Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Addict Biol ; 22(4): 1010-1021, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27001273

ABSTRACT

Addiction is a chronic brain disorder that progressively invades all aspects of personal life. Accordingly, addiction to opiates severely impairs interpersonal relationships, and the resulting social isolation strongly contributes to the severity and chronicity of the disease. Uncovering new therapeutic strategies that address this aspect of addiction is therefore of great clinical relevance. We recently established a mouse model of heroin addiction in which, following chronic heroin exposure, 'abstinent' mice progressively develop a strong and long-lasting social avoidance phenotype. Here, we explored and compared the efficacy of two pharmacological interventions in this mouse model. Because clinical studies indicate some efficacy of antidepressants on emotional dysfunction associated with addiction, we first used a chronic 4-week treatment with the serotonergic antidepressant fluoxetine, as a reference. In addition, considering prodepressant effects recently associated with kappa opioid receptor signaling, we also investigated the kappa opioid receptor antagonist norbinaltorphimine (norBNI). Finally, we assessed whether fluoxetine and norBNI could reverse abstinence-induced social avoidance after it has established. Altogether, our results show that two interspaced norBNI administrations are sufficient both to prevent and to reverse social impairment in heroin abstinent animals. Therefore, kappa opioid receptor antagonism may represent a useful approach to alleviate social dysfunction in addicted individuals.


Subject(s)
Antidepressive Agents, Second-Generation/therapeutic use , Behavior, Animal/drug effects , Grooming/drug effects , Heroin Dependence/physiopathology , Narcotic Antagonists/therapeutic use , Receptors, Opioid, kappa/antagonists & inhibitors , Animals , Disease Models, Animal , Fluoxetine/therapeutic use , Heroin/pharmacology , Male , Mice , Mice, Inbred C57BL , Naltrexone/analogs & derivatives , Naltrexone/therapeutic use , Social Behavior , Time , Treatment Outcome
2.
Nat Genet ; 25(2): 195-200, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10835636

ABSTRACT

The role of the opioid system in controlling pain, reward and addiction is well established, but its role in regulating other emotional responses is poorly documented in pharmacology. The mu-, delta- and kappa- opioid receptors (encoded by Oprm, Oprd1 and Oprk1, respectively) mediate the biological activity of opioids. We have generated Oprd1-deficient mice and compared the behavioural responses of mice lacking Oprd1, Oprm (ref. 6) and Oprk1 (ref. 7) in several models of anxiety and depression. Our data show no detectable phenotype in Oprk1-/- mutants, suggesting that kappa-receptors do not have a role in this aspect of opioid function; opposing phenotypes in Oprm-/- and Oprd1-/- mutants which contrasts with the classical notion of similar activities of mu- and delta-receptors; and consistent anxiogenic- and depressive-like responses in Oprd1-/- mice, indicating that delta-receptor activity contributes to improvement of mood states. We conclude that the Oprd1-encoded receptor, which has been proposed to be a promising target for the clinical management of pain, should also be considered in the treatment of drug addiction and other mood-related disorders.


Subject(s)
Anxiety/metabolism , Depression/metabolism , Gene Deletion , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Animals , Anxiety/genetics , Binding Sites , Darkness , Depression/genetics , Electroshock , Female , Light , Male , Mice , Mice, Knockout , Motor Activity/drug effects , Naloxone/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/metabolism , Naltrexone/pharmacology , Narcotic Antagonists/metabolism , Narcotic Antagonists/pharmacology , Pain Threshold/drug effects , Phenotype , Receptors, Opioid, delta/deficiency , Receptors, Opioid, delta/genetics , Receptors, Opioid, kappa/deficiency , Receptors, Opioid, kappa/genetics , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/deficiency , Receptors, Opioid, mu/genetics , Sex Characteristics , Swimming
3.
Eur J Neurosci ; 27(11): 2973-84, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18588537

