Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Neurobiol Pain ; 7: 100039, 2020.
Article in English | MEDLINE | ID: mdl-31909296

ABSTRACT

Previous studies have shown that oral administration of the NMDAR modulator NYX-2925 alleviates pain in several animal models of neuropathic pain and this appears to be through mPFC, but not spinal, mediated mechanisms. While much is known about the impact of neuropathic pain on NMDAR-mediated signaling in the spinal cord, limited studies have focused on the brain. In the current study, we assess signaling changes associated with NMDAR-mediated plasticity in the mPFC and the impact of NYX-2925 administration on the normalization of these signaling changes. We found a decrease in activated Src levels in the mPFC of animals with chronic constriction injury (CCI) of the sciatic nerve. While Src mediated activation of NMDARs was also decreased in CCI animals, the main NMDAR phosphorylation site of CAMKII was not affected. This is in opposition to what has been found in the spinal cord, where both Src and CAMKII activation are increased. Oral administration of NYX-2925 restored levels of activated Src and Src phosphorylation sites on GluN2A and GluN2B in the mPFC, with no effect on activated CAMKII levels. The analgesic effect of NYX-2925 appears dependent on this restoration of Src activation in the mPFC, as co-administering Src activation inhibitors prevented the NYX-2925 analgesic effect. Overall, these data suggest that NMDAR-mediated signaling plays a key role in neuropathic pain, albeit in different directions in the spinal cord vs. the mPFC. Furthermore, the analgesic effect of NYX-2925 appears to involve a restoration of NMDAR-mediated signaling in the mPFC.

2.
J Neurochem ; 152(5): 523-541, 2020 03.
Article in English | MEDLINE | ID: mdl-31376158

ABSTRACT

N-methyl-d-aspartate receptors (NMDARs) mediate both physiological and pathophysiological processes, although selective ligands lack broad clinical utility. NMDARs are composed of multiple subunits, but N-methyl-d-aspartate receptor subunit 2 (GluN2) is predominately responsible for functional heterogeneity. Specifically, the GluN2A- and GluN2B-containing subtypes are enriched in adult hippocampus and cortex and impact neuronal communication via dynamic trafficking into and out of the synapse. We sought to understand if ((2S, 3R)-3-hydroxy-2-((R)-5-isobutyryl-1-oxo-2,5-diazaspiro[3,4]octan-2-yl) butanamide (NYX-2925), a novel NMDAR modulator, alters synaptic levels of GluN2A- or GluN2B-containing NMDARs. Low-picomolar NYX-2925 increased GluN2B colocalization with the excitatory post-synaptic marker post-synaptic density protein 95 (PSD-95) in rat primary hippocampal neurons within 30 min. Twenty-four hours following oral administration, 1 mg/kg NYX-2925 increased GluN2B in PSD-95-associated complexes ex vivo, and low-picomolar NYX-2925 regulated numerous trafficking pathways in vitro. Because the NYX-2925 concentration that increases synaptic GluN2B was markedly below that which enhances long-term potentiation (mid-nanomolar), we sought to elucidate the basis of this effect. Although NMDAR-dependent, NYX-2925-mediated colocalization of GluN2B with PSD-95 occurred independent of ion flux, as colocalization increased in the presence of either the NMDAR channel blocker (5R,10S)-(-)-5-Methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine hydrogen maleate or glycine site antagonist 7-chlorokynurenic acid. Moreover, while mid-nanomolar NYX-2925 concentrations, which do not increase synaptic GluN2B, enhanced calcium transients, functional plasticity was only enhanced by picomolar NYX-2925. Thus, NYX-2925 concentrations that increase synaptic GluN2B facilitated the chemical long-term potentiation induced insertion of synaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor GluA1 subunit levels. Basal (unstimulated by chemical long-term potentiation) levels of synaptic GluA1 were only increased by mid-nanomolar NYX-2925. These data suggest that NYX-2925 facilitates homeostatic plasticity by initially increasing synaptic GluN2B via metabotropic-like NMDAR signaling. Cover Image for this issue: doi: 10.1111/jnc.14735.


Subject(s)
Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Spiro Compounds/pharmacology , Synapses/metabolism , Animals , Hippocampus/drug effects , Hippocampus/metabolism , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Rats , Rats, Sprague-Dawley , Receptors, AMPA/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Signal Transduction/drug effects , Synapses/drug effects
3.
Sleep ; 42(10)2019 10 09.
Article in English | MEDLINE | ID: mdl-31504971

