Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 810
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 180(4): 666-676.e13, 2020 02 20.
Article in English | MEDLINE | ID: mdl-32084339

ABSTRACT

The mystery of general anesthesia is that it specifically suppresses consciousness by disrupting feedback signaling in the brain, even when feedforward signaling and basic neuronal function are left relatively unchanged. The mechanism for such selectiveness is unknown. Here we show that three different anesthetics have the same disruptive influence on signaling along apical dendrites in cortical layer 5 pyramidal neurons in mice. We found that optogenetic depolarization of the distal apical dendrites caused robust spiking at the cell body under awake conditions that was blocked by anesthesia. Moreover, we found that blocking metabotropic glutamate and cholinergic receptors had the same effect on apical dendrite decoupling as anesthesia or inactivation of the higher-order thalamus. If feedback signaling occurs predominantly through apical dendrites, the cellular mechanism we found would explain not only how anesthesia selectively blocks this signaling but also why conscious perception depends on both cortico-cortical and thalamo-cortical connectivity.


Subject(s)
Anesthetics, General/pharmacology , Cerebral Cortex/drug effects , Pyramidal Cells/drug effects , Animals , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Cholinergic Antagonists/pharmacology , Consciousness , Dendrites/drug effects , Dendrites/physiology , Excitatory Amino Acid Antagonists/pharmacology , Feedback, Physiological , Female , Male , Mice , Pyramidal Cells/physiology , Synaptic Transmission , Thalamus/cytology , Thalamus/drug effects , Thalamus/physiology
2.
Brain ; 147(6): 2169-2184, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38662500

ABSTRACT

Approximately 22% of Alzheimer's disease (AD) patients suffer from seizures, and the co-occurrence of seizures and epileptiform activity exacerbates AD pathology and related cognitive deficits, suggesting that seizures may be a targetable component of AD progression. Given that alterations in neuronal excitatory:inhibitory (E:I) balance occur in epilepsy, we hypothesized that decreased markers of inhibition relative to those of excitation would be present in AD patients. We similarly hypothesized that in 5XFAD mice, the E:I imbalance would progress from an early stage (prodromal) to later symptomatic stages and be further exacerbated by pentylenetetrazol (PTZ) kindling. Post-mortem AD temporal cortical tissues from patients with or without seizure history were examined for changes in several markers of E:I balance, including levels of the inhibitory GABAA receptor, the sodium potassium chloride cotransporter 1 (NKCC1) and potassium chloride cotransporter 2 (KCC2) and the excitatory NMDA and AMPA type glutamate receptors. We performed patch-clamp electrophysiological recordings from CA1 neurons in hippocampal slices and examined the same markers of E:I balance in prodromal 5XFAD mice. We next examined 5XFAD mice at chronic stages, after PTZ or control protocols, and in response to chronic mTORC1 inhibitor rapamycin, administered following kindled seizures, for markers of E:I balance. We found that AD patients with comorbid seizures had worsened cognitive and functional scores and decreased GABAA receptor subunit expression, as well as increased NKCC1/KCC2 ratios, indicative of depolarizing GABA responses. Patch clamp recordings of prodromal 5XFAD CA1 neurons showed increased intrinsic excitability, along with decreased GABAergic inhibitory transmission and altered glutamatergic neurotransmission, indicating that E:I imbalance may occur in early disease stages. Furthermore, seizure induction in prodromal 5XFAD mice led to later dysregulation of NKCC1/KCC2 and a reduction in GluA2 AMPA glutamate receptor subunit expression, indicative of depolarizing GABA receptors and calcium permeable AMPA receptors. Finally, we found that chronic treatment with the mTORC1 inhibitor, rapamycin, at doses we have previously shown to attenuate seizure-induced amyloid-ß pathology and cognitive deficits, could also reverse elevations of the NKCC1/KCC2 ratio in these mice. Our data demonstrate novel mechanisms of interaction between AD and epilepsy and indicate that targeting E:I balance, potentially with US Food and Drug Administration-approved mTOR inhibitors, hold therapeutic promise for AD patients with a seizure history.


