Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Discov Ment Health ; 3(1): 19, 2023 Oct 10.
Article in English | MEDLINE | ID: mdl-37861869

ABSTRACT

Positive and negative emotional states in rats can be studied by investigating ultrasonic vocalizations (USVs). Positive affect in rats is indexed by 50 kHz hedonic USVs, and negative affect is indexed by 22 kHz aversive calls. We examined the relationship of emotional states in rats using medial prefrontal cortex (MPFC) quantitative electroencephalograms (qEEG) and found that hedonic USVs were associated with active wake qEEG (high alpha/low delta power), and aversive USVs occurred with groggy wake qEEG (low alpha/high delta). Further, alpha frequency electrical stimulation of the MPFC induces hedonic calls and reward-seeking behavior, whereas delta frequency stimulation produces aversive calls and avoidance behavior. The brain region responsible for generating motor output for USVs, the periaqueductal gray (PAG), shows a motor-evoked potential that is temporally locked to the alpha (hedonic) and delta (aversive) motor-evoked potential. Closed-loop alpha frequency electrical stimulation could prevent delta qEEG and aversive USVs. At the neuronal circuit level, the alpha rhythm was associated with synaptic long-term potentiation (LTP) in the cortex, whereas the delta rhythm was associated with synaptic depotentiation (LTD) in the cortex. At the pharmacological level, NMDAR and growth factor modulation regulated these forms of neuroplasticity. At the single neuron level, excitatory neurons show increased activity in response to alpha frequencies and decreased activity during delta frequencies. In humans, the feeling of joy increased alpha and decreased delta power in frontal scalp qEEG, and the opposite response was seen for sadness. Thus, the synchronization of alpha/delta oscillations through the neuronal circuit responsible for emotional expression coordinates emotional behavior, and the switch between active wake/positive affect and groggy wake/negative affect is under the control of an LTP- LTD synaptic plasticity mechanism.

2.
Mol Psychiatry ; 28(3): 1101-1111, 2023 03.
Article in English | MEDLINE | ID: mdl-36481930

ABSTRACT

We developed an IGFBP2-mimetic peptide fragment, JB2, and showed that it promotes basal synaptic structural and functional plasticity in cultured neurons and mice. We demonstrate that JB2 directly binds to dendrites and synapses, and its biological activity involves NMDA receptor activation, gene transcription and translation, and IGF2 receptors. It is not IGF1 receptor-dependent. In neurons, JB2 induced extensive remodeling of the membrane phosphoproteome. Synapse and cytoskeletal regulation, autism spectrum disorder (ASD) risk factors, and a Shank3-associated protein network were significantly enriched among phosphorylated and dephosphorylated proteins. Haploinsufficiency of the SHANK3 gene on chromosome 22q13.3 often causes Phelan-McDermid Syndrome (PMS), a genetically defined form of autism with profound deficits in motor behavior, sensory processing, language, and cognitive function. We identified multiple disease-relevant phenotypes in a Shank3 heterozygous mouse and showed that JB2 rescued deficits in synaptic function and plasticity, learning and memory, ultrasonic vocalizations, and motor function; it also normalized neuronal excitability and seizure susceptibility. Notably, JB2 rescued deficits in the auditory evoked response latency, alpha peak frequency, and steady-state electroencephalography response, measures with direct translational value to human subjects. These data demonstrate that JB2 is a potent modulator of neuroplasticity with therapeutic potential for the treatment of PMS and ASD.


Subject(s)
Autism Spectrum Disorder , Chromosome Disorders , Humans , Mice , Animals , Autism Spectrum Disorder/genetics , Nerve Tissue Proteins/genetics , Chromosome Deletion , Chromosome Disorders/genetics , Peptides/genetics , Disease Models, Animal , Neuronal Plasticity , Chromosomes, Human, Pair 22/metabolism , Microfilament Proteins/genetics
3.
Int J Neuropsychopharmacol ; 25(12): 979-991, 2022 12 12.
Article in English | MEDLINE | ID: mdl-35882204

