Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 181(9): 1421-1437, 2024 May.
Article in English | MEDLINE | ID: mdl-38044332

ABSTRACT

BACKGROUND AND PURPOSE: Chronic pain remains a major clinical problem that needs effective therapeutic agents. Glutamate delta 1 (GluD1) receptors and the protein cerebellin 1 (Cbln1) are down-regulated in the central amygdala (CeA) in models of inflammatory and neuropathic pain. One treatment with Cbln1, intracerebroventricularly (ICV) or in CeA, normalized GluD1 and reduced AMPA receptor expression, resulting in lasting (7-10 days) pain relief. Unlike many CNS-targeting biological agents, the structure of Cbln1 suggests potential blood-brain barrier penetration. Here, we have tested whether systemic administration of Cbln1 provides analgesic effects via action in the CNS. EXPERIMENTAL APPROACH: Analgesic effects of intravenous recombinant Cbln1 was assessed in complete Freund's adjuvant inflammatory pain model in mice. GluD1 knockout and a mutant form of Cbln1 were used. KEY RESULTS: A single intravenous injection of Cbln1 mitigated nocifensive and averse behaviour in both inflammatory and neuropathic pain models. This effect of Cbln1 was dependent on GluD1 receptors and required binding to the amino terminal domain of GluD1. Time course of analgesic effect was similar to previously reported ICV and intra-CeA injection. GluD1 in both spinal cord and CeA was down -regulated in the inflammatory pain model, whereas GluD1 expression in spinal cord but not in CeA, was partly normalized by intravenous Cbln1. Importantly, recombinant Cbln1 was detected in the synaptoneurosomes in spinal cord but not in the CeA. CONCLUSIONS AND IMPLICATIONS: Our results describe a novel mechanism by which systemic Cbln1 induces analgesia potentially by central actions involving normalization of signalling by spinal cord GluD1 receptors.


Subject(s)
Chronic Pain , Nerve Tissue Proteins , Neuralgia , Mice , Animals , Chronic Pain/drug therapy , Glutamic Acid , Receptors, Glutamate , Neuralgia/drug therapy , Analgesics/therapeutic use
2.
Neurobiol Dis ; 181: 106117, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37031803

ABSTRACT

Thalamic regulation of cortical function is important for several behavioral aspects including attention and sensorimotor control. This region has also been studied for its involvement in seizure activity. Among the NMDA receptor subunits GluN2C and GluN2D are particularly enriched in several thalamic nuclei including nucleus reticularis of the thalamus (nRT). We have previously found that GluN2C deletion does not have a strong influence on the basal excitability and burst firing characteristics of reticular thalamus neurons. Here we find that GluN2D ablation leads to reduced depolarization-induced spike frequency and reduced hyperpolarization-induced rebound burst firing in nRT neurons. Furthermore, reduced inhibitory neurotransmission was observed in the ventrobasal thalamus (VB). A model with preferential downregulation of GluN2D from parvalbumin (PV)-positive neurons was generated. Conditional deletion of GluN2D from PV neurons led to a decrease in excitability and burst firing. In addition, reduced excitability and burst firing was observed in the VB neurons together with reduced inhibitory neurotransmission. Finally, young mice with GluN2D downregulation in PV neurons showed significant resistance to pentylenetetrazol-induced seizure and differences in sensitivity to isoflurane anesthesia but were normal in other behaviors. Conditional deletion of GluN2D from PV neurons also affected expression of other GluN2 subunits and GABA receptor in the nRT. Together, these results identify a unique role of GluN2D-containing receptors in the regulation of thalamic circuitry and seizure susceptibility which is relevant to mutations in GRIN2D gene found to be associated with pediatric epilepsy.


Subject(s)
Receptors, N-Methyl-D-Aspartate , Thalamus , Animals , Mice , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Seizures/metabolism , Synaptic Transmission , Thalamic Nuclei/metabolism , Thalamus/metabolism
3.
Biol Psychiatry ; 94(4): 297-309, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37004850

ABSTRACT

BACKGROUND: Parvalbumin interneuron (PVI) activity synchronizes the medial prefrontal cortex circuit for normal cognitive function, and its impairment may contribute to schizophrenia (SZ). NMDA receptors in PVIs participate in these activities and form the basis for the NMDA receptor hypofunction hypothesis of SZ. However, the role of the GluN2D subunit, which is enriched in PVIs, in regulating molecular networks relevant to SZ is unknown. METHODS: Using electrophysiology and a mouse model with conditional deletion of GluN2D from PVIs (PV-GluN2D knockout [KO]), we examined the cell excitability and neurotransmission in the medial prefrontal cortex. Histochemical, RNA sequencing analysis and immunoblotting were conducted to understand molecular mechanisms. Behavioral analysis was conducted to test cognitive function. RESULTS: PVIs in the medial prefrontal cortex were found to express putative GluN1/2B/2D receptors. In a PV-GluN2D KO model, PVIs were hypoexcitable, whereas pyramidal neurons were hyperexcitable. Excitatory neurotransmission was higher in both cell types in PV-GluN2D KO, whereas inhibitory neurotransmission showed contrasting changes, which could be explained by reduced somatostatin interneuron projections and increased PVI projections. Genes associated with GABA (gamma-aminobutyric acid) synthesis, vesicular release, and uptake as well as those involved in formation of inhibitory synapses, specifically GluD1-Cbln4 and Nlgn2, and regulation of dopamine terminals were downregulated in PV-GluN2D KO. SZ susceptibility genes including Disc1, Nrg1, and ErbB4 and their downstream targets were also downregulated. Behaviorally, PV-GluN2D KO mice showed hyperactivity and anxiety behavior and deficits in short-term memory and cognitive flexibility. CONCLUSIONS: These findings demonstrate that GluN2D in PVIs serves as a point of convergence of pathways involved in the regulation of GABAergic synapses relevant to SZ.


