Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurosci ; 36(5): 1669-81, 2016 Feb 03.
Article in English | MEDLINE | ID: mdl-26843648

ABSTRACT

The ventrolateral periaqueductal gray (vlPAG) is a key structure in the descending pain modulatory circuit. Activation of the circuit occurs via disinhibition of GABAergic inputs onto vlPAG output neurons. In these studies, we tested the hypothesis that GABAergic inhibition is increased during persistent inflammation, dampening activation of the descending circuit from the vlPAG. Our results indicate that persistent inflammation induced by Complete Freund's adjuvant (CFA) modulates GABA signaling differently in male and female rats. CFA treatment results in increased presynaptic GABA release but decreased high-affinity tonic GABAA currents in female vlPAG neurons. These effects are not observed in males. The tonic currents in the vlPAG are dependent on GABA transporter activity and are modulated by agonists that activate GABAA receptors containing the δ subunit. The GABAA δ agonist THIP (gaboxadol) induced similar amplitude currents in naive and CFA-treated rats. In addition, a positive allosteric modulator of the GABAA δ subunit, DS2 (4-chloro-N-[2-(2-thienyl)imidazo[1,2-a]pyridin-3-yl]benzamide), increased tonic currents. These results indicate that GABAA δ receptors remain on the cell surface but are less active in CFA-treated female rats. In vivo behavior studies showed that morphine induced greater antinociception in CFA-treated females that was reversed with microinjections of DS2 directly into the vlPAG. DS2 did not affect morphine antinociception in naive or CFA-treated male rats. Together, these data indicate that sex-specific adaptations in GABAA receptor signaling modulate opioid analgesia in persistent inflammation. Antagonists of GABAA δ receptors may be a viable strategy for reducing pain associated with persistent inflammation, particularly in females. SIGNIFICANCE STATEMENT: These studies demonstrate that GABA signaling is modulated in the ventrolateral periaqueductal gray by persistent inflammation differently in female and male rats. Our results indicate that antagonists or negative allosteric modulators of GABAA δ receptors may be an effective strategy to alleviate chronic inflammatory pain and promote opioid antinociception, especially in females.


Subject(s)
Chronic Pain/physiopathology , Periaqueductal Gray/physiology , Receptors, GABA-A/physiology , Sex Characteristics , Signal Transduction/physiology , Animals , Chronic Pain/etiology , Dose-Response Relationship, Drug , Female , GABA Agonists/pharmacology , Hot Temperature/adverse effects , Inflammation/pathology , Inflammation/physiopathology , Male , Pain Measurement/drug effects , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , gamma-Aminobutyric Acid/pharmacology
2.
Mol Pharmacol ; 88(2): 347-56, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26013542

ABSTRACT

There is ongoing debate about the role of G protein-coupled receptor kinases (GRKs) in agonist-induced desensitization of the µ-opioid receptor (MOPr) in brain neurons. In the present paper, we have used a novel membrane-permeable, small-molecule inhibitor of GRK2 and GRK3, Takeda compound 101 (Cmpd101; 3-[[[4-methyl-5-(4-pyridyl)-4H-1,2,4-triazole-3-yl] methyl] amino]-N-[2-(trifuoromethyl) benzyl] benzamidehydrochloride), to study the involvement of GRK2/3 in acute agonist-induced MOPr desensitization. We observed that Cmpd101 inhibits the desensitization of the G protein-activated inwardly-rectifying potassium current evoked by receptor-saturating concentrations of methionine-enkephalin (Met-Enk), [d-Ala(2), N-MePhe(4), Gly-ol(5)]-enkephalin (DAMGO), endomorphin-2, and morphine in rat and mouse locus coeruleus (LC) neurons. In LC neurons from GRK3 knockout mice, Met-Enk-induced desensitization was unaffected, implying a role for GRK2 in MOPr desensitization. Quantitative analysis of the loss of functional MOPrs following acute agonist exposure revealed that Cmpd101 only partially reversed MOPr desensitization. Inhibition of extracellular signal-regulated kinase 1/2, protein kinase C, c-Jun N-terminal kinase, or GRK5 did not inhibit the Cmpd101-insensitive component of desensitization. In HEK 293 cells, Cmpd101 produced almost complete inhibition of DAMGO-induced MOPr phosphorylation at Ser(375), arrestin translocation, and MOPr internalization. Our data demonstrate a role for GRK2 (and potentially also GRK3) in agonist-induced MOPr desensitization in the LC, but leave open the possibility that another, as yet unidentified, mechanism of desensitization also exists.


