Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
iScience ; 27(5): 109782, 2024 May 17.
Article En | MEDLINE | ID: mdl-38711449

Ten-eleven translocation (TET) proteins are DNA dioxygenases that mediate active DNA demethylation. TET3 is the most highly expressed TET protein in thymic developing T cells. TET3, either independently or in cooperation with TET1 or TET2, has been implicated in T cell lineage specification by regulating DNA demethylation. However, TET-deficient mice exhibit complex phenotypes, suggesting that TET3 exerts multifaceted roles, potentially by interacting with other proteins. We performed liquid chromatography with tandem mass spectrometry in primary developing T cells to identify TET3 interacting partners in endogenous, in vivo conditions. We discover TET3 interacting partners. Our data establish that TET3 participates in a plethora of fundamental biological processes, such as transcriptional regulation, RNA polymerase elongation, splicing, DNA repair, and DNA replication. This resource brings in the spotlight emerging functions of TET3 and sets the stage for systematic studies to dissect the precise mechanistic contributions of TET3 in shaping T cell biology.

2.
J Neurosci ; 42(42): 7900-7920, 2022 10 19.
Article En | MEDLINE | ID: mdl-36261270

Neuronal activity initiates signaling cascades that culminate in diverse outcomes including structural and functional neuronal plasticity, and metabolic changes. While studies have revealed activity-dependent neuronal cell type-specific transcriptional changes, unbiased quantitative analysis of cell-specific activity-induced dynamics in newly synthesized proteins (NSPs) synthesis in vivo has been complicated by cellular heterogeneity and a relatively low abundance of NSPs within the proteome in the brain. Here we combined targeted expression of mutant MetRS (methionine tRNA synthetase) in genetically defined cortical glutamatergic neurons with tight temporal control of treatment with the noncanonical amino acid, azidonorleucine, to biotinylate NSPs within a short period after pharmacologically induced seizure in male and female mice. By purifying peptides tagged with heavy or light biotin-alkynes and using direct tandem mass spectrometry detection of biotinylated peptides, we quantified activity-induced changes in cortical glutamatergic neuron NSPs. Seizure triggered significant changes in ∼300 NSPs, 33% of which were decreased by seizure. Proteins mediating excitatory and inhibitory synaptic plasticity, including SynGAP1, Pak3, GEPH1, Copine-6, and collybistin, and DNA and chromatin remodeling proteins, including Rad21, Smarca2, and Ddb1, are differentially synthesized in response to activity. Proteins likely to play homeostatic roles in response to activity, such as regulators of proteastasis, intracellular ion control, and cytoskeleton remodeling proteins, are activity induced. Conversely, seizure decreased newly synthetized NCAM, among others, suggesting that seizure induced degradation. Overall, we identified quantitative changes in the activity-induced nascent proteome from genetically defined cortical glutamatergic neurons as a strategy to discover downstream mediators of neuronal plasticity and generate hypotheses regarding their function.SIGNIFICANCE STATEMENT Activity-induced neuronal and synaptic plasticity are mediated by changes in the protein landscape, including changes in the activity-induced newly synthesized proteins; however, identifying neuronal cell type-specific nascent proteome dynamics in the intact brain has been technically challenging. We conducted an unbiased proteomic screen from which we identified significant activity-induced changes in ∼300 newly synthesized proteins in genetically defined cortical glutamatergic neurons within 20 h after pharmacologically induced seizure. Bioinformatic analysis of the dynamic nascent proteome indicates that the newly synthesized proteins play diverse roles in excitatory and inhibitory synaptic plasticity, chromatin remodeling, homeostatic mechanisms, and proteasomal and metabolic functions, extending our understanding of the diversity of plasticity mechanisms.


Amino Acyl-tRNA Synthetases , Proteome , Male , Female , Mice , Animals , Proteome/metabolism , Proteomics/methods , Biotin/metabolism , Neurons/metabolism , Neuronal Plasticity/physiology , Amino Acids/metabolism , Methionine/metabolism , Alkynes/metabolism , Seizures/metabolism , Amino Acyl-tRNA Synthetases/genetics , Amino Acyl-tRNA Synthetases/metabolism , Neural Cell Adhesion Molecules/metabolism , ras GTPase-Activating Proteins/metabolism
3.
J Neurosci ; 42(19): 4042-4052, 2022 05 11.
Article En | MEDLINE | ID: mdl-35396330

Retinal ganglion cells (RGCs) die after optic nerve trauma or in degenerative disease. However, acute changes in protein expression that may regulate RGC response to injury are not fully understood, and detailed methods to quantify new protein synthesis have not been tested. Here, we develop and apply a new in vivo quantitative measure of newly synthesized proteins to examine changes occurring in the retina after optic nerve injury. Azidohomoalanine, a noncanonical amino acid, was injected intravitreally into the eyes of rodents of either sex with or without optic nerve injury. Isotope variants of biotin-alkyne were used for quantitative BONCAT (QBONCAT) mass spectrometry, allowing identification of protein synthesis and transport rate changes in more than 1000 proteins at 1 or 5 d after optic nerve injury. In vitro screening showed several newly synthesized proteins regulate axon outgrowth in primary neurons in vitro This novel approach to targeted quantification of newly synthesized proteins after injury uncovers a dynamic translational response within broader proteostasis regulation and enhances our understanding of the cellular response to injury.SIGNIFICANCE STATEMENT Optic nerve injury results in death and degeneration of retinal ganglion cells and their axons. The specific cellular response to injury, including changes in new protein synthesis, is obscured by existing proteins and protein degradation. In this study, we introduce QBONCAT to isolate and quantify acute protein synthesis and subsequent transport between cellular compartments. We identify novel candidate protein effectors of the regenerative response and uncover their regulation of axon growth in vitro, validating the utility of QBONCAT for the discovery of novel regulatory and therapeutic candidates after optic nerve injury.


Optic Nerve Injuries , Axons/metabolism , Humans , Nerve Regeneration/physiology , Optic Nerve Injuries/metabolism , Retina/metabolism , Retinal Ganglion Cells/metabolism
4.
Elife ; 112022 03 08.
Article En | MEDLINE | ID: mdl-35259089

Many neurons in the adult central nervous system, including retinal ganglion cells (RGCs), degenerate and die after injury. Early axon protein and organelle trafficking failure is a key component in many neurodegenerative disorders yet changes to axoplasmic transport in disease models have not been quantified. We analyzed early changes in the protein 'transportome' from RGC somas to their axons after optic nerve injury and identified transport failure of an anterograde motor protein Kif5a early in RGC degeneration. We demonstrated that manipulating Kif5a expression affects anterograde mitochondrial trafficking in RGCs and characterized axon transport in Kif5a knockout mice to identify proteins whose axon localization was Kif5a-dependent. Finally, we found that knockout of Kif5a in RGCs resulted in progressive RGC degeneration in the absence of injury. Together with expression data localizing Kif5a to human RGCs, these data identify Kif5a transport failure as a cause of RGC neurodegeneration and point to a mechanism for future therapeutics.


Optic Nerve Injuries , Animals , Axonal Transport , Axons/metabolism , Kinesins/genetics , Mice , Mice, Inbred C57BL , Nerve Regeneration , Retinal Ganglion Cells/metabolism
5.
Cell Rep ; 38(4): 110287, 2022 01 25.
Article En | MEDLINE | ID: mdl-35081342

Intercellular transfer of toxic proteins between neurons is thought to contribute to neurodegenerative disease, but whether direct interneuronal protein transfer occurs in the healthy brain is not clear. To assess the prevalence and identity of transferred proteins and the cellular specificity of transfer, we biotinylated retinal ganglion cell proteins in vivo and examined biotinylated proteins transported through the rodent visual circuit using microscopy, biochemistry, and mass spectrometry. Electron microscopy demonstrated preferential transfer of biotinylated proteins from retinogeniculate inputs to excitatory lateral geniculate nucleus (LGN) neurons compared with GABAergic neurons. An unbiased mass spectrometry-based screen identified ∼200 transneuronally transported proteins (TNTPs) isolated from the visual cortex. The majority of TNTPs are present in neuronal exosomes, and virally expressed TNTPs, including tau and ß-synuclein, were detected in isolated exosomes and postsynaptic neurons. Our data demonstrate transfer of diverse endogenous proteins between neurons in the healthy intact brain and suggest that TNTP transport may be mediated by exosomes.


