Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Neurobiol Dis ; 89: 55-64, 2016 May.
Article in English | MEDLINE | ID: mdl-26804029

ABSTRACT

To investigate whether the endogenous neuropeptide nociceptin/orphanin FQ (N/OFQ) contributes to the death of dopamine neurons in Parkinson's disease, we undertook a genetic and a pharmacological approach using NOP receptor knockout (NOP(-/-)) mice, and the selective and potent small molecule NOP receptor antagonist (-)-cis-1-methyl-7-[[4-(2,6-dichlorophenyl)piperidin-1-yl]methyl]-6,7,8,9-tetrahydro-5H-benzocyclohepten-5-ol (SB-612111). Stereological unbiased methods were used to estimate the total number of dopamine neurons in the substantia nigra of i) NOP(-/-) mice acutely treated with the parkinsonian neurotoxin 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine (MPTP), ii) naïve mice subacutely treated with MPTP, alone or in combination with SB-612111, iii) rats injected with a recombinant adeno-associated viral (AAV) vector overexpressing human mutant p.A53T α-synuclein, treated with vehicle or SB-612111. NOP(-/-) mice showed a 50% greater amount of nigral dopamine neurons spared in response to acute MPTP compared to controls, which was associated with a milder motor impairment. SB-612111, given 4 days after MPTP treatment to mimic the clinical condition, prevented the loss of nigral dopamine neurons and striatal dopaminergic terminals caused by subacute MPTP. SB-612111, administered a week after the AAV injections in a clinically-driven protocol, also increased by 50% both the number of spared nigral dopamine neurons and striatal dopamine terminals, and prevented accompanying motor deficits induced by α-synuclein. We conclude that endogenous N/OFQ contributes to dopamine neuron loss in pathogenic and etiologic models of Parkinson's disease through NOP receptor-mediated mechanisms. NOP receptor antagonists might prove effective as disease-modifying agents in Parkinson's disease, through the rescue of degenerating nigral dopamine neurons and/or the protection of the healthy ones.


Subject(s)
Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Receptors, Opioid/metabolism , Substantia Nigra/metabolism , Substantia Nigra/pathology , Animals , Cycloheptanes/administration & dosage , Dopaminergic Neurons/drug effects , Gene Deletion , Locomotion/drug effects , MPTP Poisoning , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Narcotic Antagonists/administration & dosage , Parkinsonian Disorders/genetics , Piperidines/administration & dosage , Rats , Rats, Sprague-Dawley , Receptors, Opioid/genetics , Substantia Nigra/drug effects , Nociceptin Receptor
2.
Mol Neurobiol ; 53(10): 7284-7297, 2016 12.
Article in English | MEDLINE | ID: mdl-26687234

ABSTRACT

Nociceptin/orphanin FQ (N/OFQ) is an opioid-like neuropeptide that binds and signals through a G-protein-coupled receptor called the N/OFQ peptide (NOP) receptor. N/OFQ and the NOP receptor are expressed in the midbrain and have been implicated in the pathogenesis of Parkinson's disease (PD). Genetic removal of the N/OFQ precursor partially protects midbrain dopaminergic neurons from 1-methyl-4-phenylpyridine-induced toxicity, suggesting that endogenous N/OFQ may be detrimental to dopaminergic neurons. However, whether N/OFQ directly affects the survival and growth of dopaminergic neurons is unknown. Here, we show that N/OFQ has a detrimental effect on the survival of dopaminergic neurons and the growth of their axons in primary cultures of the E14 rat ventral mesencephalon. N/OFQ potentiates the effects of the neurotoxins 6-hydroxydopamine and 1-methyl-4-phenylpyridinium through p38-MAPK signalling. We also show that like α-synuclein, there is a significant reduction in N/OFQ messenger RNA (mRNA) expression in the midbrain of patients with Parkinson's disease. These results demonstrate for the first time that N/OFQ is detrimental to the survival and growth of dopaminergic neurons and that its expression is altered in the midbrain of patients with Parkinson's disease.


