Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Mol Brain ; 14(1): 144, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34544455

ABSTRACT

Astrocytes express a plethora of G protein-coupled receptors (GPCRs) that are crucial for shaping synaptic activity. Upon GPCR activation, astrocytes can respond with transient variations in intracellular Ca2+. In addition, Ca2+-dependent and/or Ca2+-independent release of gliotransmitters can occur, allowing them to engage in bidirectional neuron-astrocyte communication. The development of designer receptors exclusively activated by designer drugs (DREADDs) has facilitated many new discoveries on the roles of astrocytes in both physiological and pathological conditions. They are an excellent tool, as they can target endogenous GPCR-mediated intracellular signal transduction pathways specifically in astrocytes. With increasing interest and accumulating research on this topic, several discrepancies on astrocytic Ca2+ signalling and astrocyte-mediated effects on synaptic plasticity have emerged, preventing a clear-cut consensus about the downstream effects of DREADDs in astrocytes. In the present study, we performed a side-by-side evaluation of the effects of bath application of the DREADD agonist, clozapine-N-oxide (10 µM), on Gq- and Gi-DREADD activation in mouse CA1 hippocampal astrocytes. In doing so, we aimed to avoid confounding factors, such as differences in experimental procedures, and to directly compare the actions of both DREADDs on astrocytic intracellular Ca2+ dynamics and synaptic plasticity in acute hippocampal slices. We used an adeno-associated viral vector approach to transduce dorsal hippocampi of male, 8-week-old C57BL6/J mice, to drive expression of either the Gq-DREADD or Gi-DREADD in CA1 astrocytes. A viral vector lacking the DREADD construct was used to generate controls. Here, we show that agonism of Gq-DREADDs, but not Gi-DREADDs, induced consistent increases in spontaneous astrocytic Ca2+ events. Moreover, we demonstrate that both Gq-DREADD as well as Gi-DREADD-mediated activation of CA1 astrocytes induces long-lasting synaptic potentiation in the hippocampal CA1 Schaffer collateral pathway in the absence of a high frequency stimulus. Moreover, we report for the first time that astrocytic Gi-DREADD activation is sufficient to elicit de novo potentiation. Our data demonstrate that activation of either Gq or Gi pathways drives synaptic potentiation through Ca2+-dependent and Ca2+-independent mechanisms, respectively.


Subject(s)
Astrocytes/physiology , CA1 Region, Hippocampal/physiology , Calcium Signaling/physiology , Clozapine/analogs & derivatives , Designer Drugs/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Astrocytes/drug effects , CA1 Region, Hippocampal/cytology , Clozapine/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , Genetic Vectors/pharmacology , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/physiology , Receptors, G-Protein-Coupled/drug effects
2.
Commun Biol ; 4(1): 662, 2021 06 02.
Article in English | MEDLINE | ID: mdl-34079054

ABSTRACT

Pathological impulsivity is a debilitating symptom of multiple psychiatric diseases with few effective treatment options. To identify druggable receptors with anti-impulsive action we developed a systematic target discovery approach combining behavioural chemogenetics and gene expression analysis. Spatially restricted inhibition of three subdivisions of the prefrontal cortex of mice revealed that the anterior cingulate cortex (ACC) regulates premature responding, a form of motor impulsivity. Probing three G-protein cascades with designer receptors, we found that the activation of Gi-signalling in layer-5 pyramidal cells (L5-PCs) of the ACC strongly, reproducibly, and selectively decreased challenge-induced impulsivity. Differential gene expression analysis across murine ACC cell-types and 402 GPCRs revealed that - among Gi-coupled receptor-encoding genes - Grm2 is the most selectively expressed in L5-PCs while alternative targets were scarce. Validating our approach, we confirmed that mGluR2 activation reduced premature responding. These results suggest Gi-coupled receptors in ACC L5-PCs as therapeutic targets for impulse control disorders.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Gyrus Cinguli/cytology , Gyrus Cinguli/physiology , Pyramidal Cells/physiology , Animals , Clozapine/analogs & derivatives , Clozapine/pharmacology , Female , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Gene Expression/drug effects , Gyrus Cinguli/drug effects , Humans , Impulsive Behavior/drug effects , Impulsive Behavior/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pyramidal Cells/cytology , Pyramidal Cells/drug effects , Receptors, Metabotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/physiology , Signal Transduction
3.
Nat Commun ; 8(1): 743, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28963507

ABSTRACT

Inactivation of opioid receptors limits the therapeutic efficacy of morphine-like analgesics and mediates the long duration of kappa opioid antidepressants by an uncharacterized, arrestin-independent mechanism. Here we use an iterative, discovery-based proteomic approach to show that following opioid administration, peroxiredoxin 6 (PRDX6) is recruited to the opioid receptor complex by c-Jun N-terminal kinase (JNK) phosphorylation. PRDX6 activation generates reactive oxygen species via NADPH oxidase, reducing the palmitoylation of receptor-associated Gαi in a JNK-dependent manner. Selective inhibition of PRDX6 blocks Gαi depalmitoylation, prevents the enhanced receptor G-protein association and blocks acute analgesic tolerance to morphine and kappa opioid receptor inactivation in vivo. Opioid stimulation of JNK also inactivates dopamine D2 receptors in a PRDX6-dependent manner. We show that the loss of this lipid modification distorts the receptor G-protein association, thereby preventing agonist-induced guanine nucleotide exchange. These findings establish JNK-dependent PRDX6 recruitment and oxidation-induced Gαi depalmitoylation as an additional mechanism of Gαi-G-protein-coupled receptor inactivation.Opioid receptors are important modulators of nociceptive pain. Here the authors show that opioid receptor activation recruits peroxiredoxin 6 (PRDX6) to the receptor-Gαi complex by c-Jun N-terminal kinase, resulting in Gαi depalmitoylation and enhanced receptor-Gαi association.