Subject(s)
Alzheimer Disease , Mice, Transgenic , Seizures , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Seizures/metabolism , Seizures/physiopathology , Mice , Male , Humans , Female , Pentylenetetrazole/toxicity , Aged , Disease Models, Animal , Kindling, Neurologic/drug effects , Aged, 80 and over
3.
Cell Mol Life Sci ; 81(1): 36, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38214768

ABSTRACT

N-methyl-D-aspartate receptors (NMDARs) play a critical role in normal brain function, and variants in genes encoding NMDAR subunits have been described in individuals with various neuropsychiatric disorders. We have used whole-cell patch-clamp electrophysiology, fluorescence microscopy and in-silico modeling to explore the functional consequences of disease-associated nonsense and frame-shift variants resulting in the truncation of GluN2A or GluN2B C-terminal domain (CTD). This study characterizes variant NMDARs and shows their reduced surface expression and synaptic localization, altered agonist affinity, increased desensitization, and reduced probability of channel opening. We also show that naturally occurring and synthetic steroids pregnenolone sulfate and epipregnanolone butanoic acid, respectively, enhance NMDAR function in a way that is dependent on the length of the truncated CTD and, further, is steroid-specific, GluN2A/B subunit-specific, and GluN1 splice variant-specific. Adding to the previously described effects of disease-associated NMDAR variants on the receptor biogenesis and function, our results improve the understanding of the molecular consequences of NMDAR CTD truncations and provide an opportunity for the development of new therapeutic neurosteroid-based ligands.


Subject(s)
Neurosteroids , Receptors, N-Methyl-D-Aspartate , Humans , Electrophysiological Phenomena , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
4.
J Neurosci ; 43(17): 3028-3041, 2023 04 26.
Article in English | MEDLINE | ID: mdl-36931707

ABSTRACT

Transcription factors have a pivotal role in synaptic plasticity and the associated modification of neuronal networks required for memory formation and consolidation. The nuclear receptors subfamily 4 group A (Nr4a) have emerged as possible modulators of hippocampal synaptic plasticity and cognitive functions. However, the molecular and cellular mechanisms underlying Nr4a2-mediated hippocampal synaptic plasticity are not completely known. Here, we report that neuronal activity enhances Nr4a2 expression and function in cultured mouse hippocampal neurons (both sexes) by an ionotropic glutamate receptor/Ca2+/cAMP response element-binding protein/CREB-regulated transcription factor 1 (iGluR/Ca2+/CREB/CRTC1) pathway. Nr4a2 activation mediates BDNF production and increases expression of iGluRs, thereby affecting LTD at CA3-CA1 synapses in acute mouse hippocampal slices (both sexes). Together, our results indicate that the iGluR/Ca2+/CREB/CRTC1 pathway mediates activity-dependent expression of Nr4a2, which is involved in glutamatergic synaptic plasticity by increasing BDNF and synaptic GluA1-AMPARs. Therefore, Nr4a2 activation could be a therapeutic approach for brain disorders associated with dysregulated synaptic plasticity.SIGNIFICANCE STATEMENT A major factor that regulates fast excitatory synaptic transmission and plasticity is the modulation of synaptic AMPARs. However, despite decades of research, the underlying mechanisms of this modulation remain poorly understood. Our study identified a molecular pathway that links neuronal activity with AMPAR modulation and hippocampal synaptic plasticity through the activation of Nr4a2, a member of the nuclear receptor subfamily 4. Since several compounds have been described to activate Nr4a2, our study not only provides mechanistic insights into the molecular pathways related to hippocampal synaptic plasticity and learning, but also identifies Nr4a2 as a potential therapeutic target for pathologic conditions associated with dysregulation of glutamatergic synaptic function.


