Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Schizophr Bull ; 49(4): 881-892, 2023 07 04.
Article in English | MEDLINE | ID: mdl-36454006

ABSTRACT

BACKGROUND: It remains unknown why ~30% of patients with psychotic disorders fail to respond to treatment. Previous genomic investigations of treatment-resistant psychosis have been inconclusive, but some evidence suggests a possible link between rare disease-associated copy number variants (CNVs) and worse clinical outcomes in schizophrenia. Here, we identified schizophrenia-associated CNVs in patients with treatment-resistant psychotic symptoms and then compared the prevalence of these CNVs to previously published schizophrenia cases not selected for treatment resistance. METHODS: CNVs were identified using chromosomal microarray (CMA) and whole exome sequencing (WES) in 509 patients with treatment-resistant psychosis (a lack of clinical response to ≥3 adequate antipsychotic medication trials over at least 5 years of psychiatric hospitalization). Prevalence of schizophrenia-associated CNVs in this sample was compared to that in a previously published large schizophrenia cohort study. RESULTS: Integrating CMA and WES data, we identified 47 cases (9.2%) with at least one CNV of known or possible neuropsychiatric risk. 4.7% (n = 24) carried a known neurodevelopmental risk CNV. The prevalence of well-replicated schizophrenia-associated CNVs was 4.1%, with duplications of the 16p11.2 and 15q11.2-q13.1 regions, and deletions of the 22q11.2 chromosomal region as the most frequent CNVs. Pairwise loci-based analysis identified duplications of 15q11.2-q13.1 to be independently associated with treatment resistance. CONCLUSIONS: These findings suggest that CNVs may uniquely impact clinical phenotypes beyond increasing risk for schizophrenia and may potentially serve as biological entry points for studying treatment resistance. Further investigation will be necessary to elucidate the spectrum of phenotypic characteristics observed in adult psychiatric patients with disease-associated CNVs.


Subject(s)
Psychotic Disorders , Schizophrenia , Humans , Cohort Studies , DNA Copy Number Variations/genetics , Prevalence , Psychotic Disorders/drug therapy , Psychotic Disorders/epidemiology , Psychotic Disorders/genetics , Schizophrenia/drug therapy , Schizophrenia/epidemiology , Schizophrenia/genetics , Genetic Predisposition to Disease
2.
Mol Psychiatry ; 28(1): 475-482, 2023 01.
Article in English | MEDLINE | ID: mdl-36380236

ABSTRACT

Tandem repeat expansions (TREs) are associated with over 60 monogenic disorders and have recently been implicated in complex disorders such as cancer and autism spectrum disorder. The role of TREs in schizophrenia is now emerging. In this study, we have performed a genome-wide investigation of TREs in schizophrenia. Using genome sequence data from 1154 Swedish schizophrenia cases and 934 ancestry-matched population controls, we have detected genome-wide rare (<0.1% population frequency) TREs that have motifs with a length of 2-20 base pairs. We find that the proportion of individuals carrying rare TREs is significantly higher in the schizophrenia group. There is a significantly higher burden of rare TREs in schizophrenia cases than in controls in genic regions, particularly in postsynaptic genes, in genes overlapping brain expression quantitative trait loci, and in brain-expressed genes that are differentially expressed between schizophrenia cases and controls. We demonstrate that TRE-associated genes are more constrained and primarily impact synaptic and neuronal signaling functions. These results have been replicated in an independent Canadian sample that consisted of 252 schizophrenia cases of European ancestry and 222 ancestry-matched controls. Our results support the involvement of rare TREs in schizophrenia etiology.