Subject(s)
Analgesics, Opioid/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , JNK Mitogen-Activated Protein Kinases/drug effects , Peroxiredoxin VI/drug effects , Receptors, Dopamine D2/drug effects , Animals , Benzeneacetamides/pharmacology , Drug Tolerance , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Fentanyl/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Morphine/pharmacology , NADPH Oxidases/drug effects , NADPH Oxidases/metabolism , Peroxiredoxin VI/metabolism , Phosphorylation , Pyrrolidines/pharmacology , Reactive Oxygen Species/metabolism , Receptors, Dopamine D2/metabolism , Receptors, Opioid/drug effects , Receptors, Opioid/metabolism , Receptors, Opioid, kappa/drug effects , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/metabolism
4.
J Neurochem ; 127(3): 329-41, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23906478

ABSTRACT

It remains unclear how opioid receptors (δ, µ, κ) are implicated in mechanisms controlling differentiation, cell proliferation, and survival. Opioid receptors are coupled to Gi/Go proteins and recent findings have shown that opioid receptors can form a multicomponent signaling complex, consisting of members of G protein and the signal transducer and activator of transcription (STAT)5B. We thus wondered whether activation of the opioid receptors could direct differentiation and neurite outgrowth through a molecular pathway involving STAT5B and other signaling intermediates. We demonstrate that prolonged δ-opioid receptor (δ-OR) activation with opioid agonists induces STAT5B phosphorylation in Neuro-2A cells. Moreover, [D-Ser2, Leu5, Thr6]-enkephalin-activation of δ-OR triggers neurite outgrowth and neuronal survival; these effects are blocked by the selective antagonist naltrindole, by treatment with pertussis toxin, and after expression of a dominant negative mutant of STAT5B (DN-STAT5B), suggesting that the signaling pathway participating in this mechanism involves Gi/o proteins and p-STAT5B. Additional studies have shown that while [D-Ser(2) , Leu(5) , Thr(6) ]-enkephalin exposure of neuroblastoma cells induces a marked increase in the differentiation marker proteins, ßIII-tubulin (Tuj-1), synaptophysin, and neural cell adhesion molecule, over-expression of the DN-STAT5B attenuated significantly their expression levels. Taken together, our findings demonstrate that δ-OR activation leads to a number of neurotropic events via a Gαi/o-linked and STAT5B-dependent manner. We propose a novel signalling pathway for δ-opioid receptor (δ-ΟR)-mediated neurotropic events. STAT5B interacts with the δ-ΟR and upon prolonged receptor activation phosphorylates STAT5B in a Gi/Go dependent manner leading to increased neuronal survival, neurite outgrowth and differentiation. These findings contribute to a better understanding of the molecular and cellular events following δ-OR activation and suggest a possible neuroprotective role opioids could exert.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , Neurons/drug effects , Receptors, Opioid, delta/metabolism , STAT5 Transcription Factor/physiology , Signal Transduction/drug effects , Animals , Blotting, Western , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Survival/physiology , Enkephalins/metabolism , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Mice , Neurites/drug effects , Neurogenesis/physiology , Phosphorylation , Receptors, Opioid, delta/drug effects
5.
Biol Psychiatry ; 74(2): 90-8, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23332355

ABSTRACT

BACKGROUND: In rodents, drugs of abuse induce locomotor hyperactivity, and repeating injections enhance this response. This effect, called behavioral sensitization, persists months after the last administration. It has been shown that behavioral sensitization to amphetamine develops parallel to an increased release of norepinephrine (NE) in the prefrontal cortex (PFC). METHODS: Rats and mice were repeatedly treated with amphetamine (1 or 2 mg/kg intraperitoneally, respectively) to obtain sensitized animals. The NE release in the PFC was measured by microdialysis in freely moving mice (n = 55). Activity of locus coeruleus (LC) noradrenergic neurons was determined in anaesthetized rats (n = 15) by in vivo extracellular electrophysiology. The α2A-adrenergic autoreceptor (α2A-AR) expression was assessed by autoradiography on brain slices, and Gαi proteins expression was measured by western blot analysis of LC punches. RESULTS: In sensitized rats LC neurons had a higher spontaneous firing rate, and clonidine-an α2A-adrenergic agonist-inhibited LC neuronal firing less efficiently than in control animals. Clonidine also induced lower levels of NE release in the PFC of sensitized mice. This desensitization was maintained by a lower density of Gαi1 and Gαi2 proteins in the LC of sensitized mice rather than weaker α2A-AR expression. Behavioral sensitization was facilitated by α2A-AR antagonist, efaroxan, during amphetamine injections and abolished by clonidine treatment. CONCLUSIONS: Our data indicate that noradrenergic inhibitory feedback is impaired for at least 1 month in rats and mice repeatedly treated with amphetamine. This work highlights the key role of noradrenergic autoreceptor signaling in the persistent modifications induced by repeated amphetamine administration.


