Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Cell Rep ; 38(9): 110435, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35235798

ABSTRACT

Apolipoprotein E transports lipids and couples metabolism between astrocytes and neurons. The E4 variant (APOE4) affects these functions and represents a genetic predisposition for Alzheimer's disease, but the molecular mechanisms remain elusive. We show that ApoE produces different types of lipoproteins via distinct lipidation pathways. ApoE forms high-density lipoprotein (HDL)-like, cholesterol-rich particles via the ATP-binding cassette transporter 1 (ABCA1), a mechanism largely unaffected by ApoE polymorphism. Alternatively, ectopic accumulation of fat in astrocytes, a stress-associated condition, redirects ApoE toward the assembly and secretion of triacylglycerol-rich lipoproteins, a process boosted by the APOE4 variant. We demonstrate in vitro that ApoE can detect triacylglycerol in membranes and spontaneously assemble lipoprotein particles (10-20 nm) rich in unsaturated triacylglycerol, and that APOE4 has remarkable properties behaving as a strong triacylglycerol binder. We propose that fatty APOE4 astrocytes have reduced ability to clear toxic fatty acids from the extracellular milieu, because APOE4 reroutes them back to secretion.


Subject(s)
Apolipoprotein E4 , Astrocytes , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Apolipoproteins E/metabolism , Astrocytes/metabolism , Protein Isoforms/metabolism , Triglycerides/metabolism
2.
CPT Pharmacometrics Syst Pharmacol ; 7(11): 759-770, 2018 11.
Article in English | MEDLINE | ID: mdl-30207429

ABSTRACT

Alzheimer disease (AD) is a devastating neurodegenerative disorder with high unmet medical need. Drug development is hampered by limited understanding of the disease and its driving factors. Quantitative Systems Pharmacology (QSP) modeling provides a comprehensive quantitative framework to evaluate the relevance of biological mechanisms in the context of disease and to predict the efficacy of novel treatments. Here, we report a QSP model for AD with a particular focus on investigating the relevance of dysregulation of cholesterol and sphingolipids. We show that our model captures the modulation of several biomarkers in subjects with AD, as well as the response to pharmacological interventions. We evaluate the impact of targeting the sphingosine-1-phosphate 5 receptor (S1PR5) as a potential novel treatment option for AD, and model predictions increase our confidence in this novel disease pathway. Future applications for the QSP model are in validation of further targets and identification of potential treatment response biomarkers.


Subject(s)
Alzheimer Disease/drug therapy , Sphingolipids/metabolism , Aged , Alzheimer Disease/metabolism , Animals , Case-Control Studies , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Rats, Sprague-Dawley , Rats, Wistar , Reproducibility of Results
3.
Hum Mol Genet ; 27(14): 2490-2501, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29688337

ABSTRACT

Huntington's disease (HD) is the most common neurodegenerative disorder for which no effective cure is yet available. Although several agents have been identified to provide benefits so far, the number of therapeutic options remains limited with only symptomatic treatment available. Over the past few years, we have demonstrated that sphingolipid-based approaches may open the door to new and more targeted treatments for the disease. In this study, we investigated the therapeutic potential of stimulating sphingosine-1-phosphate (S1P) receptor 5 by the new selective agonist A-971432 (provided by AbbVie) in R6/2 mice, a widely used HD animal model. Chronic administration of low-dose (0.1 mg/kg) A-971432 slowed down the progression of the disease and significantly prolonged lifespan in symptomatic R6/2 mice. Such beneficial effects were associated with activation of pro-survival pathways (BDNF, AKT and ERK) and with reduction of mutant huntingtin aggregation. A-971432 also protected blood-brain barrier (BBB) homeostasis in the same mice. Interestingly, when administered early in the disease, before any overt symptoms, A-971432 completely protected HD mice from the classic progressive motor deficit and preserved BBB integrity. Beside representing a promising strategy to take into consideration for the development of alternative therapeutic options for HD, selective stimulation of S1P receptor 5 may be also seen as an effective approach to target brain vasculature defects in the disease.