Subject(s)
Axons/metabolism , Dopaminergic Neurons/cytology , Dopaminergic Neurons/enzymology , Mesencephalon/cytology , Opioid Peptides/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , 1-Methyl-4-phenylpyridinium , Animals , Axons/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dopaminergic Neurons/drug effects , Humans , Neurotoxins/toxicity , Oxidopamine , Parkinson Disease/enzymology , Parkinson Disease/genetics , Parkinson Disease/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Nociceptin
3.
ACS Chem Neurosci ; 6(6): 911-9, 2015 Jun 17.
Article in English | MEDLINE | ID: mdl-25844489

ABSTRACT

Many current therapies target G protein coupled receptors (GPCR), transporters, or ion channels. In addition to directly targeting these proteins, disrupting the protein-protein interactions that localize or regulate their function could enhance selectivity and provide unique pharmacologic actions. Regulators of G protein signaling (RGS) proteins, especially RGS4, play significant roles in epilepsy and Parkinson's disease. Thiadiazolidinone (TDZD) inhibitors of RGS4 are nanomolar potency blockers of the biochemical actions of RGS4 in vitro. Here, we demonstrate the substantial selectivity (8- to >5000-fold) of CCG-203769 for RGS4 over other RGS proteins. It is also 300-fold selective for RGS4 over GSK-3ß, another target of this class of chemical scaffolds. It does not inhibit the cysteine protease papain at 100 µM. CCG-203769 enhances Gαq-dependent cellular Ca(2+) signaling in an RGS4-dependent manner. TDZD inhibitors also enhance Gαi-dependent δ-OR inhibition of cAMP production in SH-SY-5Y cells, which express endogenous receptors and RGS4. Importantly, CCG-203769 potentiates the known RGS4 mechanism of Gαi-dependent muscarinic bradycardia in vivo. Furthermore, it reverses raclopride-induced akinesia and bradykinesia in mice, a model of some aspects of the movement disorder in Parkinson's disease. A broad assessment of compound effects revealed minimal off-target effects at concentrations necessary for cellular RGS4 inhibition. These results expand our understanding of the mechanism and specificity of TDZD RGS inhibitors and support the potential for therapeutic targeting of RGS proteins in Parkinson's disease and other neural disorders.


Subject(s)
Antiparkinson Agents/pharmacology , RGS Proteins/antagonists & inhibitors , Animals , Bradycardia/drug therapy , Bradycardia/physiopathology , Calcium/metabolism , Carbachol/pharmacology , Cell Line, Tumor , Cholinergic Agonists/pharmacology , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Activity/physiology , Papain/metabolism , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/physiopathology , RGS Proteins/metabolism , Raclopride , Rats, Sprague-Dawley
4.
Mater Sci Eng C Mater Biol Appl ; 48: 294-300, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25579926

ABSTRACT

This paper describes the production, characterization and in vivo activity of lipid nanocarriers (LN) containing a levodopa prodrug (LD-PD) with therapeutic potential in Parkinson's disease. LD is the mainstay of the pharmacotherapy of Parkinson's disease. However, after a good initial response, motor fluctuations, dyskinesia and loss of efficacy, develop over time, partly due to oscillations in plasma and brain levels of the drug. LD-PD was produced with the aim of prolonging the pharmacological activity of LD. To improve solubility, and simultaneously provide a long lasting release and therapeutic efficacy, the prodrug was formulated in tristearin/lecithin LN. The obtained formulation was homogeneous in particle size and remained stable for up to 2months from preparation. For the three different tested LD concentrations, namely 1.25, 2.5 and 5.0mg/ml, the morphological characterization revealed no substantial differences between unloaded and LD-PD loaded LN. The calorimetric test showed an interaction between the lipid phase and the loaded prodrug. In vitro studies using the dialysis method and enzymatic degradation procedure showed that the LD-PD loaded LN provided a controlled prodrug release. Finally, two behavioural tests specific to akinesia (bar test) or akinesia/bradykinesia (drag test) performed in 6-hydroxydopamine hemilesioned mice (a model of Parkinson's disease) demonstrated that the LD-PD loaded LN attenuated parkinsonian disabilities, showing a slightly reduced maximal efficacy but a longer lasting action (up to 24h) than an equal dose of LD. We conclude that LD-PD loaded LN may represent a future LD formulation useful in Parkinson's disease therapy.


Subject(s)
Antiparkinson Agents/administration & dosage , Drug Carriers/chemistry , Levodopa/administration & dosage , Lipids/chemistry , Prodrugs/chemistry , Animals , Antiparkinson Agents/chemistry , Calorimetry, Differential Scanning , Cryoelectron Microscopy , Disease Models, Animal , Drug Carriers/administration & dosage , Lecithins/chemistry , Levodopa/pharmacokinetics , Mice, Inbred C57BL , Parkinson Disease/drug therapy , Prodrugs/administration & dosage , Prodrugs/chemical synthesis , Spectroscopy, Fourier Transform Infrared , Triglycerides/chemistry
5.
Neuropharmacology ; 72: 126-38, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23643745