ABSTRACT

Addiction develops from the gradual adaptation of the brain to chronic drug exposure, and involves genetic reprogramming of neuronal function. The central extended amygdala (EAc) is a network formed by the central amygdala and the bed nucleus of the stria terminalis. This key site controls drug craving and seeking behaviors, and has not been investigated at the gene regulation level. We used Affymetrix microarrays to analyze transcriptional activity in the murine EAc, with a focus on mu-opioid receptor-associated events because these receptors mediate drug reward and dependence. We identified 132 genes whose expression is regulated by a chronic escalating morphine regimen in the EAc from wild-type but not mu-opioid receptor knockout mice. These modifications are mostly EAc-specific. Gene ontology analysis reveals an overrepresentation of neurogenesis, cell growth and signaling protein categories. A separate quantitative PCR analysis of genes in the last of these groups confirms the dysregulation of both orphan (Gpr88) and known (DrD1A, Adora2A, Cnr1, Grm5, Gpr6) G protein-coupled receptors, scaffolding (PSD95, Homer1) and signaling (Sgk, Cap1) proteins, and neuropeptides (CCK, galanin). These transcriptional modifications do not occur following a single morphine injection, and hence result from long-term adaptation to excessive mu receptor activation. Proteins encoded by these genes are classically associated with spine modules function in other brain areas, and therefore our data suggest a remodeling of EAc circuits at sites where glutamatergic and monoaminergic afferences interact. Together, mu receptor-dependent genes identified in this study potentially contribute to drug-induced neural plasticity, and provide a unique molecular repertoire towards understanding drug craving and relapse.


Subject(s)
Amygdala/drug effects , Analgesics, Opioid/pharmacology , Gene Expression Regulation/drug effects , Neuronal Plasticity/drug effects , Receptors, Opioid, mu/agonists , Transcriptional Activation/drug effects , Amygdala/metabolism , Animals , Gene Expression Regulation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphine/pharmacology , Neural Pathways/drug effects , Neural Pathways/metabolism , Neuronal Plasticity/genetics , Oligonucleotide Array Sequence Analysis , Opioid-Related Disorders/genetics , Opioid-Related Disorders/metabolism , Opioid-Related Disorders/physiopathology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Opioid, mu/metabolism , Septal Nuclei/drug effects , Septal Nuclei/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptional Activation/genetics
4.
Neuroscience ; 156(4): 950-65, 2008 Oct 28.
Article in English | MEDLINE | ID: mdl-18786617

ABSTRACT

The central extended amygdala (EAc) is an ensemble of highly interconnected limbic structures of the anterior brain, and forms a cellular continuum including the bed nucleus of the stria terminalis (BNST), the central nucleus of the amygdala (CeA) and the nucleus accumbens shell (AcbSh). This neural network is a key site for interactions between brain reward and stress systems, and has been implicated in several aspects of drug abuse. In order to increase our understanding of EAc function at the molecular level, we undertook a genome-wide screen (Affymetrix) to identify genes whose expression is enriched in the mouse EAc. We focused on the less-well known BNST-CeA areas of the EAc, and identified 121 genes that exhibit more than twofold higher expression level in the EAc compared with whole brain. Among these, 43 genes have never been described to be expressed in the EAc. We mapped these genes throughout the brain, using non-radioactive in situ hybridization, and identified eight genes with a unique and distinct rostro-caudal expression pattern along AcbSh, BNST and CeA. Q-PCR analysis performed in brain and peripheral organ tissues indicated that, with the exception of one (Spata13), all these genes are predominantly expressed in brain. These genes encode signaling proteins (Adora2, GPR88, Arpp21 and Rem2), a transcription factor (Limh6) or proteins of unknown function (Rik130, Spata13 and Wfs1). The identification of genes with enriched expression expands our knowledge of EAc at a molecular level, and provides useful information to toward genetic manipulations within the EAc.


Subject(s)
Amygdala/physiology , Gene Expression Profiling/methods , Gene Expression/physiology , Nerve Tissue Proteins/genetics , Amygdala/anatomy & histology , Animals , Brain Mapping , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Oligonucleotide Array Sequence Analysis
5.
Ann N Y Acad Sci ; 1129: 175-84, 2008.
Article in English | MEDLINE | ID: mdl-18591478

ABSTRACT

The lateral hypothalamus (LH) is a brain structure that controls hedonic properties of both natural rewards and drugs of abuse. Mu opioid receptors are known to mediate drug reward, but whether overstimulation of these receptors impacts on LH function has not been studied. Here we have used a genome-wide microarray approach to identify LH responses to chronic mu opioid receptor activation at the transcriptional level. We have subjected wild-type and mu opioid receptor knockout mice to an escalating morphine regimen, which produces severe physical dependence in wild-type but not mutant animals. We have analyzed gene profiles in LH samples using the 430A.2 Affymetrix array and identified a set of 25 genes whose expression is altered by morphine in wild-type mice only. The regulation was confirmed for a subset of these genes using real-time quantitative PCR on samples from independent treatments. Altered expression of aquaporin 4, apolipoprotein D, and prostaglandin synthase is indicative of modified LH physiology. The regulation of two signaling genes (the serum glucocorticoid kinase and the regulator of G protein signaling 4) suggests that neurotransmission is altered in LH circuitry. Finally, the downregulation of apelin may indicate a potential role for this neuropeptide in opioid signaling and hedonic homeostasis. Altogether, our study shows that chronic mu opioid receptor stimulation induces gene expression plasticity in the LH and provides a unique collection of mu opioid receptor-dependent genes that potentially contribute to alter reward processes in addictive diseases.