Subject(s)
Blood-Brain Barrier/drug effects , Huntingtin Protein/genetics , Huntington Disease/drug therapy , Protein Aggregation, Pathological/drug therapy , Receptors, Lysosphingolipid/genetics , Animals , Brain/drug effects , Brain/pathology , Brain-Derived Neurotrophic Factor/genetics , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/genetics , Gene Expression Regulation/drug effects , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Mice , Mice, Transgenic , Motor Activity/drug effects , Protein Aggregation, Pathological/physiopathology , Proto-Oncogene Proteins c-akt/genetics , Receptors, Lysosphingolipid/agonists
4.
Neuropharmacology ; 101: 358-69, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26471422

ABSTRACT

The GABA(B) receptor has been indicated as a promising target for multiple CNS-related disorders. Baclofen, a prototypical orthosteric agonist, is used clinically for the treatment of spastic movement disorders, but is associated with unwanted side-effects, such as sedation and motor impairment. Positive allosteric modulators (PAM), which bind to a topographically-distinct site apart from the orthosteric binding pocket, may provide an improved side-effect profile while maintaining baclofen-like efficacy. GABA, the major inhibitory neurotransmitter in the CNS, plays an important role in the etiology and treatment of seizure disorders. Baclofen is known to produce anticonvulsant effects in the DBA/2J mouse audiogenic seizure test (AGS), suggesting it may be a suitable assay for assessing pharmacodynamic effects. Little is known about the effects of GABA(B) PAMs, however. The studies presented here sought to investigate the AGS test as a pharmacodynamic (PD) screening model for GABA(B) PAMs by comparing the profile of structurally diverse PAMs to baclofen. GS39783, rac-BHFF, CMPPE, A-1295120 (N-(3-(4-(4-chloro-3-fluorobenzyl)-6-methoxy-3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)phenyl)acetamide), and A-1474713 (N-(3-(4-(4-chlorobenzyl)-3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)-yl)phenyl)acetamide) all produced robust, dose-dependent anticonvulsant effects; a similar profile was observed with baclofen. Pre-treatment with the GABA(B) antagonist SCH50911 completely blocked the anticonvulsant effects of baclofen and CMPPE in the AGS test, indicating such effects are likely mediated by the GABA(B) receptor. In addition to the standard anticonvulsant endpoint of the AGS test, video tracking software was employed to assess potential drug-induced motor side-effects during the acclimation period of the test. This analysis was sensitive to detecting drug-induced changes in total distance traveled, which was used to establish a therapeutic index (TI = hypoactivity/anticonvulsant effects). Calculated TIs for A-1295120, CMPPE, rac-BHFF, GS39783, and A-1474713 were 5.31x, 5.00x, 4.74x, 3.41x, and 1.83x, respectively, whereas baclofen was <1. The results presented here suggest the DBA/2J mouse AGS test is a potentially useful screening model for detecting PD effects of GABA(B) PAMs and can provide an initial read-out on target-related motor side-effects. Furthermore, an improved TI was observed for PAMs compared to baclofen, indicating the PAM approach may be a viable therapeutic alternative to baclofen.


Subject(s)
Anticonvulsants/chemistry , Anticonvulsants/therapeutic use , Baclofen/therapeutic use , Seizures/drug therapy , Acoustic Stimulation/adverse effects , Allosteric Regulation/drug effects , Allosteric Site/drug effects , Animals , Animals, Newborn , Cyclopentanes/pharmacology , Drug Interactions , GABA Agonists/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , Male , Mice , Mice, Inbred DBA , Morpholines/pharmacology , Motor Activity/drug effects , Protein Binding/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Seizures/etiology , Sulfur Isotopes/pharmacokinetics
5.
J Med Chem ; 58(23): 9154-70, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26509640