Subject(s)
Benzamides/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinase 3/metabolism , Locus Coeruleus/drug effects , Receptors, Opioid, mu/metabolism , Animals , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar
3.
Eur J Neurosci ; 36(12): 3636-42, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23002724

ABSTRACT

There is considerable controversy over whether µ-opioid receptor (MOPr) desensitization is homologous or heterologous and over the mechanisms underlying such desensitization. In different cell types MOPr desensitization has been reported to involve receptor phosphorylation by various kinases, including G-protein-coupled receptor kinases (GRKs), second messenger and other kinases as well as perturbation of the MOPr effector pathway by GRK sequestration of G protein ßγ subunits or ion channel modulation. Here we report that in brainstem locus coeruleus (LC) neurons prepared from relatively mature rats (5-8 weeks old) rapid MOPr desensitization induced by the high-efficacy opioid peptides methionine enkephalin and DAMGO was homologous and not heterologous to α(2)-adrenoceptors and somatostatin SST(2) receptors. Given that these receptors all couple through G proteins to the same set of G-protein inwardly rectifying (GIRK) channels it is unlikely therefore that in mature neurons MOPr desensitization involves G protein ßγ subunit sequestration or ion channel modulation. In contrast, in slices from immature animals (less than postnatal day 20), MOPr desensitization was observed to be heterologous and could be downstream of the receptor. Heterologous MOPr desensitization was not dependent on protein kinase C or c-Jun N-terminal kinase activity, but the change from heterologous to homologous desensitization with age was correlated with a decrease in the expression levels of GRK2 in the LC and other brain regions. The observation that the mechanisms underlying MOPr desensitization change with neuronal development is important when extrapolating to the mature brain results obtained from experiments on expression systems, cell lines and immature neuronal preparations.


Subject(s)
Action Potentials , Receptors, Opioid, mu/metabolism , Animals , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Enkephalin, Methionine/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 2/metabolism , GTP-Binding Proteins/metabolism , Gene Expression , Locus Coeruleus/growth & development , Locus Coeruleus/physiology , Male , Mitogen-Activated Protein Kinase 8/metabolism , Neurons/metabolism , Neurons/physiology , Neurotransmitter Agents/pharmacology , Protein Kinase C/metabolism , Protein Subunits/metabolism , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Somatostatin/metabolism
4.
J Biol Chem ; 281(45): 34515-24, 2006 Nov 10.
Article in English | MEDLINE | ID: mdl-16982618

ABSTRACT

In this study we investigated the mechanisms responsible for MAP kinase ERK1/2 activation following agonist activation of endogenous mu opioid receptors (MOR) normally expressed in cultured striatal neurons. Treatment with the MOR agonist fentanyl caused significant activation of ERK1/2 in neurons derived from wild type mice. Fentanyl effects were blocked by the opioid antagonist naloxone and were not evident in neurons derived from MOR knock-out (-/-) mice. In contrast, ERK1/2 activation by fentanyl was not evident in neurons from GRK3-/- mice or neurons pretreated with small inhibitory RNA for arrestin3. Consistent with this observation, treatment with the opiate morphine (which is less able to activate arrestin) did not elicit ERK1/2 activation in wild type neurons; however, transfection of arrestin3-(R170E) (a dominant positive form of arrestin that does not require receptor phosphorylation for activation) enabled morphine activation of ERK1/2. In addition, activation of ERK1/2 by fentanyl and morphine was rescued in GRK3-/- neurons following transfection with dominant positive arrestin3-(R170E). The activation of ERK1/2 appeared to be selective as p38 MAP kinase activation was not increased by either fentanyl or morphine treatment in neurons from wild type, MOR-/-, or GRK3-/- mice. In addition, U0126 (a selective inhibitor of MEK kinase responsible for ERK phosphorylation) blocked ERK1/2 activation by fentanyl. These results support the hypothesis that MOR activation of ERK1/2 requires opioid receptor phosphorylation by GRK3 and association of arrestin3 to initiate the cascade resulting in ERK1/2 phosphorylation in striatal neurons.


Subject(s)
Arrestins/metabolism , Corpus Striatum/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/metabolism , Receptors, Opioid, mu/physiology , beta-Adrenergic Receptor Kinases/physiology , Animals , Arrestins/antagonists & inhibitors , Arrestins/genetics , Cells, Cultured , Corpus Striatum/cytology , Corpus Striatum/ultrastructure , Enzyme Activation/drug effects , Fentanyl/pharmacology , G-Protein-Coupled Receptor Kinase 3 , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, Knockout , Naloxone/pharmacology , Narcotic Antagonists , Neurons/cytology , Neurons/ultrastructure , Phosphorylation/drug effects , RNA, Small Interfering/pharmacology , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , beta-Adrenergic Receptor Kinases/genetics
6.
J Biol Chem ; 281(26): 18081-9, 2006 Jun 30.
Article in English | MEDLINE | ID: mdl-16648139

ABSTRACT

AtT-20 cells expressing the wild-type kappa opioid receptor (KOR) increased phospho-p38 MAPK following treatment with the kappa agonist U50,488. The increase was blocked by the kappa antagonist norbinaltorphimine and not evident in untransfected cells. In contrast, U50,488 treatment of AtT-20 cells expressing KOR having alanine substituted for serine-369 (KSA) did not increase phospho-p38. Phosphorylation of serine 369 in the KOR carboxyl terminus by G-protein receptor kinase 3 (GRK3) was previously shown to be required for receptor desensitization, and the results suggest that p38 MAPK activation by KOR may require arrestin recruitment. This hypothesis was tested by transfecting arrestin3-(R170E), a dominant positive form of arrestin that does not require receptor phosphorylation for activation. AtT-20 cells expressing both KSA and arrestin3-(R170E) responded to U50,488 treatment with an increase in phospho-p38 consistent with the hypothesis. Primary cultured astrocytes (glial fibrillary acidic protein-positive) and neurons (gamma-aminobutyric acid-positive) isolated from mouse striata also responded to U50,488 by increasing phospho-p38 immunolabeling. p38 activation was not evident in either striatal astrocytes or neurons isolated from KOR knock-out mice or GRK3 knock-out mice. Astrocytes pretreated with small interfering RNA for arrestin3 were also unable to activate p38 in response to U50,488 treatment. Furthermore, in striatal neurons, the kappa-mediated phospho-p38 labeling was colocalized with arrestin3. These findings suggest that KOR may activate p38 MAPK in brain by a GRK3 and arrestin-dependent mechanism.


Subject(s)
Arrestins/metabolism , Astrocytes/metabolism , Neurons/metabolism , Receptors, Opioid, kappa/metabolism , beta-Adrenergic Receptor Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Analgesics, Non-Narcotic/pharmacology , Animals , Astrocytes/cytology , Cells, Cultured , Corpus Striatum/cytology , G-Protein-Coupled Receptor Kinase 3 , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Neurons/cytology , Phosphorylation , Rats , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/antagonists & inhibitors , Signal Transduction/physiology
7.
J Biol Chem ; 277(18): 15729-35, 2002 May 03.
Article in English | MEDLINE | ID: mdl-11861651

ABSTRACT

Receptor desensitization by G-protein receptor kinases (GRK) and arrestins is likely to be an important component underlying the development of tolerance to opioid drugs. Reconstitution of this process in Xenopus oocytes revealed distinct differences in the kinetics of GRK and arrestin regulation of the closely related opioid receptors mu (MOR), delta (DOR), and kappa (KOR). We demonstrated that under identical conditions, GRK and arrestin-dependent desensitization of MOR proceeds dramatically slower than that of DOR. Furthermore, GRK3 phosphorylation sites required for opioid receptor desensitization also greatly differ. The determinants for DOR and KOR desensitization reside in the carboxyl-terminal tail, whereas MOR depends on Thr-180 in the second intracellular loop. Although this later finding might indicate an inefficient phosphorylation of MOR Thr-180, increasing the amount of arrestin expressed greatly increased the rate of MOR desensitization to a rate comparable with that of DOR. Similarly, coexpression of a constitutively active arrestin 2(R169E) with MOR and DOR desensitized both receptors in an agonist-dependent, GRK-independent manner at rates that were indistinguishable. Together, these data suggest that it is the activation of arrestin, rather than its binding, that is the rate-limiting step in MOR desensitization. In addition, mutation of Thr-161 in DOR, homologous to MOR Thr-180, significantly inhibited the faster desensitization of DOR. These results suggest that DOR desensitization involves phosphorylation of both the carboxyl-terminal tail and the second intracellular loop that together leads to a more efficient activation of arrestin and thus faster desensitization.


Subject(s)
Arrestins/metabolism , Receptors, Opioid, delta/physiology , Receptors, Opioid, mu/physiology , Amino Acid Sequence , Amino Acid Substitution , Animals , Arrestin , Arrestins/genetics , Cloning, Molecular , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Enkephalin, D-Penicillamine (2,5)-/pharmacology , G-Protein-Coupled Receptor Kinase 3 , GTP-Binding Proteins/metabolism , Kinetics , Mice , Models, Molecular , Mutagenesis, Site-Directed , Oocytes/physiology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Protein Conformation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Opioid, delta/drug effects , Receptors, Opioid, delta/genetics , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/genetics , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Threonine , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...