Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Mol Psychiatry ; 27(12): 4905-4917, 2022 12.
Article in English | MEDLINE | ID: mdl-36127430

ABSTRACT

Freezing is a conserved defensive behaviour that constitutes a major stress-coping mechanism. Decades of research have demonstrated a role of the amygdala, periaqueductal grey and hypothalamus as core actuators of the control of fear responses, including freezing. However, the role that other modulatory sites provide to this hardwired scaffold is not known. Here, we show that freezing elicited by exposure to electrical foot shocks activates laterodorsal tegmentum (LDTg) GABAergic neurons projecting to the VTA, without altering the excitability of cholinergic and glutamatergic LDTg neurons. Selective chemogenetic silencing of this inhibitory projection, but not other LDTg neuronal subtypes, dampens freezing responses but does not prevent the formation of conditioned fear memories. Conversely, optogenetic-activation of LDTg GABA terminals within the VTA drives freezing responses and elicits bradycardia, a common hallmark of freezing. Notably, this aversive information is subsequently conveyed from the VTA to the amygdala via a discrete GABAergic pathway. Hence, we unveiled a circuit mechanism linking LDTg-VTA-amygdala regions, which holds potential translational relevance for pathological freezing states such as post-traumatic stress disorders, panic attacks and social phobias.


Subject(s)
Amygdala , Periaqueductal Gray , Freezing , Periaqueductal Gray/metabolism , Amygdala/physiology , GABAergic Neurons
2.
Neurobiol Dis ; 118: 117-128, 2018 10.
Article in English | MEDLINE | ID: mdl-30003950

ABSTRACT

Amyloid-ß is a peptide released by synapses in physiological conditions and its pathological accumulation in brain structures necessary for memory processing represents a key toxic hallmark underlying Alzheimer's disease. The oligomeric form of Amyloid-ß (Aßο) is now believed to represent the main Amyloid-ß species affecting synapse function. Yet, the exact molecular mechanism by which Aßο modifies synapse function remains to be fully elucidated. There is accumulating evidence that glucocorticoid receptors (GRs) might participate in Aßο generation and activity in the brain. Here, we provide evidence for an acute functional cross-talk between Aß and GRs at hippocampal excitatory synapses. Using live imaging and biochemical analysis of post-synaptic densities (PSD) in cultured hippocampal neurons, we show that synthetic Aßo (100 nM) increases GR levels in spines and PSD. Also, in these cultured neurons, blocking GRs with two different GR antagonists prevents Aßo-mediated PSD95 increase within the PSD. By analyzing long-term potentiation (LTP) and long-term depression (LTD) in ex vivo hippocampal slices after pharmacologically blocking GR, we also show that GR signaling is necessary for Aßo-mediated LTP impairment, but not Aßo-mediated LTD induction. The necessity of neuronal GRs for Aßo-mediated LTP was confirmed by genetically removing GRs in vivo from CA1 neurons using conditional GR mutant mice. These results indicate a tight functional interplay between GR and Aß activities at excitatory synapses.


Subject(s)
Amyloid beta-Peptides/physiology , Excitatory Postsynaptic Potentials/physiology , Hippocampus/physiology , Peptide Fragments/physiology , Receptor Cross-Talk/physiology , Receptors, Glucocorticoid/physiology , Synapses/physiology , Animals , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
3.
Biol Psychiatry ; 95(8): 774-784, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-37804900