Subject(s)
Gene Expression Regulation , Hypothalamus/metabolism , Receptors, Opioid, mu/genetics , Animals , Gene Expression Regulation/drug effects , Hypothalamus/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphine/pharmacology , Polymerase Chain Reaction , Receptors, Opioid, mu/metabolism , Reproducibility of Results
6.
Neuroscience ; 146(3): 1182-92, 2007 May 25.
Article in English | MEDLINE | ID: mdl-17395390

ABSTRACT

Large-scale transcriptome analysis in the brain is a powerful approach to identify novel genes of potential interest toward understanding cerebral organization and function. We utilized the microarray technology to measure expression levels of about 24,000 genes and expressed sequence tags in mouse hippocampus, frontal cortex and striatum. Using expression profile obtained from whole brain as a reference, we categorized the genes into groups of genes either enriched in, or restricted to, one of the three areas of interest. We found enriched genes for each target area. Further, we identified 14 genes in the category of genes restricted to the striatum, among which were the orphan G protein-coupled receptor GPR88 and retinoic acid receptor-beta. These two genes were already reported to be selectively expressed in the striatum, thus validating our experimental approach. We selected 6 striatal-restricted genes, as well as 10 striatal-enriched candidates, that were previously undescribed. We analyzed their expression by in situ hybridization analysis in the brain, and quantitative RT-PCR in both brain and peripheral organs. Two of these unknown genes displayed a notable expression pattern. The striatal-restricted gene H3076B11 shows uniform expression throughout and uniquely in the striatum, representing a genuine striatal marker. The striatal-enriched gene 4833421E05Rik is preferentially expressed in the rostral striatum, and is also abundant in kidney, liver and lung. These two genes may contribute to some of the many striatal-controlled behaviors, including initiation of movement, habit formation, or reward and motivation.


Subject(s)
Brain Chemistry/genetics , Gene Expression Profiling , Neostriatum/metabolism , Nerve Tissue Proteins/biosynthesis , Animals , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Data Interpretation, Statistical , In Situ Hybridization , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Oligonucleotide Array Sequence Analysis , RNA/biosynthesis , RNA/genetics , Receptors, Retinoic Acid/biosynthesis , Receptors, Retinoic Acid/genetics , Reverse Transcriptase Polymerase Chain Reaction
7.
Genes Brain Behav ; 16(3): 313-327, 2017 03.
Article in English | MEDLINE | ID: mdl-27762100

ABSTRACT

DNA methylation is a major epigenetic process which regulates the accessibility of genes to the transcriptional machinery. In the present study, we investigated whether modifying the global DNA methylation pattern in the brain would alter cocaine intake by rats, using the cocaine self-administration test. The data indicate that treatment of rats with the DNA methyltransferase inhibitors 5-aza-2'-deoxycytidine (dAZA) and zebularine enhanced the reinforcing properties of cocaine. To obtain some insights about the underlying neurobiological mechanisms, a genome-wide methylation analysis was undertaken in the prefrontal cortex of rats self-administering cocaine and treated with or without dAZA. The study identified nearly 189 000 differentially methylated regions (DMRs), about half of them were located inside gene bodies, while only 9% of DMRs were found in the promoter regions of genes. About 99% of methylation changes occurred outside CpG islands. Gene expression studies confirmed the inverse correlation usually observed between increased methylation and transcriptional activation when methylation occurs in the gene promoter. This inverse correlation was not observed when methylation took place inside gene bodies. Using the literature-based Ingenuity Pathway Analysis, we explored how the differentially methylated genes were related. The analysis showed that increase in cocaine intake by rats in response to DNA methyltransferase inhibitors underlies plasticity mechanisms which mainly concern axonal growth and synaptogenesis as well as spine remodeling. Together with the Akt/PI3K pathway, the Rho-GTPase family was found to be involved in the plasticity underlying the effect of dAZA on the observed behavioral changes.