ABSTRACT

A combined pharmacological and genetic approach was undertaken to investigate the contribution of endogenous dopamine to the motor actions of nociceptin/orphanin FQ (N/OFQ) receptor (NOP receptor) ligands. Motor activity was evaluated by a battery of behavioural tests in mice. The involvement of the various DA receptor subtypes in the motor effects of N/OFQ and NOP receptor antagonists was evaluated pharmacologically, using D1/D5 (SCH23390), D2/D3 (raclopride, amisulpride) and D3 (S33084) receptor antagonists, and by using D2 receptor knockout mice. Low doses of N/OFQ and NOP receptor antagonists promoted movement whereas higher doses inhibited it. Motor facilitation was selectively prevented by raclopride while motor inhibition was prevented by amisulpride. Amisulpride also attenuated the hypolocomotion induced by the D2/D3 receptor agonist pramipexole and dopamine precursor l-3,4-dihydroxyphenylalanine, whereas raclopride (and S33084) worsened it. To dissect out the contribution of pre- and postsynaptic D2 receptors, mice lacking the D2 receptor (D2R(-/-)) or its long isoform (D2L(-/-)) were used. Motor facilitation induced by N/OFQ and NOP receptor antagonists was lost in D2R(-/-) and D2L(-/-) mice whereas motor inhibition induced by NOP receptor antagonists (and pramipexole) was lost in D2R(-/-) but preserved in D2L(-/-) mice. N/OFQ-induced hypolocomotion was observed in both genotypes. We demonstrate that motor actions of NOP receptor ligands rely on the modulation of endogenous dopamine. Motor facilitation induced by NOP receptor antagonists as well as low dose N/OFQ is mediated through D2L postsynaptic receptors whereas motor inhibition observed with higher doses of N/OFQ occurs by direct inhibition of mesencephalic DA neurons. Motor inhibition seen with high doses of NOP receptor antagonists appears to be mediated through the D2 presynaptic autoreceptors. These data confirm that endogenous N/OFQ is a powerful modulator of dopamine transmission in vivo and that the effects of NOP receptor antagonists on motor function reflect the blockade of this endogenous N/OFQ tone.


Subject(s)
Motor Activity/drug effects , Motor Activity/genetics , Opioid Peptides/metabolism , Opioid Peptides/pharmacology , Receptors, Dopamine D2/metabolism , Analgesics, Opioid/pharmacology , Animals , Carbamates/pharmacology , Catalepsy/chemically induced , Catalepsy/genetics , Dopamine Agents/pharmacology , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Levodopa/pharmacology , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Morphine/pharmacology , Opioid Peptides/genetics , Receptors, Dopamine D2/genetics , Rotarod Performance Test , Nociceptin
6.
ACS Chem Neurosci ; 4(5): 808-16, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23611155

ABSTRACT

Dual probe microdialysis was used to investigate whether GluN2A and GluN2B NMDA receptor subunits regulate striatal output pathways under dyskinetic conditions. The preferential GluN2A antagonist NVP-AAM077 perfused in the dopamine-depleted striatum of 6-hydroxydopamine hemilesioned dyskinetic rats reduced GABA and glutamate levels in globus pallidus whereas the selective GluN2B antagonist Ro 25-6981 elevated glutamate without affecting pallidal GABA. Moreover, intrastriatal NVP-AAM077 did not affect GABA but elevated glutamate levels in substantia nigra reticulata whereas Ro 25-6981 elevated GABA and reduced nigral glutamate. To investigate whether GluN2A and GluN2B NMDA receptor subunits are involved in motor pathways underlying dyskinesia expression, systemic NVP-AAM077 and Ro 25-6981 were tested for their ability to attenuate levodopa-induced abnormal involuntary movements. NVP-AAM077 failed to prevent dyskinesia while Ro 25-6981 mildly attenuated it. We conclude that in the dyskinetic striatum, striatal GluN2A subunits tonically stimulate the striato-pallidal pathway whereas striatal GluN2B subunits tonically inhibit striato-nigral projections. Moreover, GluN2A subunits are not involved in dyskinesia expression whereas GluN2B subunits minimally contribute to it.


Subject(s)
Corpus Striatum/metabolism , Dopamine/metabolism , Glutamic Acid/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Corpus Striatum/drug effects , Dopamine Agents/adverse effects , Dyskinesia, Drug-Induced/etiology , Globus Pallidus/drug effects , Globus Pallidus/metabolism , Glutamic Acid/drug effects , Levodopa/adverse effects , Male , Microdialysis , Neostriatum/drug effects , Neostriatum/metabolism , Oxidopamine/adverse effects , Phenols/pharmacology , Piperidines/pharmacology , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Substantia Nigra/drug effects , Substantia Nigra/metabolism , gamma-Aminobutyric Acid/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...