ABSTRACT

BACKGROUND: Social status in humans, generally reflected by socioeconomic status, has been associated, when constrained, with heightened vulnerability to pathologies including psychiatric diseases. Social hierarchy in mice translates into individual and interdependent behavioral strategies of animals within a group. The rules leading to the emergence of a social organization are elusive, and detangling the contribution of social status from other factors, whether environmental or genetic, to normal and pathological behaviors remains challenging. METHODS: We investigated the mechanisms shaping the emergence of a social hierarchy in isogenic C57BL/6 mice raised in groups of 4 using conditional mutant mouse models and chemogenetic manipulation of dopamine midbrain neuronal activity. We further studied the evolution of behavioral traits and the vulnerability to psychopathological-like phenotypes according to the social status of the animals. RESULTS: Higher sociability predetermined higher social hierarchy in the colony. Upon hierarchy establishment, higher-ranked mice showed increased anxiety and better cognitive abilities in a working memory task. Strikingly, the higher-ranked mice displayed a reduced activity of dopaminergic neurons within the ventral tegmental area, paired with a decreased behavioral response to cocaine and a decreased vulnerability to depressive-like behaviors following repeated social defeats. The pharmacogenetic inhibition of this neuronal population and the genetic inactivation of glucocorticoid receptor signaling in dopamine-sensing brain areas that resulted in decreased dopaminergic activity promoted accession to higher social ranks. CONCLUSIONS: Dopamine activity and its modulation by the stress response shapes social organization in mice, potentially linking interindividual and social status differences in vulnerability to psychopathologies.


Subject(s)
Dopaminergic Neurons , Mental Disorders , Humans , Mice , Animals , Dopamine , Hierarchy, Social , Mice, Inbred C57BL , Ventral Tegmental Area
4.
Neuropsychopharmacology ; 47(9): 1587-1596, 2022 08.
Article in English | MEDLINE | ID: mdl-35459925

ABSTRACT

There are about twice as many women as men who experience depression during their lifetime. Although life circumstances and especially exposure to stressful situations constitute a major risk factor to develop depression, the underlying mechanisms have yet to be unraveled. We employed the chronic social defeat procedure to elicit depressive-like symptoms in females and ketamine to validate the model. We performed ex-vivo patch clamp recordings to assess cellular adaptations and used pharmacological agents to dissect these deregulations. Chronic social defeat exposure triggers a hyperactivity of VTA putative dopamine (DA) neurons in females susceptible to stress but not resilient ones. This hyperactivity was fully reversed by a single administration of ketamine. In virally-identified brain circuits of both susceptible and resilient females, we found a hypercholinergic tone to the VTA arising from the laterodorsal tegmentum. Application of puffs of nicotine revealed a decreased sensitivity of DA neurons in resilient mice when compared to naive or susceptible ones. The in vivo acute administration of the positive allosteric modulator for α7 nicotinic acetylcholine receptors (nAChRs) not only increased susceptibility to stress by enhancing activity of VTA DA neurons, but also triggered a switch in phenotype from resilient to susceptible. Our data unravel dysregulations of VTA DA neurons activity exclusively in females exhibiting depressive-like symptoms and identify VTA nAChRs as key molecular substrates that exacerbate susceptibility to stress.


Subject(s)
Ketamine , Receptors, Nicotinic , Animals , Dopamine , Dopaminergic Neurons/physiology , Female , Humans , Mice , Receptors, Nicotinic/genetics , Ventral Tegmental Area/metabolism
5.
Nat Commun ; 13(1): 1807, 2022 04 04.
Article in English | MEDLINE | ID: mdl-35379786

ABSTRACT

Enduring behavioral changes upon stress exposure involve changes in gene expression sustained by epigenetic modifications in brain circuits, including the mesocorticolimbic pathway. Brahma (BRM) and Brahma Related Gene 1 (BRG1) are ATPase subunits of the SWI/SNF complexes involved in chromatin remodeling, a process essential to enduring plastic changes in gene expression. Here, we show that in mice, social defeat induces changes in BRG1 nuclear distribution. The inactivation of the Brg1/Smarca4 gene within dopamine-innervated regions or the constitutive inactivation of the Brm/Smarca2 gene leads to resilience to repeated social defeat and decreases the behavioral responses to cocaine without impacting midbrain dopamine neurons activity. Within striatal medium spiny neurons, Brg1 gene inactivation reduces the expression of stress- and cocaine-induced immediate early genes, increases levels of heterochromatin and at a global scale decreases chromatin accessibility. Altogether these data demonstrate the pivotal function of SWI/SNF complexes in behavioral and transcriptional adaptations to salient environmental challenges.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin , Adenosine Triphosphatases/metabolism , Animals , Cell Line, Tumor , Mice , Reward
6.
Neuropharmacology ; 190: 108534, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33781778