Subject(s)
Amphetamine/pharmacology , Autoreceptors/metabolism , Central Nervous System Sensitization/drug effects , Motor Activity/drug effects , Norepinephrine/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Animals , Autoreceptors/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Locus Coeruleus/drug effects , Locus Coeruleus/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley
6.
J Pharmacol Exp Ther ; 344(1): 179-88, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23097213

ABSTRACT

Drugs targeting G-protein-coupled receptors (GPCRs) make up more than 25% of all prescribed medicines. The ability of GPCRs to form heteromers with unique signaling properties suggests an entirely new and unexplored pool of drug targets. However, current in vitro assays are ill equipped to detect heteromer-selective compounds. We have successfully adapted an approach, using fusion proteins of GPCRs and chimeric G proteins, to create an in vitro screening assay (in human embryonic kidney cells) in which only activated heteromers are detectable. Here we show that this assay can demonstrate heteromer-selective G-protein bias as well as measure transinhibition. Using this assay, we reveal that the δ-opioid receptor agonist ADL5859, which is currently in clinical trials, has a 10-fold higher potency against δ-opioid receptor homomers than δ/µ-opioid receptor heteromers (pEC(50) = 6.7 ± 0.1 versus 5.8 ± 0.2). The assay enables the screening of large compound libraries to identify heteromer-selective compounds that could then be used in vivo to determine the functional role of heteromers and develop potential therapeutic agents.


Subject(s)
Receptors, G-Protein-Coupled/physiology , Signal Transduction/drug effects , Amino Acid Sequence , Benzamides/pharmacology , Calcium/metabolism , Calcium Signaling , Cloning, Molecular , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enkephalin, Leucine/pharmacology , Enzyme-Linked Immunosorbent Assay , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gs/drug effects , GTP-Binding Protein alpha Subunits, Gs/metabolism , HEK293 Cells , Humans , Microscopy, Fluorescence , Molecular Sequence Data , Mutant Chimeric Proteins/chemistry , Mutant Chimeric Proteins/genetics , Piperazines/pharmacology , Polymerase Chain Reaction , Receptors, G-Protein-Coupled/genetics , Receptors, Opioid, delta/drug effects , Receptors, Opioid, delta/genetics , Recombinant Fusion Proteins
7.
Am J Physiol Heart Circ Physiol ; 302(8): H1645-54, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22307674

ABSTRACT

L-type Ca(2+) channels (LTCCs) play an essential role in the excitation-contraction coupling of ventricular myocytes. We previously found that t-tubular (TT) LTCC current density was halved by the activation of protein phosphatase (PP)1 and/or PP2A, whereas surface sarcolemmal (SS) LTCC current density was increased by the inhibition of PP1 and/or PP2A activity in failing ventricular myocytes of mice chronically treated with isoproterenol (ISO mice). In the present study, we examined the possible involvement of inhibitory heterotrimeric G proteins (G(i/o)) in these abnormalities by chronically administrating pertussis toxin (PTX) to ISO mice (ISO + PTX mice). Compared with ISO mice, ISO + PTX mice exhibited significantly higher fractional shortening of the left ventricle. The expression level of Gα(i2) proteins was not altered by the treatment of mice with ISO and/or PTX. ISO + PTX myocytes had normal TT and SS LTCC current densities because they had higher and lower availability and/or open probability of TT and SS LTCCs than ISO myocytes, respectively. A selective PKA inhibitor, H-89, did not affect LTCC current densities in ISO + PTX myocytes. A selective PP2A inhibitor, fostriecin, did not affect SS or TT current density in control or ISO + PTX myocytes but significantly increased TT but not SS LTCC current density in ISO myocytes. These results indicate that chronic receptor-mediated activation of G(i/o) in vivo decreases basal TT LTCC activity by activating PP2A and increases basal SS LTCC activity by inhibiting PP1 without modulating PKA in heart failure.


Subject(s)
Calcium Channels, L-Type/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Heart Failure/enzymology , Microtubules/metabolism , Phosphoprotein Phosphatases/metabolism , Sarcolemma/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Adrenergic beta-Agonists/pharmacology , Algorithms , Animals , Blood Pressure/drug effects , Calcium Channel Agonists/pharmacology , Calcium Channels, L-Type/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , Heart Failure/diagnostic imaging , Heart Rate/drug effects , Isoproterenol/pharmacology , Male , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Microtubules/drug effects , Myocardium/pathology , Myocytes, Cardiac/physiology , Patch-Clamp Techniques , Pertussis Toxin/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Receptors, G-Protein-Coupled/drug effects , Sarcolemma/drug effects , Ultrasonography
8.
Mol Pharmacol ; 79(3): 488-98, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21088225