ABSTRACT

STUDY OBJECTIVES: The present studies examine the effects of NMDAR activation by NYX-2925 diurnal rhythmicity of both sleep and wake as well as emotion. METHODS: Twenty-four-hour sleep EEG recordings were obtained in sleep-deprived and non-sleep-deprived rats. In addition, the day-night cycle of both activity and mood was measured using home cage ultrasonic-vocalization recordings. RESULTS: NYX-2925 significantly facilitated non-REM (NREM) sleep during the lights-on (sleep) period, and this effect persisted for 3 days following a single dose in sleep-deprived rats. Sleep-bout duration and REM latencies were increased without affecting total REM sleep, suggesting better sleep quality. In addition, delta power during wake was decreased, suggesting less drowsiness. NYX-2925 also rescued learning and memory deficits induced by sleep deprivation, measured using an NMDAR-dependent learning task. Additionally, NYX-2925 increased positive affect and decreased negative affect, primarily by facilitating the transitions from sleep to rough-and-tumble play and back to sleep. In contrast to NYX-2925, the NMDAR antagonist ketamine acutely (1-4 hours post-dosing) suppressed REM and non-REM sleep, increased delta power during wake, and blunted the amplitude of the sleep-wake activity rhythm. DISCUSSION: These data suggest that NYX-2925 could enhance behavioral plasticity via improved sleep quality as well as vigilance during wake. As such, the facilitation of sleep by NYX-2925 has the potential to both reduce symptom burden on neurological and psychiatric disorders as well as serve as a biomarker for drug effects through restoration of sleep architecture.


Subject(s)
Affect/physiology , Circadian Rhythm/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Sleep Deprivation/physiopathology , Sleep/physiology , Spiro Compounds/pharmacology , Affect/drug effects , Animals , Circadian Rhythm/drug effects , Electroencephalography/methods , Male , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/agonists , Sleep/drug effects , Sleep Deprivation/drug therapy , Spiro Compounds/therapeutic use , Wakefulness/drug effects , Wakefulness/physiology
4.
Neuroreport ; 30(13): 863-866, 2019 09 04.
Article in English | MEDLINE | ID: mdl-31373964

ABSTRACT

In humans, chronic pain is often expressed as a spontaneous emotional response which can lead to fragmented sleep. Rat 50-kHz and 20-kHz ultrasonic vocalizations are well-established measures of positive and negative emotional states, respectively. The rat chronic constriction injury model was used to induce chronic pain, and ultrasonic vocalizations were measured in both the heterospecific rough-and-tumble play (i.e. tickling) test as well as during 24-hour home cage recordings. Rates of hedonic 50-kHz ultrasonic vocalizations during the non-stimulus periods of the tickling test, as well as the rewarding value of tickling, were reduced in chronic constriction injury rats compared to sham controls. In the 24-hour home cage recording study, chronic constriction injury animals showed a reduced amplitude in circadian activity, as well as reduced hedonic 50-kHz ultrasonic vocalizations and increased evoked and spontaneous aversive 20-kHz ultrasonic vocalizations. These data demonstrate that rat ultrasonic vocalizations can be used to capture core symptoms of chronic pain and may be useful in the elucidation of the neuronal mechanisms that underlie the affective component of pain.


Subject(s)
Chronic Pain/physiopathology , Chronic Pain/psychology , Emotions/physiology , Ultrasonic Waves , Vocalization, Animal/physiology , Animals , Male , Rats , Rats, Sprague-Dawley , Sciatic Neuropathy/physiopathology , Sciatic Neuropathy/psychology
5.
Int J Neuropsychopharmacol ; 22(3): 247-259, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30544218

ABSTRACT

BACKGROUND: Modulation of glutamatergic synaptic transmission by N-methyl-D-aspartate receptors can produce rapid and sustained antidepressant effects. Rapastinel (GLYX-13), initially described as a N-methyl-D-aspartate receptor partial glycine site agonist, exhibits rapid antidepressant effect in rodents without the accompanying dissociative effects of N-methyl-D-aspartate receptor antagonists. METHODS: The relationship between rapastinel's in vitro N-methyl-D-aspartate receptor pharmacology and antidepressant efficacy was determined by brain microdialysis and subsequent pharmacological characterization of therapeutic rapastinel concentrations in N-methyl-D-aspartate receptor-specific radioligand displacement, calcium mobilization, and medial prefrontal cortex electrophysiology assays. RESULTS: Brain rapastinel concentrations of 30 to 100 nM were associated with its antidepressant-like efficacy and enhancement of N-methyl-D-aspartate receptor-dependent neuronal intracellular calcium mobilization. Modulation of N-methyl-D-aspartate receptors by rapastinel was independent of D-serine concentrations, and glycine site antagonists did not block rapastinel's effect. In rat medial prefrontal cortex slices, 100 nM rapastinel increased N-methyl-D-aspartate receptor-mediated excitatory postsynaptic currents and enhanced the magnitude of long-term potentiation without any effect on miniature EPSCs or paired-pulse facilitation responses, indicating postsynaptic action of rapastinel. A critical amino acid within the NR2 subunit was identified as necessary for rapastinel's modulatory effect. CONCLUSION: Rapastinel brain concentrations associated with antidepressant-like activity directly enhance medial prefrontal cortex N-methyl-D-aspartate receptor activity and N-methyl-D-aspartate receptor-mediated synaptic plasticity in vitro. At therapeutic concentrations, rapastinel directly enhances N-methyl-D-aspartate receptor activity through a novel site independent of the glycine coagonist site. While both rapastinel and ketamine physically target N-methyl-D-aspartate receptors, the 2 molecules have opposing actions on N-methyl-D-aspartate receptors. Modest positive modulation of N-methyl-D-aspartate receptors by rapastinel represents a novel pharmacological approach to promote well-tolerated, rapid, and sustained improvements in mood disorders.


Subject(s)
Antidepressive Agents/administration & dosage , Antidepressive Agents/metabolism , Cerebral Cortex/metabolism , Oligopeptides/administration & dosage , Oligopeptides/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Cells, Cultured , Cerebral Cortex/drug effects , Dose-Response Relationship, Drug , Drug Partial Agonism , Male , Microdialysis/methods , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/agonists , Treatment Outcome
6.
Int J Neuropsychopharmacol ; 21(3): 242-254, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29099938

ABSTRACT

Background: N-methyl-D-aspartate receptors are one member of a family of ionotropic glutamate receptors that play a pivotal role in synaptic plasticity processes associated with learning and have become attractive therapeutic targets for diseases such as depression, anxiety, schizophrenia, and neuropathic pain. NYX-2925 ((2S, 3R)-3-hydroxy-2-((R)-5-isobutyryl-1-oxo-2,5-diazaspiro[3.4]octan-2-yl)butanamide) is one member of a spiro-ß-lactam-based chemical platform that mimics some of the dipyrrolidine structural features of rapastinel (formerly GLYX-13: threonine-proline-proline-threonine) and is distinct from known N-methyl-D-aspartate receptor agonists or antagonists such as D-cycloserine, ketamine, MK-801, kynurenic acid, or ifenprodil. Methods: The in vitro and in vivo pharmacological properties of NYX-2925 were examined. Results: NYX-2925 has a low potential for "off-target" activity, as it did not exhibit any significant affinity for a large panel of neuroactive receptors, including hERG receptors. NYX-2925 increased MK-801 binding to human N-methyl-D-aspartate receptor NR2A-D subtypes expressed in HEK cells and enhanced N-methyl-D-aspartate receptor current and long-term potentiation (LTP) in rat hippocampal slices (100-500 nM). Single dose ex vivo studies showed increased metaplasticity in a hippocampal LTP paradigm and structural plasticity 24 hours after administration (1 mg/kg p.o.). Significant learning enhancement in both novel object recognition and positive emotional learning paradigms were observed (0.01-1 mg/kg p.o.), and these effects were blocked by the N-methyl-D-aspartate receptor antagonist CPP. NYX-2925 does not show any addictive or sedative/ataxic side effects and has a therapeutic index of >1000. NYX-2925 (1 mg/kg p.o.) has a cerebrospinal fluid half-life of 1.2 hours with a Cmax of 44 nM at 1 hour. Conclusions: NYX-2925, like rapastinel, activates an NMDA receptor-mediated synaptic plasticity process and may have therapeutic potential for a variety of NMDA receptor-mediated central nervous system disorders.


Subject(s)
Excitatory Amino Acid Agents/pharmacology , Memory/drug effects , Neuronal Plasticity/drug effects , Oligopeptides/pharmacology , Animals , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Emotions/drug effects , Excitatory Amino Acid Agents/cerebrospinal fluid , Excitatory Amino Acid Agents/chemistry , HEK293 Cells , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Learning/drug effects , Learning/physiology , Male , Memory/physiology , Molecular Structure , Neuronal Plasticity/physiology , Oligopeptides/cerebrospinal fluid , Oligopeptides/chemistry , Pyramidal Cells/cytology , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Pyrazines/pharmacology , Rats , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
7.
Neuroreport ; 28(17): 1122-1126, 2017 Dec 06.
Article in English | MEDLINE | ID: mdl-28957945

ABSTRACT

Positive emotions have been shown to induce resilience to stress in humans, as well as increase cognitive abilities (learning, memory, and problem solving) and improve overall health. In rats, frequency modulated 50-kHz ultrasonic vocalizations (hedonic 50 kHz) reflect a positive affective state and are best elicited by rough-and-tumble play. A well-established rat chronic unpredictable stress paradigm was used to produce a robust and long-lasting decrease in positive affect, increase in negative affect, and learned helplessness in Sprague-Dawley rats. Rough-and-tumble play (3 min every 3 days) reversed stress-induced effects of chronic unpredictable stress in the Porsolt forced swim test, novelty-induced hypophagia, sucrose preference, and ultrasonic vocalization assays compared with a light touch control group. These data demonstrate that positive affect induces resilience to stress effects in rats, and that rough-and-tumble play can be used to explore the biological basis of resilience that may lead to the development of new therapeutics for stress-related disorders.


Subject(s)
Resilience, Psychological , Social Behavior , Stress, Psychological , Affect , Animals , Dietary Sucrose , Feeding Behavior , Helplessness, Learned , Male , Motor Activity , Rats, Sprague-Dawley , Taste Perception , Ultrasonics , Uncertainty , Vocalization, Animal
8.
Int J Neuropsychopharmacol ; 20(6): 476-484, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28158790

ABSTRACT

Background: Posttraumatic stress disorder is an anxiety disorder characterized by deficits in the extinction of aversive memories. Insulin-like growth factor 1 (IGF1) is the only growth factor that has shown anxiolytic and antidepressant properties in human clinical trials. In animal studies, insulin-like growth factor binding protein 2 (IGFBP2) shows both IGF1-dependent and IGF1-independent pharmacological effects, and IGFBP2 expression is upregulated by rough-and-tumble play that induces resilience to stress. Methods: IGFBP2 was evaluated in Porsolt, contextual fear conditioning, and chronic unpredictable stress models of posttraumatic stress disorder. The dependence of IGFBP2 effects on IGF1- and AMPA-receptor activation was tested using selective receptor antagonists. Dendritic spine morphology was measured in the dentate gyrus and the medial prefrontal cortex 24 hours after in vivo dosing. Results: IGFBP2 was 100 times more potent than IGF1 in the Porsolt test. Unlike IGF1, effects of IGFBP2 were not blocked by the IGF1-receptor antagonist JB1, or by the AMPA-receptor antagonist 2,3-Dioxo-6-nitro-1,2,3,4 tetrahydrobenzo[f]quinoxaline-7-sulfonamide (NBQX) in the Porsolt test. IGFBP2 (1 µg/kg) and IGF1 (100 µg/kg i.v.) each facilitated contextual fear extinction and consolidation. Using a chronic unpredictable stress paradigm, IGFBP2 reversed stress-induced effects in the Porsolt, novelty-induced hypophagia, sucrose preference, and ultrasonic vocalization assays. IGFBP2 also increased mature dendritic spine densities in the medial prefrontal cortex and hippocampus 24 hours postdosing. Conclusions: These data suggest that IGFBP2 has therapeutic-like effects in multiple rat models of posttraumatic stress disorder via a novel IGF1 receptor-independent mechanism. These data also suggest that the long-lasting effects of IGFBP2 may be due to facilitation of structural plasticity at the dendritic spine level. IGFBP2 and mimetics may have therapeutic potential for the treatment of posttraumatic stress disorder.


Subject(s)
Dendritic Spines/drug effects , Dentate Gyrus/drug effects , Insulin-Like Growth Factor Binding Protein 2/pharmacology , Prefrontal Cortex/drug effects , Psychotropic Drugs/pharmacology , Stress Disorders, Post-Traumatic/drug therapy , Animals , Dendritic Spines/metabolism , Dendritic Spines/pathology , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Fear/drug effects , Fear/physiology , Insulin-Like Growth Factor Binding Protein 2/administration & dosage , Insulin-Like Growth Factor I/administration & dosage , Insulin-Like Growth Factor I/metabolism , Learning/drug effects , Learning/physiology , Male , Memory Consolidation/drug effects , Memory Consolidation/physiology , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Rats, Sprague-Dawley , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/metabolism , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/metabolism , Stress Disorders, Post-Traumatic/metabolism , Stress Disorders, Post-Traumatic/pathology
9.
Curr Neuropharmacol ; 15(1): 35-46, 2017.
Article in English | MEDLINE | ID: mdl-26955966

ABSTRACT

BACKGROUND: Depression affects over 120 million individuals of all ages and is the leading cause of disability worldwide. The lack of objective diagnostic criteria, together with the heterogeneity of the depressive disorder itself, makes it challenging to develop effective therapies. The accumulation of preclinical data over the past 20 years derived from a multitude of models using many divergent approaches, has fueled the resurgence of interest in targeting glutamatergic neurotransmission for the treatment of major depression. OBJECTIVE: The emergence of mechanistic studies are advancing our understanding of the molecular underpinnings of depression. While clearly far from complete and conclusive, they offer the potential to lead to the rational design of more specific therapeutic strategies and the development of safer and more effective rapid acting, long lasting antidepressants. METHODS: The development of comprehensive omics-based approaches to the dysregulation of synaptic transmission and plasticity that underlies the core pathophysiology of MDD are reviewed to illustrate the fundamental elements. RESULTS: This review frames the rationale for the conceptualization of depression as a "pathway disease". As such, it culminates in the call for the development of novel state-of-the-art "-omics approaches" and neurosystems biological techniques necessary to advance our understanding of spatiotemporal interactions associated with targeting glutamatergic-triggered signaling in the CNS. CONCLUSION: These technologies will enable the development of novel psychiatric medications specifically targeted to impact specific, critical intracellular networks in a more focused manner and have the potential to offer new dimensions in the area of translational neuropsychiatry.


Subject(s)
Antidepressive Agents/therapeutic use , Depressive Disorder, Major/drug therapy , Gene Expression Regulation/drug effects , Neuronal Plasticity/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Antidepressive Agents/pharmacology , Depressive Disorder, Major/pathology , Humans
10.
Curr Neuropharmacol ; 15(1): 47-56, 2017.
Article in English | MEDLINE | ID: mdl-26997507

ABSTRACT

BACKGROUND: Rapastinel (GLYX-13) is a NMDA receptor modulator with glycine-site partial agonist properties. It is a robust cognitive enhancer and shows rapid and long-lasting antidepressant properties in both animal models and in humans. METHODS: Rapastinel was derived from a monoclonal antibody, B6B21, is a tetrapeptide (threonine-proline-proline-threonine-amide) obtained from amino acid sequence information obtained from sequencing one of the hypervariable regions of the light chain of B6B21. The in-vivo and in-vitro pharmacology of rapastinel was examined. RESULTS: Rapastinel was found to be a robust cognitive enhancer in a variety of learning and memory paradigms and shows marked antidepressant-like properties in multiple models including the forced swim (Porsolt), learned helplessness and chronic unpredictable stress. Rapastinel's rapid-acting antidepressant properties appear to be mediated by its ability to activate NMDA receptors leading to enhancement in synaptic plasticity processes associated with learning and memory. This is further substantiated by the increase in mature dendritic spines found 24 hrs after rapastinel treatment in both the rat dentate gyrus and layer five of the medial prefrontal cortex. Moreover, ex vivo LTP studies showed that the effects of rapastinel persisted at least two weeks post-dosing. CONCLUSION: These data suggest that rapastinel has significant effects on metaplasticity processes that may help explain the long lasting antidepressant effects of rapastinel seen in the human clinical trial results.


Subject(s)
Depression/drug therapy , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Age Factors , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Depression/pathology , Depression/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Exploratory Behavior/drug effects , Long-Term Potentiation/drug effects , Maze Learning/drug effects , Memory/drug effects , Neuronal Plasticity/drug effects , Oligopeptides/chemistry , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Swimming , Synapses/drug effects , Synapses/ultrastructure , Vocalization, Animal/drug effects
11.
Glycobiology ; 26(12): 1271-1283, 2016 12.
Article in English | MEDLINE | ID: mdl-27510958

ABSTRACT

The mechanism of transcriptional silencing of ST6Gal1 in gliomas has not yet been elucidated. Multiple independent promoters govern the expression of the ST6Gal I gene. Here, we investigated whether epigenetic abnormalities involving DNA methylation affect ST6Gal1 expression. Transcript-specific qRT-PCR following exposure of glioma cell lines to 5-aza-2'-deoxycytidine (5-aza-dC), a DNA methyltransferase inhibitor, resulted in the re-expression of the normally quiescent ST6Gal1 mRNA driven exclusively by the P3 promoter sequence. The P3 promoter-specific transcription start site (TSS) was delineated by primer extension and core promoter sequences and associated functional transcription elements identified by deletion analysis utilizing chloramphenicol acetyltransferase reporter constructs. Minimal promoter activity was found to reside within the first 100 bp of the TSS and maximal activity was controlled by functional AP2 binding sites residing between 400 and 500 bp upstream of the initiation site. As altered AP2 binding was not directly associated with AP2 availability, these analyses demonstrate that ST6Gal1 transcription is regulated by DNA methylation within core promoter regions, ultimately by determining critical transcription factor accessibility within these regions. Transcriptional reactivation of ST6Gal1 expression by 5-aza-dC resulted in increased cell surface α2,6 sialoglycoconjugate expression, increased α2,6 sialylation of ß1 integrin, and decreased adhesion to fibronectin substrate: functional correlates of decreased invasivity. The effects of global hypomethylation are not glycome-wide. Focused glycotranscriptomic analyses of three invasive glioma cell lines following 5-aza-dC treatment demonstrated the modulation of select glycogene transcripts. Taken together, these results demonstrate that epigenetic modulation of ST6Gal1 expression plays a key role in the glioma phenotype in vitro and that that therapeutic approaches targeting elements of the epigenetic machinery for the treatment of human glioblastoma are warranted.


Subject(s)
Antigens, CD/genetics , DNA Methylation/genetics , Glioma/genetics , Sialyltransferases/genetics , Cell Line, Tumor , Glioma/pathology , Humans , Phenotype , Promoter Regions, Genetic/genetics
12.
Int J Neuropsychopharmacol ; 19(2)2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26374350

ABSTRACT

BACKGROUND: Growth factors play an important role in regulating neurogenesis and synapse formation and may be involved in regulating the antidepressant response to conventional antidepressants. To date, Insulin-like growth factor I (IGFI) is the only growth factor that has shown antidepressant properties in human clinical trials. However, its mechanism of action remains unclear. METHODS: The antidepressant-like effect of a single IV dose of IGFI was determined using a chronic unpredictable stress paradigm in the rat Porsolt, sucrose preference, novelty-induced hypophagia, and ultrasonic vocalization models. The dependence of the medial prefrontal cortex for these effects was determined by direct medial prefrontal cortex injection followed by Porsolt testing as well as IGFI receptor activation in the medial prefrontal cortex following an optimal IV antidepressant-like dose of IGFI. The effect of IGFI on synaptic transmission and long-term potentiation (LTP) of synaptic strength was assessed in the hippocampus and medial prefrontal cortex. The dependence of these effects on IGFI and AMPA receptor activation and protein synthesis were also determined. RESULTS: IGFI produced a rapid-acting and long-lasting antidepressant-like effect in each of the depression models. These effects were blocked by IGFI and AMPA receptor antagonists, and medial prefrontal cortex was localized. IGFI robustly increased synaptic strength in the hippocampus and medial prefrontal cortex and these effects were IGFI receptor and protein synthesis-dependent but N-methyl-d-aspartate receptor independent. IGFI also robustly facilitated hippocampal metaplasticity 24 hours postdosing. CONCLUSIONS: These data support the conclusion that the antidepressant-like effects of IGFI are mediated by a persistent, LTP-like enhancement of synaptic strength requiring both IGFIR activation and ongoing protein synthesis.


Subject(s)
Antidepressive Agents/administration & dosage , Hippocampus/physiology , Insulin-Like Growth Factor I/administration & dosage , Long-Term Potentiation/physiology , Prefrontal Cortex/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Dose-Response Relationship, Drug , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Male , Microinjections , Organ Culture Techniques , Prefrontal Cortex/drug effects , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
13.
J Proteome Res ; 14(9): 3932-9, 2015 Sep 04.
Article in English | MEDLINE | ID: mdl-26185906

ABSTRACT

Bone marrow-derived human mesenchymal stem cells (BM-hMSCs) have the innate ability to migrate or home toward and engraft in tumors such as glioblastoma (GBM). Because of this unique property of BM-hMSCs, we have explored their use for cell-mediated therapeutic delivery for the advancement of GBM treatment. Extravasation, the process by which blood-borne cells­such as BM-hMSCs­enter the tissue, is a highly complex process but is heavily dependent upon glycosylation for glycan-glycan and glycan-protein adhesion between the cell and endothelium. However, in a translationally significant preclinical glioma stem cell xenograft (GSCX) model of GBM, BM-hMSCs demonstrate unequal tropism toward these tumors. We hypothesized that there may be differences in the glycan compositions between the GSCXs that elicit homing ("attractors") and those that do not ("non-attractors") that facilitate or impede the engraftment of BM-hMSCs in the tumor. In this study, glycotranscriptomic analysis revealed significant heterogeneity within the attractor phenotype and the enrichment of high mannose type N-glycan biosynthesis in the non-attractor phenotype. Orthogonal validation with topical PNGase F deglycosylation on the tumor regions of xenograft tissue, followed by nLC-ESI-MS, confirmed the presence of increased high mannose type N-glycans in the non-attractors. Additional evidence provided by our glycomic study revealed the prevalence of terminal sialic acid-containing N-glycans in non-attractors and terminal galactose and N-acetyl-glucosamine N-glycans in attractors. Our results provide the first evidence for differential glycomic profiles in attractor and non-attractor GSCXs and extend the scope of molecular determinates in BM-hMSC homing to glioma.


Subject(s)
Gene Expression Profiling/methods , Glioma/metabolism , Glycomics/methods , Mesenchymal Stem Cells/metabolism , Polysaccharides/metabolism , Animals , Glycosylation , Heterografts , Humans , Male , Mannose/metabolism , Mice , Mice, Nude , Polysaccharides/analysis , Polysaccharides/chemistry
14.
Behav Brain Res ; 294: 177-85, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26210936

ABSTRACT

Rapastinel (GLYX-13) is a NMDA receptor modulator with glycine-site partial agonist properties. It is a robust cognitive enhancer and shows rapid and long-lasting antidepressant properties in both animal models and in humans. Contextual fear extinction (CFE) in rodents has been well characterized and used extensively as a model to study the neurobiological mechanisms of post-traumatic stress disorder (PTSD). Since CFE is NMDA receptor modulated and neural circuitry in the medial prefrontal cortex (MPFC) regulates both depression and PTSD, studies were undertaken to examine the effects of rapastinel for its therapeutic potential in PTSD and to use rapastinel as a tool to study its underlying glutamatergic mechanisms. A 21-day chronic mild unpredictable stress (CUS) rat model was used to model depression and PTSD. The effects of CUS alone compared to No CUS controls, and the effects of rapastinel (3 mg/kg IV) on CUS-treated animals were examined. The effect of rapastinel was first assessed using CUS-treated rats in three depression models, Porsolt, sucrose preference, and novelty-induced hypophagia tests, and found to produce a complete reversal of the depressive-like state in each model. Rapastinel was then assessed in a MPFC-dependent positive emotional learning paradigm and in CFE and again a reversal of the impairments induced by CUS treatment was observed. Both synaptic plasticity and metaplasticity, as measured by the induction of long-term potentiation in rat MPFC slice preparations, was found to be markedly impaired in CUS-treated animals. This impairment was reversed when CUS-treated rats were administered rapastinel and tested 24 h later. Transcriptomic analysis of MPFC mRNA expression in CUS-treated rats corroborated the link between rapastinel's behavioral effects and synaptic plasticity. A marked enrichment in both the LTP and LTD connectomes in rapastinel-treated CUS rats was observed compared to CUS-treated controls. The effects of rapastinel on depression models, PEL, and most importantly on CFE demonstrate the therapeutic potential of rapastinel for the treatment of PTSD. Moreover, rapastinel appears to elicit its therapeutic effects through a NMDA receptor-mediated, LTP-like, metaplasticity process in the MPFC.