ABSTRACT

S1P5 is one of 5 receptors for sphingosine-1-phosphate and is highly expressed on endothelial cells within the blood-brain barrier, where it maintains barrier integrity in in vitro models (J. Neuroinflamm. 2012, 9, 133). Little more is known about the effects of S1P5 modulation due to the absence of tool molecules with suitable selectivity and drug-like properties. We recently reported that molecule A-971432 (Harris, 2010) (29 in this paper) is highly efficacious in reversing lipid accumulation and age-related cognitive decline in rats (Van der Kam , , AAIC 2014). Herein we describe the development of a series of selective S1P5 agonists that led to the identification of compound 29, which is highly selective for S1P5 and has excellent plasma and CNS exposure after oral dosing in preclinical species. To further support its suitability for in vivo studies of S1P5 biology, we extensively characterized 29, including confirmation of its selectivity in pharmacodynamic assays of S1P1 and S1P3 function in rats. In addition, we found that 29 improves blood-brain barrier integrity in an in vitro model and reverses age-related cognitive decline in mice. These results suggest that S1P5 agonism is an innovative approach with potential benefit in neurodegenerative disorders involving lipid imbalance and/or compromised blood-brain barrier such as Alzheimer's disease or multiple sclerosis.


Subject(s)
Azetidinecarboxylic Acid/analogs & derivatives , Benzene Derivatives/chemistry , Benzene Derivatives/pharmacology , Neurodegenerative Diseases/drug therapy , Receptors, Lysosphingolipid/agonists , Administration, Oral , Animals , Azetidinecarboxylic Acid/administration & dosage , Azetidinecarboxylic Acid/chemistry , Azetidinecarboxylic Acid/pharmacokinetics , Azetidinecarboxylic Acid/pharmacology , Benzene Derivatives/administration & dosage , Benzene Derivatives/pharmacokinetics , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Cell Line , Cognitive Aging , Dogs , Female , Humans , Macaca fascicularis , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Rats , Rats, Sprague-Dawley , Receptors, Lysosphingolipid/metabolism
6.
J Neuroinflammation ; 9: 133, 2012 Jun 20.
Article in English | MEDLINE | ID: mdl-22715976

ABSTRACT

BACKGROUND: The sphingosine 1-phosphate (S1P) receptor modulator FTY720P (Gilenya®) potently reduces relapse rate and lesion activity in the neuroinflammatory disorder multiple sclerosis. Although most of its efficacy has been shown to be related to immunosuppression through the induction of lymphopenia, it has been suggested that a number of its beneficial effects are related to altered endothelial and blood-brain barrier (BBB) functionality. However, to date it remains unknown whether brain endothelial S1P receptors are involved in the maintenance of the function of the BBB thereby mediating immune quiescence of the brain. Here we demonstrate that the brain endothelial receptor S1P5 largely contributes to the maintenance of brain endothelial barrier function. METHODS: We analyzed the expression of S1P5 in human post-mortem tissues using immunohistochemistry. The function of S1P5 at the BBB was assessed in cultured human brain endothelial cells (ECs) using agonists and lentivirus-mediated knockdown of S1P5. Subsequent analyses of different aspects of the brain EC barrier included the formation of a tight barrier, the expression of BBB proteins and markers of inflammation and monocyte transmigration. RESULTS: We show that activation of S1P5 on cultured human brain ECs by a selective agonist elicits enhanced barrier integrity and reduced transendothelial migration of monocytes in vitro. These results were corroborated by genetically silencing S1P5 in brain ECs. Interestingly, functional studies with these cells revealed that S1P5 strongly contributes to brain EC barrier function and underlies the expression of specific BBB endothelial characteristics such as tight junctions and permeability. In addition, S1P5 maintains the immunoquiescent state of brain ECs with low expression levels of leukocyte adhesion molecules and inflammatory chemokines and cytokines through lowering the activation of the transcription factor NFκB. CONCLUSION: Our findings demonstrate that S1P5 in brain ECs contributes to optimal barrier formation and maintenance of immune quiescence of the barrier endothelium.