Subject(s)
Parvalbumins , Schizophrenia , Animals , Mice , Interneurons/physiology , Mice, Knockout , Nerve Tissue Proteins/metabolism , Parvalbumins/metabolism , Prefrontal Cortex/metabolism , Receptor, ErbB-4/metabolism , Schizophrenia/genetics , Schizophrenia/metabolism
4.
Sci Adv ; 8(29): eabo6574, 2022 Jul 22.
Article in English | MEDLINE | ID: mdl-35867797

ABSTRACT

Cocaine-associated memories induce cravings and interfere with the ability of users to cease cocaine use. Reducing the strength of cue-drug memories by facilitating extinction may have therapeutic value for the treatment of cocaine addiction. Here, we demonstrate the expression of GluN1/2A/2C NMDA receptor currents in astrocytes in the nucleus accumbens core. Selective ablation of GluN1 subunit from astrocytes in the nucleus accumbens enhanced extinction of cocaine preference memory but did not affect cocaine conditioning or reinstatement. Repeated cocaine exposure up-regulated GluN2C subunit expression and increased astrocytic NMDA receptor currents. Furthermore, intra-accumbal inhibition of GluN2C/2D-containing receptors and GluN2C subunit deletion facilitated extinction of cocaine memory. Cocaine-induced neuroadaptations including dendritic spine maturation and AMPA receptor recruitment were absent in GluN2C knockout mice. Impaired retention of cocaine preference memory in GluN2C knockout mice was restored by exogenous administration of recombinant glypican 4. Together, these results identify a previously unknown astrocytic GluN2C-containing NMDA receptor mechanism underlying maintenance of cocaine preference memory.

5.
Pharmacol Res ; 178: 106144, 2022 04.
Article in English | MEDLINE | ID: mdl-35304260

ABSTRACT

The glutamate delta family of receptors is composed of GluD1 and GluD2 and serve as synaptic organizers. We have previously demonstrated several autism-like molecular and behavioral phenotypes including an increase in dendritic spines in GluD1 knockout mice. Based on previous reports we evaluated whether disruption of autophagy mechanisms may account for these phenotypes. Mouse model with conditional deletion of GluD1 from excitatory neurons in the corticolimbic regions was utilized. GluD1 loss led to overactive Akt-mTOR pathway, higher p62 and a lower LC3-II/LC3-I ratio in the somatosensory cortex suggesting reduced autophagy. Excitatory elements were increased in number but had immature phenotype based on puncta size, lower AMPA subunit GluA1 expression and impaired development switch from predominantly GluN2B to mixed GluN2A/GluN2B subunit expression. Overactive Akt-mTOR signaling and impaired autophagy was also observed in dorsal striatum upon conditional ablation of GluD1 and in the prefrontal cortex and hippocampus in constitutive knockout. Finally, cognitive deficits in novel object recognition test and fear conditioning were observed in mice with conditional ablation of GluD1 from the corticolimbic regions. Together, these results demonstrate a novel function of GluD1 in the regulation of autophagy pathway which may underlie autism phenotypes and is relevant to the genetic association of GluD1 coding, GRID1 gene with autism and other developmental disorders.


Subject(s)
Glutamic Acid , Receptors, Glutamate , Somatosensory Cortex , Animals , Autophagy , Mice , Mice, Knockout , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Glutamate/genetics , Receptors, Glutamate/metabolism , Somatosensory Cortex/metabolism , Synapses/physiology , TOR Serine-Threonine Kinases/metabolism
6.
Cells ; 10(10)2021 10 03.
Article in English | MEDLINE | ID: mdl-34685624

ABSTRACT

Chronic pain is a debilitating condition involving neuronal dysfunction, but the synaptic mechanisms underlying the persistence of pain are still poorly understood. We found that the synaptic organizer glutamate delta 1 receptor (GluD1) is expressed postsynaptically at parabrachio-central laterocapsular amygdala (PB-CeLC) glutamatergic synapses at axo-somatic and punctate locations on protein kinase C δ -positive (PKCδ+) neurons. Deletion of GluD1 impairs excitatory neurotransmission at the PB-CeLC synapses. In inflammatory and neuropathic pain models, GluD1 and its partner cerebellin 1 (Cbln1) are downregulated while AMPA receptor is upregulated. A single infusion of recombinant Cbln1 into the central amygdala led to sustained mitigation of behavioral pain parameters and normalized hyperexcitability of central amygdala neurons. Cbln2 was ineffective under these conditions and the effect of Cbln1 was antagonized by GluD1 ligand D-serine. The behavioral effect of Cbln1 was GluD1-dependent and showed lateralization to the right central amygdala. Selective ablation of GluD1 from the central amygdala or injection of Cbln1 into the central amygdala in normal animals led to changes in averse and fear-learning behaviors. Thus, GluD1-Cbln1 signaling in the central amygdala is a teaching signal for aversive behavior but its sustained dysregulation underlies persistence of pain. Significance statement: Chronic pain is a debilitating condition which involves synaptic dysfunction, but the underlying mechanisms are not fully understood. Our studies identify a novel mechanism involving structural synaptic changes in the amygdala caused by impaired GluD1-Cbln1 signaling in inflammatory and neuropathic pain behaviors. We also identify a novel means to mitigate pain in these conditions using protein therapeutics.