ABSTRACT

The chemokine receptor CCR2, which has been implicated in a variety of inflammatory, autoimmune, and cardiovascular conditions, binds several natural chemokine ligands. Here, we assessed the recruitment of ß-arrestin to CCR2 in response to these ligands using bioluminescence resonance energy transfer technology. Compared with CCL2, which was considered as a full agonist, other CCR2 ligands were partial agonists with reduced efficacy and potency. Agonist potencies were not a function of their affinity for CCR2. Efficacy of arrestin recruitment matched that of agonist-induced CCR2 internalization. Although the potency and efficacy rank orders of the ligands in arrestin recruitment were similar to those observed for Gα(i1) activation, arrestin recruitment was at least in part resistant to Gα(i/o)-inactivating pertussis toxin, suggesting partial independence from Gα(i/o). The degree of pertussis toxin resistance of arrestin recruitment was different between the chemokines. Moreover, qualitative differences between the arrestin responses to the different ligands were identified in the stability of the response: although CCL7-induced arrestin recruitment had a half-life of less than 15 min, CCL8 and CCL13 induced stable CCR2-arrestin interactions. Finally, the ligands stabilized different conformations of the CCR2 homodimer. Our results support the validity of models for receptor-ligand interactions in which different ligands stabilize different receptor conformations also for endogenous receptor ligands, with corresponding implications for drug development targeting CCR2.


Subject(s)
Arrestins/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Receptors, CCR2/metabolism , Signal Transduction , Adenylyl Cyclases/metabolism , Dose-Response Relationship, Drug , Endocytosis/drug effects , Flow Cytometry , GTP-Binding Protein alpha Subunits/drug effects , GTP-Binding Protein alpha Subunits/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , HEK293 Cells , Humans , Ligands , Pertussis Toxin/pharmacology , Protein Binding , Radioligand Assay , Receptors, CCR2/drug effects , Signal Transduction/drug effects , beta-Arrestins
9.
Eur J Pharmacol ; 653(1-3): 32-40, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21156170

ABSTRACT

The present study investigated the mechanisms involved in the antinociception produced by the triterpene 3ß, 6ß, 16ß-trihydroxylup-20(29)-ene (TTHL) in mice. TTHL administered by intra-gastric (i.g.) gavage inhibited glutamate-induced nociception with an ID(50) of 19.0 (13.2-27.5) mg/kg. This action started 60 min (inhibition of: 59±6%) after i.g. administration and remained significant up to 6h (inhibition of 37±6%). Moreover, TTHL inhibited both phases of formalin induced pain. The antinociception of TTHL was reversed by the pre-administration of naloxone (1mg/kg; non-selective opioid receptor antagonist), CTOP (1mg/kg; selective µ-opioid receptor antagonist), nor-binaltorphimine (1mg/kg; selective κ-opioid receptor antagonist), naltrindol (3mg/kg; selective δ-opioid receptor antagonist), p-chlorophenylalanine methyl ester (100mg/kg for 4 consecutive days; inhibitor of serotonin synthesis), WAY100635 (0.5mg/kg; selective 5-HT(1A) receptor antagonist) and ketanserin (0.3mg/kg; selective 5-HT(2A) receptor antagonist) but not by L-arginine (600 mg/kg; nitric oxide precursor) or ondansetron (0.5mg/kg; 5-HT(3) receptor antagonist). Furthermore, the TTHL antinociception was prevented by intrathecal (i.t.) pre-treatment with pertussis toxin (0.5 µg/site; inactivator of G(i/o) protein), charybdotoxin (250 pg/site; blocker of large-conductance calcium-gated K(+) channels), tetraethylammonium (1 µg/site; blocker of voltage-gated K(+) channels) and glibenclamide (80 µg/site; blocker of ATP-gated K(+) channels) but not by apamin (50 ng/site; blocker of small-conductance calcium-gated K(+) channels). The antinociception of TTHL was not it associated with locomotor impairment or sedation. These results showed that TTHL presented a pronounced antinociceptive effect, which is dependent on opioid and serotonergic systems, G(i/o) protein activation and the opening of specific K(+) channels.


Subject(s)
Analgesics/pharmacology , Pain/drug therapy , Triterpenes/pharmacology , Analgesics/administration & dosage , Analgesics/toxicity , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Inhibitory Concentration 50 , Male , Mice , Motor Activity/drug effects , Pain/physiopathology , Potassium Channels/drug effects , Potassium Channels/metabolism , Receptors, Opioid/drug effects , Receptors, Opioid/metabolism , Receptors, Serotonin/drug effects , Receptors, Serotonin/metabolism , Time Factors , Triterpenes/administration & dosage , Triterpenes/toxicity
10.
J Neurochem ; 113(4): 1046-59, 2010 May.
Article in English | MEDLINE | ID: mdl-20236221