ABSTRACT

Stress has been acknowledged as one of the main risk factors for the onset of psychiatric disorders. Social stress is the most common type of stressor encountered in our daily lives. Uncovering the molecular determinants of the effect of stress on the brain would help understanding the complex maladaptations that contribute to pathological stress-related mental states. We examined molecular changes in the reward system following social defeat stress in mice, as increasing evidence implicates this system in sensing stressful stimuli. Following acute or chronic social defeat stress, the activation (i.e. phosphorylation) of extracellular signal-regulated kinases ERK1 and ERK2 (pERK1/2), markers of synaptic plasticity, was monitored in sub-regions of the reward system. We employed pharmacological antagonists and inhibitory DREADD to dissect the sequence of events controlling pERK1/2 dynamics. The nucleus accumbens (NAc) showed marked increases in pERK1/2 following both acute and chronic social stress compared to the dorsal striatum. Increases in pERK1/2 required dopamine D1 receptors and GluN2B-containing NMDA receptors. Paraventricular thalamic glutamatergic inputs to the NAc are required for social stress-induced pERK1/2. The molecular adaptations identified here could contribute to the long-lasting impact of stress on the brain and may be targeted to counteract stress-related psychopathologies.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neostriatum/metabolism , Nucleus Accumbens/metabolism , Receptors, Dopamine D1/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Stress, Psychological/metabolism , Animals , MAP Kinase Signaling System , Mice , Midline Thalamic Nuclei/metabolism , Receptors, Dopamine/metabolism , Receptors, Glutamate/metabolism
7.
Front Synaptic Neurosci ; 13: 799274, 2021.
Article in English | MEDLINE | ID: mdl-34970134

ABSTRACT

Drug addiction is defined as a compulsive pattern of drug-seeking- and taking- behavior, with recurrent episodes of abstinence and relapse, and a loss of control despite negative consequences. Addictive drugs promote reinforcement by increasing dopamine in the mesocorticolimbic system, which alters excitatory glutamate transmission within the reward circuitry, thereby hijacking reward processing. Within the reward circuitry, the striatum is a key target structure of drugs of abuse since it is at the crossroad of converging glutamate inputs from limbic, thalamic and cortical regions, encoding components of drug-associated stimuli and environment, and dopamine that mediates reward prediction error and incentive values. These signals are integrated by medium-sized spiny neurons (MSN), which receive glutamate and dopamine axons converging onto their dendritic spines. MSN primarily form two mostly distinct populations based on the expression of either DA-D1 (D1R) or DA-D2 (D2R) receptors. While a classical view is that the two MSN populations act in parallel, playing antagonistic functional roles, the picture seems much more complex. Herein, we review recent studies, based on the use of cell-type-specific manipulations, demonstrating that dopamine differentially modulates dendritic spine density and synapse formation, as well as glutamate transmission, at specific inputs projecting onto D1R-MSN and D2R-MSN to shape persistent pathological behavioral in response to drugs of abuse. We also discuss the identification of distinct molecular events underlying the detrimental interplay between dopamine and glutamate signaling in D1R-MSN and D2R-MSN and highlight the relevance of such cell-type-specific molecular studies for the development of innovative strategies with potential therapeutic value for addiction. Because drug addiction is highly prevalent in patients with other psychiatric disorders when compared to the general population, we last discuss the hypothesis that shared cellular and molecular adaptations within common circuits could explain the co-occurrence of addiction and depression. We will therefore conclude this review by examining how the nucleus accumbens (NAc) could constitute a key interface between addiction and depression.

8.
Sci Adv ; 7(43): eabg5970, 2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34669474

ABSTRACT

Addictive drugs increase dopamine in the nucleus accumbens (NAc), where it persistently shapes excitatory glutamate transmission and hijacks natural reward processing. Here, we provide evidence, from mice to humans, that an underlying mechanism relies on drug-evoked heteromerization of glutamate N-methyl-d-aspartate receptors (NMDAR) with dopamine receptor 1 (D1R) or 2 (D2R). Using temporally controlled inhibition of D1R-NMDAR heteromerization, we unraveled their selective implication in early phases of cocaine-mediated synaptic, morphological, and behavioral responses. In contrast, preventing D2R-NMDAR heteromerization blocked the persistence of these adaptations. Interfering with these heteromers spared natural reward processing. Notably, we established that D2R-NMDAR complexes exist in human samples and showed that, despite a decreased D2R protein expression in the NAc, individuals with psychostimulant use disorder display a higher proportion of D2R forming heteromers with NMDAR. These findings contribute to a better understanding of molecular mechanisms underlying addiction and uncover D2R-NMDAR heteromers as targets with potential therapeutic value.