Subject(s)
Central Amygdaloid Nucleus/metabolism , Chronic Pain/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Glutamate/metabolism , Signal Transduction , Synapses/metabolism , Animals , Behavior, Animal , Chronic Pain/complications , Chronic Pain/physiopathology , Disease Models, Animal , Down-Regulation , Female , Inflammation/complications , Inflammation/pathology , Male , Mice, Knockout , Nociception/drug effects , Rats , Recombinant Proteins/pharmacology , Synaptic Transmission
7.
Int J Neuropsychopharmacol ; 24(11): 907-919, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34363482

ABSTRACT

BACKGROUND: Enhancement of N-methyl-D-aspartate (NMDA) receptor function using glycine-site agonist D-cycloserine is known to facilitate fear extinction, providing a means to augment cognitive behavioral therapy in anxiety disorders. A novel class of glycine-site agonists has recently been identified, and we have found that the prototype, AICP, is more effective than D-cycloserine in modulating neuronal function. METHODS: Using novel glycine-site agonist AICP, local infusion studies, and genetic models, we elucidated the role of GluN2C-containing receptors in fear extinction. RESULTS: We tested the effect of intracerebroventricular injection of AICP on fear extinction and found a robust facilitation of fear extinction. This effect was dependent on GluN2C subunit, consistent with superagonist action of AICP at GluN2C-containing receptors. Local infusion studies in wild-type and GluN2C knockout mice suggested that AICP produces its effect via GluN2C-containing receptors in the basolateral amygdala (BLA). Furthermore, consistent with astrocytic expression of GluN2C subunit in the amygdala, we found that AICP did not facilitate fear extinction in mice with conditional deletion of obligatory GluN1 subunit from astrocytes. Importantly, chemogenetic activation of astrocytes in the basolateral amygdala facilitated fear extinction. Acutely, AICP was found to facilitate excitatory neurotransmission in the BLA via presynaptic GluN2C-dependent mechanism. Immunohistochemical studies suggest that AICP-mediated facilitation of fear extinction involves synaptic insertion of α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor GluA1 subunit. CONCLUSION: These results identify a unique role of astrocytic NMDA receptors composed of GluN2C subunit in extinction of conditioned fear memory and demonstrate that further development of recently identified superagonists of GluN2C-containing receptors may have utility for anxiety disorders.


Subject(s)
Amygdala/drug effects , Astrocytes/metabolism , Extinction, Psychological/drug effects , Fear/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Basolateral Nuclear Complex/metabolism , Conditioning, Psychological/drug effects , Cycloserine/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Mice , Receptors, AMPA/metabolism , Synaptic Transmission/drug effects , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism
8.
Mol Neurobiol ; 58(10): 4787-4801, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34173171

ABSTRACT

Glutamate delta-1 receptor (GluD1) is a member of the ionotropic glutamate receptor family expressed at excitatory synapses and functions as a synaptogenic protein by interacting with presynaptic neurexin. We have previously shown that GluD1 plays a role in the maintenance of excitatory synapses in a region-specific manner. Loss of GluD1 leads to reduced excitatory neurotransmission in medium spiny neurons (MSNs) in the dorsal striatum, but not in the ventral striatum (both core and shell of the nucleus accumbens (NAc)). Here, we found that GluD1 loss leads to reduced inhibitory neurotransmission in MSNs of the NAc core as evidenced by a reduction in the miniature inhibitory postsynaptic current frequency and amplitude. Presynaptic effect of GluD1 loss was further supported by an increase in paired pulse ratio of evoked inhibitory responses indicating reduced release probability. Furthermore, analysis of GAD67 puncta indicated a reduction in the number of putative inhibitory terminals. The changes in mIPSC were independent of cannabinoid or dopamine signaling. A role of feed-forward inhibition was tested by selective ablation of GluD1 from PV neurons which produced modest reduction in mIPSCs. Behaviorally, local ablation of GluD1 from NAc led to hypolocomotion and affected anxiety- and depression-like behaviors. When GluD1 was ablated from the dorsal striatum, several behavioral phenotypes were altered in opposite manner compared to GluD1 ablation from NAc. Our findings demonstrate that GluD1 regulates inhibitory neurotransmission in the NAc by a combination of pre- and postsynaptic mechanisms which is critical for motor control and behaviors relevant to neuropsychiatric disorders.


Subject(s)
Anxiety/metabolism , Glutamate Dehydrogenase/biosynthesis , Inhibitory Postsynaptic Potentials/physiology , Neural Inhibition/physiology , Nucleus Accumbens/metabolism , Synaptic Transmission/physiology , Animals , Anxiety/genetics , Excitatory Amino Acid Antagonists/pharmacology , Glutamate Dehydrogenase/antagonists & inhibitors , Glutamate Dehydrogenase/genetics , Inhibitory Postsynaptic Potentials/drug effects , Locomotion/drug effects , Locomotion/physiology , Male , Mice , Mice, Knockout , Neural Inhibition/drug effects , Nucleus Accumbens/drug effects , Social Interaction/drug effects , Synaptic Transmission/drug effects
9.
Neurobiol Dis ; 150: 105254, 2021 03.
Article in English | MEDLINE | ID: mdl-33421565

ABSTRACT

Globus pallidus externa (GPe) is a nucleus in the basal ganglia circuitry involved in the control of movement. Recent studies have demonstrated a critical role of GPe cell types in Parkinsonism. Specifically increasing the function of parvalbumin (PV) neurons in the GPe has been found to facilitate motor function in a mouse model of Parkinson's disease (PD). The knowledge of contribution of NMDA receptors to GPe function is limited. Here, we demonstrate that fast spiking neurons in the GPe express NMDA receptor currents sensitive to GluN2C/GluN2D-selective inhibitors and glycine site agonist with higher efficacy at GluN2C-containing receptors. Furthermore, using a novel reporter model, we demonstrate the expression of GluN2C subunits in PV neurons in the GPe which project to subthalamic nuclei. GluN2D subunit was also found to localize to PV neurons in GPe. Ablation of GluN2C subunit does not affect spontaneous firing of fast spiking neurons. In contrast, facilitating the function of GluN2C-containing receptors using glycine-site NMDA receptor agonists, D-cycloserine (DCS) or AICP, increased the spontaneous firing frequency of PV neurons in a GluN2C-dependent manner. Finally, we demonstrate that local infusion of DCS or AICP into the GPe improved motor function in a mouse model of PD. Together, these results demonstrate that GluN2C-containing receptors and potentially GluN2D-containing receptors in the GPe may serve as a therapeutic target for alleviating motor dysfunction in PD and related disorders.


Subject(s)
Globus Pallidus/metabolism , Movement/physiology , Neurons/metabolism , Parkinsonian Disorders/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Cycloserine/pharmacology , Disease Models, Animal , Globus Pallidus/cytology , Mice , Motor Activity , Movement/drug effects , Parkinsonian Disorders/physiopathology , Parvalbumins/metabolism , Patch-Clamp Techniques , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Subthalamic Nucleus
10.
J Comp Neurol ; 529(7): 1703-1718, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33084025

ABSTRACT

The glutamate receptor delta 1 (GluD1) is strongly expressed in the striatum. Knockout of GluD1 expression in striatal neurons elicits cognitive deficits and disrupts the thalamostriatal system in mice. To understand the potential role of GluD1 in the primate striatum, we compared the cellular and subcellular localization of striatal GluD1 immunoreactivity (GluD1-IR) in mice and monkeys. In both species, striatal GluD1-IR displayed a patchy pattern of distribution in register with the striosome/matrix compartmentation, but in an opposite fashion. While GluD1 was more heavily expressed in the striosomes than the matrix in the monkey caudate nucleus, the opposite was found in the mouse striatum. At the electron microscopic level, GluD1-IR was preferentially expressed in dendritic shafts (47.9 ± 1.2%), followed by glia (37.7 ± 2.5%), and dendritic spines (14.3 ± 2.6%) in the matrix of the mouse striatum. This pattern was not statistically different from the labeling in the striosome and matrix compartments of the monkey caudate nucleus, with the exception of a small amount of GluD1-positive unmyelinated axons and axon terminals in the primate striatum. Immunogold staining revealed synaptic and perisynaptic GluD1 labeling at putative axo-dendritic and axo-spinous glutamatergic synapses, and intracellular labeling on the surface of mitochondria. Confocal microscopy showed that GluD1 is preferentially colocalized with thalamic over cortical terminals in both the striosome and matrix compartments. These data provide the anatomical substrate for a deeper understanding of GluD1 regulation of striatal glutamatergic synapses, but also suggest possible extrasynaptic, glial, and mitochondrial GluD1 functions.


Subject(s)
Corpus Striatum/metabolism , Receptors, Glutamate/metabolism , Animals , Macaca mulatta , Male , Mice
11.
Neurobiol Dis ; 137: 104746, 2020 04.
Article in English | MEDLINE | ID: mdl-31945419

ABSTRACT

Impaired behavioral flexibility and repetitive behavior is a common phenotype in autism and other neuropsychiatric disorders, but the underlying synaptic mechanisms are poorly understood. The trans-synaptic glutamate delta (GluD)-Cerebellin 1-Neurexin complex, critical for synapse formation/maintenance, represents a vulnerable axis for neuropsychiatric diseases. We have previously found that GluD1 deletion results in reversal learning deficit and repetitive behavior. In this study, we show that selective ablation of GluD1 from the dorsal striatum impairs behavioral flexibility in a water T-maze task. We further found that striatal GluD1 is preferentially found in dendritic shafts, and more frequently associated with thalamic than cortical glutamatergic terminals suggesting localization to projections from the thalamic parafascicular nucleus (Pf). Conditional deletion of GluD1 from the striatum led to a selective loss of thalamic, but not cortical, terminals, and reduced glutamatergic neurotransmission. Optogenetic studies demonstrated functional changes at thalamostriatal synapses from the Pf, but no effect on the corticostriatal system, upon ablation of GluD1 in the dorsal striatum. These studies suggest a novel molecular mechanism by which genetic variations associated with neuropsychiatric disorders may impair behavioral flexibility, and reveal a unique principle by which GluD1 subunit regulates forebrain circuits.


Subject(s)
Behavior, Animal/physiology , Corpus Striatum/metabolism , Receptors, Glutamate/metabolism , Thalamus/metabolism , Animals , Corpus Striatum/physiology , Female , Male , Mice , Neurogenesis/physiology , Synapses/physiology , Synaptic Transmission/physiology , Thalamus/physiopathology
12.
Mol Pharmacol ; 2019 Jun 03.
Article in English | MEDLINE | ID: mdl-31160332

ABSTRACT

The GluN2C subunit of the NMDA receptor is enriched in the neurons in nucleus reticularis of the thalamus (nRT), but its role in regulating their function is not well understood. We found that deletion of GluN2C subunit did not affect spike frequency in response to depolarizing current injection or hyperpolarization-induced rebound burst firing of nRT neurons. D-cycloserine or CIQ (GluN2C/GluN2D positive allosteric modulator) did not affect the depolarization-induced spike frequency in nRT neurons. A newly identified highly potent and efficacious co-agonist of GluN1/GluN2C NMDA receptors, AICP, was found to reduce the spike frequency and burst firing of nRT neurons in wildtype but not GluN2C knockout. This effect was potentially due to facilitation of GluN2C-containing receptors because inhibition of NMDA receptors by AP5 did not affect spike frequency in nRT neurons. We evaluated the effect of intracerebroventricular injection of AICP. AICP did not affect basal locomotion or prepulse inhibition but facilitated MK-801-induced hyperlocomotion. This effect was observed in wildtype but not in GluN2C knockout mice demonstrating that AICP produces GluN2C-selective effects in vivo Using a chemogenetic approach we examined the role of nRT in this behavioral effect. Gq or Gi coupled DREADDs were selectively expressed in nRT neurons using cre-dependent viral vectors and PV-Cre mouse line. We found that similar to AICP effect, activation of Gq but not Gi coupled DREADD facilitated MK-801-induced hyperlocomotion. Together, these results identify a unique role of GluN2C-containing receptors in the regulation of nRT neurons and suggest GluN2C-selective in vivo targeting of NMDA receptors by AICP. SIGNIFICANCE STATEMENT: The nucleus reticularis of the thalamus composed of GABAergic neurons is termed as guardian of the gateway and is an important regulator of corticothalamic communication which may be impaired in autism, non-convulsive seizures and other conditions. We found that strong facilitation of tonic activity of GluN2C subtype of NMDA receptors using AICP, a newly identified glycine-site agonist of NMDA receptors, modulates the function of reticular thalamus neurons. AICP was also able to produce GluN2C-dependent behavioral effects in vivo. Together, these finding identify a novel mechanism and a pharmacological tool to modulate activity of reticular thalamic neurons in disease states.

13.
Sci Rep ; 9(1): 7572, 2019 05 20.
Article in English | MEDLINE | ID: mdl-31110197

ABSTRACT

The GluN2C- and GluN2D-containing NMDA receptors are distinct from GluN2A- and GluN2B-containing receptors in many aspects including lower sensitivity to Mg2+ block and lack of desensitization. Recent studies have highlighted the unique contribution of GluN2C and GluN2D subunits in various aspects of neuronal and circuit function and behavior, however a direct comparison of the effect of ablation of these subunits in mice on pure background strain has not been conducted. Using knockout-first strains for the GRIN2C and GRIN2D produced on pure C57BL/6N strain, we compared the effect of partial or complete ablation of GluN2C and GluN2D subunit on various behaviors relevant to mental disorders. A large number of behaviors described previously in GluN2C and GluN2D knockout mice were reproduced in these mice, however, some specific differences were also observed possibly representing strain effects. We also examined the response to NMDA receptor channel blockers in these mouse strains and surprisingly found that unlike previous reports GluN2D knockout mice were not resistant to phencyclidine-induced hyperlocomotion. Interestingly, the GluN2C knockout mice showed reduced sensitivity to phencyclidine-induced hyperlocomotion. We also found that NMDA receptor channel blocker produced a deficit in prepulse inhibition which was prevented by a GluN2C/2D potentiator in wildtype and GluN2C heterozygous mice but not in GluN2C knockout mice. Together these results demonstrate a unique role of GluN2C subunit in schizophrenia-like behaviors.


Subject(s)
Gene Deletion , Receptors, N-Methyl-D-Aspartate/genetics , Schizophrenia/genetics , Animals , Anxiety/genetics , Depression/genetics , Disease Models, Animal , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Schizophrenia/chemically induced
14.
Transl Psychiatry ; 8(1): 219, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30315226

ABSTRACT

Cocaine exposure induces plasticity of glutamatergic synapses of medium spiny neurons (MSNs) in the nucleus accumbens (NAc), which has been proposed to contribute to its addictive behavior. The mechanisms underlying cocaine-induced plasticity are not fully understood. The orphan glutamate delta-1 (GluD1) receptor is a member of the ionotropic glutamate receptor family but does not function as a typical ligand-gated ion channel. Instead it serves a synaptogenic function by interacting with presynaptic Neurexin protein. Recent neuroanatomical studies have demonstrated enriched expression of GluD1 in the NAc but its role in reward behavior, MSN function, and drug-induced plasticity remains unknown. Using a combination of constitutive and conditional GluD1 KO models, we evaluated the effect of GluD1 ablation on cocaine-conditioned place preference (CPP) and cocaine-induced structural and functional plasticity. GluD1 KO mice showed higher cocaine CPP. Selective ablation of GluD1 from striatal neurons but not cortico-limbic excitatory neurons reproduced higher CPP. Higher cocaine preference in GluD1 KO correlated with an increase in spine density, greater maturation of dendritic spines, and basally upregulated spine-regulating active cofilin. GluD1 loss did not affect basal excitatory neurotransmission or plasticity but masked the generation of cocaine-induced silent synapses. Finally, loss of GluD1 increased the GluN2B subunit contribution to NMDA receptor currents in MSNs and a partial agonist of GluN2B-containing NMDA receptors normalized the higher active cofilin and cocaine preference in GluD1 KO mice. Together, these findings demonstrate a critical role of GluD1 in controlling susceptibility to cocaine preference and cocaine-induced plasticity by modulating NMDA receptor subunit contribution.


Subject(s)
Cocaine/administration & dosage , Neuronal Plasticity , Neurons/physiology , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiology , Receptors, Glutamate/physiology , Animals , Dendrites/physiology , Drug-Seeking Behavior , Excitatory Postsynaptic Potentials , Male , Mice, Knockout , Receptor, Metabotropic Glutamate 5/physiology , Receptors, Glutamate/genetics , Receptors, Metabotropic Glutamate/physiology , Receptors, N-Methyl-D-Aspartate/physiology
15.
Neuroscience ; 380: 49-62, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29559384

ABSTRACT

Hypofunction of NMDA receptors in parvalbumin (PV)-positive interneurons has been proposed as a potential mechanism for cortical abnormalities and symptoms in schizophrenia. GluN2C-containing receptors have been linked to this hypothesis due to the higher affinity of psychotomimetic doses of ketamine for GluN1/2C receptors. However, the precise cell-type expression of GluN2C subunit remains unknown. We describe the expression of the GluN2C subunit using a novel EGFP reporter model. We observed EGFP(GluN2C) localization in PV-positive neurons in the nucleus reticularis of the thalamus, globus pallidus externa and interna, ventral pallidum and substantia nigra. In contrast, EGFP(GluN2C)-expressing cells did not co-localize with PV-positive neurons in the cortex, striatum, hippocampus or amygdala. Instead, EGFP(GluN2C) expression in these regions co-localized with an astrocytic marker. We confirmed functional expression of GluN2C-containing receptors in the PV-neurons in substantia nigra and cortical astrocytes using electrophysiology. GluN2C was found to be enriched in several first-order and higher order thalamic nuclei. Interestingly, we found that a previous GluN2C ß-gal reporter model excluded expression from PV-neurons and certain thalamic nuclei but exhibited expression in the retrosplenial cortex. GluN2C's unique distribution in neuronal and non-neuronal cells in a brain region-specific manner raises interesting questions regarding the role of GluN2C-containing receptors in the central nervous system.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/biosynthesis , Animals , Gene Knock-In Techniques/methods , Green Fluorescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Parvalbumins/metabolism , Receptors, N-Methyl-D-Aspartate/analysis
16.
Addict Biol ; 22(2): 291-302, 2017 Mar.
Article in English | MEDLINE | ID: mdl-26549324

ABSTRACT

Although dysregulation of the dopaminergic mesolimbic system is generally considered central to addiction, the involvement of other circuits is increasingly being appreciated. An interaction between locus coeruleus (LC) noradrenergic neurons and the posterior ventral tegmental area (pVTA) dopaminergic system, in the processing of drug-triggered reward, has been suggested, but not demonstrated in behaving animals. Herein, we try to tease out the precise role of noradrenergic neurons in the LC-VTA circuit in mediating reward and reinforcement behavior associated with ethanol. In the standard two-lever (active/inactive) operant paradigm, the rats were trained to self-administer ethanol in pVTA and subjected to pharmacological intervention. Intra-pVTA administration of phenylephrine (alpha-1 adrenoceptor agonist) increased ethanol self-administration, while prazosin and disulfiram (agents that reduce noradrenergic tone) produced opposite effects. While degeneration [N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine hydrochloride, DSP-4, intraperitoneal route] or silencing (lidocaine or muscimol, both via intra-LC route) of the LC noradrenergic neurons decreased, phenylephrine via the intra-LC route reinstated ethanol self-administration. Furthermore, lidocaine reduced ethanol self-administration, but the effect was fully attenuated by noradrenaline given directly in the pVTA. This suggests that the feedback signals from LC to pVTA are necessary to sustain the ethanol self-infusion activity. Ethanol self-administration significantly increased tyrosine hydroxylase immunoreactivity in pVTA and LC; the response was blocked by DSP-4 pre-treatment. While dopamine D1 , but not D2 , receptors were localized on noradrenergic LC neurons, pre-treatment with SCH-23390 (intra-LC) dampened the lever press activity. We suggest that two-way communications between VTA and LC regions is essential for ethanol-triggered reinforcement behavior.


Subject(s)
Adrenergic Neurons/drug effects , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Locus Coeruleus/drug effects , Reinforcement, Psychology , Ventral Tegmental Area/drug effects , Acetaldehyde Dehydrogenase Inhibitors/pharmacology , Adrenergic Agents/pharmacology , Adrenergic alpha-1 Receptor Agonists/pharmacology , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Anesthetics, Local/pharmacology , Animals , Behavior, Animal/drug effects , Benzazepines/pharmacology , Benzylamines/pharmacology , Central Nervous System Depressants/administration & dosage , Conditioning, Operant , Disulfiram/pharmacology , Dopamine Antagonists/pharmacology , Ethanol/administration & dosage , GABA-A Receptor Agonists/pharmacology , Lidocaine/pharmacology , Locus Coeruleus/metabolism , Male , Muscimol/pharmacology , Phenylephrine/pharmacology , Prazosin/pharmacology , Rats , Rats, Wistar , Receptors, Dopamine D1/antagonists & inhibitors , Reward , Self Administration , Tyrosine 3-Monooxygenase/drug effects , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/metabolism
17.
Behav Brain Res ; 317: 340-349, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27686026

ABSTRACT

Rats with electrode implanted in the lateral hypothalamus (LH)-medial forebrain bundle (MFB) area actively engage in intracranial self-stimulation (ICSS). However, the neuronal substrate that translates the electrical pulses into the neural signals, and integrates the information with mesolimbic reward system, has remained elusive. We test the hypothesis that the cocaine- and amphetamine-regulated transcript (CART) neurons in the LH-MFB area may support this function. The ICSS activity via an electrode in LH-MFB area was facilitated by CART (55-102) peptide stereotaxically injected in the lateral ventricle or posterior ventral tegmental area (pVTA), but attenuated by CART antibody. While the ICSS experience seems to activate CART cells in the LH, the pVTA showed significant increment in the CART fiber terminals on the dopamine cells, increase in tyrosine hydroxylase (TH)-immunoreactivity, and CART and synaptophysin colabeled elements. Neuronal tracing experiments revealed that CART cells of the LH-MFB region project to the pVTA. The rats with stereotaxically implanted cannulae in pVTA avidly self-infused CART (55-102) suggesting a role for the peptide in motivation, however, CART (1-39) was ineffective. CART self-infusing activity was inhibited by dopamine D1 receptors antagonist, given directly in the nucleus accumbens shell (AcbSh). The rats trained to self-administer CART (55-102) showed enhanced TH immunoreactivity in the cells of pVTA and fibers in AcbSh. We suggest that CART neurons of the LH-MFB area may play a role in conveying reward information to the mesolimbic dopamine neurons, which in turn may arouse the goal directed behavior.


Subject(s)
Hypothalamic Area, Lateral/cytology , Motivation/physiology , Nerve Tissue Proteins/pharmacology , Neural Pathways/physiology , Neurons/drug effects , Peptide Fragments/pharmacology , Ventral Tegmental Area/drug effects , Animals , Antibodies/pharmacology , Benzazepines/pharmacology , Conditioning, Operant/drug effects , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Functional Laterality , Hypothalamic Area, Lateral/physiology , Locomotion/drug effects , Male , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Neural Pathways/drug effects , Neurons/physiology , Peptide Fragments/immunology , Rats , Rats, Wistar , Self Stimulation/physiology , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/physiology
18.
Br J Pharmacol ; 173(17): 2589-99, 2016 09.
Article in English | MEDLINE | ID: mdl-27273730

ABSTRACT

BACKGROUND AND PURPOSE: Agmatine, a putative neurotransmitter, plays a vital role in learning and memory. Although it is considered an endogenous ligand of imidazoline receptors, agmatine exhibits high affinity for α-adrenoceptors, NOS and NMDA receptors. These substrates within the locus coeruleus (LC) are critically involved in learning and memory processes. EXPERIMENTAL APPROACH: The hippocampus and LC of male Wistar rat were stereotaxically cannulated for injection. Effects of agmatine, given i.p. or intra-LC, on acquisition, consolidation and retrieval of inhibitory avoidance (IA) memory were measured. The NO donor S-nitrosoglutathione, non-specific (L-NAME) and specific NOS inhibitors (L-NIL, 7-NI, L-NIO), the α2 -adrenoceptor antagonist (yohimbine) or the corresponding agonist (clonidine) were injected intra-LC before agmatine. Intra-hippocampal injections of the NMDA antagonist, MK-801 (dizocilpine), were used to modify the memory enhancing effects of agmatine, SNG and yohimbine. Expression of tyrosine hydroxylase (TH) and eNOS in the LC was assessed immunohistochemically. KEY RESULTS: Agmatine (intra-LC or i.p.) facilitated memory retrieval in the IA test. S-nitrosoglutathione potentiated, while L-NAME and L-NIO decreased, these effects of agmatine. L-NIL and 7-NI did not alter the effects of agmatine. Yohimbine potentiated, whereas clonidine attenuated, effects of agmatine within the LC. The effects of agmatine, S-nitrosoglutathione and yohimbine were blocked by intra-hippocampal MK-801. Agmatine increased the population of TH- and eNOS-immunoreactive elements in the LC. CONCLUSIONS AND IMPLICATIONS: The facilitation of memory retrieval in the IA test by agmatine is probably mediated by interactions between eNOS, NO and noradrenergic pathways in the LC.


Subject(s)
Agmatine/pharmacology , Avoidance Learning/drug effects , Locus Coeruleus/drug effects , Locus Coeruleus/metabolism , Nitric Oxide/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Agmatine/administration & dosage , Animals , Dose-Response Relationship, Drug , Male , Nitric Oxide/chemistry , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-2/chemistry , Structure-Activity Relationship
19.
Neuropharmacology ; 110(Pt A): 198-210, 2016 11.
Article in English | MEDLINE | ID: mdl-27084697

ABSTRACT

While dopamine (DA) neurons in the ventral tegmental area (VTA) drive the mesolimbic-reward pathway, confluent lines of evidence underscore the importance of transient receptor potential vanilloid (TRPV) channels as novel regulators of these neurons. Among the TRPV-subfamily, TRPV3 is of particular interest in reward, since active ingredients of flavour-enhancing spices in food serve as TRPV3 agonists and modulate DAergic neurotransmission. The nature of TRPV3 elements in the VTA and their role in driving the mesolimbic-DA-reward pathway has however, remained unexplored. We observed TRPV3 mRNA as well as TRPV3-immunoreactive neurons in the VTA of Wistar rats. We therefore explored whether these ion channels participate in modulating mesolimbic-DA reward pathway. In the posterior VTA (pVTA), 82 ± 2.6% of the TRPV3 neurons co-express tyrosine hydroxylase and 68 ± 5.5% of these neurons project to the nucleus accumbens shell (Acb shell). While ex vivo treatment of midbrain slices with TRPV3-agonist, thymol increased [Ca(2+)]i-activity in pVTA neurons, intra-pVTA injections of thymol in freely-moving, satiated rats enhanced positive reinforcement for active lever pressings in an operant chamber to self-administer sweet pellets. This behavior was attenuated by prior treatment with intra-Acb shell DA D1- and D2-like receptor antagonists. These results demonstrate a role for TRPV3 in driving mesolimbic-DA food-reward pathway, and underscores the importance of these channels in the VTA as key components processing reward.


Subject(s)
Dopamine/metabolism , Neurons/metabolism , Reward , TRPV Cation Channels/metabolism , Ventral Tegmental Area/metabolism , Animals , Calcium/metabolism , Cations, Divalent/metabolism , Cell Line , Central Nervous System Agents/pharmacology , Dietary Sucrose , Dopamine Antagonists/pharmacology , Feeding Behavior/drug effects , Feeding Behavior/physiology , Male , Mice , Neural Pathways/cytology , Neural Pathways/drug effects , Neural Pathways/metabolism , Neurons/cytology , Neurons/drug effects , Random Allocation , Rats, Wistar , Receptors, Dopamine/metabolism , Self Administration , TRPV Cation Channels/agonists , Thymol/pharmacology , Tissue Culture Techniques , Ventral Tegmental Area/cytology , Ventral Tegmental Area/drug effects
20.
Behav Brain Res ; 301: 262-72, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26771081

ABSTRACT

Parkinson's disease (PD) is an age-related disorder characterized by a progressive degeneration of dopaminergic neurons of substantia nigra (SN). The neuropeptide cocaine- and amphetamine-regulated transcript (CART) is known to closely interact with the dopamine system and regulate psychomotor activity. We screened the effectiveness of CART in reversing the symptoms of PD in a rat model. PD like condition was induced by administering 6-hydroxydopamine (6-OHDA) directly in the SN of the right side. Fifteen days later, intraperitoneal (IP) treatment with apomorphine hydrochloride to these rats, resulted in contralateral rotations in the rotation test chamber suggesting induction of PD-like symptoms. This action of apomorphine was significantly attenuated by intracerebroventricular (ICV) treatment with CART and potentiated by CART antibody. IP treatment with levodopa also produced contralateral rotation in PD induced rats, and showed anti-Parkinson-like action. Prior treatment with CART via ICV route potentiated the anti-Parkinsonian effects of levodopa, while CART antibody produced opposite effects. CART treatment per se, to PD induced rats produced ipsilateral rotations, suggesting that the peptide may promote the endogenous release of dopamine from intact neurons. While CART-immunoreactivity in arcuate nucleus, paraventricular nucleus, striatum, substantia nigra, ventral tegmental area and locus coeruleus was reduced in the PD induced rats, levodopa treatment restored the expression of CART-immunoreactivity in these nuclei. These results suggest that endogenous CART might closely interact with the dopamine containing SN-striatal pathway which is known to profoundly influence the motor system. The study underscores the importance of CART as a potential therapeutic agent in the treatment of PD.


Subject(s)
Antiparkinson Agents/pharmacology , Levodopa/pharmacology , Movement/drug effects , Nerve Tissue Proteins/pharmacology , Parkinsonian Disorders/drug therapy , Animals , Antibodies/pharmacology , Apomorphine/pharmacology , Brain/drug effects , Brain/metabolism , Brain/pathology , Dopamine Agonists/pharmacology , Dose-Response Relationship, Drug , Fluorescent Antibody Technique , Male , Movement/physiology , Nerve Tissue Proteins/immunology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Oxidopamine , Parkinsonian Disorders/pathology , Parkinsonian Disorders/physiopathology , Rats, Sprague-Dawley , Rotarod Performance Test , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...