ABSTRACT

BACKGROUND: The role of glutamatergic receptors in major depressive disorder continues to be of great interest for therapeutic development. Recent studies suggest that both negative and positive modulation of N-methyl-D-aspartate receptors (NMDAR) can produce rapid antidepressant effects. Here we report that zelquistinel, a novel NMDAR allosteric modulator, exhibits high oral bioavailability and dose-proportional exposures in plasma and the central nervous system and produces rapid and sustained antidepressant-like effects in rodents by enhancing activity-dependent, long-term synaptic plasticity. METHODS: NMDAR-mediated functional activity was measured in cultured rat brain cortical neurons (calcium imaging), hNR2A or B subtype-expressing HEK cells, and synaptic plasticity in rat hippocampal and medial prefrontal cortex slices in vitro. Pharmacokinetics were evaluated in rats following oral administration. Antidepressant-like effects were assessed in the rat forced swim test and the chronic social deficit mouse model. Target engagement and the safety/tolerability profile was assessed using phencyclidine-induced hyperlocomotion and rotarod rodent models. RESULTS: Following a single oral dose, zelquistinel (0.1-100 µg/kg) produced rapid and sustained antidepressant-like effects in the rodent depression models. Brain/ cerebrospinal fluid concentrations associated with zelquistinel antidepressant-like activity also increased NMDAR function and rapidly and persistently enhanced activity-dependent synaptic plasticity (long-term potentiation), suggesting that zelquistinel produces antidepressant-like effects by enhancing NMDAR function and synaptic plasticity. Furthermore, Zelquistinel inhibited phencyclidine (an NMDAR antagonist)-induced hyperlocomotion and did not impact rotarod performance. CONCLUSIONS: Zelquistinel produces rapid and sustained antidepressant effects by positively modulating the NMDARs, thereby enhancing long-term potentiation of synaptic transmission.


Subject(s)
Depressive Disorder, Major , Receptors, N-Methyl-D-Aspartate , Animals , Rats , Mice , Depressive Disorder, Major/drug therapy , Rats, Sprague-Dawley , Antidepressive Agents/therapeutic use , Long-Term Potentiation/physiology , Phencyclidine/pharmacology
4.
Neuroreport ; 33(7): 312-319, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35594441

ABSTRACT

BACKGROUND: A novel N-methyl-D-aspartate receptor (NMDAR) allosteric modulator, rapastinel (RAP, formerly GLYX-13), elicits long-lasting antidepressant-like effects by enhancing long-term potentiation (LTP) of synaptic transmission. RAP elicits these effects by binding to a unique site in the extracellular region of the NMDAR complex, transiently enhancing NMDAR-gated current in pyramidal neurons of both hippocampus and medial prefrontal cortex. METHODS: We compared efficacy of RAP in modulating Schaffer collateral-evoked NMDAR-currents as a function of kinetics of the Ca2+ chelator in the intracellular solution, using whole-cell patch-clamp recordings. The intracellular solution contained either the slow Ca2+ chelator EGTA [3,12-bis(carboxymethyl)-6,9-dioxa-3,12-diazatetradecane-1,14-dioic acid, 0.5 mmol/l] or the 40-500-fold kinetically faster, more selective Ca2+ chelator BAPTA {2,2',2″,2‴-[ethane-1,2-diylbis(oxy-2,1-phenylenenitrilo)] tetraacetic acid, 5 mmol/l}. NMDAR-gated currents were pharmacologically isolated by bath application of the 2-amino-3-(3-hydroxy-5-methyl-isoxazol-4-yl)propanoic acid receptor antagonist 6-nitro-2,3-dioxo-1,2,3,4-tetrahydrobenzo[f]quinoxaline-7-sulfonamide (10 µmol/l) plus the GABA receptor blocker bicuculline (20 µmol/l). RESULTS: When the slow Ca2+ chelator EGTA was in the intracellular solution, RAP elicited significant enhancement of NMDAR-gated current at a 1 µmol/l concentration, and significantly reduced current at 10 µmol/l. In contrast, when recording with the 40-500-fold kinetically faster, more selective Ca2+ chelator BAPTA, NMDAR current increased in magnitude by 84% as BAPTA washed into the cell, and the enhancement of NMDAR current by 1 µmol/l RAP was completely blocked. Interestingly, the reduction in NMDAR current from 10 µmol/l RAP was not affected by the presence of BAPTA in the recording pipette, indicating that this effect is mediated by a different mechanism. CONCLUSION: Extracellular binding of RAP to the NMDAR produces a novel, long-range reduction in affinity of the Ca2+ inactivation site on the NMDAR C-terminus accessible to the intracellular space. This action underlies enhancement in NMDAR-gated conductance elicited by RAP.


Subject(s)
Calcium , Receptors, N-Methyl-D-Aspartate , Chelating Agents/pharmacology , Egtazic Acid/pharmacology , Hippocampus/physiology , Oligopeptides
5.
Neuron ; 110(4): 627-643.e9, 2022 02 16.
Article in English | MEDLINE | ID: mdl-34921780

ABSTRACT

Although many neuronal membrane proteins undergo proteolytic cleavage, little is known about the biological significance of neuronal ectodomain shedding (ES). Here, we show that the neuronal sheddome is detectable in human cerebrospinal fluid (hCSF) and is enriched in neurodevelopmental disorder (NDD) risk factors. Among shed synaptic proteins is the ectodomain of CNTNAP2 (CNTNAP2-ecto), a prominent NDD risk factor. CNTNAP2 undergoes activity-dependent ES via MMP9 (matrix metalloprotease 9), and CNTNAP2-ecto levels are reduced in the hCSF of individuals with autism spectrum disorder. Using mass spectrometry, we identified the plasma membrane Ca2+ ATPase (PMCA) extrusion pumps as novel CNTNAP2-ecto binding partners. CNTNAP2-ecto enhances the activity of PMCA2 and regulates neuronal network dynamics in a PMCA2-dependent manner. Our data underscore the promise of sheddome analysis in discovering neurobiological mechanisms, provide insight into the biology of ES and its relationship with the CSF, and reveal a mechanism of regulation of Ca2+ homeostasis and neuronal network synchrony by a shed ectodomain.


Subject(s)
Autism Spectrum Disorder , Membrane Proteins , Nerve Tissue Proteins , Plasma Membrane Calcium-Transporting ATPases , Autism Spectrum Disorder/cerebrospinal fluid , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Cell Membrane/metabolism , Homeostasis , Humans , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Plasma Membrane Calcium-Transporting ATPases/cerebrospinal fluid , Plasma Membrane Calcium-Transporting ATPases/genetics , Plasma Membrane Calcium-Transporting ATPases/metabolism , Signal Transduction
6.
Brain Sci ; 11(11)2021 Oct 23.
Article in English | MEDLINE | ID: mdl-34827389

ABSTRACT

Since the realization that human emotional experiences and behavior evolved from mammalian ancestors and are evolutionary continuations of animal emotional behavior [...].

7.
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
8.
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
9.
Psychopharmacology (Berl) ; 236(12): 3687-3693, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31392357

ABSTRACT

BACKGROUND: NYX-2925 is a novel N-methyl-D-aspartate receptor (NMDAR) modulator that has been shown to facilitate both NMDAR-dependent long-term potentiation (LTP) in vitro and learning and memory in vivo. OBJECTIVE: The present studies examine the effects of NYX-2925 on NMDAR-dependent auditory LTP (aLTP) in vivo. METHODS: NMDAR-dependent aLTP and NMDAR-dependent auditory mismatch negativity (MMN) was measured, as well as changes in resting-state qEEG power. RESULTS: NYX-2925 (1, 10 mg/kg PO) increased aLTP 1 h after auditory tetanus measured by the post- minus pre-tetanus difference waveform 140-180 ms post tone onset. NYX-2925 (0.1, 1 mg/kg PO) facilitated MMN measured by the difference waveform (i.e., deviant minus standard tones). NYX-2925 (0.1, 1, 10 mg/kg PO) also enhanced resting-state alpha qEEG power. Conversely, the NMDAR glutamate site antagonist CPP (10 mg/kg IP) reduces alpha power and MMN and produces an opposite effect as NYX-2925 on aLTP. CONCLUSIONS: Together, these data suggest that the activation of the NMDAR by NYX-2925 enhances synaptic plasticity in vivo, which may both reduce symptoms of neurological disorders and serve as a biomarker for drug effects. This is the first demonstration of a long-lasting (1-h post-tetanus) effect of NMDAR modulation on synaptic plasticity processes in vivo using a noninvasive technique in freely behaving animals.


