Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Expert Opin Investig Drugs ; 32(12): 1113-1121, 2023.
Article in English | MEDLINE | ID: mdl-37994870

ABSTRACT

INTRODUCTION: Successful phase 3 trials of KarXT in people with schizophrenia herald a new era of treating the disorder with drugs that do not target the dopamine D2 receptor. The active component of KarXT is xanomeline, a muscarinic (CHRM) M1 and M4 agonist, making muscarinic receptors a viable target for treating schizophrenia. AREAS COVERED: This review covers the process of taking drugs that activate the muscarinic M1 and M4 receptors from conceptualization to the clinic and details the mechanisms by which activating the CHRM1 and 4 can affect the broad spectrum of symptoms experienced by people with schizophrenia. EXPERT OPINION: Schizophrenia is a syndrome which means drugs that activate muscarinic M1 and M4 receptors, as was the case for antipsychotic drugs acting on the dopamine D2 receptor, will not give optimal outcomes in everyone within the syndrome. Thus, it would be ideal to identify people who are responsive to drugs activating the CHRM1 and 4. Given knowledge of the actions of these receptors, it is possible treatment non-response could be restricted to sub-groups within the syndrome who have deficits in cortical CHRM1 or those with one of the cognitive endophenotypes that may be identifiable by changes in the blood transcriptome.


Subject(s)
Antipsychotic Agents , Schizophrenia , Humans , Schizophrenia/drug therapy , Muscarinic Agonists/pharmacology , Muscarinic Agonists/therapeutic use , Receptor, Muscarinic M4/agonists , Receptor, Muscarinic M4/genetics , Receptor, Muscarinic M4/therapeutic use , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Receptors, Dopamine D2/therapeutic use , Receptor, Muscarinic M1
2.
J Alzheimers Dis ; 85(1): 323-330, 2022.
Article in English | MEDLINE | ID: mdl-34806612

ABSTRACT

BACKGROUND: Central nervous system disruption of cholinergic (ACh) signaling, which plays a major role in cognitive processes, is well documented in dementia with Lewy bodies (DLB) and Alzheimer's disease (AD). The expression of muscarinic ACh receptors type 1 and 4 (CHRM1 and CHRM4) has been reported to be altered in the brain of DLB patients. OBJECTIVE: We aim to assess the peripheral gene expression of CHRM1 and 4 in DLB as a possible marker as compared to AD and healthy control (HC) subjects. METHODS: Peripheral blood mononuclear cells were collected from 21 DLB, 13 AD, and 8 HC matched subjects. RT-PCR was performed to estimate gene expression of CHRM1 and CHRM4. RESULTS: Peripheral CHRM1 expression was higher and CHRM4 was lower in DLB and AD compared to HC, whereas both CHRM1 and CHRM4 levels were higher in AD compared to DLB patients. Receiver operating characteristics curves, with logistic regression analysis, showed that combining peripheral CHRM1 and CHRM4 levels, DLB and AD subjects were classified with an accuracy of 76.0%. CONCLUSION: Alterations of peripheral CHRM1 and CHRM4 was found in both AD and DLB patients as compared to HC. CHRM1 and CHRM4 gene expression resulted to be lower in DLB patients compared to AD. In the future, peripheral CHRM expression could be studied as a possible marker of neurodegenerative conditions associated with cholinergic deficit and a possible marker of response to acetylcholinesterase inhibitors.


Subject(s)
Alzheimer Disease/metabolism , Lewy Body Disease/metabolism , Receptor, Muscarinic M1/metabolism , Receptor, Muscarinic M4/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Case-Control Studies , Diagnosis, Differential , Female , Humans , Lewy Body Disease/genetics , Logistic Models , Male , ROC Curve , Receptor, Muscarinic M1/genetics , Receptor, Muscarinic M4/genetics
3.
Biomolecules ; 11(7)2021 07 08.
Article in English | MEDLINE | ID: mdl-34356625

ABSTRACT

Monoamine oxidases (MAOs) and muscarinic acetylcholine receptors (mAChRs) are considered important therapeutic targets for Parkinson's disease (PD). Lipophilic tanshinones are major phytoconstituents in the dried roots of Salvia miltiorrhiza that have demonstrated neuroprotective effects against dopaminergic neurotoxins and the inhibition of MAO-A. Since MAO-B inhibition is considered an effective therapeutic strategy for PD, we tested the inhibitory activities of three abundant tanshinone congeners against recombinant human MAO (hMAO) isoenzymes through in vitro experiments. In our study, tanshinone I (1) exhibited the highest potency against hMAO-A, followed by tanshinone IIA and cryptotanshinone, with an IC50 less than 10 µM. They also suppressed hMAO-B activity, with an IC50 below 25 µM. Although tanshinones are known to inhibit hMAO-A, their enzyme inhibition mechanism and binding sites have yet to be investigated. Enzyme kinetics and molecular docking studies have revealed the mode of inhibition and interactions of tanshinones during enzyme inhibition. Proteochemometric modeling predicted mAChRs as possible pharmacological targets of 1, and in vitro functional assays confirmed the selective M4 antagonist nature of 1 (56.1% ± 2.40% inhibition of control agonist response at 100 µM). These findings indicate that 1 is a potential therapeutic molecule for managing the motor dysfunction and depression associated with PD.