Subject(s)
Blood-Brain Barrier/cytology , Blood-Brain Barrier/immunology , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Immunity, Cellular , Receptors, Lysosphingolipid/physiology , Aged, 80 and over , Blood-Brain Barrier/metabolism , Cell Line , Cells, Cultured , Endothelium, Vascular/metabolism , Gene Knockdown Techniques , Humans , Immunity, Cellular/genetics , Lentivirus/genetics , Male , Receptors, Lysosphingolipid/deficiency , Receptors, Lysosphingolipid/genetics
7.
Pharmacology ; 87(5-6): 286-96, 2011.
Article in English | MEDLINE | ID: mdl-21577043

ABSTRACT

The rewarding effects of drugs of abuse are often studied by means of the conditioned place preference (CPP) paradigm. CPP is one of the most widely used models in behavioral pharmacology, yet its theoretical underpinnings are not well understood, and there are very few studies on the methodological and theoretical aspects of this model. An important drawback of the classical CPP paradigm is that it often does not show dose-dependent results. The persistence of the conditioned response, i.e. the time required until the CPP effect is extinct, may be related to the strength of conditioning, which in turn may be related to the rewarding efficacy of a drug. Resistance to extinction may therefore be a useful additional measure to quantify the rewarding effect of drugs. In the present study we examined the persistence of drug-environment associations after conditioning with morphine (1, 3 and 10 mg/kg i.p.), oxycodone (0.3, 1 and 3 mg/kg i.p.) and heroin (0.05, 0.25 and 0.5 mg/kg i.p.) by repeated retesting in the CPP apparatus (15-min sessions, 5 days/week) until the rats reached extinction (i.e. less than 55% preference over 3 consecutive sessions). Following an unbiased CPP protocol, morphine, oxycodone and heroin induced CPP with minimal effective doses of 3, 1 and 0.25 mg/kg, respectively, and with similar effect sizes for each CPP-inducing dose. The number of sessions required for extinction was positively correlated with the dose of the drug (experiment 1: 18 and 45 sessions for 3 and 10 mg/kg morphine, and 19 and 27 sessions for 1 and 3 mg/kg oxycodone; experiment 2: 12 and 24 sessions for 3 and 10 mg/kg morphine, and 10 and 14 sessions for 0.25 and 0.5 mg/kg heroin). These findings suggest that the use of an extinction paradigm can extend the quantitative assessment of the rewarding effect of drugs - however, within certain limits only. The present paradigm appears to be less suited for comparing the rewarding efficacy of different drugs due to great test-retest variability. Finally, the additional potential gain of information using this paradigm has to be weighed against the considerably large amount of additional time and effort.


Subject(s)
Analgesics, Opioid/pharmacology , Conditioning, Psychological/drug effects , Extinction, Psychological/drug effects , Animals , Dose-Response Relationship, Drug , Heroin/pharmacology , Male , Morphine/pharmacology , Oxycodone/pharmacology , Rats , Rats, Sprague-Dawley , Substance-Related Disorders
8.
Eur J Pain ; 15(3): 299-305, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20801699

ABSTRACT

It was previously shown that morphine more potently reduces the affective as compared to the sensory component of nociception, and this effect is independent of morphine's rewarding properties. Here we investigated whether this finding can be generalized to other classes of anti-nociceptive drugs. The effect of oxycodone (0-10 mg/kg, i.p.), tramadol (0-10 mg/kg, i.p.), ibuprofen (0-300 mg/kg, i.p.) and pregabalin (0-31.6 mg/kg, i.p.) on negative affect and mechanical hypersensitivity accompanying carrageenan-induced (0.5% intraplantar) inflammatory nociception was assessed using conditioned place aversion (CPA) and Randall Selitto paw pressure test, respectively. The rewarding effect of these drugs was assessed using conditioned place preference (CPP). All four anti-nociceptive drugs dose-dependently reduced carrageenan-induced CPA and mechanical hypersensitivity. Furthermore all drugs induced CPP, except for ibuprofen. Similar to morphine, oxycodone and tramadol showed a large dissociation of anti-aversive versus anti-nociceptive potency, i.e. 10 times more potent against the affective versus the sensory component of nociception. Oxycodone and tramadol were 30 and 10 times more potent to produce CPP in animals under normal versus painful conditions. Ibuprofen and pregabalin also showed a dissociation of anti-aversive and anti-nociceptive potency, but less pronounced (i.e. three times more potent against the affective component). However, pregabalin showed no dissociation between rewarding potency under normal versus painful conditions. Taken together, these data suggest that the dissociation of rewarding potency in animals under normal versus painful conditions is limited to drugs with an opioid mechanism of action, while the dissociation of anti-aversive and anti-nociceptive potency applies to anti-nociceptive drugs with different mechanisms of action.