Subject(s)
Oligopeptides/pharmacology , Prefrontal Cortex/drug effects , Psychotropic Drugs/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Stress Disorders, Post-Traumatic/drug therapy , Animals , Chronic Disease , Depressive Disorder/drug therapy , Depressive Disorder/physiopathology , Disease Models, Animal , Excitatory Amino Acid Agents/pharmacology , Learning/drug effects , Learning/physiology , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Memory/drug effects , Memory/physiology , Prefrontal Cortex/physiopathology , Rats, Sprague-Dawley , Stress Disorders, Post-Traumatic/physiopathology , Stress, Psychological , Tissue Culture Techniques , Transcriptome/drug effects , Transcriptome/physiology , Uncertainty
15.
J Proteome Res ; 14(9): 3415-31, 2015 Sep 04.
Article in English | MEDLINE | ID: mdl-26076068

ABSTRACT

This paper summarizes the recent activities of the Chromosome-Centric Human Proteome Project (C-HPP) consortium, which develops new technologies to identify yet-to-be annotated proteins (termed "missing proteins") in biological samples that lack sufficient experimental evidence at the protein level for confident protein identification. The C-HPP also aims to identify new protein forms that may be caused by genetic variability, post-translational modifications, and alternative splicing. Proteogenomic data integration forms the basis of the C-HPP's activities; therefore, we have summarized some of the key approaches and their roles in the project. We present new analytical technologies that improve the chemical space and lower detection limits coupled to bioinformatics tools and some publicly available resources that can be used to improve data analysis or support the development of analytical assays. Most of this paper's content has been compiled from posters, slides, and discussions presented in the series of C-HPP workshops held during 2014. All data (posters, presentations) used are available at the C-HPP Wiki (http://c-hpp.webhosting.rug.nl/) and in the Supporting Information.


Subject(s)
Chromosome Mapping , Proteins/genetics , Proteome , Chromatography, Liquid , Genomics , Humans , Proteins/chemistry , Tandem Mass Spectrometry
16.
J Proteome Res ; 14(2): 603-8, 2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25369122

ABSTRACT

We describe the utility of integrated strategies that employ both translation of ENCODE data and major proteomic technology pillars to improve the identification of the "missing proteins", novel proteoforms, and PTMs. On one hand, databases in combination with bioinformatic tools are efficiently utilized to establish microarray-based transcript analysis and supply rapid protein identifications in clinical samples. On the other hand, sequence libraries are the foundation of targeted protein identification and quantification using mass spectrometric and immunoaffinity techniques. The results from combining proteoENCODEdb searches with experimental mass spectral data indicate that some alternative splicing forms detected at the transcript level are in fact translated to proteins. Our results provide a step toward the directives of the C-HPP initiative and related biomedical research.


Subject(s)
Proteome/chemistry , Humans , Protein Isoforms/chemistry
17.
Expert Opin Investig Drugs ; 23(2): 243-54, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24251380

ABSTRACT

INTRODUCTION: The N-methyl-d-aspartate receptor-ionophore complex plays a key role in learning and memory and has efficacy in animals and humans with affective disorders. GLYX-13 is an N-methyl-d-aspartate receptor (NMDAR) glycine-site functional partial agonist and cognitive enhancer that also shows rapid antidepressant activity without psychotomimetic side effects. AREAS COVERED: The authors review the mechanism of action of GLYX-13 that was investigated in preclinical studies and evaluated in clinical studies. Specifically, the authors review its pharmacology, pharmacokinetics, and drug safety that were demonstrated in clinical studies. EXPERT OPINION: NMDAR full antagonists can produce rapid antidepressant effects in treatment-resistant subjects; however, they are often accompanied by psychotomimetic effects that make chronic use outside of a clinical trial inpatient setting problematic. GLYX-13 appears to exert its antidepressant effects in the frontal cortex via NMDAR-triggered synaptic plasticity. Understanding the mechanistic underpinning of GLYX-13's antidepressant action should provide both novel insights into the role of the glutamatergic system in depression and identify new targets for therapeutic development.


Subject(s)
Antidepressive Agents/therapeutic use , Depression/drug therapy , Excitatory Amino Acid Agonists/therapeutic use , Oligopeptides/therapeutic use , Receptors, N-Methyl-D-Aspartate/agonists , Animals , Antidepressive Agents/pharmacokinetics , Antidepressive Agents/pharmacology , Cognition/drug effects , Excitatory Amino Acid Agonists/pharmacokinetics , Excitatory Amino Acid Agonists/pharmacology , Glycine/metabolism , Humans , Oligopeptides/pharmacokinetics , Oligopeptides/pharmacology , Treatment Outcome
18.
J Proteome Res ; 13(1): 191-9, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24266786

ABSTRACT

One subproject within the global Chromosome 19 Consortium is to define chromosome 19 gene and protein expression in glioma-derived cancer stem cells (GSCs). Chromosome 19 is notoriously linked to glioma by 1p/19q codeletions, and clinical tests are established to detect that specific aberration. GSCs are tumor-initiating cells and are hypothesized to provide a repository of cells in tumors that can self-replicate and be refractory to radiation and chemotherapeutic agents developed for the treatment of tumors. In this pilot study, we performed RNA-Seq, label-free quantitative protein measurements in six GSC lines, and targeted transcriptomic analysis using a chromosome 19-specific microarray in an additional six GSC lines. The data have been deposited to the ProteomeXchange with identifier PXD000563. Here we present insights into differences in GSC gene and protein expression, including the identification of proteins listed as having no or low evidence at the protein level in the Human Protein Atlas, as correlated to chromosome 19 and GSC subtype. Furthermore, the upregulation of proteins downstream of adenovirus-associated viral integration site 1 (AAVS1) in GSC11 in response to oncolytic adenovirus treatment was demonstrated. Taken together, our results may indicate new roles for chromosome 19, beyond the 1p/19q codeletion, in the future of personalized medicine for glioma patients.


Subject(s)
Brain Neoplasms/metabolism , Chromosomes, Human, Pair 19 , Glioma/metabolism , Neoplastic Stem Cells/metabolism , Proteome , Transcriptome , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Glioma/genetics , Glioma/pathology , Humans , Neoplastic Stem Cells/pathology
19.
Neuropsychopharmacology ; 39(1): 5-23, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23800968

ABSTRACT

Although advances in psychotherapy have been made in recent years, drug discovery for brain diseases such as schizophrenia and mood disorders has stagnated. The need for new biomarkers and validated therapeutic targets in the field of neuropsychopharmacology is widely unmet. The brain is the most complex part of human anatomy from the standpoint of number and types of cells, their interconnections, and circuitry. To better meet patient needs, improved methods to approach brain studies by understanding functional networks that interact with the genome are being developed. The integrated biological approaches--proteomics, transcriptomics, metabolomics, and glycomics--have a strong record in several areas of biomedicine, including neurochemistry and neuro-oncology. Published applications of an integrated approach to projects of neurological, psychiatric, and pharmacological natures are still few but show promise to provide deep biological knowledge derived from cells, animal models, and clinical materials. Future studies that yield insights based on integrated analyses promise to deliver new therapeutic targets and biomarkers for personalized medicine.


Subject(s)
Neuropharmacology/methods , Psychopharmacology/methods , Systems Biology/methods , Animals , Biomarkers , Gene Expression Profiling/methods , Glycomics/methods , Humans , Metabolomics/methods , Models, Biological , Proteomics/methods
20.
Mol Vis ; 19: 1538-53, 2013.
Article in English | MEDLINE | ID: mdl-23878504

ABSTRACT

PURPOSE: This study investigated changes in the transcript levels of genes related to glutamate neurotransmission and transport as diabetes progresses in the Long-Evans rat retina. Transcript levels of vascular endothelial growth factor (VEGF), erythropoietin, and insulin-like growth factor binding protein 3 (IGFBP3) were also measured due to their protective effects on the retinal vasculature and neurons. METHODS: Diabetes was induced in Long-Evans rats with a single intraperitoneal (IP) injection of streptozotocin (STZ; 65 mg/kg) in sodium citrate buffer. Rats with blood glucose >300 mg/dl were deemed diabetic. Age-matched controls received a single IP injection of sodium citrate buffer only. The retinas were dissected at 4 and 12 weeks after induction of diabetes, and mRNA and protein were extracted from the left and right retinas of each rat, respectively. Gene expression was analyzed using quantitative real-time reverse-transcription PCR. Enzyme-linked immunosorbent assay was used to quantify the concentration of VEGF protein in each retina. Statistical significance was determined using 2×2 analysis of variance followed by post-hoc analysis using Fisher's protected least squares difference. RESULTS: Transcript levels of two ionotropic glutamate receptor subunits and one glutamate transporter increased after 4 weeks of diabetes. In contrast, 12 weeks of diabetes decreased the transcript levels of several genes, including two glutamate transporters, four out of five N-methyl-D-aspartate (NMDA) receptor subunits, and all five kainate receptor subunits. Diabetes had a greater effect on gene expression of NMDA and kainate receptor subunits than on the α-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) receptor subunits, for which only GRIA4 significantly decreased after 12 weeks. VEGF protein levels were significantly increased in 4-week diabetic rats compared to age-matched control rats whereas the increase was not significant after 12 weeks. Transcript levels of VEGF and VEGF receptors were unchanged with diabetes. Erythropoietin and IGFBP3 mRNA levels significantly increased at both time points, and IGFBP2 mRNA levels increased after 12 weeks. CONCLUSIONS: Diabetes caused significant changes in the transcriptional expression of genes related to ionotropic glutamate neurotransmission, especially after 12 weeks. Most genes with decreased transcript levels after 12 weeks were expressed by retinal ganglion cells, which include glutamate transporters and ionotropic glutamate receptors. Two genes expressed by retinal ganglion cells but unrelated to glutamate neurotransmission, γ-synuclein (SNCG) and adenosine A1 receptor (ADORA1), also had decreased mRNA expression after 12 weeks. These findings may indicate ganglion cells were lost as diabetes progressed in the retina. Decreased expression of the glutamate transporter SLC1A3 would lead to decreased removal of glutamate from the extracellular space, suggesting that diabetes impairs this function of Müller cells. These findings suggest that ganglion cells were lost due to glutamate excitotoxicity. The changes at 12 weeks occurred without significant changes in retinal VEGF protein or mRNA, although higher VEGF protein levels at 4 weeks may be an early protective response. Increased transcript levels of erythropoietin and IGFBP3 may also be a protective response.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/physiopathology , Gene Expression Regulation , Glutamic Acid/metabolism , Retina/metabolism , Synaptic Transmission/genetics , Animals , Biological Transport/genetics , Blood Glucose/metabolism , Body Weight/genetics , Diabetes Mellitus, Experimental/blood , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Long-Evans , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Retina/pathology , Streptozocin , Transcriptome/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...