Subject(s)
Abietanes , Monoamine Oxidase Inhibitors , Monoamine Oxidase , Phenanthrenes , Receptor, Muscarinic M4 , Salvia miltiorrhiza/chemistry , Abietanes/chemistry , Abietanes/pharmacology , Animals , CHO Cells , Cricetulus , Humans , Monoamine Oxidase/chemistry , Monoamine Oxidase/genetics , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/chemistry , Monoamine Oxidase Inhibitors/pharmacology , Phenanthrenes/chemistry , Phenanthrenes/pharmacology , Receptor, Muscarinic M4/antagonists & inhibitors , Receptor, Muscarinic M4/chemistry , Receptor, Muscarinic M4/genetics , Receptor, Muscarinic M4/metabolism
4.
J Neurosci ; 41(34): 7267-7277, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34272313

ABSTRACT

Adaptive reward-related decision making requires accurate prospective consideration of the specific outcome of each option and its current desirability. Often this information must be inferred based on the presence of predictive environmental events. The basolateral amygdala (BLA) and medial orbitofrontal cortex (mOFC) are two key nodes in the circuitry supporting such outcome expectations, but very little is known about the function of direct connections between these regions. Here, in male rats, we first anatomically confirmed the existence of bidirectional, direct projections between the mOFC and BLA and found that BLA projections to mOFC are largely distinct from those to lateral OFC (lOFC). Next, using pathway-specific chemogenetic inhibition and the outcome-selective Pavlovian-to-instrumental transfer and devaluation tests, we interrogated the function of the bidirectional mOFC-BLA connections in reward-directed behavior. We found evidence that the mOFC→BLA pathway mediates the use of environmental cues to understand which specific reward is predicted, information needed to infer which action to choose, and how desirable that reward is to ensure adaptive responses to the cue. By contrast, the BLA→mOFC pathway is not needed to use the identity of an expected reward to guide choice but does mediate adaptive responses to cues based on the current desirability of the reward they predict. These functions differ from those we previously identified for the lOFC-BLA circuit. Collectively, the data reveal the mOFC-BLA circuit as critical for the cue-dependent reward outcome expectations that influence adaptive behavior and decision making.SIGNIFICANCE STATEMENT To make good decisions we evaluate how advantageous a particular course of action would be. This requires understanding what rewarding outcomes can be expected and how desirable they currently are. Such prospective considerations are critical for adaptive decision making but disrupted in many psychiatric diseases. Here, we reveal that direct connections between the medial orbitofrontal cortex and basolateral amygdala mediate these functions. These findings are especially important in light of evidence of dysfunction in this circuit in substance use disorder and mental illnesses marked by poor decision making.


Subject(s)
Adaptation, Psychological/physiology , Basolateral Nuclear Complex/physiology , Choice Behavior/physiology , Cues , Decision Making/physiology , Neural Pathways/physiology , Prefrontal Cortex/physiology , Reward , Acoustic Stimulation , Animals , Axonal Transport , Conditioning, Classical/drug effects , Dependovirus/genetics , Extinction, Psychological , Fluorescent Dyes/analysis , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Image Processing, Computer-Assisted , Male , Rats , Rats, Long-Evans , Receptor, Muscarinic M4/genetics , Receptor, Muscarinic M4/physiology , Recombinant Proteins/metabolism
5.
J Psychopharmacol ; 35(1): 31-39, 2021 01.
Article in English | MEDLINE | ID: mdl-33143542

ABSTRACT

BACKGROUND: Clozapine has heterogenous efficacy in enhancing working memory in schizophrenia. We have previously hypothesized that this is due to opposing effects of clozapine and its metabolite, N-desmethylclozapine, at the muscarinic M1 receptor and demonstrated that a lower clozapine/N-desmethylclozapine ratio is associated with better working memory than clozapine or N-desmethylclozapine levels alone. AIMS: In this study, we expanded the above hypothesis to explore whether genetic variation in the cholinergic receptor muscarinic 1 gene, encoding the M1 receptor, affects the relationship between clozapine/N-desmethylclozapine and working memory. Further, we explored whether CYP1A2 gene variants affect the ratio of clozapine/N-desmethylclozapine and by this, working memory performance. METHODS: We evaluated two functionally significant single nucleotide polymorphisms, rs1942499 and rs2075748, in cholinergic receptor muscarinic 1, with the haplotype T-A associated with lower transcriptional activity than the haplotype C-G. Further, we examined CYP1A2 *1F, with *1F/*1F conferring high inducibility in the presence of smoking. RESULTS: In a sample of 30 patients with schizophrenia on clozapine monotherapy, clozapine/N-desmethylclozapine was correlated with working memory only in non-carriers of the haplotype T-A of the cholinergic receptor muscarinic 1 gene. Interaction of CYP1A2 genotype and smoking status significantly affected clozapine concentrations, but there were no significant effects of CYP1A2 genotype and smoking status on the relationship between clozapine/N-desmethylclozapine on working memory. CONCLUSIONS: Our finding that the relationship between clozapine/N-desmethylclozapine and working memory is specific to patients with potentially higher transcription of M1 receptor (i.e. non-carriers of the haplotype T-A of cholinergic receptor muscarinic 1) supports a cholinergic mechanism underlying this relationship.