Subject(s)
Autism Spectrum Disorder , Schizophrenia , Humans , Schizophrenia/genetics , Genome-Wide Association Study , Canada , Gene Frequency , Genetic Predisposition to Disease/genetics
4.
J Neurosci ; 41(45): 9466-9481, 2021 11 10.
Article in English | MEDLINE | ID: mdl-34642214

ABSTRACT

TSNARE1, which encodes the protein tSNARE1, is a high-confidence gene candidate for schizophrenia risk, but nothing is known about its cellular or physiological function. We identified the major gene products of TSNARE1 and their cytoplasmic localization and function in endosomal trafficking in cortical neurons. We validated three primary isoforms of TSNARE1 expressed in human brain, all of which encode a syntaxin-like Qa SNARE domain. RNA-sequencing data from adult and fetal human brain suggested that the majority of tSNARE1 lacks a transmembrane domain that is thought to be necessary for membrane fusion. Biochemical data demonstrate that tSNARE1 can compete with Stx12 for incorporation into an endosomal SNARE complex, supporting its possible role as an inhibitory SNARE. Live-cell imaging in cortical neurons from mice of both sexes demonstrated that brain tSNARE1 isoforms localized to the endosomal network. The most abundant brain isoform, tSNARE1c, localized most frequently to Rab7+ late endosomes, and endogenous tSNARE1 displayed a similar localization in human neural progenitor cells and neuroblastoma cells. In mature rat neurons from both sexes, tSNARE1 localized to the dendritic shaft and dendritic spines, supporting a role for tSNARE1 at the postsynapse. Expression of either tSNARE1b or tSNARE1c, which differ only in their inclusion or exclusion of an Myb-like domain, delayed the trafficking of the dendritic endosomal cargo Nsg1 into late endosomal and lysosomal compartments. These data suggest that tSNARE1 regulates endosomal trafficking in cortical neurons, likely by negatively regulating early endosomal to late endosomal trafficking.SIGNIFICANCE STATEMENT Schizophrenia is a severe and polygenic neuropsychiatric disorder. Understanding the functions of high-confidence candidate genes is critical toward understanding how their dysfunction contributes to schizophrenia pathogenesis. TSNARE1 is one of the high-confidence candidate genes for schizophrenia risk, yet nothing was known about its cellular or physiological function. Here we describe the major isoforms of TSNARE1 and their cytoplasmic localization and function in the endosomal network in cortical neurons. Our results are consistent with the hypothesis that the majority of brain tSNARE1 acts as a negative regulator to endolysosomal trafficking.


Subject(s)
Cerebral Cortex/metabolism , Endosomes/metabolism , Neurons/metabolism , SNARE Proteins/metabolism , Schizophrenia/metabolism , Animals , Female , Humans , Male , Mice , Mice, Inbred C57BL , Protein Isoforms/metabolism , Protein Transport/physiology , Rats , Rats, Sprague-Dawley
5.
Schizophr Res ; 224: 195-197, 2020 10.
Article in English | MEDLINE | ID: mdl-32943312

ABSTRACT

The 3q29 deletion is a rare copy number variant associated with neurodevelopmental and psychiatric disorders, including a >40-fold increased risk for schizophrenia. Current understanding of the clinical phenotype is derived primarily from published cases of patients in childhood or early adolescence. Symptoms include mild to moderate learning disability, developmental delay, facial dysmorphism, microcephaly, ocular disorders, and gastrointestinal abnormalities. There is, however, a lack of detailed longitudinal case studies describing 3q29 deletion syndrome in adults with psychosis. In this case report, we describe the lifetime clinical portrait of a 57-year-old woman with 3q29 deletion syndrome, treatment-resistant psychotic symptoms, multiple medical comorbidities, and a previously unreported co-occurrence of early-onset dementia.


Subject(s)
Dementia , Intellectual Disability , Psychotic Disorders , Adolescent , Adult , Child , Chromosome Deletion , Developmental Disabilities/genetics , Female , Humans , Intellectual Disability/genetics , Middle Aged , Psychotic Disorders/genetics
6.
Nat Commun ; 11(1): 1842, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32296054

ABSTRACT

Despite considerable progress in schizophrenia genetics, most findings have been for large rare structural variants and common variants in well-imputed regions with few genes implicated from exome sequencing. Whole genome sequencing (WGS) can potentially provide a more complete enumeration of etiological genetic variation apart from the exome and regions of high linkage disequilibrium. We analyze high-coverage WGS data from 1162 Swedish schizophrenia cases and 936 ancestry-matched population controls. Our main objective is to evaluate the contribution to schizophrenia etiology from a variety of genetic variants accessible to WGS but not by previous technologies. Our results suggest that ultra-rare structural variants that affect the boundaries of topologically associated domains (TADs) increase risk for schizophrenia. Alterations in TAD boundaries may lead to dysregulation of gene expression. Future mechanistic studies will be needed to determine the precise functional effects of these variants on biology.


