Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
J Extracell Vesicles ; 13(4): e12439, 2024 Apr.
Article En | MEDLINE | ID: mdl-38647111

Our previous findings demonstrated that astrocytic HIF-1α plays a major role in HIV-1 Tat-mediated amyloidosis which can lead to Alzheimer's-like pathology-a comorbidity of HIV-Associated Neurocognitive Disorders (HAND). These amyloids can be shuttled in extracellular vesicles, and we sought to assess whether HIV-1 Tat stimulated astrocyte-derived EVs (ADEVs) containing the toxic amyloids could result in neuronal injury in vitro and in vivo. We thus hypothesized that blocking HIF-1α could likely mitigate HIV-1 Tat-ADEV-mediated neuronal injury. Rat hippocampal neurons when exposed to HIV-1 Tat-ADEVs carrying the toxic amyloids exhibited amyloid accumulation and synaptodendritic injury, leading to functional loss as evidenced by alterations in miniature excitatory post synaptic currents. The silencing of astrocytic HIF-1α not only reduced the biogenesis of ADEVs, as well as amyloid cargos, but also ameliorated neuronal synaptodegeneration. Next, we determined the effect of HIV-1 Tat-ADEVs carrying amyloids in the hippocampus of naive mice brains. Naive mice receiving the HIV-1 Tat-ADEVs, exhibited behavioural changes, and Alzheimer's 's-like pathology accompanied by synaptodegeneration. This impairment(s) was not observed in mice injected with HIF-1α silenced ADEVs. This is the first report demonstrating the role of amyloid-carrying ADEVs in mediating synaptodegeneration leading to behavioural changes associated with HAND and highlights the protective role of HIF-1α.


Astrocytes , Extracellular Vesicles , HIV-1 , Hippocampus , Hypoxia-Inducible Factor 1, alpha Subunit , Neurons , Extracellular Vesicles/metabolism , Animals , Astrocytes/metabolism , Mice , Rats , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , HIV-1/metabolism , Hippocampus/metabolism , Neurons/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Humans , Neurocognitive Disorders/metabolism , Neurocognitive Disorders/etiology , HIV Infections/metabolism , HIV Infections/complications , Male , AIDS Dementia Complex/metabolism
2.
Neurobiol Dis ; 181: 106117, 2023 06 01.
Article En | MEDLINE | ID: mdl-37031803

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.


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 En | MEDLINE | ID: mdl-37004850

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.


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.
J Neurosci ; 43(12): 2075-2089, 2023 03 22.
Article En | MEDLINE | ID: mdl-36810227

Resident cochlear macrophages rapidly migrate into the inner hair cell synaptic region and directly contact the damaged synaptic connections after noise-induced synaptopathy. Eventually, such damaged synapses are spontaneously repaired, but the precise role of macrophages in synaptic degeneration and repair remains unknown. To address this, cochlear macrophages were eliminated using colony stimulating factor 1 receptor (CSF1R) inhibitor, PLX5622. Sustained treatment with PLX5622 in CX3CR1 GFP/+ mice of both sexes led to robust elimination of resident macrophages (∼94%) without significant adverse effects on peripheral leukocytes, cochlear function, and structure. At 1 day (d) post noise exposure of 93 or 90 dB SPL for 2 hours, the degree of hearing loss and synapse loss were comparable in the presence and absence of macrophages. At 30 d after exposure, damaged synapses appeared repaired in the presence of macrophages. However, in the absence of macrophages, such synaptic repair was significantly reduced. Remarkably, on cessation of PLX5622 treatment, macrophages repopulated the cochlea, leading to enhanced synaptic repair. Elevated auditory brainstem response thresholds and reduced auditory brainstem response Peak 1 amplitudes showed limited recovery in the absence of macrophages but recovered similarly with resident and repopulated macrophages. Cochlear neuron loss was augmented in the absence of macrophages but showed preservation with resident and repopulated macrophages after noise exposure. While the central auditory effects of PLX5622 treatment and microglia depletion remain to be investigated, these data demonstrate that macrophages do not affect synaptic degeneration but are necessary and sufficient to restore cochlear synapses and function after noise-induced synaptopathy.SIGNIFICANCE STATEMENT The synaptic connections between cochlear inner hair cells and spiral ganglion neurons can be lost because of noise over exposure or biological aging. This loss may represent the most common causes of sensorineural hearing loss also known as hidden hearing loss. Synaptic loss results in degradation of auditory information, leading to difficulty in listening in noisy environments and other auditory perceptual disorders. We demonstrate that resident macrophages of the cochlea are necessary and sufficient to restore synapses and function following synaptopathic noise exposure. Our work reveals a novel role for innate-immune cells, such as macrophages in synaptic repair, that could be harnessed to regenerate lost ribbon synapses in noise- or age-linked cochlear synaptopathy, hidden hearing loss, and associated perceptual anomalies.


Hair Cells, Auditory, Inner , Hearing Loss, Noise-Induced , Male , Female , Animals , Mice , Hair Cells, Auditory, Inner/physiology , Acoustic Stimulation/adverse effects , Auditory Threshold/physiology , Cochlea/metabolism , Synapses/physiology , Evoked Potentials, Auditory, Brain Stem/physiology , Macrophages/metabolism
5.
Pharmacol Res ; 178: 106144, 2022 04.
Article En | MEDLINE | ID: mdl-35304260

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.


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.
Article En | MEDLINE | ID: mdl-36812097

Aim: Activation of microglial NLRP3 inflammasome is an essential contributor to neuroinflammation underlying HIV-associated neurological disorders (HAND). Under pathological conditions, microglia-derived-EVs (MDEVs) can affect neuronal functions by delivering neurotoxic mediators to recipient cells. However, the role of microglial NLRP3 in mediating neuronal synaptodendritic injury has remained unexplored to date. In the present study, we sought to assess the regulatory role of HIV-1 Tat induced microglial NLRP3 in neuronal synaptodendritic injury. We hypothesized that HIV-1 Tat mediated microglia EVs carrying significant levels of NLRP3 contribute to the synaptodendritic injury, thereby affecting the maturation of neurons. Methods: To understand the cross-talk between microglia and neuron, we isolated EVs from BV2 and human primary microglia (HPM) cells with or without NLRP3 depletion using siNLRP3 RNA. EVs were isolated by differential centrifugation, characterized by ZetaView nanoparticle tracking analysis, electron microscopy, and western blot analysis for exosome markers. Purified EVs were exposed to primary rat neurons isolated from E18 rats. Along with green fluorescent protein (GFP) plasmid transfection, immunocytochemistry was performed to visualize neuronal synaptodendritic injury. Western blotting was employed to measure siRNA transfection efficiency and the extent of neuronal synaptodegeneration. Images were captured in confocal microscopy, and subsequently, Sholl analysis was performed for analyzing dendritic spines using neuronal reconstruction software Neurolucida 360. Electrophysiology was performed on hippocampal neurons for functional assessment. Results: Our findings demonstrated that HIV-1 Tat induced expression of microglial NLRP3 and IL1ß, and further that these were packaged in microglial exosomes (MDEV) and were also taken up by the neurons. Exposure of rat primary neurons to microglial Tat-MDEVs resulted in downregulation of synaptic proteins- PSD95, synaptophysin, excitatory vGLUT1, as well as upregulation of inhibitory proteins- Gephyrin, GAD65, thereby implicating impaired neuronal transmissibility. Our findings also showed that Tat-MDEVs not only caused loss of dendritic spines but also affected numbers of spine sub-types- mushroom and stubby. Synaptodendritic injury further affected functional impairment as evidenced by the decrease in miniature excitatory postsynaptic currents (mEPSCs). To assess the regulatory role of NLRP3 in this process, neurons were also exposed to Tat-MDEVs from NLRP3 silenced microglia. Tat-MDEVs from NLRP3 silenced microglia exerted a protective role on neuronal synaptic proteins, spine density as well as mEPSCs. Conclusion: In summary, our study underscores the role of microglial NLRP3 as an important contributor to Tat-MDEV mediated synaptodendritic injury. While the role of NLRP3 in inflammation is well-described, its role in EV-mediated neuronal damage is an interesting finding, implicating it as a target for therapeutics in HAND.

7.
Neuropharmacology ; 201: 108818, 2021 12 15.
Article En | MEDLINE | ID: mdl-34610288

N-methyl-d-aspartate (NMDA) receptors (NMDARs) are a subtype of ionotropic glutamate receptor with important roles in CNS function. Since excessive NMDAR activity can lead to neuronal cell death and epilepsy, there is interest in developing NMDAR negative allosteric modulators (NAMs) as neuroprotective agents. In this study, we characterize the inhibitory properties of a novel NMDAR antagonist, UBP792. This compound displays partial subtype-selectivity by having a varied maximal inhibition of GluN2A-, GluN2B-, GluN2C-, and GluN2D-containing receptors (52%, 70%, 87%, 89%, respectively) with IC50s 4-10 µM. UBP792 inhibited NMDAR responses by reducing l-glutamate and glycine potencies and efficacies. Consistent with non-competitive inhibition, increasing agonist concentrations 30-fold did not reduce UBP792 potency. UBP792 inhibition was also not competitive with the structurally-related positive allosteric modulator (PAM) UBP684. UBP792 activity was voltage-independent, unaffected by GluN1's exon-5, and reduced at low pH (except for GluN1/GluN2A receptors which were more sensitive at acidic pH). UBP792 binding appeared independent of agonist binding and may be entering the plasma membrane to gain access to its binding site. Inhibition by UBP792 is reduced when the ligand-binding domain (LBD) of the GluN2 subunit, but not that of the GluN1 subunit, is cross-linked in the closed-cleft, activated conformation. Thus, UBP792 may be inhibiting by stabilizing an open GluN2-LBD cleft associated with channel inactivation or by stabilizing downstream closed channel conformations allosterically-coupled to the GluN2-LBD. These findings further expand the repertoire displayed by NMDAR NAMs thus expanding the opportunities for developing NMDAR modulators with the most appropriate selectivity and physiological actions for specific therapeutic indications.


Carboxylic Acids , Naphthalenes , Neuroprotective Agents , Receptors, N-Methyl-D-Aspartate , Animals , Allosteric Regulation , Binding Sites , Carboxylic Acids/chemistry , Carboxylic Acids/pharmacology , Dose-Response Relationship, Drug , Glutamic Acid/metabolism , Glycine , Naphthalenes/chemistry , Naphthalenes/pharmacology , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Oocytes , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Xenopus
8.
Cells ; 10(10)2021 10 03.
Article En | MEDLINE | ID: mdl-34685624

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.


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
9.
Mol Neurobiol ; 58(10): 4787-4801, 2021 Oct.
Article En | MEDLINE | ID: mdl-34173171

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.


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
10.
Neurobiol Dis ; 150: 105254, 2021 03.
Article En | MEDLINE | ID: mdl-33421565

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.


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
11.
Neurobiol Dis ; 137: 104746, 2020 04.
Article En | MEDLINE | ID: mdl-31945419

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.


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

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.


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
13.
J Ethnopharmacol ; 202: 97-102, 2017 Apr 18.
Article En | MEDLINE | ID: mdl-28315457

ETHNOPHARMACOLOGICAL RELEVANCE: Root powder of Achyranthes aspera Linn. (A. aspera) belongs to family Amaranthaceae is used in Indian traditional medicine for the management of epilepsy and its efficacy is widely acclaimed among the different rural communities. AIM OF THE STUDY: The present study was aimed to establish the possible anticonvulsant effect of A. aspera methanolic root extract using acute anticonvulsant models and to evaluate the acute toxicity and neurotoxic potential A. aspera extract. MATERIAL AND METHODS: A. aspera methanolic extract was standardized with respect to betaine using HPTLC. The maximal electroshock (MES), pentylenetetrazol (PTZ), picrotoxin and bicuculline induced seizure models were used to evaluate the anticonvulsant potential of standardized A. aspera root extract. The GABA content in cortex and hippocampus of extract treated mice was evaluated using HPLC. Moreover, the animals were also evaluated for acute toxicity study and neurotoxicity test. RESULTS: A significant enhancement in the seizure threshold was observed by A. aspera extract (5 and 10mg/kg) treated mice in PTZ, picrotoxin and bicuculline models as compared to saline treated mice respectively, whereas the extract failed to show protection in MES induced seizures. Moreover, A. aspera treatment (5 and 10mg/kg) significantly enhances the GABA levels in hippocampus and cortex as compared to saline treated group. A. aspera root extract was devoid of any sign of acute toxicity as well as neurotoxicity. CONCLUSIONS: A. aspera root extract exhibits significant anticonvulsant effect by facilitation of GABAergic neurotransmission in the brain.


Achyranthes/chemistry , Achyranthes/toxicity , Anticonvulsants/pharmacology , Neurotoxins/toxicity , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Electroshock , Hippocampus/drug effects , Hippocampus/metabolism , India , Male , Medicine, Traditional , Mice , Plant Extracts/pharmacology , Plant Extracts/toxicity , Plant Roots/chemistry , Postural Balance/drug effects , Powders , Seizures/chemically induced , Seizures/prevention & control , gamma-Aminobutyric Acid/metabolism
14.
Biomed Pharmacother ; 86: 271-278, 2017 Feb.
Article En | MEDLINE | ID: mdl-28006753

The present study investigated the pharmacological effect of agmatine in Complete Freud Adjuvant (CFA) induced arthritis and cachexia in rats. The rats were injected with CFA (0.1ml/rat) to induced symptoms of arthritis. Day 8 onwards of CFA administration, rats were injected daily with agmatine for next 7days, and arthritis score, body weights and food intake were monitored daily (g). Since cachexia is known to produce severe inflammation, malnutrition and inhibition of albumin gene expression, we have also monitored the total proteins, albumin, TNF-α and IL-6 levels in arthritic rats and its modulation by agmatine. In the present study, CFA treated rats showed a progressive reduction in both food intake and body weight. In addition analysis of blood serum of arthritis animals showed a significant reduction in proteins and albumin and significant elevation in tumor necrosis factor (TNF)-α and Interleukins (IL)-6. Chronic agmatine (20-40mg/kg, ip) treatment not only attenuated the signs of arthritis but also reverses anorexia and body weight loss in CFA treated rats. In addition, agmatine restored total protein and albumin and reduces TNF-α and IL-6 levels in arthritis rats. These results suggest that agmatine administration can prevent the body weights loss and symptoms of arthritis via inhibition of inflammatory cytokines.


Agmatine/therapeutic use , Arthritis, Experimental/drug therapy , Cachexia/drug therapy , Agmatine/pharmacology , Animals , Arthritis, Experimental/pathology , Body Weight/drug effects , Body Weight/physiology , Cachexia/pathology , Inflammation/drug therapy , Inflammation/pathology , Male , Rats , Rats, Sprague-Dawley
15.
Nat Prod Rep ; 31(11): 1585-611, 2014 Nov.
Article En | MEDLINE | ID: mdl-25051191

In silico approaches have been widely recognised to be useful for drug discovery. Here, we consider the significance of available databases of medicinal plants and chemo- and bioinformatics tools for in silico drug discovery beyond the traditional use of folk medicines. This review contains a practical example of the application of combined chemo- and bioinformatics methods to study pleiotropic therapeutic effects (known and novel) of 50 medicinal plants from Traditional Indian Medicine.


Drug Discovery , Medicine, Traditional , Plants, Medicinal/chemistry , Computational Biology , Databases, Factual , Molecular Structure
16.
Comput Biol Med ; 47: 1-6, 2014 Apr.
Article En | MEDLINE | ID: mdl-24503467

BACKGROUND: Exploration of therapeutic mechanism is an integral part of medicinal plant based drug discovery for better understanding of pharmacological behavior of these agents. But, its study using conventional hit and trial wet laboratory experiments proves to be very tedious, time consuming and expensive, thus encouraging development of in silico techniques. Hence, an in silico technique has been devised using a computer software Prediction of Activity Spectra for Substances (PASS) to study pharmacodynamics of medicinal plants. The technique has been presented with a case study using Ficus religiosa L. (Moraceae) in which its anticonvulsant mechanism has been elucidated with PASS and further validated experimentally. METHODS: Pentylenetetrazol (PTZ)-induced convulsion test was used to study the anticonvulsant effect of standardized bark extract of F. religiosa. Thereafter, structure of all the reported bioactive metabolites in the bark was subjected to PASS software to obtain biological activity spectrum of each compound. The mechanism signifying anticonvulsant effect was selected from the spectrum and was further validated using in vitro test. RESULTS AND DISCUSSION: The extract showed significant anticonvulsant effect in PTZ test. PASS analysis showed a high activity score for GABA aminotransferase (GABA-AT) inhibitory effect of the bioactive metabolites present in the bark. In vitro GABA-AT enzyme assay results were in concordance with the predicted mechanism by PASS for the observed anticonvulsant effect, as the extract showed potent inhibition of the enzyme. The results of present study showed the in silico technique to be useful for elucidation of unknown therapeutic mechanisms of medicinal plants.


Computational Biology/methods , Computer Simulation , Models, Biological , Models, Statistical , Plant Extracts/pharmacology , 4-Aminobutyrate Transaminase , Analysis of Variance , Animals , Anticonvulsants/chemistry , Anticonvulsants/pharmacology , Convulsants/toxicity , Ficus , Male , Mice , Models, Chemical , Pentylenetetrazole/toxicity , Plant Extracts/chemistry , Plants, Medicinal , Reproducibility of Results , Seizures/chemically induced
17.
Eur Neuropsychopharmacol ; 24(6): 955-63, 2014 Jun.
Article En | MEDLINE | ID: mdl-24461723