Subject(s)
Cocaine-Related Disorders/genetics , Cocaine/administration & dosage , DNA Methylation , DNA Modification Methylases/antagonists & inhibitors , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Brain/metabolism , Cocaine-Related Disorders/enzymology , Cocaine-Related Disorders/metabolism , CpG Islands , Cytidine/analogs & derivatives , Cytidine/pharmacology , DNA/genetics , DNA Modification Methylases/metabolism , Decitabine , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic , Genome , Male , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic , Rats , Rats, Wistar , Reinforcement, Psychology , Self Administration
8.
Neuromuscul Disord ; 3(5-6): 515-8, 1993.
Article in English | MEDLINE | ID: mdl-8186703

ABSTRACT

In the present study, monoclonal antibodies raised against the C-terminal domain of dystrophin were used to identify and characterize Dp71 from the central nervous system. It was observed that the expression of Dp71 gradually increases from the embryo stage until the adult. Subcellular distribution analysis indicates that Dp71 is mainly recovered in synaptic plasma membranes, microsomes and at a lesser extent in synaptic vesicles and mitochondria. The amino acid composition and N-terminal sequence of bovine brain Dp71 were determined. Moreover, we found that this protein is glycosylated.


Subject(s)
Brain Chemistry , Dystrophin/analogs & derivatives , Dystrophin/analysis , Animals , Antibodies, Monoclonal , Blotting, Western , Cattle , Electrophoresis, Polyacrylamide Gel , Microsomes/chemistry , Mitochondria/chemistry , Molecular Weight , Rats , Subcellular Fractions/chemistry , Synaptic Membranes/chemistry , Synaptic Vesicles/chemistry
9.
J Neuroimmunol ; 147(1-2): 121-2, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14741441

ABSTRACT

The implication of opioid receptors in immune response has been studied using mu-, delta- and kappa-opioid receptor knockout mice. The mutant animals were compared to their wild-type (WT) counterparts for antibody (Ab) response to the prototype Ag keyhole limpet hemocyanin (KLH). Kappa-receptor deficient mice displayed higher Ab titers for either total Ig, IgM, IgG1 or IgG2a isotypes, whereas mu and delta animals behaved as wild-type mice. Therefore, endogenous kappa-receptor activation would tonically inhibit Ab response. Opioid receptor deficient mice were also used to investigate the immunosuppressive action of naltrindole, a delta-opioid receptor antagonist, shown earlier to inhibit graft rejection and the allogeneic mixed lymphocyte reaction (MLR) in vitro. Naltrindole and two related compounds inhibited MLR performed with lymphocytes from wild-type and delta-opioid receptor knockout mice. These compounds also suppressed MLR assayed with cells from triple mu/delta/kappa-opioid receptor mutants. We therefore demonstrate that naltrindole immunosuppressive activity is not mediated by any of the three mu-, delta- or kappa-opioid receptors, but by a target which remains to be discovered.


Subject(s)
Antibodies/immunology , Lymphocytes/immunology , Mice, Knockout/immunology , Naltrexone/analogs & derivatives , Receptors, Opioid/metabolism , Analysis of Variance , Animals , Antibody Formation , Graft Rejection , Immunosuppression Therapy , In Vitro Techniques , Lymphocyte Culture Test, Mixed , Mice , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Receptors, Opioid/genetics , Receptors, Opioid/immunology
10.
Neuroscience ; 106(3): 469-80, 2001.
Article in English | MEDLINE | ID: mdl-11591451

ABSTRACT

The opioid receptor-like 1 (ORL1) receptor shares a high degree of sequence homology with the classical mu-, delta- and kappa-opioid receptors and a functional mutual opposition between these receptors has been suggested. To further address this possible interaction we have used mu-, delta- and kappa-opioid receptor knockout mice to determine autoradiographically if there are any changes in the number or distribution of the ORL1 receptor, labelled with [(3)H]nociceptin, in the brains of mice deficient in each of the opioid receptors. An up-regulation of ORL1 expression was observed across all brain regions in delta-knockouts with cortical regions typically showing a 15-30% increase in binding that was most marked in heterozygous mice. In contrast, ORL1 receptor expression was down-regulated in virtually all brain structures in heterozygous kappa-knockouts although the magnitude of this change was not as great as for the delta-knockouts. No significant alterations in ORL1 receptor expression were observed across brain regions in mu-receptor knockout mice and there were no qualitative differences in ORL1 receptor expression in any groups. These data suggest there are interactions between the ORL1 system and the classical opioid receptors and that the interactions are receptor-specific. The greater differences observed in heterozygous mice suggest that these interactions might be most relevant when there is only partial loss of receptor function.