Cell Communication/physiology , Exosomes/metabolism , Neurons/metabolism , Visual Cortex/metabolism , Animals , Neuroanatomical Tract-Tracing Techniques , Proteomics , Rats , Rats, Wistar , Visual Pathways/metabolism , Xenopus
6.
Cell Rep ; 28(7): 1935-1947.e5, 2019 08 13.
Article En | MEDLINE | ID: mdl-31412257

The brain processes information and generates cognitive and motor outputs through functions of spatially organized proteins in different types of neurons. More complete knowledge of proteins and their distributions within neuronal compartments in intact circuits would help in the understanding of brain function. We used unbiased in vivo protein labeling with intravitreal NHS-biotin for discovery and analysis of endogenous axonally transported proteins in the visual system using tandem mass spectrometric proteomics, biochemistry, and both light and electron microscopy. Purification and proteomic analysis of biotinylated peptides identified ∼1,000 proteins transported from retinal ganglion cells into the optic nerve and ∼575 biotinylated proteins recovered from presynaptic compartments of lateral geniculate nucleus and superior colliculus. Approximately 360 biotinylated proteins were differentially detected in the two retinal targets. This study characterizes axonally transported proteins in the healthy adult visual system by analyzing proteomes from multiple compartments of retinal ganglion cell projections in the intact brain.


Axonal Transport , Optic Nerve/metabolism , Presynaptic Terminals/metabolism , Proteome/analysis , Proteome/metabolism , Retinal Ganglion Cells/metabolism , Visual Pathways/metabolism , Animals , Male , Optic Nerve/cytology , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/cytology , Visual Pathways/cytology
7.
Proc Natl Acad Sci U S A ; 116(32): 16086-16094, 2019 08 06.
Article En | MEDLINE | ID: mdl-31320591

Exosomes are thought to be released by all cells in the body and to be involved in intercellular communication. We tested whether neural exosomes can regulate the development of neural circuits. We show that exosome treatment increases proliferation in developing neural cultures and in vivo in dentate gyrus of P4 mouse brain. We compared the protein cargo and signaling bioactivity of exosomes released by hiPSC-derived neural cultures lacking MECP2, a model of the neurodevelopmental disorder Rett syndrome, with exosomes released by isogenic rescue control neural cultures. Quantitative proteomic analysis indicates that control exosomes contain multiple functional signaling networks known to be important for neuronal circuit development. Treating MECP2-knockdown human primary neural cultures with control exosomes rescues deficits in neuronal proliferation, differentiation, synaptogenesis, and synchronized firing, whereas exosomes from MECP2-deficient hiPSC neural cultures lack this capability. These data indicate that exosomes carry signaling information required to regulate neural circuit development.


Exosomes/metabolism , Nerve Net/metabolism , Neurogenesis , Action Potentials , Animals , Cell Count , Cell Differentiation , Cell Proliferation , Cells, Cultured , Dentate Gyrus/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Methyl-CpG-Binding Protein 2/deficiency , Methyl-CpG-Binding Protein 2/metabolism , Mice , Neurons/cytology , Neurons/metabolism , Signal Transduction , Spheroids, Cellular/cytology , Synapses/metabolism
8.
Elife ; 72018 02 07.
Article En | MEDLINE | ID: mdl-29412139

Experience-dependent synaptic plasticity refines brain circuits during development. To identify novel protein synthesis-dependent mechanisms contributing to experience-dependent plasticity, we conducted a quantitative proteomic screen of the nascent proteome in response to visual experience in Xenopus optic tectum using bio-orthogonal metabolic labeling (BONCAT). We identified 83 differentially synthesized candidate plasticity proteins (CPPs). The CPPs form strongly interconnected networks and are annotated to a variety of biological functions, including RNA splicing, protein translation, and chromatin remodeling. Functional analysis of select CPPs revealed the requirement for eukaryotic initiation factor three subunit A (eIF3A), fused in sarcoma (FUS), and ribosomal protein s17 (RPS17) in experience-dependent structural plasticity in tectal neurons and behavioral plasticity in tadpoles. These results demonstrate that the nascent proteome is dynamic in response to visual experience and that de novo synthesis of machinery that regulates RNA splicing and protein translation is required for experience-dependent plasticity.