Subject(s)
Genome-Wide Association Study/methods , Schizophrenia/genetics , Brain/metabolism , Exome/genetics , Genome, Human/genetics , High-Throughput Nucleotide Sequencing , Humans , Male , Nervous System/metabolism , Quality Control , Sequence Analysis, DNA
7.
Transl Psychiatry ; 10(1): 42, 2020 01 28.
Article in English | MEDLINE | ID: mdl-32066678

ABSTRACT

The 15q11.2 BP1-BP2 (Burnside-Butler) deletion is a rare copy number variant impacting four genes (NIPA1, NIPA2, CYFIP1, and TUBGCP5), and carries increased risks for developmental delay, intellectual disability, and neuropsychiatric disorders (attention-deficit/hyperactivity disorder, autism, and psychosis). In this case report (supported by extensive developmental information and medication history), we present the complex clinical portrait of a 44-year-old woman with 15q11.2 BP1-BP2 deletion syndrome and chronic, treatment-resistant psychotic symptoms who has resided nearly her entire adult life in a long-term state psychiatric institution. Diagnostic and treatment implications are discussed.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Intellectual Disability , Psychotic Disorders , Adult , Chromosome Aberrations , Chromosomes, Human, Pair 15/genetics , DNA Copy Number Variations , Female , Humans , Intellectual Disability/genetics , Psychotic Disorders/genetics
8.
North Carol Law Rev ; 97(5): 1359-1398, 2019.
Article in English | MEDLINE | ID: mdl-31871365

ABSTRACT

Gene-environment interactions play a key role in how psychiatric disorders manifest and develop. Psychiatric genetics researchers are making progress in identifying genomic correlates of many disorders. And recently, the field of genetics has given rise to a technology that many claim will revolutionize the biological sciences and propel the field into a transformative phase: the powerful gene-editing tool known as CRISPR-Cas9. This Article illustrates which psychiatric conditions are likely to make an attractive target for CRISPR as the technology evolves and CRISPR therapies becomes a viable tool to manage or prevent disorders in a clinical setting. We examine the potential scientific and clinical challenges of applying CRISPR in the mental health context, along with the regulatory, ethical, and legal issues that might arise as a consequence of these applications.

10.
Sci Rep ; 8(1): 3840, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29497149

ABSTRACT

Clozapine-N-oxide (CNO) has long been the ligand of choice for selectively activating Designer Receptors Exclusively Activated by Designer Drugs (DREADDs). However, recent studies have challenged the long-held assertion that CNO is otherwise pharmacologically inert. The present study aimed to 1) determine whether CNO is reverse-metabolized to its parent compound clozapine in mice (as has recently been reported in rats), and 2) determine whether CNO exerts clozapine-like interoceptive stimulus effects in rats and/or mice. Following administration of 10.0 mg/kg CNO, pharmacokinetic analyses replicated recent reports of back-conversion to clozapine in rats and revealed that this phenomenon also occurs in mice. In rats and mice trained to discriminate 1.25 mg/kg clozapine from vehicle, CNO (1.0-20.0 mg/kg) produced partial substitution for the clozapine stimulus on average, with full substitution being detected in some individual animals of both species at doses frequently used to activate DREADDs. The present demonstration that CNO is converted to clozapine and exerts clozapine-like behavioral effects in both mice and rats further emphasizes the need for appropriate control groups in studies employing DREADDs, and highlights the utility of the drug discrimination procedure as a tool with which to screen the off-target effects of novel DREADD agonists.