ABSTRACT

Studies in animal models of Parkinson's disease have revealed that degeneration of noradrenaline neurons is involved in the motor deficits. Several types of adrenoceptors are highly expressed in neostriatal neurons. However, the selective actions of these receptors on striatal signaling pathways have not been characterized. In this study, we investigated the role of adrenoceptors in the regulation of dopamine/dopamine- and cAMP-regulated phosphoprotein of M(r) 32 kDa (DARPP-32) signaling by analyzing DARPP-32 phosphorylation at Thr34 [protein kinase A (PKA)-site] in mouse neostriatal slices. Activation of beta(1)-adrenoceptors induced a rapid and transient increase in DARPP-32 phosphorylation. Activation of alpha(2)-adrenoceptors also induced a rapid and transient increase in DARPP-32 phosphorylation, which subsequently decreased below basal levels. In addition, activation of alpha(2)-adrenoceptors attenuated, and blockade of alpha(2)-adrenoceptors enhanced dopamine D(1) and adenosine A(2A) receptor/DARPP-32 signaling. Chemical lesioning of noradrenergic neurons mimicked the effects of alpha(2)-adrenoceptor blockade. Under conditions of alpha(2)-adrenoceptor blockade, the dopamine D(2) receptor-induced decrease in DARPP-32 phosphorylation was attenuated. Our data demonstrate that beta(1)- and alpha(2)-adrenoceptors regulate DARPP-32 phosphorylation in neostriatal neurons. G(i) activation by alpha(2)-adrenoceptors antagonizes G(s)/PKA signaling mediated by D(1) and A(2A) receptors in striatonigral and striatopallidal neurons, respectively, and thereby enhances D(2) receptor/G(i) signaling in striatopallidal neurons. alpha(2)-Adrenoceptors may therefore be a therapeutic target for the treatment of Parkinson's disease.


Subject(s)
Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Dopamine/metabolism , Neostriatum/metabolism , Neurons/metabolism , Receptors, Adrenergic/metabolism , Adrenergic alpha-2 Receptor Antagonists , Animals , Catalytic Domain/drug effects , Catalytic Domain/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Male , Mice , Mice, Inbred C57BL , Neostriatum/drug effects , Neurons/drug effects , Organ Culture Techniques , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Phosphorylation/drug effects , Receptor, Adenosine A2A/drug effects , Receptor, Adenosine A2A/metabolism , Receptors, Adrenergic/drug effects , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Adrenergic, beta-1/drug effects , Receptors, Adrenergic, beta-1/metabolism , Receptors, Dopamine D2/drug effects , Receptors, Dopamine D2/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Threonine/metabolism
11.
Biochem Pharmacol ; 79(10): 1506-15, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20097180

ABSTRACT

BLT2, a low-affinity leukotriene B4 (LTB4) receptor, is a member of the G-protein coupled receptor (GPCR) family and is involved in the pathogenesis of inflammatory diseases such as asthma. Despite its clinical implications, however, no pharmacological inhibitors are available. In the present study, we screened for small molecules that interfere with the interaction between the third intracellular loop region of BLT2 (BLT2iL3) and the Galphai3 protein subunit (Galphai3), using a high-throughput screening (HTS) assay with a library of 1040 FDA-approved drugs and bioactive compounds. We identified two small molecules-purpurin [1,2,4-trihydroxy-9,10-anthraquinone; IC50 = 1.6 microM for BLT2] and chloranil [tetrachloro-1,4-benzoquinone; IC50 = 0.42 microM for BLT2]-as specific BLT2-blocking agents. We found that blockade of the BLT2iL3-Galphai3 interaction by these small molecules inhibited the BLT2-downstream signaling cascade. For example, BLT2-signaling to phosphoinositide-3 kinase (PI3K)/Akt phosphorylation was completely abolished by these molecules. Furthermore, we observed that these small molecules blocked LTB4-induced chemotaxis by inhibiting the BLT2-PI3K/Akt-downstream, Rac1-reactive oxygen species-dependent pathway. Taken together, our results show that purpurin and chloranil interfere with the interaction between BLT2iL3 and Galphai3 and thus block the biological functions of BLT2 (e.g., chemotaxis). The present findings suggest a potential application of purpurin and chloranil as pharmacological therapeutic agents against BLT2-associated inflammatory human diseases.


Subject(s)
Anthraquinones/pharmacology , Chemotaxis/drug effects , Chloranil/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Receptors, Leukotriene B4/antagonists & inhibitors , Animals , Anthraquinones/metabolism , CHO Cells , Cell Survival , Chloranil/metabolism , Cricetinae , Cricetulus , Electrophoresis, Polyacrylamide Gel , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , Immunoblotting , Signal Transduction/drug effects , Signal Transduction/physiology , Surface Plasmon Resonance , rac1 GTP-Binding Protein/drug effects , rac1 GTP-Binding Protein/physiology
12.
J Headache Pain ; 10(6): 435-40, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19756945

ABSTRACT

A hypofunctionality of Gi proteins has been found in migraine patients. The fixed combination of indomethacin, prochlorperazine and caffeine (Indoprocaf) is a drug of well-established use in the acute treatment of migraine and tension-type headache. The aim of this study was to investigate if Indoprocaf was able to exert its central antinociceptive action when Gi proteins activity is abolished by pertussis toxin (PTX), compared to its single active ingredients and to sumatriptan. The mice model of abdominal constriction test induced by an i.p. injection of a 0.6% solution of acetic acid was used. The study showed that Indoprocaf (a fixed combination of indomethacin 1 mg/kg, prochlorperazine 1 mg/kg and caffeine 3 mg/kg, s.c.) and sumatriptan (20 mg/kg, s.c.) exert their central antinociceptive action independently from the Gi proteins. In addition, the antinociceptive efficacy of Indoprocaf in this study was statistically superior to that of sumatriptan. This study also showed that the single active ingredients of Indoprocaf, indomethacin (1 mg/kg, s.c.), prochlorperazine (1 mg/kg, s.c.) and caffeine (3 mg/kg, s.c.), were able to exert their central antinociceptive action independently from the Gi proteins. However, Indoprocaf at analgesic doses was able to abolish almost completely the abdominal constrictions, with a statistically higher efficacy compared to the single active ingredients, showing an important synergic effect of Indoprocaf. This synergic effect was evident not only when Gi proteins activity was abolished by PTX, but also under control condition, when Gi proteins were active. This study suggests that the central antinociceptive action induced by antimigraine drugs is independent from Gi proteins.