Subject(s)
Analgesics/pharmacology , Conditioning, Psychological/drug effects , Ibuprofen/pharmacology , Oxycodone/pharmacology , Pain/drug therapy , Tramadol/pharmacology , gamma-Aminobutyric Acid/analogs & derivatives , Analgesics/therapeutic use , Animals , Carrageenan , Dose-Response Relationship, Drug , Ibuprofen/therapeutic use , Inflammation/chemically induced , Male , Oxycodone/therapeutic use , Pain/chemically induced , Pain Measurement/drug effects , Pregabalin , Rats , Rats, Sprague-Dawley , Tramadol/therapeutic use , gamma-Aminobutyric Acid/pharmacology , gamma-Aminobutyric Acid/therapeutic use
9.
Addict Biol ; 16(1): 108-15, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20579001

ABSTRACT

We have recently reported that the metabotropic glutamate receptor 5 antagonist 2-methyl-6-(phenylethynyl)-pyridine (MPEP) potentiates acquisition of conditioned place preference (CPP) induced by heroin and ketamine. The present study investigated to what extent this effect of MPEP can be generalized to other classes of drugs, such as the stimulants nicotine and cocaine, and to drugs that produce CPP in the rat despite a lack of abuse potential in humans, such as buspirone and clonidine. Adult male Sprague Dawley rats were subjected to a standard unbiased CPP protocol (six conditioning sessions lasting 20 minutes for nicotine and 40 minutes for the other compounds). Rats were conditioned with either nicotine (0.05-0.2 mg/kg, subcutaneously), cocaine [1-10 mg/kg, intraperitoneally (i.p.)], buspirone (0.3-3 mg/kg, i.p.) or clonidine (0.2-0.6 mg/kg, i.p.) in combination with MPEP (0 or 10 mg/kg, i.p.). For nicotine and cocaine, the minimal effective dose to induce CPP was lowered by pre-treatment with MPEP. While buspirone and clonidine did not induce CPP when given alone (i.e. combined with MPEP vehicle), both compounds induced CPP after pre-treatment with MPEP. It is concluded that MPEP consistently potentiates acquisition of drug-induced reward, independent of the mechanism of action of the co-administered drug. We suggest that the proposed anti-abuse effect of MPEP may be due to a substitution-like effect.


Subject(s)
Choice Behavior/drug effects , Conditioning, Classical/drug effects , Illicit Drugs/pharmacology , Pyridines/pharmacology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Social Environment , Substance-Related Disorders/physiopathology , Animals , Brain/drug effects , Brain/physiopathology , Buspirone/pharmacology , Clonidine/pharmacology , Cocaine/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Injections, Intraperitoneal , Injections, Subcutaneous , Male , Nicotine/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5
10.
Eur J Pharmacol ; 607(1-3): 114-20, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19326478