Subject(s)
Electroencephalography/methods , Neuronal Plasticity/drug effects , Receptors, N-Methyl-D-Aspartate/physiology , Spiro Compounds/pharmacology , Translational Research, Biomedical/methods , Animals , Electroencephalography/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Learning/drug effects , Learning/physiology , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Memory/drug effects , Memory/physiology , Neuronal Plasticity/physiology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/agonists
10.
J Pharmacol Exp Ther ; 366(3): 485-497, 2018 09.
Article in English | MEDLINE | ID: mdl-29986951

ABSTRACT

NYX-2925 [(2S,3R)-3-hydroxy-2-((R)-5-isobutyryl-1-oxo-2,5-diazaspiro[3.4]octan-2-yl)butanamide] is a novel N-methyl-d-aspartate (NMDA) receptor modulator that is currently being investigated in phase 2 clinical studies for the treatment of painful diabetic peripheral neuropathy and fibromyalgia. Previous studies demonstrated that NYX-2925 is a member of a novel class of NMDA receptor-specific modulators that affect synaptic plasticity processes associated with learning and memory. Studies here examined NYX-2925 administration in rat peripheral chronic constriction nerve injury (CCI) and streptozotocin-induced diabetic mechanical hypersensitivity. Additionally, NYX-2925 was examined in formalin-induced persistent pain model and the tail flick test of acute nociception. Oral administration of NYX-2925 resulted in rapid and long-lasting analgesia in both of the neuropathic pain models and formalin-induced persistent pain, but was ineffective in the tail flick model. The analgesic effects of NYX-2925 were blocked by the systemic administration of NMDA receptor antagonist 3-(2-carboxypiperazin-4-yl)propyl-1-phosphonic acid. Microinjection of NYX-2925 into the medial prefrontal cortex of CCI rats resulted in analgesic effects similar to those observed following systemic administration, whereas intrathecal administration of NYX-2925 was ineffective. In CCI animals, NYX-2925 administration reversed deficits seen in a rat model of rough-and-tumble play. Thus, it appears that NYX-2925 may have therapeutic potential for the treatment of neuropathic pain, and the data presented here support the idea that NYX-2925 may act centrally to ameliorate pain and modulate negative affective states associated with chronic neuropathic pain.


Subject(s)
Analgesics/pharmacology , Neuralgia/drug therapy , Neuralgia/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Spiro Compounds/pharmacology , Analgesics/therapeutic use , Animals , Disease Models, Animal , Male , Rats , Rats, Sprague-Dawley , Spiro Compounds/therapeutic use , Vocalization, Animal/drug effects
11.
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
12.
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
13.
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
14.
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
15.
Curr Neuropharmacol ; 15(1): 3-10, 2017.
Article in English | MEDLINE | ID: mdl-27102428

ABSTRACT

BACKGROUND: Positive emotions have been shown to induce resilience to depression and anxiety 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 USVs) reflect a positive affective state and are best elicited by rough-and-tumble play. METHODS: The effect of positive affect induced by rough-and tumble play was examined on models of depression and learning and memory. The molecular and pharmacological basis of play induced positive affect was also examined. RESULTS: Rough-and-tumble play induced Hedonic 50-kHz USVs, lead to resilience to depression and anxiety, and facilitation of learning and memory. These effects are mediated, in part, by increased NMDAR expression and activation in the medial prefrontal cortex. CONCLUSIONS: We hypothesize that positive affect induces resilience to depression by facilitating NMDAR-dependent synaptic plasticity in the medial prefrontal cortex. Targeting MPFC synaptic plasticity may lead to novel treatments for depression.