Subject(s)
Brain-Derived Neurotrophic Factor , Receptors, AMPA , Male , Female , Mice , Animals , Receptors, AMPA/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Neuronal Plasticity/physiology , Hippocampus/physiology , Learning , Synapses/physiology , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Transcription Factors/metabolism
5.
J Neurochem ; 168(5): 719-727, 2024 May.
Article in English | MEDLINE | ID: mdl-38124277

ABSTRACT

The excitatory neurotransmitter glutamate has a role in neuronal migration and process elongation in the central nervous system (CNS). The effects of chronic glutamate hyperactivity on vesicular and protein transport within CNS neurons, that is, processes necessary for neurite growth, have not been examined previously. In this study, we measured the effects of lifelong hyperactivity of glutamate neurotransmission on axoplasmic transport in CNS neurons. We compared wild-type (wt) to transgenic (Tg) mice over-expressing the glutamate dehydrogenase gene Glud1 in CNS neurons and exhibiting increases in glutamate transmitter formation, release, and synaptic activation in brain throughout the lifespan. We found that Glud1 Tg as compared with wt mice exhibited increases in the rate of anterograde axoplasmic transport in neurons of the hippocampus measured in brain slices ex vivo, and in olfactory neurons measured in vivo. We also showed that the in vitro pharmacologic activation of glutamate synapses in wt mice led to moderate increases in axoplasmic transport, while exposure to selective inhibitors of ion channel forming glutamate receptors very significantly suppressed anterograde transport, suggesting a link between synaptic glutamate receptor activation and axoplasmic transport. Finally, axoplasmic transport in olfactory neurons of Tg mice in vivo was partially inhibited following 14-day intake of ethanol, a known suppressor of axoplasmic transport and of glutamate neurotransmission. The same was true for transport in hippocampal neurons in slices from Glud1 Tg mice exposed to ethanol for 2 h ex vivo. In conclusion, endogenous activity at glutamate synapses regulates and glutamate synaptic hyperactivity increases intraneuronal transport rates in CNS neurons.


Subject(s)
Glutamate Dehydrogenase , Mice, Transgenic , Neurons , Receptors, Glutamate , Animals , Mice , Glutamate Dehydrogenase/metabolism , Glutamate Dehydrogenase/genetics , Neurons/metabolism , Neurons/drug effects , Receptors, Glutamate/metabolism , Axonal Transport/drug effects , Axonal Transport/physiology , Glutamic Acid/metabolism , Hippocampus/metabolism , Mice, Inbred C57BL
6.
Annu Rev Neurosci ; 39: 257-76, 2016 07 08.
Article in English | MEDLINE | ID: mdl-27145911

ABSTRACT

Addiction is a disease of altered behavior. Addicts use drugs compulsively and will continue to do so despite negative consequences. Even after prolonged periods of abstinence, addicts are at risk of relapse, particularly when cues evoke memories that are associated with drug use. Rodent models mimic many of the core components of addiction, from the initial drug reinforcement to cue-associated relapse and continued drug intake despite negative consequences. Rodent models have also enabled unprecedented mechanistic insight into addiction, revealing plasticity of glutamatergic synaptic transmission evoked by the strong activation of mesolimbic dopamine-a defining feature of all addictive drugs-as a neural substrate for these drug-adaptive behaviors. Cell type-specific optogenetic manipulations have allowed both identification of the relevant circuits and design of protocols to reverse drug-evoked plasticity and to establish links of causality with drug-adaptive behaviors. The emergence of a circuit model for addiction will open the door for novel therapies, such as deep brain stimulation.


Subject(s)
Behavior, Addictive/physiopathology , Substance-Related Disorders/physiopathology , Animals , Brain/physiopathology , Dopamine/pharmacology , Humans , Neuronal Plasticity/drug effects , Synaptic Transmission/drug effects
7.
Chembiochem ; 25(6): e202300813, 2024 03 15.
Article in English | MEDLINE | ID: mdl-38227784

ABSTRACT

AMPA glutamate receptors (AMPARs) play a pivotal role in excitatory neurotransmission, particularly in the hippocampus where the TARP γ-8 subunit is enriched and serves as a target for emerging anti-epileptic drugs. To enable in vivo visualization of TARP γ-8 distribution and expression by positron emission tomography (PET), this study focuses on the development of novel 18 F-labeled TARP γ-8 inhibitors and their corresponding precursors, stemming from the azabenzimidazole scaffold. The resulting radioligands [18 F]TARP-2204 and [18 F]TARP-2205 were successfully synthesized with acceptable radiochemical yield, high molar activity, and excellent radiochemical purity. In vitro autoradiography demonstrates high level of specific binding of [18 F]TARP-2205 to TARP γ-8 in both rat and nonhuman primate brain tissues. However, unexpected radiodefluorination in PET imaging studies of rodents emphasizes the need for further structural refinement. This work serves as an excellent starting point for the development of future 18 F-labeled TARP γ-8 PET tracers, offering valuable insights into medicinal chemistry design, radiosynthesis and subsequent PET evaluation.


Subject(s)
Positron-Emission Tomography , Receptors, AMPA , Rats , Animals , Receptors, AMPA/metabolism , Positron-Emission Tomography/methods , Hippocampus
8.
Cell Mol Life Sci ; 80(11): 345, 2023 Nov 03.
Article in English | MEDLINE | ID: mdl-37921875

ABSTRACT

AMPA receptors are members of the glutamate receptor family and mediate a fast component of excitatory synaptic transmission at virtually all central synapses. Thus, their functional characteristics are a critical determinant of brain function. We evaluate intolerance of each GRIA gene to genetic variation using 3DMTR and report here the functional consequences of 52 missense variants in GRIA1-4 identified in patients with various neurological disorders. These variants produce changes in agonist EC50, response time course, desensitization, and/or receptor surface expression. We predict that these functional and localization changes will have important consequences for circuit function, and therefore likely contribute to the patients' clinical phenotype. We evaluated the sensitivity of variant receptors to AMPAR-selective modulators including FDA-approved drugs to explore potential targeted therapeutic options.


Subject(s)
Nervous System Diseases , Humans , Nervous System Diseases/genetics , Synaptic Transmission/physiology , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Synapses/metabolism
9.
Cell Mol Life Sci ; 80(2): 42, 2023 Jan 16.
Article in English | MEDLINE | ID: mdl-36645496

ABSTRACT

N-methyl-D-aspartate receptors (NMDARs) play vital roles in normal brain functions (i.e., learning, memory, and neuronal development) and various neuropathological conditions, such as epilepsy, autism, Parkinson's disease, Alzheimer's disease, and traumatic brain injury. Endogenous neuroactive steroids such as 24(S)-hydroxycholesterol (24(S)-HC) have been shown to influence NMDAR activity, and positive allosteric modulators (PAMs) derived from 24(S)-hydroxycholesterol scaffold can also enhance NMDAR function. This study describes the structural determinants and mechanism of action for 24(S)-hydroxycholesterol and two novel synthetic analogs (SGE-550 and SGE-301) on NMDAR function. We also show that these agents can mitigate the altered function caused by a set of loss-of-function missense variants in NMDAR GluN subunit-encoding GRIN genes associated with neurological and neuropsychiatric disorders. We anticipate that the evaluation of novel neuroactive steroid NMDAR PAMs may catalyze the development of new treatment strategies for GRIN-related neuropsychiatric conditions.


Subject(s)
Alzheimer Disease , Nervous System Diseases , Neurosteroids , Humans , Receptors, N-Methyl-D-Aspartate/metabolism , Neurosteroids/pharmacology , Neurosteroids/therapeutic use , Hydroxycholesterols/pharmacology , Hydroxycholesterols/therapeutic use , Nervous System Diseases/drug therapy , Nervous System Diseases/genetics , Alzheimer Disease/drug therapy , Steroids/pharmacology , Allosteric Regulation/physiology
10.
Cell Mol Life Sci ; 80(4): 110, 2023 Mar 31.
Article in English | MEDLINE | ID: mdl-37000222

ABSTRACT

The short pre-M1 helix within the S1-M1 linker (also referred to as the pre-M1 linker) between the agonist-binding domain (ABD, S1) and the M1 transmembrane helix of the NMDA receptor (NMDAR) is devoid of missense variants within the healthy population but is a locus for de novo pathogenic variants associated with neurological disorders. Several de novo variants within this helix have been identified in patients presenting early in life with intellectual disability, developmental delay, and/or epilepsy. In this study, we evaluated functional properties for twenty variants within the pre-M1 linker in GRIN1, GRIN2A, and GRIN2B genes, including six novel missense variants. The effects of pre-M1 variants on agonist potency, sensitivity to endogenous allosteric modulators, response time course, channel open probability, and surface expression were assessed. Our data indicated that virtually all of the variants evaluated altered channel function, and multiple variants had profound functional consequences, which may contribute to the neurological conditions in the patients harboring the variants in this region. These data strongly suggest that the residues within the pre-M1 helix play a key role in channel gating and are highly intolerant to genetic variation.