ABSTRACT

We recently reported that the mGlu5 receptor antagonist 2-methyl-6-(phenylethynyl)pyridine (MPEP) reduces intravenous self-administration of ketamine and, to a lesser extent, heroin in rats. We also found that MPEP potentiates conditioned place preference induced by these drugs, suggesting that the reduction of self-administration results from an MPEP-induced potentiation of the rewarding effect of the self-administered drug. The aim of the present study was to examine whether MPEP has intrinsic positive reinforcing and rewarding effects. In experiment 1, rats were trained to self-administer either ketamine [0.5 mg/kg/infusion, 2 h sessions, fixed-ratio (FR) 3] or heroin (0.05 mg/kg/infusion, 1 h sessions, FR 10), followed by a number of substitution sessions with MPEP (1 mg/kg/infusion) or saline. In experiment 2, drug-naïve rats were allowed to acquire intravenous self-administration of MPEP (1 mg/kg/infusion, 2 h sessions, FR 3) or saline. In experiment 3, rats were subjected to a single-trial unbiased conditioned place preference protocol with MPEP (0.3-10 mg/kg i.v., 20 min conditioning). It was found that (1) substitution with MPEP in rats which had learned to self-administer ketamine or heroin resulted in stable self-administration behavior, whereas substitution with saline resulted in a typical extinction profile, (2) drug-naïve rats learned to self-administer MPEP, but not saline, and self-administration remained stable for at least 7 sessions, and (3) MPEP induced dose-dependent place preference with a minimal effective dose of 3 mg/kg. These data clearly demonstrate that MPEP has (weak) positive reinforcing and rewarding effects when administered i.v.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Heroin/administration & dosage , Ketamine/administration & dosage , Pyridines/pharmacology , Animals , Conditioning, Operant/drug effects , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/administration & dosage , Infusions, Intravenous , Male , Pyridines/administration & dosage , Rats , Rats, Long-Evans , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Reinforcement Schedule , Reward , Self Administration , Time Factors
11.
Eur J Pharmacol ; 606(1-3): 94-101, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19210976

ABSTRACT

The mGlu(5) receptor antagonist 2-methyl-6-(phenylethynyl)pyridine (MPEP) has been shown to reduce intravenous self-administration of ketamine and, to a limited extent, heroin in rats. We investigated whether MPEP affects the rewarding effect of ketamine and heroin as assessed in a conditioned place preference (CPP) paradigm. Sprague Dawley rats were subjected to a standard unbiased CPP protocol. Rats were conditioned with either ketamine or heroin (0.316-31.6 and 0.0125-0.5 mg/kg i.p., respectively), in combination with MPEP (10 mg/kg, i.p.) or its vehicle. The effect of MPEP (10 mg/kg) on the duration of extinction and on reinstatement of ketamine- and heroin-induced CPP was also examined. Ketamine and heroin induced CPP with a minimal effective dose (MED) of 10 mg/kg and 0.25 mg/kg, respectively. MPEP (1-31.6 mg/kg) did not induce CPP by itself; however, co-treatment with MPEP resulted in a 10-fold and 5-fold leftward shift in the MED of ketamine and heroin for inducing CPP, respectively. MPEP slowed extinction of ketamine-induced CPP, but not of heroin-induced CPP, and once extinction was achieved, was able to reinstate CPP in both groups. These findings indicate that a moderate dose of MPEP (10 mg/kg i.p.) potentiates, rather than attenuates, the rewarding effect of ketamine and heroin. Moreover, these data suggest that the attenuating effect of MPEP on ketamine and heroin intravenous self-administration is due to an increase, rather than a decrease, of the rewarding/reinforcing effect of these compounds.


Subject(s)
Conditioning, Psychological/drug effects , Extinction, Psychological/drug effects , Heroin/pharmacology , Ketamine/pharmacology , Pyridines/pharmacology , Reward , Animals , Dose-Response Relationship, Drug , Male , Rats , Rats, Sprague-Dawley
12.
Pain ; 136(3): 373-379, 2008 Jun.
Article in English | MEDLINE | ID: mdl-17825490

ABSTRACT

Pain is generally considered to have a sensory and an affective component. Clinical research has suggested that morphine more potently attenuates the affective component as compared to the sensory component. Because preclinical nociception models typically focus on the sensory component of nociception, and do not assess the affective component, it is unclear whether this potency difference of morphine can also be found in preclinical models. We therefore adapted the place conditioning paradigm to investigate negative affect accompanying carrageenan-induced (0.5% intraplantar) inflammatory nociception in rats. We found that carrageenan produced clear conditioned place aversion (CPA). Morphine (0.01-10mg/kg i.p.) dose-dependently reduced carrageenan-induced CPA with a minimal effective dose (MED) of 0.03mg/kg. Since morphine has a rewarding effect by itself, morphine-induced conditioned place preference (CPP) was also investigated. Morphine induced CPP with a MED of 1mg/kg, suggesting that the rewarding effect of morphine was not responsible for reducing carrageenan-induced CPA. We also demonstrated that morphine reduced carrageenan-induced mechanical nociception as assessed in the Randall Selitto paradigm with a MED of 1mg/kg. It is concluded that the CPA model allows for an assessment of the negative affective component of carrageenan-induced nociception. Moreover, morphine was able to reduce the affective component of nociception at doses that did not affect the sensory component of nociception, and this effect was not due to its rewarding properties. The fact that this finding mirrors the clinical situation validates the use of the CPA model for assessing the affective component of nociception.