Neuronal Plasticity , Proteome/analysis , Superior Colliculi/physiology , Visual Perception , Animals , Computational Biology , Protein Interaction Maps , Xenopus
9.
J Proteome Res ; 13(9): 3966-78, 2014 Sep 05.
Article En | MEDLINE | ID: mdl-25117199

Mass spectrometric strategies to identify protein subpopulations involved in specific biological functions rely on covalently tagging biotin to proteins using various chemical modification methods. The biotin tag is primarily used for enrichment of the targeted subpopulation for subsequent mass spectrometry (MS) analysis. A limitation of these strategies is that MS analysis does not easily discriminate unlabeled contaminants from the labeled protein subpopulation under study. To solve this problem, we developed a flexible method that only relies on direct MS detection of biotin-tagged proteins called "Direct Detection of Biotin-containing Tags" (DiDBiT). Compared with conventional targeted proteomic strategies, DiDBiT improves direct detection of biotinylated proteins ∼200 fold. We show that DiDBiT is applicable to several protein labeling protocols in cell culture and in vivo using cell permeable NHS-biotin and incorporation of the noncanonical amino acid, azidohomoalanine (AHA), into newly synthesized proteins, followed by click chemistry tagging with biotin. We demonstrate that DiDBiT improves the direct detection of biotin-tagged newly synthesized peptides more than 20-fold compared to conventional methods. With the increased sensitivity afforded by DiDBiT, we demonstrate the MS detection of newly synthesized proteins labeled in vivo in the rodent nervous system with unprecedented temporal resolution as short as 3 h.


Biotin/analogs & derivatives , Proteins/analysis , Proteome/analysis , Proteomics/methods , Succinimides/chemistry , Tandem Mass Spectrometry/methods , Animals , Biotin/chemistry , Biotin/metabolism , HEK293 Cells , Humans , Male , Proteins/chemistry , Proteins/metabolism , Proteome/chemistry , Proteome/metabolism , Rats , Rats, Wistar , Succinimides/metabolism
10.
Cell Rep ; 6(4): 737-47, 2014 Feb 27.
Article En | MEDLINE | ID: mdl-24529705

Neural plasticity requires protein synthesis, but the identity of newly synthesized proteins generated in response to plasticity-inducing stimuli remains unclear. We used in vivo bio-orthogonal noncanonical amino acid tagging (BONCAT) with the methionine analog azidohomoalanine (AHA) combined with the multidimensional protein identification technique (MudPIT) to identify proteins that are synthesized in the tadpole brain over 24 hr. We induced conditioning-dependent plasticity of visual avoidance behavior, which required N-methyl-D-aspartate (NMDA) and Ca(2+)-permeable α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, αCaMKII, and rapid protein synthesis. Combining BONCAT with western blots revealed that proteins including αCaMKII, MEK1, CPEB, and GAD65 are synthesized during conditioning. Acute synthesis of CPEB during conditioning is required for behavioral plasticity as well as conditioning-induced synaptic and structural plasticity in the tectal circuit. We outline a signaling pathway that regulates protein-synthesis-dependent behavioral plasticity in intact animals, identify newly synthesized proteins induced by visual experience, and demonstrate a requirement for acute synthesis of CPEB in plasticity.


Avoidance Learning , Brain/metabolism , Gene Expression Regulation, Developmental , Neuronal Plasticity , Transcription Factors/metabolism , Xenopus Proteins/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Animals , Brain/growth & development , Brain/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Conditioning, Classical , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Larva/metabolism , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , Proteome/genetics , Proteome/metabolism , Transcription Factors/genetics , Visual Perception , Xenopus , Xenopus Proteins/genetics , mRNA Cleavage and Polyadenylation Factors/genetics
11.
Curr Opin Neurobiol ; 23(6): 997-1004, 2013 Dec.
Article En | MEDLINE | ID: mdl-23998929

Exosomes are small extracellular vesicles that mediate intercellular signaling in the brain without requiring direct contact between cells. Although exosomes have been shown to play a role in neurological diseases and in response to nerve trauma, a role for exosome-mediated signaling in brain development and function has not yet been demonstrated. Here we review data building a case for exosome function in the brain.