Subject(s)
Brain/metabolism , Pain/metabolism , Receptors, Opioid, delta/deficiency , Receptors, Opioid, kappa/deficiency , Receptors, Opioid, mu/deficiency , Receptors, Opioid/metabolism , Animals , Brain/cytology , Brain/drug effects , Brain Mapping , Down-Regulation/genetics , Female , Male , Mice , Mice, Knockout , Opioid Peptides/antagonists & inhibitors , Opioid Peptides/metabolism , Opioid Peptides/pharmacokinetics , Pain/physiopathology , Radioligand Assay , Receptors, Opioid/drug effects , Receptors, Opioid, delta/genetics , Receptors, Opioid, kappa/genetics , Receptors, Opioid, mu/genetics , Reference Values , Tritium/pharmacokinetics , Up-Regulation/genetics , Nociceptin Receptor , Nociceptin
11.
Eur J Pharmacol ; 414(2-3): 189-95, 2001 Mar 02.
Article in English | MEDLINE | ID: mdl-11239918

ABSTRACT

Despite ample pharmacological evidence for the existence of multiple mu-, delta- and kappa-opioid receptor subtypes, only three genes encoding mu-(MOR), delta-(DOR) and kappa-(KOR) opioid receptor have been cloned. The KOR gene encodes kappa(1)-sites, which specifically bind arylacetamide compounds, and the possible existence of kappa-opioid receptor subtypes derived from another kappa-opioid-receptor gene, yet to be characterized, remains a very contentious issue. kappa(2)-Opioid receptors are described as binding sites typically labelled by the non-selective benzomorphan ligand [3H]bremazocine in the presence of mu-, delta- and kappa(1)-opioid receptor blocking ligands. To investigate the genetic origin of kappa(2)-opioid receptors, we have carried out homogenate binding experiments with [3H]bremazocine in brains of single MOR-, DOR-, KOR- and double MOR/DOR-deficient mice. Scatchard analysis showed that 68+/-12% of the binding sites arise from the MOR gene, 27+/-1% from the DOR gene and 14.5+/-0.2% from the KOR gene, indicating that the three known genes account for total [3H]bremazocine binding. Experiments in the presence of mu-, delta- and kappa(1)-opioid receptor suppressor ligands further showed that non-kappa(1)-opioid receptor labelling can be accounted for by binding to both the mu- and delta-opioid receptors. Finally, [3H]bremazocine binding experiments performed on brain membranes from the triple MOR/DOR/KOR-deficient mice revealed a complete absence of binding sites, confirming definitively that no additional gene is required to explain the total population of [3H]bremazocine binding sites. Altogether the data show that the putative kappa(2)-opioid receptors are in fact a mixed population of KOR, DOR and predominantly MOR gene products.


Subject(s)
Analgesics/metabolism , Benzomorphans/metabolism , Brain/metabolism , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Animals , Mice , Mice, Knockout , Receptors, Opioid, delta/deficiency , Receptors, Opioid, kappa/deficiency , Receptors, Opioid, mu/deficiency
12.
Brain Res Bull ; 54(6): 655-60, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11403992

ABSTRACT

Orphanin FQ/nociceptin (OFQ/N) has been shown to modulate nociception, responses to stress and anxiety. We investigated OFQ/N function in human immune cells. We find that monocytic U937, T lymphocytic CEM, and MOLT-4 cell lines express OFQ/N binding sites at levels comparable to that of human SH-SY5Y neuroblastoma cells. We show that OFQ/N receptors are functionally coupled to G proteins in these cells. Finally OFQ/N decreases proliferation of phytohemagglutinin-stimulated peripheral blood mononuclear cells in vitro at doses ranging from 10(-13) to 10(-8) M. Thus, our data suggest that OFQ/N and OFQ/N receptor may act as an immunomodulatory system.


Subject(s)
Cell Line/metabolism , Immune System/metabolism , Leukocytes, Mononuclear/metabolism , Opioid Peptides/metabolism , Receptors, Opioid/metabolism , Adult , Animals , Binding Sites/drug effects , Binding Sites/physiology , CHO Cells/drug effects , CHO Cells/metabolism , Cell Division/drug effects , Cell Division/physiology , Cell Line/cytology , Cell Line/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cricetinae , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , Humans , Immune System/cytology , Immune System/drug effects , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Neuroblastoma , Opioid Peptides/immunology , Opioid Peptides/pharmacology , Phytohemagglutinins/pharmacology , Radioligand Assay , Receptors, Opioid/drug effects , Receptors, Opioid/immunology , Sulfur Radioisotopes/pharmacology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Nociceptin Receptor , Nociceptin
13.
J Pharmacol Exp Ther ; 313(1): 410-21, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15590769