Subject(s)
Affect/drug effects , Carrageenan , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Morphine/administration & dosage , Touch/drug effects , Analgesics, Opioid/administration & dosage , Animals , Dose-Response Relationship, Drug , Male , Rats , Rats, Sprague-Dawley
13.
Behav Pharmacol ; 18(8): 717-24, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17989509

ABSTRACT

The mGluR5 antagonist 2-methyl-6-(phenylethynyl) pyridine (MPEP) may be beneficial for drug abuse treatment, as it has been found to reduce self-administration of ethanol, nicotine and cocaine in preclinical models. This study investigated whether this finding can be extended to dissociative anaesthetics and opioids. Long Evans rats were trained to intravenously self-administer ketamine (0.5 mg/kg/infusion, 2 h sessions, fixed ratio 3) or heroin (0.05 mg/kg/infusion, 1 h sessions, fixed ratio 10). After reaching stable responding, the effect of MPEP pretreatment (1.25-20 mg/kg, intraperitoneal; t=-30 min) on intravenous self-administration (IVSA) of each compound was investigated. Behavioural specificity of MPEP on IVSA was assessed using a food-reinforcement procedure. IVSA of ketamine was dose-dependently reduced by MPEP pretreatment, with a minimal effective dose of 5 mg/kg and a 75% reduction at the highest dose tested. IVSA of heroin was only modestly reduced by the highest dose of MPEP (20% reduction). Food-reinforced behaviour was not altered by MPEP, either given alone or in combination with ketamine or heroin, indicating that the effect in the IVSA paradigm was behaviourally specific. It is suggested that the differential effect of MPEP on IVSA of ketamine and heroin is related to the particular class of the self-administered drug or its relative reinforcing efficacy.


Subject(s)
Heroin/administration & dosage , Ketamine/administration & dosage , Pyridines/pharmacology , Animals , Conditioning, Operant/drug effects , Injections, Intravenous , Male , Rats , Rats, Long-Evans , Self Administration
14.
Behav Pharmacol ; 17(4): 331-40, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16914951

ABSTRACT

Differences in cocaine self-administration can be attributed to differences in the rewarding value that cocaine has for the individual. An ongoing debate, however, exists whether a high rewarding or a low rewarding value leads to an increase in self-administration. To investigate which of these two alternatives is correct, we investigated the occurrence of cocaine-induced conditioned place preference in apomorphine susceptible and apomorphine unsusceptible rats. We have recently shown that under specific environmental conditions (challenged-not habituated to the environment-as measured by distance travelled) apomorphine susceptible rats consistently self-administer more cocaine than apomorphine unsusceptible rats do. As conditioned place preference allows the assessment of the rewarding value of cocaine, we investigated the expression of cocaine-induced conditioned place preference in apomorphine susceptible and apomorphine unsusceptible rats under the same conditions as the self-administration experiments in order to establish whether the rewarding value of cocaine is greater or smaller in challenged apomorphine susceptible rats than in challenged apomorphine unsusceptible rats. The data clearly showed that challenged apomorphine susceptible rats had a preference for the cocaine-paired compartment with lower doses of cocaine (10 mg/kg) than challenged apomorphine unsusceptible rats. Apomorphine unsusceptible rats expressed conditioned place preference only with the highest dose tested (20 mg/kg). On the basis of these data, we concluded that the rewarding value that cocaine has in challenged apomorphine susceptible rats is greater than that in challenged apomorphine unsusceptible rats. It is suggested that challenged apomorphine susceptible rats self-administer more of a lower dose of cocaine than challenged apomorphine unsusceptible rats do, because the rewarding value of cocaine is greater in challenged apomorphine susceptible rats than in challenged apomorphine unsusceptible rats.