Brain/growth & development , Cell Communication/physiology , Exosomes/metabolism , Neural Pathways/growth & development , Neurogenesis/physiology , Animals , Brain/metabolism , Humans , Neural Pathways/physiology
12.
J Alzheimers Dis ; 17(4): 773-86, 2009.
Article En | MEDLINE | ID: mdl-19542617

Synapse loss occurs early in Alzheimer's disease (AD) and is considered the best pathological correlate of cognitive decline. Ephrins and Eph receptors are involved in regulation of excitatory neurotransmission and play a role in cytoskeleton remodeling. We asked whether alterations in Eph receptors could underlie cognitive impairment in an AD mouse model overexpressing human amyloid-beta protein precursor (hA beta PP) with familial mutations (hA beta PP swe-ind mice). We found that EphA4 and EphB2 receptors were reduced in the hippocampus before the development of impaired object recognition and spatial memory. Similar results were obtained in another line of transgenic A beta PP mice, Tg2576. A reduction in Eph receptor levels was also found in postmortem hippocampal tissue from patients with incipient AD. At the time of onset of memory decline inhA beta PP swe-ind mice, no change in surface expression of AMPA or NMDA receptor subunits was apparent, but we found changes in Eph-receptor downstream signaling, in particular a decrease in membrane-associated phosho-cofilin levels that may cause cytoskeletal changes and disrupted synaptic activity. Consistent with this finding, Eph receptor activation in cell culture increased phosho-cofilin levels. The results suggest that alterations in Eph receptors may play a role in synaptic dysfunction in the hippocampus leading to cognitive impairment in a model of AD.


Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Cognition , Hippocampus/metabolism , Memory , Receptor, EphA4/metabolism , Receptor, EphB2/metabolism , Actin Depolymerizing Factors/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/genetics , Animals , Blotting, Western , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuropsychological Tests , Postmortem Changes , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Recognition, Psychology , Space Perception , Time Factors
13.
Neurobiol Dis ; 33(3): 369-78, 2009 Mar.
Article En | MEDLINE | ID: mdl-19101630

Transgenic mice expressing mutant human amyloid precursor protein (APP) develop an age-dependent amyloid pathology and memory deficits, but no overt neuronal loss. Here, in mice overexpressing wild-type human APP (hAPP(wt)) we found an early memory impairment, particularly in the water maze and to a lesser extent in the object recognition task, but beta-amyloid peptide (Abeta(42)) was barely detectable in the hippocampus. In these mice, hAPP processing was basically non-amyloidogenic, with high levels of APP carboxy-terminal fragments, C83 and APP intracellular domain. A tau pathology with an early increase in the levels of phosphorylated tau in the hippocampus, a likely consequence of enhanced ERK1/2 activation, was also observed. Furthermore, these mice presented a loss of synapse-associated proteins: PSD95, AMPA and NMDA receptor subunits and phosphorylated CaMKII. Importantly, signs of neurodegeneration were found in the hippocampal CA1 subfield and in the entorhinal cortex that were associated to a marked loss of MAP2 immunoreactivity. Conversely, in mice expressing mutant hAPP, high levels of Abeta(42) were found in the hippocampus, but no signs of neurodegeneration were apparent. The results support the notion of Abeta-independent pathogenic pathways in Alzheimer's disease.


Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Cognition/physiology , Hippocampus/metabolism , Hippocampus/pathology , Maze Learning , Memory , Peptide Fragments/metabolism , Receptors, Cell Surface/metabolism , Recognition, Psychology , Amyloid beta-Protein Precursor/genetics , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Disks Large Homolog 4 Protein , Guanylate Kinases , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Mutation , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Phosphorylation , Protease Nexins , Receptors, AMPA/metabolism , Receptors, Cell Surface/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/physiology , tau Proteins/metabolism
14.
J Neurochem ; 94(4): 884-95, 2005 Aug.
Article En | MEDLINE | ID: mdl-16092936

Stimulation of hippocampal 5-HT(1A) receptors impairs memory retention. The highly selective 5-HT(1A) antagonist, WAY-100635, prevents the cognitive deficits induced not only by 5-HT(1A) stimulation but also by cholinergic or NMDA receptor blockade. On this basis, the effects of WAY-100635 on molecular events associated with memory storage were explored. In rat hippocampus, WAY-100635 produced a rapid increase in phosphorylated Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) and in Ca(2+)-independent CaMKII and protein kinase A (PKA) enzyme activity. This increase was followed a few hours later by an enhanced membrane expression of AMPA receptor subunits, especially of the GluR1 subunit phosphorylated at the CaMKII site, pGluR1(Ser831). The same qualitative effects were found with the weaker 5-HT(1A) antagonist NAN-190. The effects of both antagonists were no longer apparent in rats with a previous 5-HT depletion induced by the tryptophan hydroxylase inhibitor p-chlorophenylalanine (PCPA), suggesting that 5-HT(1A) receptor blockade removes the tonic inhibition of 5-HT through 5-HT(1A) receptor stimulation on excitatory hippocampal neurons, with the consequent increase in PKA activity. In addition, administration of WAY-100635 potentiated the learning-specific increase in the hippocampus of phospho-CaMKII, Ca(2+)-independent CaMKII activity, as well as the phosphorylation of either the CaMKII or the PKA site on the AMPA receptor GluR1 subunit. This study suggests that blockade of hippocampal 5-HT(1A) receptors favours molecular events critically involved in memory formation, and provides an in vivo molecular basis for the proposed utility of 5-HT(1A) receptor antagonists in the treatment of cognitive disorders.


Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Membrane/metabolism , Hippocampus/metabolism , Memory/physiology , Receptors, AMPA/metabolism , Serotonin 5-HT1 Receptor Antagonists , Animals , Avoidance Learning/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Cyclic AMP-Dependent Protein Kinases/metabolism , Male , Phosphoprotein Phosphatases/metabolism , Piperazines/pharmacology , Protein Isoforms/metabolism , Pyridines/pharmacology , Rats , Rats, Wistar , Serotonin Antagonists/pharmacology
15.
Bioorg Med Chem ; 12(19): 5181-91, 2004 Oct 01.
Article En | MEDLINE | ID: mdl-15351401

A series of new mixed benzimidazole-arylpiperazine derivatives were designed by incorporating in general structure III the pharmacophoric elements of 5-HT(1A) and 5-HT(3) receptors. Compounds 1-11 were synthesized and evaluated for binding affinity at both serotoninergic receptors, all of them exhibiting high 5-HT(3)R affinity (K(i)=10-62nM), and derivatives with an o-alkoxy group in the arylpiperazine ring showing nanomolar affinity for the 5-HT(1A)R (K(i)=18-150nM). Additionally, all the synthesized compounds were selective over alpha(1)-adrenergic and dopamine D(2) receptors (K(i)>1000-10,000nM). Compound 3 was selected for further pharmacological characterization due to its interesting binding profile as mixed 5-HT(1A)/5-HT(3) ligand with high affinity for both receptors (5-HT(1A): K(i)=18.0nM, 5-HT(3): K(i)=27.2nM). In vitro and in vivo findings suggest that this compound acts as a partial agonist at 5-HT(1A)Rs and as a 5-HT(3)R antagonist. This novel mixed 5-HT(1A)/5-HT(3) ligand was also effective in preventing the cognitive deficits induced by muscarinic receptor blockade in a passive avoidance learning test, suggesting a potential interest in the treatment of cognitive dysfunction.


Benzimidazoles/chemical synthesis , Piperazines/chemical synthesis , Serotonin Antagonists/chemical synthesis , Animals , Avoidance Learning/drug effects , Benzimidazoles/pharmacology , Cognition Disorders/drug therapy , Guanosine 5'-O-(3-Thiotriphosphate)/chemistry , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Guinea Pigs , Hippocampus/metabolism , Hypothermia/chemically induced , Ligands , Male , Mice , Muscle Contraction/drug effects , Piperazines/pharmacology , Rats , Rats, Wistar , Serotonin 5-HT1 Receptor Antagonists , Serotonin 5-HT3 Receptor Antagonists , Serotonin Antagonists/pharmacology , Structure-Activity Relationship
16.
Bioorg Med Chem Lett ; 13(19): 3177-80, 2003 Oct 06.
Article En | MEDLINE | ID: mdl-12951088

A series of new benzimidazole-arylpiperazine derivatives III were designed, synthesized and evaluated for binding affinity at serotoninergic 5-HT(1A) and 5-HT(3) receptors. Compound IIIc was identified as a novel mixed 5-HT(1A)/5-HT(3) ligand with high affinity for both serotonin receptors and excellent selectivity over alpha(1)-adrenergic and dopamine D(2) receptors. This compound was characterized as a partial agonist at 5-HT(1A)Rs and a 5-HT(3)R antagonist, and was effective in preventing the cognitive deficits induced by muscarinic receptor blockade in a passive avoidance learning test.


Benzimidazoles/metabolism , Piperazines/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, Serotonin, 5-HT3/metabolism , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacology , Drug Design , Ligands , Mice , Piperazines/chemical synthesis , Piperazines/pharmacology , Protein Binding/drug effects , Protein Binding/physiology , Rats
...