9.
Microbiome ; 9(1): 157, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34238386

ABSTRACT

BACKGROUND: Autism spectrum disorders (ASD) are associated with dysregulation of the microbiota-gut-brain axis, changes in microbiota composition as well as in the fecal, serum, and urine levels of microbial metabolites. Yet a causal relationship between dysregulation of the microbiota-gut-brain axis and ASD remains to be demonstrated. Here, we hypothesized that the microbial metabolite p-Cresol, which is more abundant in ASD patients compared to neurotypical individuals, could induce ASD-like behavior in mice. RESULTS: Mice exposed to p-Cresol for 4 weeks in drinking water presented social behavior deficits, stereotypies, and perseverative behaviors, but no changes in anxiety, locomotion, or cognition. Abnormal social behavior induced by p-Cresol was associated with decreased activity of central dopamine neurons involved in the social reward circuit. Further, p-Cresol induced changes in microbiota composition and social behavior deficits could be transferred from p-Cresol-treated mice to control mice by fecal microbiota transplantation (FMT). We also showed that mice transplanted with the microbiota of p-Cresol-treated mice exhibited increased fecal p-Cresol excretion, compared to mice transplanted with the microbiota of control mice. In addition, we identified possible p-Cresol bacterial producers. Lastly, the microbiota of control mice rescued social interactions, dopamine neurons excitability, and fecal p-Cresol levels when transplanted to p-Cresol-treated mice. CONCLUSIONS: The microbial metabolite p-Cresol induces selectively ASD core behavioral symptoms in mice. Social behavior deficits induced by p-Cresol are dependant on changes in microbiota composition. Our study paves the way for therapeutic interventions targeting the microbiota and p-Cresol production to treat patients with ASD. Video abstract.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Gastrointestinal Microbiome , Animals , Autistic Disorder/etiology , Cresols , Fecal Microbiota Transplantation , Humans , Mice
10.
Handb Exp Pharmacol ; (192): 173-207, 2009.
Article in English | MEDLINE | ID: mdl-19184650

ABSTRACT

Nicotine achieves its psychopharmacological effects by interacting with nicotinic acetylcholine receptors (nAChRs) in the brain. There are numerous subtypes of nAChR that differ in their properties, including their sensitivity to nicotine, permeability to calcium and propensity to desensitise. The nAChRs are differentially localised to different brain regions and are found on presynaptic terminals as well as in somatodendritic regions of neurones. Through their permeability to cations, these ion channel proteins can influence both neuronal excitability and cell signalling mechanisms, and these various responses can contribute to the development or maintenance of dependence. However, many questions and uncertainties remain in our understanding of these events and their relevance to tobacco addiction. In this chapter, we briefly overview the fundamental characteristics of nAChRs that are germane to nicotine's effects and then consider the cellular responses to acute and chronic nicotine, with particular emphasis on dopamine systems because they have been the most widely studied in the context of nicotine dependence. Where appropriate, methodological aspects are critically reviewed.


Subject(s)
Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Receptors, Nicotinic/drug effects , Animals , Brain/drug effects , Brain/metabolism , Dopamine/metabolism , Gene Expression Regulation/drug effects , Humans , Neurotransmitter Agents/metabolism , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Receptors, Nicotinic/metabolism , Signal Transduction/drug effects
11.
Cell Rep ; 29(2): 317-331.e5, 2019 Oct 08.
Article in English | MEDLINE | ID: mdl-31597094

ABSTRACT

There is a growing consensus that Alzheimer's disease (AD) involves failure of the homeostatic machinery, which underlies the firing stability of neural circuits. What are the culprits leading to neuron firing instability? The amyloid precursor protein (APP) is central to AD pathogenesis, and we recently showed that its intracellular domain (AICD) could modify synaptic signal integration. We now hypothesize that AICD modifies neuron firing activity, thus contributing to the disruption of memory processes. Using cellular, electrophysiological, and behavioral techniques, we show that pathological AICD levels weaken CA1 neuron firing activity through a gene-transcription-dependent mechanism. Furthermore, increased AICD production in hippocampal neurons modifies oscillatory activity, specifically in the γ-frequency range, and disrupts spatial memory task. Collectively, our data suggest that AICD pathological levels, observed in AD mouse models and in human patients, might contribute to progressive neuron homeostatic failure, driving the shift from normal aging to AD.