Subject(s)
Clozapine/analogs & derivatives , Designer Drugs/pharmacology , Designer Drugs/pharmacokinetics , Animals , Clozapine/administration & dosage , Clozapine/metabolism , Clozapine/pharmacology , Designer Drugs/metabolism , Female , Male , Mice , Mice, Inbred Strains , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
11.
PLoS One ; 11(3): e0150602, 2016.
Article in English | MEDLINE | ID: mdl-26963248

ABSTRACT

RATIONALE: The sacred lotus (Nelumbo nucifera) contains many phytochemicals and has a history of human use. To determine which compounds may be responsible for reported psychotropic effects, we used in silico predictions of the identified phytochemicals. Nuciferine, an alkaloid component of Nelumbo nucifera and Nymphaea caerulea, had a predicted molecular profile similar to antipsychotic compounds. Our study characterizes nuciferine using in vitro and in vivo pharmacological assays. METHODS: Nuciferine was first characterized in silico using the similarity ensemble approach, and was followed by further characterization and validation using the Psychoactive Drug Screening Program of the National Institute of Mental Health. Nuciferine was then tested in vivo in the head-twitch response, pre-pulse inhibition, hyperlocomotor activity, and drug discrimination paradigms. RESULTS: Nuciferine shares a receptor profile similar to aripiprazole-like antipsychotic drugs. Nuciferine was an antagonist at 5-HT2A, 5-HT2C, and 5-HT2B, an inverse agonist at 5-HT7, a partial agonist at D2, D5 and 5-HT6, an agonist at 5-HT1A and D4 receptors, and inhibited the dopamine transporter. In rodent models relevant to antipsychotic drug action, nuciferine blocked head-twitch responses and discriminative stimulus effects of a 5-HT2A agonist, substituted for clozapine discriminative stimulus, enhanced amphetamine induced locomotor activity, inhibited phencyclidine (PCP)-induced locomotor activity, and rescued PCP-induced disruption of prepulse inhibition without induction of catalepsy. CONCLUSIONS: The molecular profile of nuciferine was similar but not identical to that shared with several approved antipsychotic drugs suggesting that nuciferine has atypical antipsychotic-like actions.


Subject(s)
Antipsychotic Agents/chemistry , Antipsychotic Agents/pharmacology , Aporphines/chemistry , Aporphines/pharmacology , Serotonin 5-HT1 Receptor Agonists/chemistry , Serotonin 5-HT1 Receptor Agonists/pharmacology , Animals , Behavior, Animal/drug effects , HEK293 Cells , Humans , Mice , Receptors, Dopamine D4/agonists
12.
Neuropsychopharmacology ; 41(5): 1404-15, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26383016

ABSTRACT

Elucidating how the brain's serotonergic network mediates diverse behavioral actions over both relatively short (minutes-hours) and long period of time (days-weeks) remains a major challenge for neuroscience. Our relative ignorance is largely due to the lack of technologies with robustness, reversibility, and spatio-temporal control. Recently, we have demonstrated that our chemogenetic approach (eg, Designer Receptors Exclusively Activated by Designer Drugs (DREADDs)) provides a reliable and robust tool for controlling genetically defined neural populations. Here we show how short- and long-term activation of dorsal raphe nucleus (DRN) serotonergic neurons induces robust behavioral responses. We found that both short- and long-term activation of DRN serotonergic neurons induce antidepressant-like behavioral responses. However, only short-term activation induces anxiogenic-like behaviors. In parallel, these behavioral phenotypes were associated with a metabolic map of whole brain network activity via a recently developed non-invasive imaging technology DREAMM (DREADD Associated Metabolic Mapping). Our findings reveal a previously unappreciated brain network elicited by selective activation of DRN serotonin neurons and illuminate potential therapeutic and adverse effects of drugs targeting DRN neurons.