In the present study, modulation of anxiolytic action of agmatine by neuropeptide Y (NPY) in the central nucleus of amygdala (CeA) is evaluated employing Vogel's conflict test (VCT) in rats. The intra-CeA administration of agmatine (0.6 and 1.2µmol/rat), NPY (10 and 20pmol/rat) or NPY Y1/Y5 receptors agonist [Leu(31), Pro(34)]-NPY (30 and 60pmol/rat) significantly increased the number of punished drinking licks following 15min of treatment. Combination treatment of subeffective dose of NPY (5pmol/rat) or [Leu(31), Pro(34)]-NPY (15pmol/rat) and agmatine (0.3µmol/rat) produced synergistic anxiolytic-like effect. However, intra-CeA administration of selective NPY Y1 receptor antagonist, BIBP3226 (0.25 and 0.5mmol/rat) produced anxiogenic effect. In separate set of experiment, pretreatment with BIBP3226 (0.12mmol/rat) reversed the anxiolytic effect of agmatine (0.6µmol/rat). Furthermore, we evaluated the effect of intraperitoneal injection of agmatine (40mg/kg) on NPY-immunoreactivity in the nucleus accumbens shell (AcbSh), lateral part of bed nucleus of stria terminalis (BNSTl) and CeA. While agmatine treatment significantly decreased the fibers density in BNSTl, increase was noticed in AcbSh. In addition, agmatine reduced NPY-immunoreactive cells in the AcbSh and CeA. Immunohistochemical data suggest the enhanced transmission of NPY from the AcbSh and CeA. Taken together, this study suggests that agmatine produced anxiolytic effect which might be regulated via modulation of NPYergic system particularly in the CeA.


Agmatine/pharmacology , Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , Central Amygdaloid Nucleus/drug effects , Neuropeptide Y/metabolism , Animals , Anxiety/pathology , Anxiety/physiopathology , Arginine/analogs & derivatives , Arginine/pharmacology , Cell Count , Central Amygdaloid Nucleus/pathology , Central Amygdaloid Nucleus/physiopathology , Immunohistochemistry , Male , Motor Activity/drug effects , Motor Activity/physiology , Neuropeptide Y/analogs & derivatives , Neuropeptide Y/pharmacology , Neuropsychological Tests , Nucleus Accumbens/drug effects , Nucleus Accumbens/pathology , Nucleus Accumbens/physiopathology , Photomicrography , Rats, Sprague-Dawley , Receptors, Neuropeptide Y/antagonists & inhibitors , Receptors, Neuropeptide Y/metabolism , Septal Nuclei/drug effects , Septal Nuclei/pathology , Septal Nuclei/physiopathology
18.
Pharmacol Biochem Behav ; 100(3): 398-403, 2012 Jan.
Article En | MEDLINE | ID: mdl-21989253

The effect of agmatine in preclinical behavioral tests of schizophrenia has been examined in rodents. Agmatine at the doses of 40 and 80 mg/kg blocked conditioned avoidance responding, attenuated apomorphine induced climbing, diminished amphetamine and ketamine hyperlocomotor activity and augmented plasma prolactin levels. Pretreatment of animals with 20 mg/kg of agmatine potentiated the inhibitory effect of haloperidol (0.1 mg/kg, ip) and olanzepine (0.5 mg/kg, ip) in conditioned avoidance response test and apomorphine induced climbing. Agmatine alone at the doses tested here did not induce any cataleptic behavior in mice. However significant catalepsy was exhibited when agmatine (80 mg/kg, ip) was injected to mice pretreated with 5-HT1A receptor antagonist, WAY100, 635. These results indicate that agmatine via regulation of brain dopaminergic signaling modulates dopamine mediated behaviors. The alteration in the levels of endogenous agmatine may contribute to the genesis of psychosis and development of drugs that enhance endogenous agmatine content may be better therapeutic approach to treat schizophrenia with low incidences of extra pyramidal side effects.


Agmatine/therapeutic use , Antipsychotic Agents/therapeutic use , Behavior, Animal/drug effects , Schizophrenia/drug therapy , Agmatine/adverse effects , Animals , Antipsychotic Agents/adverse effects , Avoidance Learning , Catalepsy/chemically induced , Disease Models, Animal , Dopamine Agents/adverse effects , Dopamine Agents/therapeutic use , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Dyskinesia, Drug-Induced/drug therapy , Male , Mice , Molecular Targeted Therapy , Prolactin/blood , Random Allocation , Rats , Rats, Sprague-Dawley , Schizophrenia/blood , Schizophrenia/physiopathology
...