Subject(s)
Anxiety/etiology , Depression/etiology , Emotions/physiology , Neuronal Plasticity/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Anxiety/drug therapy , Depression/drug therapy , Disease Models, Animal , Emotions/drug effects , Gene Expression Regulation/drug effects , Humans , Learning/drug effects , Learning/physiology , Neuronal Plasticity/drug effects , Rats , Receptors, N-Methyl-D-Aspartate/genetics , Vocalization, Animal/drug effects , Vocalization, Animal/physiology
16.
Behav Brain Res ; 299: 105-10, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26632337

ABSTRACT

GLYX-13 (rapastinel), a tetrapeptide (Thr-Pro-Pro-Thr-amide), has been reported to have fast acting antidepressant properties in man based upon its N-methyl-D-aspartate receptor (NMDAR) glycine site functional partial agonism. Ketamine, a non-competitive NMDAR antagonist, also reported to have fast acting antidepressant properties, produces cognitive impairment in rodents and man, whereas rapastinel has been reported to have cognitive enhancing properties in rodents, without impairing cognition in man, albeit clinical testing has been limited. The goal of this study was to compare the cognitive impairing effects of rapastinel and ketamine in novel object recognition (NOR), a measure of declarative memory, in male C57BL/6J mice treated with phencyclidine (PCP), another NMDAR noncompetitive antagonist known to severely impair cognition, in both rodents and man. C57BL/6J mice given a single dose or subchronic ketamine (30 mg/kg.i.p.) showed acute or persistent deficits in NOR, respectively. Acute i.v. rapastinel (1.0 mg/kg), did not induce NOR deficit. Pre-treatment with rapastinel significantly prevented acute ketamine-induced NOR deficit. Rapastinel (1.0 mg/kg, but not 0.3 mg/kg, iv) significantly reversed both subchronic ketamine- and subchronic PCP-induced NOR deficits. Rapastinel also potentiated the atypical antipsychotic drug with antidepressant properties, lurasidone, to restore NOR in subchronic ketamine-treated mice. These findings indicate that rapastinel, unlike ketamine, does not induce a declarative memory deficit in mice, and can prevent or reverse the ketamine-induced NOR deficit. Further study is required to determine if these differences translate during clinical use of ketamine and rapastinel as fast acting antidepressant drugs and if rapastinel could have non-ionotropic effects as an add-on therapy with antipsychotic/antidepressant medications.


Subject(s)
Excitatory Amino Acid Antagonists/administration & dosage , Memory Disorders/drug therapy , Oligopeptides/administration & dosage , Animals , Cognition/drug effects , Disease Models, Animal , Ketamine , Male , Memory Disorders/chemically induced , Mice , Mice, Inbred C57BL , Phencyclidine , Recognition, Psychology/physiology
17.
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
18.
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
19.
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
20.
Behav Brain Res ; 251: 18-24, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23623884

ABSTRACT

Early childhood autism is characterized by deficits in social approach and play behaviors, socio-emotional relatedness, and communication/speech abnormalities, as well as repetitive behaviors. These core neuropsychological features of autism can be modeled in laboratory rats, and the results may be useful for drug discovery and therapeutic development. We review data that show that rats selectively bred for low rates of play-related pro-social ultrasonic vocalizations (USVs) can be used to model social deficit symptoms of autism. Low-line animals engage in less social contact time with conspecifics, show lower rates of play induced pro-social USVs, and show an increased proportion of non-frequency modulated (i.e. monotonous) ultrasonic vocalizations compared to non-selectively bred random-line animals. Gene expression patterns in the low-line animals show significant enrichment in autism-associated genes, and the NMDA receptor family was identified as a significant hub. Treatment of low-line animals with the NMDAR functional glycine site partial agonist, GLYX-13, rescued the deficits in play-induced pro-social 50-kHz USVs and reduced monotonous USVs. Since the NMDA receptor has been implicated in the genesis of autistic symptoms, it is possible that GLYX-13 may be of therapeutic value in the treatment of autism.


Subject(s)
Child Development Disorders, Pervasive/genetics , Play and Playthings , Receptors, N-Methyl-D-Aspartate/genetics , Social Behavior , Vocalization, Animal/physiology , Animals , Breeding , Child , Child Development Disorders, Pervasive/metabolism , Child Development Disorders, Pervasive/physiopathology , Humans , Rats , Receptors, N-Methyl-D-Aspartate/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...