ABSTRACT

The delta opioid receptor modulates nociceptive and emotional behaviors. This receptor has been shown to exhibit measurable spontaneous activity. Progress in understanding the biological relevance of this activity has been slow, partly due to limited characterization of compounds with intrinsic negative activity. Here, we have used constitutively active mutant (CAM) delta receptors in two different functional assays, guanosine 5'-O-(3-thio)triphosphate binding and a reporter gene assay, to test potential inverse agonism of 15 delta opioid compounds, originally described as antagonists. These include the classical antagonists naloxone, naltrindole, 7-benzylidene-naltrexone, and naltriben, a new set of naltrindole derivatives, H-Tyr-Tic-Phe-Phe-OH (TIPP) and H-Tyr-TicPsi[CH2N]Cha-Phe-OH [TICP(Psi)], as well as three 2',6'-dimethyltyrosine-1,2,3,4-tetrahydroquinoline-3-carboxylate (Dmt-Tic) peptides. A reference agonist, SNC 80 [(+)-4-[(alphaR)-alpha-((2S,5R)-4-Allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide], and inverse agonist, ICI 174864 (N,N-diallyl-Tyr-Aib-Aib-Phe-Leu), were also included. In a screen using wild-type and CAM M262T delta receptors, naltrindole (NTI) and close derivatives were mostly inactive, and TIPP behaved as an agonist, whereas Dmt-Tic-OH and N,N(CH3)2-Dmt-Tic-NH2 showed inverse agonism. The two latter compounds showed negative activity across 27 CAM receptors, suggesting that this activity was independent from the activation mechanism. These two compounds also exhibited nanomolar potencies in dose-response experiments performed on wild-type, M262T, Y308H, and C328R CAM receptors. TICP(Psi) exhibited strong inverse agonism at the Y308H receptor. We conclude that the stable N,N(CH3)2-Dmt-Tic-NH2 compound represents a useful tool to explore the spontaneous activity of delta receptors, and NTI and novel derivatives behave as neutral antagonists.


Subject(s)
Dipeptides/pharmacology , Enkephalin, Leucine/analogs & derivatives , Naltrexone/analogs & derivatives , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/antagonists & inhibitors , Alkaline Phosphatase/metabolism , Benzamides/pharmacology , Cell Line , Dose-Response Relationship, Drug , Enkephalin, Leucine/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Humans , Ligands , Mutation , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Piperazines/pharmacology , Receptors, Opioid, delta/genetics , Structure-Activity Relationship
14.
Biochem Biophys Res Commun ; 149(2): 776-83, 1987 Dec 16.
Article in English | MEDLINE | ID: mdl-3426600

ABSTRACT

Axonal transport of mitochondria is a microtubule-associated movement. Microtubule-mitochondria interactions were studied in vitro using organelles isolated from rat brain. Thanks to the ligand blotting method we were able to show two mitochondrial membrane proteins with apparent molecular masses of 30 kDa and 60 kDa that bind microtubule-associated proteins. The binding of the 30 kDa protein has an apparent Kd of 8 x 10(-8) M. Digitonin fractionation of mitochondria reveals a bimodal localization of the 30 kDa and the 60 kDa proteins within the outer membrane. The data suggest that these polypeptides could participate to the interactions observed in situ between microtubules and mitochondria.


Subject(s)
Brain/metabolism , Carrier Proteins/analysis , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Animals , Carrier Proteins/metabolism , Microtubules/metabolism , Phosphorus Radioisotopes , Rats , Rats, Inbred Strains
15.
Neurobiology (Bp) ; 4(4): 417-29, 1996.
Article in English | MEDLINE | ID: mdl-9200133

ABSTRACT

Microtubule-associated tau proteins are likely candidates to interfere with axonal transport of membranous organelles. We studied that tau proteins influenced the enzyme activity of kinesin, known to drive anterograd transport along microtubules. An in vitro reconstituted system was applied; microtubules were assembled from purified tubulin with or without tau proteins. Both types of reconstituted microtubules stimulated MgATPase activity of purified kinesin in a concentration dependent, saturable manner. The extent of maximal stimulation by tau-coated microtubules was lower than that of microtubules without tau proteins. Analysis of kinetic data, on the other hand, suggests that tau-coated microtubules apparently bind kinesin with higher affinity then microtubules not associated with tau proteins. Tau proteins, similarly to tubulin dimers, seem to bind to the heavy chain of kinesin. These data support the notion that tau proteins could act as regulators of kinesin-driven processes.