Subject(s)
Action Potentials/physiology , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , CA1 Region, Hippocampal/physiology , Neurons/physiology , Spatial Memory/physiology , Animals , Calcium Channels/metabolism , Gamma Rhythm/physiology , Humans , Male , Mice, Inbred C57BL , Models, Biological , Potassium Channels/metabolism , Protein Domains , Rats, Sprague-Dawley , Structure-Activity Relationship , Transcription, Genetic
12.
Nat Commun ; 9(1): 4449, 2018 10 25.
Article in English | MEDLINE | ID: mdl-30361503

ABSTRACT

Stressful life events are primary environmental factors that markedly contribute to depression by triggering brain cellular maladaptations. Dysregulation of ventral tegmental area (VTA) dopamine neurons has been causally linked to the appearance of social withdrawal and anhedonia, two classical manifestations of depression. However, the relevant inputs that shape these dopamine signals remain largely unknown. We demonstrate that chronic social defeat (CSD) stress, a preclinical paradigm of depression, causes marked hyperactivity of laterodorsal tegmentum (LDTg) excitatory neurons that project to the VTA. Selective chemogenetic-mediated inhibition of cholinergic LDTg neurons prevent CSD-induced VTA DA neurons dysregulation and depressive-like behaviors. Pro-depressant outcomes are replicated by pairing activation of LDTg cholinergic terminals in the VTA with a moderate stress. Prevention of CSD outcomes are recapitulated by blocking corticotropin-releasing factor receptor 1 within the LDTg. These data uncover a neuro-circuitry of depressive-like disorders and demonstrate that stress, via a neuroendocrine signal, profoundly dysregulates the LDTg.


Subject(s)
Acetylcholine/metabolism , Behavior, Animal , Depression/psychology , Dopaminergic Neurons/pathology , Mesencephalon/pathology , Pons/pathology , Stress, Psychological/complications , Animals , Chronic Disease , Corticotropin-Releasing Hormone/metabolism , Depression/pathology , Dopaminergic Neurons/metabolism , Gene Silencing , Glutamic Acid/metabolism , Male , Mice, Inbred C57BL , Models, Biological , Pontine Tegmentum/pathology , Receptors, Corticotropin-Releasing Hormone/metabolism , Signal Transduction , Ventral Tegmental Area/pathology
13.
Neuroreport ; 18(2): 121-6, 2007 Jan 22.
Article in English | MEDLINE | ID: mdl-17301675

ABSTRACT

Transcriptional regulation is central to the long-term effects of drugs of abuse. Activation of the extracellular signal-regulated kinase (ERK1/2) pathway underlies plasticity changes that accompany drug use. One target of ERK1/2 activation is the Ets-like transcription factor Elk-1. We show here that nicotine modulates Elk-1 in the rat hippocampus in a spatially and temporally specific manner. In-vitro nicotine (1 muM) activated Elk-1 in hippocampal slices. In-vivo acute nicotine (0.4 mg/kg) activated Elk-1 in the CA1 area but not in the dentate gyrus. Chronic nicotine for 14 days changed the level of total Elk-1 but not its phosphorylation state. Thus, Elk-1 regulation of transcriptional events may contribute to nicotine-induced changes in the hippocampus.


Subject(s)
Hippocampus/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Tobacco Use Disorder/metabolism , ets-Domain Protein Elk-1/metabolism , Animals , Hippocampus/metabolism , Immunohistochemistry , Male , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Tobacco Use Disorder/physiopathology , Transcription, Genetic/drug effects , Transcription, Genetic/physiology
15.
Neuropsychopharmacology ; 42(2): 512-523, 2017 01.
Article in English | MEDLINE | ID: mdl-27461084

ABSTRACT

Serotonin (5-HT) deficiency occurs in a number of brain disorders that affect cognitive function. However, a direct causal relationship between 5-HT hypo-transmission and memory and underlying mechanisms has not been established. We used mice with a constitutive depletion of 5-HT brain levels (Pet1KO mice) to analyze the contribution of 5-HT to different forms of learning and memory. Pet1KO mice exhibited a striking deficit in novel object recognition memory, a hippocampal-dependent task. No alterations were found in tasks for social recognition, procedural learning, or fear memory. Viral delivery of designer receptors exclusively activated by designer drugs was used to selectively silence the activity of 5-HT neurons in the raphe. Inhibition of 5-HT neurons in the median raphe, but not the dorsal raphe, was sufficient to impair object recognition in adult mice. In vivo electrophysiology in behaving mice showed that long-term potentiation in the hippocampus of 5-HT-deficient mice was altered, and administration of the 5-HT1A agonist 8-OHDPAT rescued the memory deficits. Our data suggest that hyposerotonergia selectively affects declarative hippocampal-dependent memory. Serotonergic projections from the median raphe are necessary to regulate object memory and hippocampal synaptic plasticity processes, through an inhibitory control mediated by 5-HT1A receptors.


Subject(s)
Hippocampus/physiology , Memory/physiology , Neuronal Plasticity , Serotonergic Neurons/physiology , Serotonin/physiology , Animals , Excitatory Postsynaptic Potentials , Female , Long-Term Potentiation , Male , Memory Consolidation/physiology , Mice, Knockout , Raphe Nuclei/physiology , Receptor, Serotonin, 5-HT1A/physiology , Recognition, Psychology/physiology , Transcription Factors/genetics
16.
J Mol Neurosci ; 30(1-2): 137-40, 2006.
Article in English | MEDLINE | ID: mdl-17192660

ABSTRACT

Neuronal nicotinic acetylcholine receptors (nAChRs) in the CNS appear to exert a predominantly modulatory influence on brain mechanisms, despite being fast-acting ligand-gated ion channels. Many nAChRs have an extrasynaptic location on somatodendritic regions or presynaptic terminals. They influence local excitability by depolarization and can initiate short- and long-term changes by interfacing with Ca2+ signaling pathways (Dajas- Bailador and Wonnacott, 2004). The modulation of neurotransmitter release by presynaptic nAChRs is well-documented (Wonnacott, 1997): Both Na+ and Ca2+ fluxes associated with nAChR activation can influence transmitter release. It is also emerging that nAChRs, especially the alpha7 subtype, can exert an indirect effect on transmitter release, through modulation of amino acid transmitters. This complex scenario facilitates transmitter cross talk, which is the subject of this short review.


Subject(s)
Corpus Striatum/physiology , Hippocampus/physiology , Receptor Cross-Talk/physiology , Receptors, Nicotinic/physiology , Animals , Glutamine/physiology , Hippocampus/drug effects , gamma-Aminobutyric Acid/physiology
17.
Eur J Pharmacol ; 536(1-2): 1-11, 2006 Apr 24.
Article in English | MEDLINE | ID: mdl-16563372

ABSTRACT

Neuronal nicotinic acetylcholine receptors subserve predominantly modulatory roles in the brain, making them attractive therapeutic targets. Natural products provide key leads in the quest for nicotinic receptor subtype-selective compounds. Cytisine, found in Leguminosae spp., binds with high affinity to alpha4beta2* nicotinic receptors. We have compared the effect of C3 and C5 halogenation of cytisine and methylcytisine (MCy) on their interaction with native rat nicotinic receptors. 3-Bromocytisine (3-BrCy) and 3-iodocytisine (3-ICy) exhibited increased binding affinity (especially at alpha7 nicotinic receptors; Ki approximately 0.1 microM) and functional potency, whereas C5-halogenation was detrimental. 3-BrCy and 3-ICy were more potent than cytisine at evoking [3H]dopamine release from striatal slices (EC50 approximately 11 nM), [3H]noradrenaline release from hippocampal slices (EC50 approximately 250 nM), increases in intracellular Ca2+ in PC12 cells and inward currents in Xenopus oocytes expressing human alpha3beta4 nicotinic receptor (EC50 approximately 2 microM). These compounds were also more efficacious than cytisine. C3-halogenation of cytisine is proposed to stabilize the open conformation of the nicotinic receptor but does not enhance subtype selectivity.


Subject(s)
Alkaloids/pharmacology , Hydrocarbons, Brominated/pharmacology , Nicotinic Agonists/pharmacology , Receptors, Nicotinic/metabolism , Alkaloids/chemistry , Alkaloids/metabolism , Animals , Azocines/chemistry , Azocines/metabolism , Azocines/pharmacology , Binding, Competitive/drug effects , Brain/drug effects , Brain/metabolism , Calcium/metabolism , Dopamine/metabolism , Dose-Response Relationship, Drug , Female , Humans , Hydrocarbons, Brominated/chemistry , Hydrocarbons, Brominated/metabolism , Male , Membrane Potentials/drug effects , Nicotine/antagonists & inhibitors , Nicotine/pharmacology , Nicotinic Agonists/chemistry , Nicotinic Agonists/metabolism , Norepinephrine/metabolism , Oocytes/drug effects , Oocytes/physiology , PC12 Cells , Quinolizines/chemistry , Quinolizines/metabolism , Quinolizines/pharmacology , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/genetics , Receptors, Nicotinic/physiology , Xenopus
18.
Br J Pharmacol ; 145(8): 1084-92, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15937519

ABSTRACT

In this study, we have examined cellular responses of neuroblastoma SH-SY5Y cells after chronic treatment with galantamine, a drug used to treat Alzheimer's disease that has a dual mechanism of action: inhibition of acetylcholinesterase and allosteric potentiation of nicotinic acetylcholine receptors (nAChR). Acute experiments confirmed that maximum potentiation of nicotinic responses occurs at 1 microM galantamine; hence this concentration was chosen for chronic treatment. Exposure to 1 microM galantamine for 4 days decreased Ca(2+) responses (by 19.8+/-3.6%) or [(3)H]noradrenaline ([(3)H]NA) release (by 23.9+/-3.3%) elicited by acute application of nicotine. KCl-evoked increases in intracellular Ca(2+) were also inhibited by 10.0+/-1.9% after 4 days' treatment with galantamine. These diminished responses are consistent with the downregulation of downstream cellular processes. Ca(2+) responses evoked by activation of muscarinic acetylcholine receptors were unaffected by chronic galantamine treatment. Exposure to the more potent acetylcholinesterase inhibitor rivastigmine (1 microM) for 4 days failed to alter nicotine-, KCl-, or muscarinic receptor-evoked increases in intracellular Ca(2+). These observations support the hypothesis that chronic galantamine exerts its effects through interaction with nAChR in this cell line. Exposure to 10 microM nicotine for 4 days produced decreases in acute nicotine- (18.0+/-3.5%) and KCl-evoked Ca(2+) responses (10.6+/-2.5%) and nicotine-evoked [(3)H]NA release (26.0+/-3.3%) that are comparable to the effects of a corresponding exposure to galantamine. Treatment with 1 microM galantamine did not alter numbers of [(3)H]epibatidine-binding sites in SH-SY5Y cells, in contrast to 62% upregulation of these sites in response to 10 microM nicotine. Thus, chronic galantamine acts at nAChR to decrease subsequent functional responses to acute stimulation with nicotine or KCl. This effect appears to be independent of the upregulation of nAChR-binding sites.


Subject(s)
Calcium/metabolism , Cell Membrane/drug effects , Cholinesterase Inhibitors/pharmacokinetics , Galantamine/pharmacology , Receptors, Nicotinic/metabolism , Binding Sites , Cell Line, Tumor , Cell Membrane/metabolism , Dose-Response Relationship, Drug , Humans , Neuroblastoma/pathology , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Potassium Chloride/pharmacology , Time Factors
19.
Neuropsychopharmacology ; 40(7): 1772-81, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25622751

ABSTRACT

The early phase of Alzheimer's disease (AD) is characterized by hippocampus-dependent memory deficits and impaired synaptic plasticity. Increasing evidence suggests that stress and dysregulation of the hypothalamo-pituitary-adrenal (HPA) axis, marked by the elevated circulating glucocorticoids, are risk factors for AD onset. How these changes contribute to early hippocampal dysfunction remains unclear. Using an elaborated version of the object recognition task, we carefully monitored alterations in key components of episodic memory, the first type of memory altered in AD patients, in early symptomatic Tg2576 AD mice. We also combined biochemical and ex vivo electrophysiological analyses to reveal novel cellular and molecular dysregulations underpinning the onset of the pathology. We show that HPA axis, circadian rhythm, and feedback mechanisms, as well as episodic memory, are compromised in this early symptomatic phase, reminiscent of human AD pathology. The cognitive decline could be rescued by subchronic in vivo treatment with RU486, a glucocorticoid receptor antagonist. These observed phenotypes were paralleled by a specific enhancement of N-Methyl-D-aspartic acid receptor (NMDAR)-dependent LTD in CA1 pyramidal neurons, whereas LTP and metabotropic glutamate receptor-dependent LTD remain unchanged. NMDAR transmission was also enhanced. Finally, we show that, as for the behavioral deficit, RU486 treatment rescues this abnormal synaptic phenotype. These preclinical results define glucocorticoid signaling as a contributing factor to both episodic memory loss and early synaptic failure in this AD mouse model, and suggest that glucocorticoid receptor targeting strategies could be beneficial to delay AD onset.


Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/pathology , Memory Disorders/etiology , Memory, Episodic , Neuronal Plasticity/genetics , Receptors, Glucocorticoid/metabolism , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Dexamethasone/therapeutic use , Disease Models, Animal , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Glucocorticoids/blood , Glucocorticoids/therapeutic use , Hippocampus/pathology , Hormone Antagonists/therapeutic use , Humans , Memory Disorders/drug therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mifepristone/therapeutic use , Mutation/genetics , Neuronal Plasticity/drug effects , Quinoxalines/pharmacology , Recognition, Psychology/drug effects , Valine/analogs & derivatives , Valine/pharmacology
20.
J Neurosci Methods ; 134(1): 65-74, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15102504

ABSTRACT

Alpha 7 subunit-containing nicotinic acetylcholine receptors (alpha7* nAChR) are involved in a variety of functions in the mammalian brain, including modulating neurotransmitter release and synaptic plasticity. Identifying the precise cellular distribution of alpha7* nAChRs with respect to the local neurochemical environment is crucial to understanding these biological roles. Current strategies for localising alpha7* nAChRs at the subcellular level have limitations. Anti-alpha7 subunit antibodies detect both assembled and unassembled subunits whereas biotinylated alphabungarotoxin (alphaBgt) only binds to assembled alpha7* nAChRs, but interpretation of labelling is marred by co-detection of endogenous tissue biotin. To overcome these problems, we have characterised a novel 1.4 nm gold alphaBgt conjugate used to directly localise alpha7* nAChR. Gold conjugation does not significantly decrease binding affinity, and gold alphaBgt specifically labels alpha7* nAChR in both unfixed and aldehyde-fixed tissue at the light and electron microscope levels, labelling being abolished in the presence of excess competing toxin. At the ultrastructural level, gold alphaBgt is associated with neuronal membranes and located at axon-dendritic synapses in the rat hippocampus CA1 stratum radiatum. These results reveal gold alphaBgt to be a valuable new tool in elucidating the functional neuroanatomy of alpha7* nAChR in the central nervous system.


Subject(s)
Brain/cytology , Bungarotoxins/analysis , Gold/analysis , Nanotechnology/methods , Receptors, Nicotinic/analysis , Animals , Brain/metabolism , Bungarotoxins/metabolism , Dose-Response Relationship, Drug , Gold/metabolism , Male , Protein Binding/physiology , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/metabolism , Subcellular Fractions/chemistry , Subcellular Fractions/metabolism , alpha7 Nicotinic Acetylcholine Receptor
SELECTION OF CITATIONS
SEARCH DETAIL