Subject(s)
Apomorphine/pharmacology , Cocaine/pharmacology , Conditioning, Operant/drug effects , Dopamine Agonists/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Animals , Dose-Response Relationship, Drug , Male , Motor Activity/drug effects , Rats , Rats, Wistar , Reward , Species Specificity
15.
Eur J Pharmacol ; 526(1-3): 251-8, 2005 Dec 05.
Article in English | MEDLINE | ID: mdl-16253227

ABSTRACT

Drug dependence and addiction is a chronic mental illness that has far reaching consequences for society in terms of economic loss, health costs and judicial problems. A crucial question in drug addiction, is what factors are involved in its aetiology, and especially what mediates the shit from use to abuse. As in most other mental illnesses, addiction can best be described using the so-called three hit model, which states that a disease results from an interaction between genetic factors, early lie events and late environmental factors. However, the precise nature of these factors still remains to be elucidated. This present review discusses the results from an animal model in which these three different hit are currently being investigated. The apomorphine susceptible (APO-SUS) and apomorphine unsusceptible (APO-UNSUS) rats, originally selected on the basis of their behavioural response to the dopaminergic agonist apomorphine, were recently found to be genetically different in the number of gene copies of a component of the gamma-secretase complex called Aph-1b. Whereas APO-UNSUS rats have three copies of the gene, APO-SUS rats have either 1 or 2 copies. In addition we have shown that these rats show differences in cocaine and alcohol self-administration, and that both early life events and late environmental factors can alter this self-administration behaviour. Thus, the data so far support the hypothesis that the APO-SUS and APO-UNSUS rats offer an interesting animal model for drug dependence in which genes and environment interact. We finally propose a theoretical model which can explain this gene-environment interaction.


Subject(s)
Apomorphine/pharmacology , Environment , Substance-Related Disorders/genetics , Amyloid Precursor Protein Secretases , Animals , Endopeptidases/genetics , Genetic Predisposition to Disease , Rats , Risk Factors , Species Specificity , Substance-Related Disorders/psychology
16.
Neuropharmacology ; 48(5): 685-95, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15814103

ABSTRACT

Research into factors that determine the propensity to self-administer cocaine has shown that stressors can determine the amount of cocaine self-administered as well as the rate of acquisition. However, the interaction between the genetic make-up of the animal and stress is unknown. This study investigated this interaction by using the genetic animal model consisting of apomorphine susceptible (APO-SUS) and unsusceptible (APO-UNSUS) rats. Animals were allowed to self-administer 0.25 mg/kg cocaine under stressful and habituated conditions. This study revealed that the amount of cocaine consumed was highly dependent on the genetic make-up of the animal as well as the amount of stress during self-administration. Under habituated circumstances the APO-UNSUS rats took far more cocaine than the APO-SUS rats. Under stressful circumstances, however, the APO-SUS rats took far more cocaine than the APO-UNSUS rats. This difference in the amount consumed by APO-SUS and APO-UNSUS rats is likely to be due to the specific neurobiological features of their dopaminergic and, possibly, noradrenergic system as well as the reactivity of their HPA-axis. It is suggested that the amount of a drug consumed and, accordingly, its addictive potential and 'drug-vulnerability' are determined by the interaction between the genetic make-up of the animals and stress, and not by either component alone.


Subject(s)
Anesthetics, Local/administration & dosage , Apomorphine/pharmacology , Cocaine/administration & dosage , Dopamine Agonists/pharmacology , Environment , Analysis of Variance , Animals , Behavior, Animal , Conditioning, Operant/drug effects , Genes/physiology , Male , Motor Activity/drug effects , Rats , Rats, Wistar , Self Administration , Stress, Psychological/drug therapy , Stress, Psychological/physiopathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...