Subject(s)
Anxiety/physiopathology , Depression/physiopathology , Dorsal Raphe Nucleus/physiology , Serotonergic Neurons/physiology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/drug effects , Brain/metabolism , Brain/physiology , Circadian Rhythm , Designer Drugs/administration & dosage , Dorsal Raphe Nucleus/drug effects , Dorsal Raphe Nucleus/metabolism , Male , Mice , Mice, Transgenic , Serotonergic Neurons/drug effects , Serotonergic Neurons/metabolism , Time Factors
13.
Nature ; 527(7579): 477-83, 2015 Nov 26.
Article in English | MEDLINE | ID: mdl-26550826

ABSTRACT

At least 120 non-olfactory G-protein-coupled receptors in the human genome are 'orphans' for which endogenous ligands are unknown, and many have no selective ligands, hindering the determination of their biological functions and clinical relevance. Among these is GPR68, a proton receptor that lacks small molecule modulators for probing its biology. Using yeast-based screens against GPR68, here we identify the benzodiazepine drug lorazepam as a non-selective GPR68 positive allosteric modulator. More than 3,000 GPR68 homology models were refined to recognize lorazepam in a putative allosteric site. Docking 3.1 million molecules predicted new GPR68 modulators, many of which were confirmed in functional assays. One potent GPR68 modulator, ogerin, suppressed recall in fear conditioning in wild-type but not in GPR68-knockout mice. The same approach led to the discovery of allosteric agonists and negative allosteric modulators for GPR65. Combining physical and structure-based screening may be broadly useful for ligand discovery for understudied and orphan GPCRs.


Subject(s)
Benzyl Alcohols/chemistry , Benzyl Alcohols/pharmacology , Drug Discovery , Lorazepam/chemistry , Lorazepam/pharmacology , Receptors, G-Protein-Coupled/metabolism , Triazines/chemistry , Triazines/pharmacology , Allosteric Regulation/drug effects , Allosteric Site , Animals , Anti-Anxiety Agents/analysis , Anti-Anxiety Agents/chemistry , Anti-Anxiety Agents/metabolism , Anti-Anxiety Agents/pharmacology , Benzyl Alcohols/analysis , Benzyl Alcohols/metabolism , Conditioning, Classical , Fear , Female , HEK293 Cells , Humans , Ligands , Lorazepam/analysis , Lorazepam/metabolism , Male , Memory/drug effects , Mice , Mice, Knockout , Models, Molecular , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/deficiency , Signal Transduction/drug effects , Triazines/analysis , Triazines/metabolism
15.
Neuropsychopharmacology ; 38(5): 854-62, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23303063

ABSTRACT

Here, we describe a newly generated transgenic mouse in which the Gs DREADD (rM3Ds), an engineered G protein-coupled receptor, is selectively expressed in striatopallidal medium spiny neurons (MSNs). We first show that in vitro, rM3Ds can couple to Gαolf and induce cAMP accumulation in cultured neurons and HEK-T cells. The rM3Ds was then selectively and stably expressed in striatopallidal neurons by creating a transgenic mouse in which an adenosine2A (adora2a) receptor-containing bacterial artificial chromosome was employed to drive rM3Ds expression. In the adora2A-rM3Ds mouse, activation of rM3Ds by clozapine-N-oxide (CNO) induces DARPP-32 phosphorylation, consistent with the known consequence of activation of endogenous striatal Gαs-coupled GPCRs. We then tested whether CNO administration would produce behavioral responses associated with striatopallidal Gs signaling and in this regard CNO dose-dependently decreases spontaneous locomotor activity and inhibits novelty induced locomotor activity. Last, we show that CNO prevented behavioral sensitization to amphetamine and increased AMPAR/NMDAR ratios in transgene-expressing neurons of the nucleus accumbens shell. These studies demonstrate the utility of adora2a-rM3Ds transgenic mice for the selective and noninvasive modulation of Gαs signaling in specific neuronal populations in vivo.This unique tool provides a new resource for elucidating the roles of striatopallidal MSN Gαs signaling in other neurobehavioral contexts.