Subject(s)
Analgesics/pharmacology , Caffeine/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Indomethacin/pharmacology , Migraine Disorders/drug therapy , Migraine Disorders/metabolism , Prochlorperazine/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Central Nervous System Stimulants/pharmacology , Cerebral Arteries/drug effects , Cerebral Arteries/metabolism , Cerebral Arteries/physiopathology , Disease Models, Animal , Dopamine Antagonists/pharmacology , Drug Combinations , Drug Evaluation, Preclinical , Drug Synergism , Male , Mice , Migraine Disorders/physiopathology , Serotonin Receptor Agonists/pharmacology , Sumatriptan/pharmacology , Treatment Outcome
13.
J Biomol Screen ; 14(3): 246-55, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19211780

ABSTRACT

G-protein-coupled receptors can couple to different signal transduction pathways in different cell types (termed cell-specific signaling) and can activate different signaling pathways depending on the receptor conformation(s) stabilized by the activating ligand (functional selectivity). These concepts offer potential for developing pathway-specific drugs that increase efficacy and reduce side effects. Despite significant interest, functional selectivity has been difficult to exploit in drug discovery, in part due to the burden of multiple assays. Cellular impedance assays use an emerging technology that can qualitatively distinguish Gs, Gi/o, and Gq signaling in a single assay and is thereby suited for studying these pharmacological concepts. Cellular impedance confirmed cell-specific Gs and Gq coupling for the melanocortin-4 receptor and dual Gi and Gs signaling with the cannabinoid-1 (CB1) receptor. The balance of Gi versus Gs signaling depended on the cell line. In CB1-HEKs, Giand Gs-like responses combined to yield a novel impedance profile demonstrating the dynamic nature of these traces. Cellspecific signaling was observed with endogenous D1 receptor in U-2 cells and SK-N-MC cells, yet the pharmacological profile of partial and full agonists was similar in both cell lines. We conclude that the dynamic impedance profile encodes valuable relative signaling information and is sufficiently robust to help evaluate cell-specific signaling and functional selectivity.


Subject(s)
Biological Assay/methods , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , CHO Cells , Cell Culture Techniques , Cell Line, Tumor , Cells, Cultured , Cricetinae , Cricetulus , Cytochalasin D/pharmacology , Dopamine Agonists/pharmacology , Dose-Response Relationship, Drug , Electric Impedance , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , GTP-Binding Protein alpha Subunits, Gs/drug effects , Humans , Inhibitory Concentration 50 , Kidney/cytology , Neuroectodermal Tumors, Primitive, Peripheral/metabolism , Neuroectodermal Tumors, Primitive, Peripheral/pathology , Osteosarcoma/metabolism , Osteosarcoma/pathology , Pertussis Toxin/pharmacology , Receptor, Muscarinic M1/metabolism , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D5/metabolism , Sensitivity and Specificity , Signal Transduction/drug effects , Signal Transduction/physiology , alpha-MSH/agonists , alpha-MSH/analogs & derivatives
14.
Pharmacology ; 82(4): 245-9, 2008.
Article in English | MEDLINE | ID: mdl-18818509

ABSTRACT

GPR35 is a Gi/o- and G16-coupled receptor abundantly expressed in gastrointestinal tissues and immune cells. Kynurenic acid (a tryptophan metabolite and ionotropic glutamate receptor antagonist) and zaprinast (a phosphodiesterase inhibitor) are GPR35 agonists. Here, we show that the chloride channel blocker 5-nitro-2-(3-phenylpropylamino) benzoic acid (NPPB) is also a GPR35 agonist. NPPB activates the GPR35-Gi/o and GPR35-G16 pathways in human embryonic kidney 293 (HEK293) cells and induces intracellular calcium mobilization in a concentration-dependent manner in HEK293 cells coexpressing human, rat or mouse GPR35 and the chimeric G protein G(qi5). These results suggest a novel pharmacological activity of NPPB and will provide useful information to search for more potent and selective GPR35 agonists.