Subject(s)
Kinesins/metabolism , Microtubules/metabolism , tau Proteins/metabolism , Animals , Brain Chemistry , Ca(2+) Mg(2+)-ATPase/isolation & purification , Ca(2+) Mg(2+)-ATPase/metabolism , Cattle , Enzyme Activation/drug effects , Enzyme Activation/physiology , Immunoblotting , Kinesins/isolation & purification , Kinesins/pharmacology , Microtubule-Associated Proteins/metabolism , Microtubules/enzymology , Protein Binding , tau Proteins/isolation & purification
16.
Anal Biochem ; 150(2): 332-6, 1985 Nov 01.
Article in English | MEDLINE | ID: mdl-4091261

ABSTRACT

Mitochondrial aspartate aminotransferase and malate dehydrogenase were purified to homogeneity from rat liver by the use of aspartate-coupled Sepharose, ion exchange, and Blue Sepharose chromatography. This procedure permits rapid preparation of these enzymes. The pI of each enzyme was determined and anomalous electrophoretic properties of aspartate aminotransferase were described.


Subject(s)
Aspartate Aminotransferases/isolation & purification , Malate Dehydrogenase/isolation & purification , Mitochondria, Liver/enzymology , Animals , Chromatography, Affinity , Electrophoresis, Polyacrylamide Gel , Isoelectric Focusing , Rats
17.
Cell Motil Cytoskeleton ; 14(3): 372-81, 1989.
Article in English | MEDLINE | ID: mdl-2510942

ABSTRACT

Two major brain microtubule-associated proteins (MAPs), MAP2 and tau, were found to be able to bind to purified rat brain mitochondria. The apparent dissociation constants of the binding of thermostable 32P-labeled MAP2 and tau are 0.9 +/- 0.04 x 10(-7) and 3.8 +/- 0.7 x 10(-7) M, respectively. 32P-labeled MAP2 and tau bound to the mitochondria can be displaced by phosphorylated, nonradioactive MAP2. The binding parameters of MAP2 prepared without heat treatment and those of the thermostable MAP2 were of the same order of magnitude. Microtubule-binding and projection domains of MAP2 were obtained by chymotryptic digestion of rat brain microtubules (Vallee, Proc. Natl. Acad. Sci. USA, 77:3206-3210, 1980). Displacement studies with these two domains show that MAP2 bound to mitochondria can be displaced by the microtubule-binding domain, whereas the projection domain does not displace MAP2. The two domains of MAP2 bind to the mitochondria with similar affinity constants; however, the Bmax for the projection domain was 10 times and 35 times lower than the Bmax of the binding of the intact MAP2 and the microtubule-binding domain, respectively. Chymotryptic digestion of MAP2 bound to the mitochondria yielded peptide fragments with molecular masses similar to those obtained by the digestion of MAP2 bound to the microtubules. The fragments corresponding to the projection domain were released into the extramitochondrial supernatant, whereas the fragments originating from the microtubule-binding domain remained bound to the mitochondria. These results suggest that MAP2 binds to mitochondria preferentially via its microtubule-binding domain.


Subject(s)
Brain/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Mitochondria/metabolism , Nerve Tissue Proteins/metabolism , Animals , Binding Sites , Chymotrypsin , Phosphorylation , Protein Binding , Rats , Rats, Inbred Strains , tau Proteins
18.
Cell Motil Cytoskeleton ; 24(4): 245-55, 1993.
Article in English | MEDLINE | ID: mdl-8097434

ABSTRACT

To explore the behaviour of microtubule-associated proteins, MAP2 and TAU in the interactions of mitochondria with microtubules, an homologous acellular system has been reconstituted with organelles isolated from rat brain. We have established a quantitative in vitro binding assay based on the cosedimentation of 125I-labeled microtubules with mitochondria. We found that binding of microtubules to mitochondria was concentration dependent and saturable. Binding was insensitive to ATP. A comparison of taxol-stabilized microtubules prepared from MAP-free tubulin or tubulin coated with TAU or MAP2 showed that the microtubule-associated proteins diminished, or reduced to background levels, the formation of complexes with mitochondria. In contrast, the amount of MAP-free taxol microtubules that cosedimented with mitochondria increased two- and six-fold when mitochondria were coated with MAP2 or TAU. These studies suggest that the two major brain MAPs could have a crosslinking or a spacing role, depending on their organelle localization.