Subject(s)
Corpus Striatum/cytology , Cyclic AMP/metabolism , GTP-Binding Protein alpha Subunits, Gs/genetics , Neurons/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Muscarinic/genetics , Adrenergic Uptake Inhibitors/pharmacology , Amphetamine/pharmacology , Animals , Animals, Newborn , Clozapine/analogs & derivatives , Clozapine/pharmacology , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Green Fluorescent Proteins/genetics , Locomotion/drug effects , Locomotion/genetics , Luminescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation/drug effects , Phosphorylation/genetics , Receptor, Adenosine A2A/genetics , Receptors, Dopamine D1/genetics , Receptors, Dopamine D2/genetics , Red Fluorescent Protein
16.
Brain Res ; 1511: 6-20, 2013 May 20.
Article in English | MEDLINE | ID: mdl-23063887

ABSTRACT

Pharmacology, in its broadest interpretation, is defined as the study of the interaction between physiological entities and drugs. In modern neuropsychopharmacology, this interaction is viewed as the drug itself on one side and signal transducer (receptor), the signal transduction cascade (effector proteins, second messengers), the cellular response (transcriptional regulation, activity modulation), the organ response (brain circuitry modulation), and, finally, the whole organism response (behavior) on the other. In other words, pharmacology has structured itself around the idea that the exogenous molecule (the drug) encodes a "signal" leading to everything on the other side including, in extreme renditions, a physiological response. The inference is that engaging a particular signal transduction pathway in a defined cell type leads inexorably to a prototypic physiological response. Thus, for instance, serotonergic activation of 5-HT(2A) receptors in rat aortic smooth muscle cells leads to an increase in intracellular Ca(++) (via IP3 release) and smooth muscle contraction (Roth et al., 1986). Here, we suggest that the invention of synthetic ligand--GPCR pairs (aka DREADDs, RASSLS, 'pharmacogenetics') permits the study of pharmacology using a shifted equation: more of the signal transduction elements moved to the left and, subsequently, under experimental control. For the purposes of disambiguation and to clarify this new interpretation as a creation of pharmacological manipulation, we present the term pharmacosynthetics to describe what has heretofore been called pharmacogenetics or chemicogenetics. This review discusses this new interpretation and reviews recent applications of the technology and considerations of the approach. This article is part of a Special Issue entitled Optogenetics (7th BRES).


Subject(s)
Neurons/drug effects , Pharmacogenetics , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Animals , Humans , Models, Biological , Neurons/physiology , Receptors, G-Protein-Coupled/metabolism
17.
Article in English | MEDLINE | ID: mdl-22207840
18.
J Pharmacol Exp Ther ; 339(1): 99-105, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21737536

ABSTRACT

Dysregulation of the 5-HT(2A) receptor is implicated in both the etiology and treatment of schizophrenia. Although the essential role of 5-HT(2A) receptors in atypical antipsychotic drug actions is widely accepted, the contribution of 5-HT(2A) down-regulation to their efficacy is not known. We hypothesized that down-regulation of cortical 5-HT(2A) receptors contributes to the therapeutic action of atypical antipsychotic drugs. To test this hypothesis, we assessed the effect of chronically administered antipsychotics (clozapine, olanzapine, and haloperidol) and several 5-HT(2A) antagonists [ketanserin, altanserin, α-(2,3-dimethoxyphenyl)-1-[2-(4-fluorophenylethyl)]-4-piperidinemethanol (M100907), α-phenyl-1-(2-phenylethyl)-4-piperidinemethano (M11939), 4-[(2Z)-3-{[2-(dimethylamino)ethoxy]amino}-3-(2-fluorophenyl)prop-2-en-1-ylidene]cyclohexa-2,5-dien-1-one (SR46349B), and pimavanserin], on the phencyclidine (PCP)-induced hyperlocomotor response and cortical 5-HT(2A) receptor levels in C57BL/6J mice. Clozapine and olanzapine, but not haloperidol, induced receptor down-regulation and attenuated PCP-induced locomotor responses. Of the selective 5-HT(2A) antagonists tested, only ketanserin caused significant receptor protein down-regulation, whereas SR46349B up-regulated 5-HT(2A) receptors and potentiated PCP-hyperlocomotion; the other 5-HT(2A) receptor antagonists were without effect. The significance of these findings with respect to atypical antipsychotic drug action is discussed.