Subject(s)
Clozapine/analogs & derivatives , Cytochrome P-450 CYP1A2/genetics , Memory, Short-Term , Receptor, Muscarinic M4/genetics , Schizophrenia , Adult , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/pharmacokinetics , Clozapine/administration & dosage , Clozapine/pharmacokinetics , Female , Humans , Male , Memory, Short-Term/drug effects , Memory, Short-Term/physiology , Pharmacogenomic Testing/methods , Pharmacogenomic Variants , Polymorphism, Single Nucleotide , Schizophrenia/diagnosis , Schizophrenia/drug therapy , Schizophrenia/genetics , Schizophrenic Psychology , Smoking/metabolism
6.
Brain Struct Funct ; 225(5): 1615-1629, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32409918

ABSTRACT

The deletion of M4 muscarinic receptors (MRs) changes biological rhythm parameters in females. Here, we searched for the mechanisms responsible for these changes. We performed biological rhythm analysis in two experiments: in experiment 1, the mice [C57Bl/6NTac (WT) and M4 MR -/- mice (KO)] were first exposed to a standard LD regime (12/12-h light/dark cycle) for 8 days and then subsequently exposed to constant darkness (for 24 h/day, DD regime) for another 16 days. In experiment 2, the mice (after the standard LD regime) were exposed to the DD regime and to one light pulse (zeitgeber time 14) on day 9. We also detected M1 MRs in brain areas implicated in locomotor biological rhythm regulation. In experiment 1, the biological rhythm activity curves differed: the period (τ, duration of diurnal cycle) was shorter in the DD regime. Moreover, the day mean, mesor (midline value), night mean and their difference were higher in KO animals. The time in which the maximal slope occurred was lower in the DD regime than in the LD regime in both WT and KO but was lower in KO than in WT mice. In experiment 2, there were no differences in biological rhythm parameters between WT and KO mice. The densities of M1 MRs in the majority of areas implicated in locomotor biological rhythm were low. A significant amount of M1 MR was found in the striatum. These results suggest that although core clock output is changed by M4 MR deletion, the structures involved in biological rhythm regulation in WT and KO animals are likely the same, and the most important areas are the striatum, thalamus and intergeniculate leaflet.


Subject(s)
Locomotion/physiology , Neostriatum/physiology , Periodicity , Receptor, Muscarinic M4/physiology , Thalamus/physiology , Actigraphy , Animals , Female , Mice, Inbred C57BL , Mice, Knockout , Receptor, Muscarinic M4/genetics
7.
Biol Psychiatry ; 88(12): 898-909, 2020 12 15.
Article in English | MEDLINE | ID: mdl-32331824

ABSTRACT

BACKGROUND: Alcohol use disorder (AUD) is a major socioeconomic burden on society, and current pharmacotherapeutic treatment options are inadequate. Aberrant alcohol use and seeking alters frontostriatal function. METHODS: We performed genome-wide RNA sequencing and subsequent quantitative polymerase chain reaction and receptor binding validation in the caudate-putamen of human AUD samples to identify potential therapeutic targets. We then back-translated our top candidate targets into a rodent model of long-term alcohol consumption to assess concordance of molecular adaptations in the rat striatum. Finally, we adopted rat behavioral models of alcohol intake and seeking to validate a potential therapeutic target. RESULTS: We found that G protein-coupled receptors were the top canonical pathway differentially regulated in individuals with AUD. The M4 muscarinic acetylcholine receptor (mAChR) was downregulated at the gene and protein levels in the putamen, but not in the caudate, of AUD samples. We found concordant downregulation of the M4 mAChR, specifically on dopamine D1 receptor-expressing medium spiny neurons in the rat dorsolateral striatum. Systemic administration of the selective M4 mAChR positive allosteric modulator, VU0467154, reduced home cage and operant alcohol self-administration, motivation to obtain alcohol, and cue-induced reinstatement of alcohol seeking in rats. Local microinjections of VU0467154 in the rat dorsolateral striatum reduced alcohol self-administration and cue-induced reinstatement of alcohol seeking. CONCLUSIONS: Collectively, these results identify the M4 mAChR as a potential therapeutic target for the treatment of AUD and the D1 receptor-positive medium spiny neurons in the dorsolateral striatum as a key site mediating the actions of M4 mAChR in relation to alcohol consumption and seeking.


Subject(s)
Alcoholism , Receptor, Muscarinic M4 , Acetylcholine , Alcoholism/drug therapy , Alcoholism/genetics , Animals , Cholinergic Agents , Humans , Rats , Receptor, Muscarinic M4/genetics , Rodentia
8.
Toxicol Appl Pharmacol ; 395: 114978, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32234387

ABSTRACT

Parasympathetic nervous system dysfunction is common in patients with liver disease. We have previously shown that muscarinic acetylcholine receptors (mAchRs) play an important role in the regulation of hepatic fibrosis and that the receptor agonists and antagonists affect hepatocyte proliferation. However, little is known about the impact of the different mAchR subtypes and associated signaling pathways on liver injury. Here, we treated the human liver cell line HL7702 with 10 mmol/L carbon tetrachloride (CCL4) to induce hepatocyte damage. We found that CCL4 treatment increased the protein levels of group I mAchRs (M1, M3, M5) but reduced the expression of group II mAchRs (M2, M4) and activated the Nrf2/ARE and MAPK signaling pathways. Although overexpression of M1, M3, or M5 led to hepatocyte damage with an intact Nrf2/ARE pathway, overexpression of M2 or M4 increased, and siRNA-mediated knockdown of either M2 or M4 decreased the protein levels of Nrf2 and its downstream target genes. Moreover, CCL4 treatment increased serum ALT levels more significantly, but only induced slight changes in the expression of mAchRs, NQO1 and HO1, while reducing the expression of M2 and M4 in liver tissues of Nrf2-/- mice compared to wild type mice. Our findings suggest that group II mAchRs, M2 and M4, activate the Nrf2/ARE signaling pathway, which regulates the expression of M2 and M4, to protect the liver from CCL4-induced injury.


Subject(s)
Antioxidant Response Elements/physiology , Liver Diseases/physiopathology , NF-E2-Related Factor 2/physiology , Receptor, Muscarinic M2/physiology , Receptor, Muscarinic M4/physiology , Receptors, Muscarinic/physiology , Signal Transduction/physiology , Animals , Carbon Tetrachloride/pharmacology , Cell Line , Chemical and Drug Induced Liver Injury/physiopathology , Gene Expression/drug effects , Gene Knockdown Techniques , Hepatocytes , Liver Diseases/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/deficiency , NF-E2-Related Factor 2/genetics , RNA, Small Interfering/pharmacology , Receptor, Muscarinic M2/genetics , Receptor, Muscarinic M4/genetics , Receptors, Muscarinic/genetics , Signal Transduction/drug effects
9.
PLoS One ; 14(12): e0226080, 2019.
Article in English | MEDLINE | ID: mdl-31805123

ABSTRACT

In early-onset generalized torsion dystonia, caused by a GAG deletion in TOR1A (DYT1), enhanced striatal cholinergic activity has been suggested to be critically involved. Previous studies have shown increased acetylcholine levels in the striatum of DYT1 knock-in (KI) mice. Ex vivo data indicated that muscarinic receptor antagonists normalize the activity of striatal cholinergic interneurons. Currently receptor subtype specific antagonists are developed for therapy, however, it is yet unknown whether the levels of targeted receptors are unaltered. In the present study, we firstly examined the expression of M1 and M4 receptors in DYT1 KI mice in comparison to wildtype mice. While no changes in mRNA were found in the motor cortex, the expression of M1 was higher in the striatum of DYT1 KI. However, M1 protein did not differ in striatum and cortex between the animal groups as shown by immunohistochemistry and western blot. M4 receptor protein, unaltered in the cortex, was slightly lower in lateral subparts of the striatum, but unchanged in somata of cholinergic interneurons and substance P immunoreactive projection neurons. Functional alterations of the cholinergic system and of aberrant striatal plasticity, demonstrated by previous studies, seem not to be related to overt changes in M1 and M4 expression. This critically informs the ongoing development of respective antagonists for therapy of dystonia.


Subject(s)
Dystonia Musculorum Deformans/genetics , Dystonia/metabolism , Gene Expression Regulation , Neostriatum/metabolism , Receptor, Muscarinic M1/genetics , Receptor, Muscarinic M4/genetics , Acetylcholine/metabolism , Animals , Disease Models, Animal , Dystonia/genetics , Feedback, Physiological , Gene Knock-In Techniques , Male , Mice , RNA, Messenger/genetics
10.
Sci Adv ; 5(4): eaaw1567, 2019 04.
Article in English | MEDLINE | ID: mdl-31001591

ABSTRACT

Designer receptors exclusively activated by designer drugs (DREADDs) derived from muscarinic receptors not only are a powerful tool to test causality in basic neuroscience but also are potentially amenable to clinical translation. A major obstacle, however, is that the widely used agonist clozapine N-oxide undergoes conversion to clozapine, which penetrates the blood-brain barrier but has an unfavorable side effect profile. Perlapine has been reported to activate DREADDs at nanomolar concentrations but is not approved for use in humans by the Food and Drug Administration or the European Medicines Agency, limiting its translational potential. Here, we report that the atypical antipsychotic drug olanzapine, widely available in various formulations, is a potent agonist of the human M4 muscarinic receptor-based DREADD, facilitating clinical translation of chemogenetics to treat central nervous system diseases.


Subject(s)
Designer Drugs/pharmacology , Olanzapine/chemistry , Olanzapine/pharmacology , Receptor, Muscarinic M4/agonists , Receptor, Muscarinic M4/genetics , Selective Serotonin Reuptake Inhibitors/chemistry , Selective Serotonin Reuptake Inhibitors/pharmacology , Computer Simulation , Designer Drugs/chemistry , High-Throughput Screening Assays , Humans , Signal Transduction
11.
Brain Behav ; 8(8): e01057, 2018 08.
Article in English | MEDLINE | ID: mdl-29978954

ABSTRACT

OBJECTIVES: M4 muscarinic receptors (MR) presumably play a role in motor coordination. Previous studies have shown different results depending on genetic background and number of backcrosses. However, no attention has been given to biorhythms. MATERIAL AND METHODS: We therefore analyzed biorhythms under a light/dark cycle obtained telemetrically in intact animals (activity, body temperature) in M4 KO mice growth on the C57Bl6 background using ChronosFit software. Studying pure effects of gene knockout in daily rhythms is especially important knowledge for pharmacological/behavioral studies in which drugs are usually tested in the morning. RESULTS: We show that M4 KO mice motor activity does not differ substantially from wild-type mice during light period while in the dark phase (mice active part of the day), the M4 KO mice reveal biorhythm changes in many parameters. Moreover, these differences are sex-dependent and are evident in females only. Mesor, night-day difference, and night value were doubled or tripled when comparing female KO versus male KO. Our in vitro autoradiography demonstrates that M4 MR proportion represents 24% in the motor cortex (MOCx), 30% in the somatosensory cortex, 50% in the striatum, 69% in the thalamus, and 48% in the intergeniculate leaflet (IGL). The M4 MR densities were negligible in the subparaventricular zone, the posterior hypothalamic area, and in the suprachiasmatic nuclei. CONCLUSIONS: We conclude that cholinergic signaling at M4 MR in brain structures such as striatum, MOCx, and probably with the important participation of IGL significantly control motor activity biorhythm. Animal activity differs in the light and dark phases, which should be taken into consideration when interpreting the results.


Subject(s)
Behavior, Animal/physiology , Brain/physiology , Motor Activity/genetics , Motor Activity/physiology , Periodicity , Receptor, Muscarinic M4/genetics , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Receptor, Muscarinic M4/deficiency , Sex Factors
12.
Alcohol Clin Exp Res ; 42(5): 879-888, 2018 05.
Article in English | MEDLINE | ID: mdl-29668112

ABSTRACT

BACKGROUND: Chronic alcohol intake leads to long-lasting changes in reward- and stress-related neuronal circuitry. The nucleus accumbens (NAc) is an integral component of this circuitry. Here, we investigate the effects of DREADDs (Designer Receptors Exclusively Activated by Designer Drugs) on neuronal activity in the NAc and binge-like drinking. METHODS: C57BL/6J mice were stereotaxically injected with AAV2 hSyn-HA hM3Dq, -hM4Di, or -eGFP bilaterally into NAc [core + shell, core or shell]. We measured clozapine-n-oxide (CNO)-induced changes in NAc activity and assessed binge-like ethanol (EtOH) or tastant/fluid intake in a limited access Drinking in the Dark (DID) schedule. RESULTS: We found that CNO increased NAc firing in hM3Dq positive cells and decreased firing in hM4Di cells, confirming the efficacy of these channels to alter neuronal activity both spatially and temporally. Increasing NAc core + shell activity decreased binge-like drinking without altering intake of other tastants. Increasing activity specifically in the NAc core reduced binge-like drinking, and decreasing activity in the NAc core increased drinking. Manipulation of NAc shell activity did not alter DID. Thus, we find that increasing activity in the entire NAc, or just the NAc core is sufficient to decrease binge drinking. CONCLUSIONS: We conclude that the reduction in EtOH drinking is not due to general malaise, altered perception of taste, or reduced calorie-seeking. Furthermore, we provide the first evidence for bidirectional control of NAc core and binge-like drinking. These findings could have promising implications for treatment.


Subject(s)
Alcohol Drinking , Clozapine/analogs & derivatives , Drinking/drug effects , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiology , Action Potentials/physiology , Adenoviridae , Animals , Clozapine/pharmacology , Female , Genetic Vectors , Mice , Mice, Transgenic , Receptor, Muscarinic M3/genetics , Receptor, Muscarinic M4/genetics
13.
J Pharmacol Exp Ther ; 365(2): 291-300, 2018 05.
Article in English | MEDLINE | ID: mdl-29523700

ABSTRACT

Mutations in the MeCP2 gene are responsible for the neurodevelopmental disorder Rett syndrome (RTT). MeCP2 is a DNA-binding protein whose abundance and ability to complex with histone deacetylase 3 is linked to the regulation of chromatin structure. Consequently, loss-of-function mutations in MeCP2 are predicted to have broad effects on gene expression. However, to date, studies in mouse models of RTT have identified a limited number of gene or pathway-level disruptions, and even fewer genes have been identified that could be considered amenable to classic drug discovery approaches. Here, we performed RNA sequencing (RNA-seq) on nine motor cortex and six cerebellar autopsy samples from RTT patients and controls. This approach identified 1887 significantly affected genes in the motor cortex and 2110 genes in the cerebellum, with a global trend toward increased expression. Pathway-level analysis identified enrichment in genes associated with mitogen-activated protein kinase signaling, long-term potentiation, and axon guidance. A survey of our RNA-seq results also identified a significant decrease in expression of the CHRM4 gene, which encodes a receptor [muscarinic acetylcholine receptor 4 (M4)] that is the subject of multiple large drug discovery efforts for schizophrenia and Alzheimer's disease. We confirmed that CHRM4 expression was decreased in RTT patients, and, excitingly, we demonstrated that M4 potentiation normalizes social and cognitive phenotypes in Mecp2+/- mice. This work provides an experimental paradigm in which translationally relevant targets can be identified using transcriptomics in RTT autopsy samples, back-modeled in Mecp2+/- mice, and assessed for preclinical efficacy using existing pharmacological tool compounds.


Subject(s)
Molecular Targeted Therapy , Receptor, Muscarinic M4/metabolism , Rett Syndrome/drug therapy , Rett Syndrome/genetics , Sequence Analysis, RNA , Animals , Autopsy , Cerebellum/metabolism , Humans , Mice , Motor Cortex/metabolism , Receptor, Muscarinic M4/genetics , Rett Syndrome/metabolism , Rett Syndrome/pathology
14.
Neuropharmacology ; 128: 492-502, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28729220

ABSTRACT

Although selective activation of the M1 muscarinic acetylcholine receptor (mAChR) subtype has been shown to improve cognitive function in animal models of neuropsychiatric disorders, recent evidence suggests that enhancing M4 mAChR function can also improve memory performance. Positive allosteric modulators (PAMs) targeting the M4 mAChR subtype have shown therapeutic potential for the treatment of multiple symptoms observed in schizophrenia, including positive and cognitive symptoms when assessed in acute preclinical dosing paradigms. Since the cholinergic system has been implicated in multiple stages of learning and memory, we evaluated the effects of repeated dosing with the highly selective M4 PAM VU0467154 on either acquisition and/or consolidation of learning and memory when dosed alone or after pharmacologic challenge with the N-methyl-d-aspartate subtype of glutamate receptors (NMDAR) antagonist MK-801. MK-801 challenge represents a well-documented preclinical model of NMDAR hypofunction that is thought to underlie some of the positive and cognitive symptoms observed in schizophrenia. In wildtype mice, 10-day, once-daily dosing of VU0467154 either prior to, or immediately after daily testing enhanced the rate of learning in a touchscreen visual pairwise discrimination task; these effects were absent in M4 mAChR knockout mice. Following a similar 10-day, once-daily dosing regimen of VU0467154, we also observed 1) improved acquisition of memory in a cue-mediated conditioned freezing paradigm, 2) attenuation of MK-801-induced disruptions in the acquisition of memory in a context-mediated conditioned freezing paradigm and 3) reversal of MK-801-induced hyperlocomotion. Comparable efficacy and plasma and brain concentrations of VU0467154 were observed after repeated dosing as those previously reported with an acute, single dose administration of this M4 PAM. Together, these studies are the first to demonstrate that cognitive enhancing and antipsychotic-like activity are not subject to the development of tolerance following repeated dosing with a selective M4 PAM in mice and further suggest that activation of M4 mAChRs may modulate both acquisition and consolidation of memory functions.


Subject(s)
Antipsychotic Agents/therapeutic use , Cognition Disorders/drug therapy , Pyridazines/therapeutic use , Receptor, Muscarinic M4/genetics , Thiophenes/therapeutic use , Allosteric Regulation/drug effects , Animals , Antipsychotic Agents/metabolism , Brain/drug effects , Cognition Disorders/etiology , Discrimination, Psychological/drug effects , Disease Models, Animal , Dizocilpine Maleate/toxicity , Dose-Response Relationship, Drug , Hyperkinesis/chemically induced , Hyperkinesis/drug therapy , Male , Memory Disorders/chemically induced , Memory Disorders/drug therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Pyridazines/metabolism , Schizophrenia/complications , Schizophrenia/drug therapy , Schizophrenia/genetics , Thiophenes/metabolism
15.
Neuron ; 96(6): 1358-1372.e4, 2017 12 20.
Article in English | MEDLINE | ID: mdl-29268098

ABSTRACT

Cholinergic regulation of dopaminergic inputs into the striatum is critical for normal basal ganglia (BG) function. This regulation of BG function is thought to be primarily mediated by acetylcholine released from cholinergic interneurons (ChIs) acting locally in the striatum. We now report a combination of pharmacological, electrophysiological, optogenetic, chemogenetic, and functional magnetic resonance imaging studies suggesting extra-striatal cholinergic projections from the pedunculopontine nucleus to the substantia nigra pars reticulata (SNr) act on muscarinic acetylcholine receptor subtype 4 (M4) to oppose cAMP-dependent dopamine receptor subtype 1 (D1) signaling in presynaptic terminals of direct pathway striatal spiny projections neurons. This induces a tonic inhibition of transmission at direct pathway synapses and D1-mediated activation of motor activity. These studies provide important new insights into the unique role of M4 in regulating BG function and challenge the prevailing hypothesis of the centrality of striatal ChIs in opposing dopamine regulation of BG output.


Subject(s)
Basal Ganglia/cytology , Cholinergic Neurons/physiology , Dopamine/metabolism , Pars Reticulata/physiology , Receptor, Muscarinic M4/metabolism , Acetylcholine/metabolism , Animals , Basal Ganglia/diagnostic imaging , Basal Ganglia/physiology , Channelrhodopsins/genetics , Channelrhodopsins/metabolism , Choline O-Acetyltransferase/metabolism , Cholinergic Agents/pharmacology , Cholinergic Neurons/drug effects , Dopamine/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Locomotion/drug effects , Locomotion/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurotransmitter Agents/pharmacology , Oxygen/blood , Pars Reticulata/cytology , Pars Reticulata/diagnostic imaging , Pedunculopontine Tegmental Nucleus/cytology , Receptor, Muscarinic M4/genetics , Receptors, Dopamine D1/genetics , Receptors, Dopamine D1/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
16.
PLoS One ; 12(12): e0188330, 2017.
Article in English | MEDLINE | ID: mdl-29211764

ABSTRACT

Rodent genomic alignment sequences support a 2-exon model for muscarinic M4 receptor. Using this model a novel N-terminal extension was discovered in the human muscarinic acetylcholine M4 receptor. An open reading frame was discovered in the human, mouse and rat with a common ATG (methionine start codon) that extended the N-terminus of the muscarinic acetylcholine M4 receptor subtype by 155 amino acids resulting in a longer variant. Transcriptional evidence for this splice variant was confirmed by RNA-Seq and RT-PCR experiments performed from human donor brain prefrontal cortices. We detected a human upstream exon indicating the translation of the mature longer M4 receptor transcript. The predicted size for the longer two-exon M4 receptor splice variant with the additional 155 amino acid N-terminal extension, designated M4L is 69.7 kDa compared to the 53 kDa canonical single exon M4 receptor (M4S). Western blot analysis from a mammalian overexpression system, and saturation radioligand binding with [3H]-NMS (N-methyl-scopolamine) demonstrated the expression of this new splice variant. Comparative pharmacological characterization between the M4L and M4S receptors revealed that both the orthosteric and allosteric binding sites for both receptors were very similar despite the addition of an N-terminal extension.


Subject(s)
RNA Splicing , Receptor, Muscarinic M4/metabolism , Animals , Base Sequence , Binding Sites , Binding, Competitive , Exons , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Mice , Polymerase Chain Reaction , Prefrontal Cortex/metabolism , Radioligand Assay , Rats , Receptor, Muscarinic M4/genetics , Sequence Analysis, RNA , Sequence Homology, Amino Acid
17.
Behav Brain Res ; 329: 75-83, 2017 06 30.
Article in English | MEDLINE | ID: mdl-28442355

ABSTRACT

Muscarinic M1/M4 receptor stimulation can reduce abuse-related effects of cocaine and may represent avenues for treating cocaine addiction. Muscarinic antagonists can mimic and enhance effects of cocaine, including discriminative stimulus (SD) effects, but the receptor subtypes mediating those effects are not known. A better understanding of the complex cocaine/muscarinic interactions is needed to evaluate and develop potential muscarinic-based medications. Here, knockout mice lacking M1, M2, or M4 receptors (M1-/-, M2-/-, M4-/-), as well as control wild-type mice and outbred Swiss-Webster mice, were trained to discriminate 10mg/kg cocaine from saline. Muscarinic receptor antagonists with no subtype selectivity (scopolamine), or preferential affinity at the M1, M2, or M4 subtype (telenzepine, trihexyphenidyl; methoctramine, AQ-RA 741; tropicamide) were tested alone and in combination with cocaine. In intact animals, antagonists with high affinity at M1/M4 receptors partially substituted for cocaine and increased the SD effect of cocaine, while M2-preferring antagonists did not substitute, and reduced the SD effect of cocaine. The cocaine-like effects of scopolamine were absent in M1-/- mice. The cocaine SD attenuating effects of methoctramine were absent in M2-/- mice and almost absent in M1-/- mice. The findings indicate that the cocaine-like SD effects of muscarinic antagonists are primarily mediated through M1 receptors, with a minor contribution of M4 receptors. The data also support our previous findings that stimulation of M1 receptors and M4 receptors can each attenuate the SD effect of cocaine, and show that this can also be achieved by blocking M2 autoreceptors, likely via increased acetylcholine release.


Subject(s)
Cocaine/administration & dosage , Conditioning, Operant/drug effects , Diamines/pharmacology , Discrimination, Psychological/drug effects , Dopamine Uptake Inhibitors/administration & dosage , Muscarinic Antagonists/pharmacology , Receptors, Muscarinic/deficiency , Analysis of Variance , Animals , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Muscarinic M1/genetics , Receptor, Muscarinic M1/metabolism , Receptor, Muscarinic M2/genetics , Receptor, Muscarinic M2/metabolism , Receptor, Muscarinic M4/genetics , Receptor, Muscarinic M4/metabolism , Receptors, Muscarinic/genetics , Trihexyphenidyl/pharmacology , Tropicamide/pharmacology
18.
Neuropharmacology ; 117: 376-386, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28257887

ABSTRACT

Dopamine- and cAMP-regulated phosphoprotein of 32 kDa (DARPP-32) play essential roles in dopamine (DA) transmission in the striatum. It is suggested that a link exists between muscarinic acetylcholine receptors (mAChRs) and DA/DARPP-32 signaling, but the molecular mechanisms mediating this relationship have not been elucidated. The predominant mAChRs subtypes in the striatum are M1 and M4. In this study, we investigated the functions of these two receptors, particularly M4, in regulating cAMP production and DARPP-32 phosphorylation in rat striatal medium spiny neurons (MSNs). We used time-resolved fluorescence resonance energy transfer, immunofluorescence confocal microscopy, and western blot assays. In cultured intact MSNs, we confirmed that muscarinic M1 and M4 receptors were highly expressed. Notably, M4 receptors were co-expressed with D1 receptors in only a portion of the cultured MSNs. The nonselective muscarinic agonist oxotremorine M (OX) slightly enhanced cAMP production, but this effect was independent of M1 or M4 receptors. However, OX directly participated in DARPP-32 phosphorylation, phosphorylating DARPP-32 at Thr75 (the CDK5 site) and concomitantly de-phosphorylating DARPP-32 at Thr34 (the PKA site) in virtually cultured MSNs, whereas APO phosphorylated DARPP-32 at both Thr34 and Thr75. The OX-induced time-dependent increase in DARPP-32 phosphorylation at Thr75 was accompanied by increased p35 and CDK5 activity. Specifically, elevated immunoreactivity for phospho-DARPP-32-Thr75 and p35 was detected in M4 receptor-expressing MSNs. Both genetic knockdown and pharmacologic inhibition of M4 receptors with MT3, an M4 receptor-selective antagonist, decreased the OX-induced DARPP-32-Thr75 phosphorylation in MSNs. These results indicate that the M4 muscarinic receptor plays a critical role in modulating phosphorylation of DARPP-32-Thr75 in MSNs. The results suggest that M4 receptor activation acts antagonistically with dopamine D1-like receptors within the striatum, and indicate that M4 receptors may be a potential target for the treatment of Parkinson's disease and other relevant central nervous system disorders.