Subject(s)
Nitrobenzoates/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , Cell Line , Chloride Channels/antagonists & inhibitors , Dose-Response Relationship, Drug , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Kidney/cytology , Kidney/metabolism , Mice , Nitrobenzoates/administration & dosage , Purinones/administration & dosage , Purinones/pharmacology , Rats
15.
Am J Physiol Cell Physiol ; 295(5): C1417-26, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18815223

ABSTRACT

Activator of G protein Signaling 1 (AGS1) and Ras homologue enriched in striatum (Rhes) define a new group of Ras-like monomeric G proteins whose signaling properties and physiological roles are just beginning to be understood. Previous results suggest that AGS1 and Rhes exhibit distinct preferences for heterotrimeric G proteins, with AGS1 selectively influencing Galphai and Rhes selectively influencing Galphas. Here, we demonstrate that AGS1 and Rhes trigger nearly identical modulation of N-type Ca(2+) channels (Ca(V)2.2) by selectively altering Galphai-dependent signaling. Whole-cell currents were recorded from HEK293 cells expressing Ca(V)2.2 and Galphai- or Galphas-coupled receptors. AGS1 and Rhes reduced basal current densities and triggered tonic voltage-dependent (VD) inhibition of Ca(V)2.2. Additionally, each protein attenuated agonist-initiated channel inhibition through Galphai-coupled receptors without reducing channel inhibition through a Galphas-coupled receptor. The above effects of AGS1 and Rhes were blocked by pertussis toxin (PTX) or by expression of a Gbetagamma-sequestering peptide (masGRK3ct). Transfection with HRas, KRas2, Rap1A-G12V, Rap2B, Rheb2, or Gem failed to duplicate the effects of AGS1 and Rhes on Ca(V)2.2. Our data provide the first demonstration that AGS1 and Rhes exhibit similar if not identical signaling properties since both trigger tonic Gbetagamma signaling and both attenuate receptor-initiated signaling by the Gbetagamma subunits of PTX-sensitive G proteins. These results are consistent with the possibility that AGS1 and Rhes modulate Ca(2+) influx through Ca(V)2.2 channels under more physiological conditions and thereby influence Ca(2+)-dependent events such as neurosecretion.


Subject(s)
Calcium Channels, N-Type/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Proteins/metabolism , Proteins/metabolism , Signal Transduction , Animals , Calcium Channels, N-Type/drug effects , Calcium Channels, N-Type/genetics , Carbachol/pharmacology , Cell Line , Cholinergic Agonists/pharmacology , Exodeoxyribonucleases , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , GTP-Binding Proteins/genetics , Humans , Membrane Potentials , Pertussis Toxin/pharmacology , Phosphoproteins , Proteins/genetics , Rabbits , Receptor, Muscarinic M2/agonists , Receptor, Muscarinic M2/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/drug effects , Transfection
17.
Pharmacology ; 82(2): 97-104, 2008.
Article in English | MEDLINE | ID: mdl-18547979

ABSTRACT

Regulators of G protein signaling (RGS) are a family of GTPase-activating proteins (GAP) that interact with heterotrimeric G proteins in the negative regulation of G-protein-coupled receptor (GPCR) signaling. RGS4, the first identified mammalian member of the RGS family, has been implicated in many GPCR signaling pathways involved in disease states. We report herein the identification of a 16-amino-acid peptide (P17) as an inhibitor of RGS4. The peptide was found by screening a random peptide library using RGS4 as 'bait' in a yeast two-hybrid system. This peptide inhibited RGS4 GAP activity on Galpha(i1)in a GTPase assay, and blocked the interaction between RGS4 and Galpha(i1)in a pull-down assay. The peptide displayed dose-dependent inhibition of RGS4 and Galpha-interacting protein (GAIP) GAP activities, yet showed no substantial effect on RGS7. Electrophysiological studies in Xenopus oocytes demonstrated that P17 attenuates RGS4 modulation of M(2) muscarinic receptor stimulation of GIRK (G-protein-mediated inwardly rectifying potassium) channels. Deletion of an arginine at the N terminus of P17 abolished its ability to inhibit RGS4 GAP activity, as did deletions of C-terminal residues. The P17 peptide showed no similarity to any known peptide sequence. Further investigation and optimization of the peptide may provide unique information for the development of RGS4 inhibitors for future therapeutic application.


Subject(s)
Peptides/pharmacology , RGS Proteins/antagonists & inhibitors , Signal Transduction/drug effects , Amino Acid Sequence , Animals , Dose-Response Relationship, Drug , Drug Design , Electrophysiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , GTP Phosphohydrolases/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Oocytes , Peptide Library , Peptides/administration & dosage , Peptides/chemistry , Receptor, Muscarinic M2/metabolism , Two-Hybrid System Techniques , Xenopus laevis
18.
Learn Mem ; 15(4): 261-70, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18391187

ABSTRACT

Cyclic AMP signaling plays a central role in regulating activity at a number of synapses in the brain. We showed previously that pairing activation of receptors that inhibit adenylate cyclase (AC) and reduce the concentration of cyclic AMP, with elevation of the concentration of cyclic GMP is sufficient to elicit a presynaptically expressed form of LTD at Schaffer collateral-CA1 synapses in the hippocampus. To directly test the role of AC inhibition and G-protein signaling in LTD at these synapses, we utilized transgenic mice that express a mutant, constitutively active inhibitory G protein, Galpha(i2), in principal neurons of the forebrain. Transgene expression of Galpha(i2) markedly enhanced LTD and impaired late-phase LTP at Schaffer collateral synapses, with no associated differences in input/output relations, paired-pulse facilitation, or NMDA receptor-gated conductances. When paired with application of a type V phosphodiesterase inhibitor to elevate the concentration of intracellular cyclic GMP, constitutively active Galpha(i2) expression converted the transient depression normally caused by this treatment to an LTD that persisted after the drug was washed out. Moreover, this effect could be mimicked in control slices by pairing type V phosphodiesterase inhibitor application with application of a PKA inhibitor. Electrophysiological recordings of spontaneous excitatory postsynaptic currents and two-photon visualization of vesicular release using FM1-43 revealed that constitutively active Galpha(i2) tonically reduced basal release probability from the rapidly recycling vesicle pool of Schaffer collateral terminals. Our findings support the hypothesis that inhibitory G-protein signaling acts presynaptically to regulate release, and, when paired with elevations in the concentration of cyclic GMP, converts a transient cyclic GMP-induced depression into a long-lasting decrease in release.