Subject(s)
Brain/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Mitochondria/metabolism , Tubulin/metabolism , tau Proteins/metabolism , Adenosine Triphosphate/metabolism , Animals , Brain Chemistry , Cell-Free System , Microtubules/drug effects , Paclitaxel/pharmacology , Rats , Rats, Wistar , Tubulin/isolation & purification
19.
Arch Biochem Biophys ; 278(1): 142-7, 1990 Apr.
Article in English | MEDLINE | ID: mdl-2321954

ABSTRACT

Recently, analysis of protein distribution in rat brain mitochondria suggested the existence of distinct cholesterol domains in the outer membrane (Dorbani et al., 1987, Arch. Biochem. Biophys. 252, 188-196) while such domains were not detected in rat liver mitochondria (Jancsik et al., 1988, Arch. Biochem. Biophys. 264, 295-301). We studied cholesterol distribution in both types of mitochondria by analyzing the kinetics of filipin-cholesterol complex formation, using the stopped-flow technique. In liver mitochondria, the kinetics are characterized by a biphasic curve which presumably corresponds to the two membranes. This was confirmed by the finding that pretreatment with digitonin abolished one of the kinetic components. Sonication of the mitochondria increased the rate of the filipin-cholesterol complex formation and also abolished one of the two components. In the case of brain mitochondria, several distinct cholesterol domains could be revealed: one of them was cholesterol-free and it was directly accessible to filipin. Two other domains were revealed by differences found in the rate of the cholesterol-filipin complex formation. It is noteworthy that only a part of the cholesterol is accessible to filipin. Sonication of mitochondria decreased the proportion of cholesterol molecules accessible to filipin. This suggests specific interactions of cholesterol with other mitochondrial components, which occur only in brain mitochondria.


Subject(s)
Antifungal Agents , Brain Chemistry , Cholesterol/analysis , Filipin , Intracellular Membranes/analysis , Membrane Lipids/analysis , Mitochondria, Liver/analysis , Mitochondria/analysis , Submitochondrial Particles/analysis , Animals , Kinetics , Liposomes , Rats
20.
J Biol Chem ; 274(26): 18574-81, 1999 Jun 25.
Article in English | MEDLINE | ID: mdl-10373467

ABSTRACT

We have investigated whether transmembrane amino acid residues Asp128 (domain III), Tyr129 (domain III) [corrected], and Tyr308 (domain VII) in the mouse delta opioid receptor play a role in receptor activation. To do so, we have used a [35S]GTPgammaS (where GTPgammaS is guanosine 5'-3-O-(thio)triphosphate) binding assay to quantify the activation of recombinant receptors transiently expressed in COS cells and compared functional responses of D128N, D128A, Y129F, Y129A, and Y308F point-mutated receptors to that of the wild-type receptor. In the absence of ligand, [35S]GTPgammaS binding was increased for every mutant receptor under study (1.6-2.6-fold), suggesting that all mutations are able to enhance constitutive activity at the receptor. In support of this finding, the inverse agonist N,N-diallyl-Tyr-Aib-Aib-Phe-Leu (where Aib represents alpha-aminobutyric acid) efficiently reduced basal [35S]GTPgammaS binding in the mutated receptor preparations. The potent agonist BW373U86 stimulated [35S]GTPgammaS binding above basal levels with similar (D128N, Y129F, and Y129A) or markedly increased (Y308F) efficacy compared with wild-type receptor. BW373U86 potency was maintained or increased. In conclusion, our results demonstrate that the mutations under study increase functional activity of the receptor. Three-dimensional modeling suggests that Asp128 (III) and Tyr308 (VII) interact with each other and that Tyr129 (III) undergoes H bonding with His278 (VI). Thus, Asp128, Tyr129, and Tyr308 may be involved in a network of interhelical bonds, which contributes to maintain the delta receptor under an inactive conformation. We suggest that the mutations weaken helix-helix interactions and generate a receptor state that favors the active conformation and/or interacts with heterotrimeric G proteins more effectively.


Subject(s)
Receptors, Opioid, delta/metabolism , Amino Acid Substitution , Animals , COS Cells , Cell Membrane/metabolism , Enkephalin, Leucine-2-Alanine/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Ligands , Mice , Models, Molecular , Mutagenesis, Site-Directed , Naltrexone/analogs & derivatives , Naltrexone/metabolism , Narcotic Antagonists/metabolism , Protein Structure, Secondary , Receptors, Opioid, delta/genetics , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL