation leads to the phosphorylation of both ERK isoforms, the exact progression of events has remained elusive. RESULTS: Our results indicate that the transphosphorylation of an endogenous epidermal growth factor receptor (EGFR) in the human embryonic kidney (HEK-293) cell line does not occur when co-expressed delta-ORs are stimulated by the delta-opioid agonist, D-Ser-Leu-enkephalin-Thr (DSLET). Moreover, neither pre-incubation of cultures with the selective EGFR antagonist, AG1478, nor down-regulation of the EGFR to a point where EGF could no longer activate ERKs had an inhibitory effect on ERK activation by DSLET. These results appear to rule out any structural or catalytic role for the EGFR in the delta-opioid-mediated MAPK cascade. To confirm these results, we used C6 glioma cells, a cell line devoid of the EGFR. In delta-OR-expressing C6 glioma cells, opioids produce a robust phosphorylation of ERK 1 and 2, whereas EGF has no stimulatory effect. Furthermore, antagonists to the RTKs that are endogenously expressed in C6 glioma cells (insulin receptor (IR) and platelet-derived growth factor receptor (PDGFR)) were unable to reduce opioid-mediated ERK activation. CONCLUSION: Taken together, these data suggest that the transactivation of resident RTKs does not appear to be required for OR-mediated ERK phosphorylation and that the tyrosine-phosphorylated delta-OR, itself, is likely to act as its own signalling scaffold.


Subject(s)
Enkephalin, Leucine/analogs & derivatives , Enkephalin, Leucine/pharmacology , MAP Kinase Signaling System/drug effects , Narcotics/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Opioid, delta/metabolism , Cell Line , Enzyme Activation , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Humans , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Receptor, Insulin/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...