Subject(s)
Adenylyl Cyclase Inhibitors , Cyclic GMP/metabolism , Enzyme Inhibitors/pharmacology , GTP-Binding Protein Regulators/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , Hippocampus , Intracellular Signaling Peptides and Proteins/pharmacology , Phosphodiesterase 5 Inhibitors , Phosphodiesterase Inhibitors/pharmacology , Receptors, Presynaptic/drug effects , Synapses/drug effects , Animals , Depression/metabolism , Depression/psychology , Disease Models, Animal , Drug Synergism , Enzyme Inhibitors/administration & dosage , Hippocampus/anatomy & histology , Hippocampus/drug effects , Hippocampus/metabolism , Intracellular Signaling Peptides and Proteins/administration & dosage , Long-Term Potentiation , Mice , Mice, Transgenic , Phosphodiesterase Inhibitors/administration & dosage
19.
Physiol Res ; 57(1): 137-139, 2008.
Article in English | MEDLINE | ID: mdl-18412515

ABSTRACT

We investigated the effect of pertussis toxin (PTX) on hypotensive response induced by acetylcholine (ACh) and bradykinin (BK) and on noradrenaline (NA)-induced pressor response in spontaneously hypertensive rats (SHR). Fifteen-week-old Wistar rats and age-matched SHR were used. Half of SHR received PTX (10 microg/kg/i.v.) and the experiments were performed 48 h later. After the anesthesia the right carotid artery was cannulated in order to record blood pressure (BP). The hypotensive response to ACh was enhanced in SHR compared to Wistar rats. After pretreatment of SHR with PTX the hypotensive response to ACh was reduced compared to untreated SHR and it was also diminished in comparison to Wistar rats. Similarly, the hypotensive response to BK was also decreased after PTX pretreatment. The pressor response to NA was increased in SHR compared to Wistar rats. NA-induced pressor response was considerably decreased after PTX pretreatment compared to untreated SHR. In conclusion, the enhancement of hypotensive and pressor responses in SHR was abolished after PTX pretreatment. Our results suggested that the activation of PTX-sensitive inhibitory G(i) proteins is involved in the regulation of integrated vasoactive responses in SHR and PTX pretreatment could be effectively used for modification of BP regulation in this type of experimental hypertension.


Subject(s)
Enzyme Inhibitors/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Hypotension/physiopathology , Norepinephrine/metabolism , Pertussis Toxin/pharmacology , Acetylcholine , Adaptation, Physiological/drug effects , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Bradykinin , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , Hemodynamics/drug effects , Hemodynamics/physiology , Hypotension/chemically induced , Male , Matched-Pair Analysis , Rats , Rats, Inbred SHR , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology
20.
J Endocrinol ; 196(1): 67-77, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18180318

ABSTRACT

Although there is significant evidence for progesterone's role as an immunomodulator, nuclear progesterone receptors have not been consistently identified in immune cells. Recently, three new putative membrane progesterone receptors (mPRs), mPRalpha, mPRbeta, and mPRgamma have been described. The objective of this study was to examine whether mPRs are expressed in peripheral blood leukocytes (PBLs) in women of reproductive age, and to further characterize them in T lymphocytes and immortalized T cells (Jurkat cells). Transcripts for mPRalpha and mPRbeta but not mPRgamma, were detected by RT-PCR in PBLs, T lymphocytes, and Jurkat cells. Western blot analysis showed the presence of the mPRalpha and mPRbeta proteins on cell membranes of T lymphocytes and Jurkat cells. Expression of the mPRalpha mRNA was upregulated in the luteal phase of the menstrual cycle in cluster of differentiation (CD)8+, but not in CD4+, T lymphocytes. Radioreceptor assays revealed specific [(3)H]progesterone binding to T- and Jurkat cell membranes (K(d) 4.25 nM) characteristic of steroid membrane receptors. Progesterone activated an inhibitory G-protein (G(i)), suggesting that mPRs are coupled to G(i) in Jurkat cells. These results suggest a potential novel mechanism for progesterone's immunoregulatory function through activation of mPRs.


Subject(s)
Cell Membrane/chemistry , GTP-Binding Proteins/drug effects , Gene Expression/drug effects , Progesterone/pharmacology , Receptors, Progesterone/genetics , T-Lymphocytes/chemistry , Adult , Cell Membrane/metabolism , Female , Flow Cytometry , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , GTP-Binding Proteins/physiology , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Immunosorbent Techniques , Jurkat Cells , Luteal Phase , Progesterone/metabolism , RNA, Messenger/analysis , Receptors, Progesterone/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sulfur Radioisotopes , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...