Subject(s)
Receptor, Serotonin, 5-HT2A/biosynthesis , Receptor, Serotonin, 5-HT2A/drug effects , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Animals , Behavior, Animal/drug effects , Benzodiazepines/pharmacology , Blotting, Western , Clozapine/pharmacology , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Haloperidol/pharmacology , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Olanzapine , Phencyclidine/pharmacology , Radioligand Assay , Receptor, Serotonin, 5-HT2A/genetics , Serotonin Receptor Agonists/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Stereotyped Behavior/drug effects , Up-Regulation/drug effects
19.
Neuropsychopharmacology ; 36(3): 638-51, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21048700

ABSTRACT

Clozapine, by virtue of its absence of extrapyramidal side effects and greater efficacy, revolutionized the treatment of schizophrenia, although the mechanisms underlying this exceptional activity remain controversial. Combining an unbiased cheminformatics and physical screening approach, we evaluated clozapine's activity at >2350 distinct molecular targets. Clozapine, and the closely related atypical antipsychotic drug olanzapine, interacted potently with a unique spectrum of molecular targets. This distinct pattern, which was not shared with the typical antipsychotic drug haloperidol, suggested that the serotonergic neuronal system was a key determinant of clozapine's actions. To test this hypothesis, we used pet1(-/-) mice, which are deficient in serotonergic presynaptic markers. We discovered that the antipsychotic-like properties of the atypical antipsychotic drugs clozapine and olanzapine were abolished in a pharmacological model that mimics NMDA-receptor hypofunction in pet1(-/-) mice, whereas haloperidol's efficacy was unaffected. These results show that clozapine's ability to normalize NMDA-receptor hypofunction, which is characteristic of schizophrenia, depends on an intact presynaptic serotonergic neuronal system.


Subject(s)
Clozapine/pharmacology , Neurons/cytology , Presynaptic Terminals/drug effects , Serotonin Antagonists/pharmacology , Serotonin/metabolism , Acoustic Stimulation/methods , Action Potentials/drug effects , Adrenergic Uptake Inhibitors/pharmacology , Amphetamines/pharmacology , Animals , Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Checkpoint Kinase 2 , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Ketanserin/pharmacokinetics , Lysine/analogs & derivatives , Lysine/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 3/metabolism , Motor Activity/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Neurons/drug effects , Neurons/physiology , Patch-Clamp Techniques/methods , Phencyclidine/pharmacology , Protein Binding/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/deficiency , Radioligand Assay/methods , Raphe Nuclei/cytology , Receptor, Serotonin, 5-HT1A/metabolism , Reflex, Startle/drug effects , Reflex, Startle/physiology , Stereotyped Behavior/drug effects , Tritium/pharmacokinetics , Tryptophan Hydroxylase/metabolism
20.
Mol Biosyst ; 6(8): 1376-80, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20532295

ABSTRACT

Recently we have perfected a chemical-genetic approach to gain precise spatio-temporal control of cellular signaling. This approach entails the cell-type specific expression of mutant G-protein coupled receptors which have been evolved to be activated by the pharmacologically inert drug-like small molecule clozapine N-oxide. We have named these mutant GPCRs DREADDs (Designer Receptors Exclusively Activated by Designer Drugs). In this paper we will first review recent applications of this technology for the remote control of neuronal and non-neuronal signaling. Next, we will also introduce new variants which could be useful for the control of cellular signaling in discrete cellular compartments. Finally, we will suggest future basic science and therapeutic applications of this general technology.


Subject(s)
Cell Physiological Phenomena , Combinatorial Chemistry Techniques/methods , Genetic Techniques , Animals , Cell Physiological Phenomena/genetics , Chemistry Techniques, Analytical/methods , Growth and Development/genetics , Growth and Development/physiology , Humans , Models, Biological , Organ Specificity/genetics , Organ Specificity/physiology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...