Subject(s)
Corpus Striatum/drug effects , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Muscarinic Agonists/pharmacology , Neurons/drug effects , Oxotremorine/pharmacology , Receptor, Muscarinic M4/metabolism , Animals , Cells, Cultured , Corpus Striatum/cytology , Corpus Striatum/metabolism , Cyclic AMP/metabolism , Gene Knockdown Techniques , Muscarinic Antagonists/pharmacology , Neurons/cytology , Neurons/metabolism , Phosphorylation/drug effects , Rats, Sprague-Dawley , Receptor, Muscarinic M1/metabolism , Receptor, Muscarinic M4/agonists , Receptor, Muscarinic M4/antagonists & inhibitors , Receptor, Muscarinic M4/genetics , Receptors, Dopamine D1/metabolism
19.
PLoS One ; 11(5): e0154374, 2016.
Article in English | MEDLINE | ID: mdl-27145133

ABSTRACT

The hippocampus is critical for the storage of new autobiographical experiences as memories. Following an initial encoding stage in the hippocampus, memories undergo a process of systems-level consolidation, which leads to greater stability through time and an increased reliance on neocortical areas for retrieval. The extent to which the retrieval of these consolidated memories still requires the hippocampus is unclear, as both spared and severely degraded remote memory recall have been reported following post-training hippocampal lesions. One difficulty in definitively addressing the role of the hippocampus in remote memory retrieval is the precision with which the entire volume of the hippocampal region can be inactivated. To address this issue, we used Designer Receptors Exclusively Activated by Designer Drugs (DREADDs), a chemical-genetic tool capable of highly specific neuronal manipulation over large volumes of brain tissue. We find that remote (>7 weeks after acquisition), but not recent (1-2 days after acquisition) contextual fear memories can be recalled after injection of the DREADD agonist (CNO) in animals expressing the inhibitory DREADD in the entire hippocampus. Our data demonstrate a time-dependent role of the hippocampus in memory retrieval, supporting the standard model of systems consolidation.


Subject(s)
Hippocampus/physiology , Mental Recall/physiology , Animals , Clozapine/analogs & derivatives , Clozapine/metabolism , Clozapine/pharmacology , Designer Drugs/metabolism , Fear/physiology , Hippocampus/drug effects , Humans , Memory, Long-Term/drug effects , Memory, Long-Term/physiology , Mental Recall/drug effects , Mice , Mice, Inbred C57BL , Receptor, Muscarinic M4/agonists , Receptor, Muscarinic M4/genetics , Receptor, Muscarinic M4/metabolism , Receptors, Drug/agonists , Receptors, Drug/genetics , Receptors, Drug/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Time Factors
20.
J Neurophysiol ; 115(6): 3008-17, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26961109

ABSTRACT

GABAergic interneurons are positioned to powerfully influence the dynamics of neural activity, yet the interneuron-mediated circuit mechanisms that control spontaneous and evoked neocortical activity remains elusive. Vasoactive intestinal peptide (VIP+) interneurons are a specialized cell class which synapse specifically on other interneurons, potentially serving to facilitate increases in cortical activity. In this study, using in vivo Ca(2+) imaging, we describe the interaction between local network activity and VIP+ cells and determine their role in modulating neocortical activity in mouse visual cortex. VIP+ cells were active across brain states including locomotion, nonlocomotion, visual stimulation, and under anesthesia. VIP+ activity correlated most clearly with the mean level of population activity of nearby excitatory neurons during all brain states, suggesting VIP+ cells enable high-excitability states in the cortex. The pharmacogenetic blockade of VIP+ cell output reduced network activity during locomotion, nonlocomotion, anesthesia, and visual stimulation, suggesting VIP+ cells exert a state-independent facilitation of neural activity in the cortex. Collectively, our findings demonstrate that VIP+ neurons have a causal role in the generation of high-activity regimes during spontaneous and stimulus evoked neocortical activity.


Subject(s)
Interneurons/physiology , Neocortex/physiology , Neural Inhibition/physiology , Vasoactive Intestinal Peptide/metabolism , Visual Cortex/cytology , Animals , Calcium/metabolism , Clozapine/analogs & derivatives , Clozapine/pharmacology , Female , Interneurons/drug effects , Locomotion/physiology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Transgenic , Nerve Net/physiology , Neural Inhibition/drug effects , Neural Inhibition/genetics , Photic Stimulation , Receptor, Muscarinic M4/genetics , Receptor, Muscarinic M4/metabolism , Somatostatin/genetics , Somatostatin/metabolism , Transduction, Genetic , Vasoactive Intestinal Peptide/genetics , gamma-Aminobutyric Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...