Subject(s)
Epilepsy , Intellectual Disability , Receptors, N-Methyl-D-Aspartate , Humans , Epilepsy/genetics , Mutation, Missense/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
11.
J Plant Res ; 137(3): 485-503, 2024 May.
Article in English | MEDLINE | ID: mdl-38448641

ABSTRACT

Plant glutamate receptor-like channels (GLRs) play important roles in plant development, immune response, defense signaling and Nitric oxide (NO) production. However, their involvement in abiotic stress responses, particularly in regulating Reactive Oxygen Species (ROS), is not well understood. This study aimed to investigate GLR-mediated NO production on ROS regulation in salt-stressed cells. To achieve this, Arabidopsis thaliana Columbia (Col-0) were treated with NaCl, glutamate antagonists [(DNQX (6,7-dinitroquinoxaline-2,3-dione and AP-5(D-2-amino-5-phosphono pentanoic acid)], and NO scavenger [cPTIO (2-(4-Carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide potassium salt)]. Salt-stressed plants in combination with DNQX and AP-5 have exhibited higher increase in lipid peroxidation (TBARS), hydrogen peroxide (H2O2) and superoxide radical (O-2) contents as compared to solely NaCl-treated plants. Furthermore, NO and total glutathione contents, and S-nitrosoglutathione reductase (GSNOR) activity decreased with these treatments. AP-5 and DNQX increased the activities of NADPH oxidase (NOX), catalase (CAT), peroxidase (POX), cell wall peroxidase (CWPOX) in salt-stressed Arabidopsis leaves. However, their activities (except NOX) were significantly inhibited by cPTIO. Conversely, the combination of NaCl and GLR antagonists, NO scavenger decreased the activities of ascorbate peroxidase (APX), superoxide dismutase (SOD), glutathione reductase (GR), dehydroascorbate reductase (DHAR) and monodehydroascorbate reductase (MDHAR) resulting in elevated GSSG levels, a low GSH/GSSG ratio, impaired ROS scavenging, excessive ROS accumulation and cell membrane damage. The findings of this study provide evidence that GLR-mediated NO plays a crucial role in improvement of the tolerance of Arabidopsis plants to salt-induced oxidative stress. It helps to maintain cellular redox homeostasis by reducing ROS accumulation and increasing the activity of SOD, GSNOR, and the ASC-GSH cycle enzymes.


Subject(s)
Arabidopsis , Nitric Oxide , Reactive Oxygen Species , Salt Stress , Arabidopsis/physiology , Arabidopsis/drug effects , Arabidopsis/metabolism , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Receptors, Glutamate/metabolism , Lipid Peroxidation/drug effects , Hydrogen Peroxide/metabolism
12.
J Appl Toxicol ; 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38828527

ABSTRACT

Glyphosate (GLY) is a pesticide that severely alters nigrostriatal dopaminergic neurotransmission, inducing great increases in dopamine release from rat dorsal striatum. This GLY-induced striatal dopamine overflow occurs through mechanisms not yet fully understood, hence the interest in evaluating the role of other neurotransmitter systems in such effects. So, the main objective of this mechanistic study was to evaluate the possible mediation of the glutamatergic, cholinergic, and nitrergic systems in the GLY-induced in vivo dopamine release from rat dorsal striatum. The extracellular dopamine levels were measured by cerebral microdialysis and HPLC with electrochemical detection. Intrastriatal administration of GLY (5 mmol/L) significantly increased the dopamine release (1102%). Pretreatment with MK-801 (50 or 400 µmol/L), a non-competitive antagonist of NMDA receptors, significantly decreased the effect of GLY (by 70% and 74%, respectively), whereas AP-5 (400 µmol/L), a competitive antagonist of NMDA receptors, or CNQX (500 µmol/L), an AMPA/kainate receptor antagonist, had no significant effect. Administration of the nitric oxide synthase inhibitors, L-nitroarginine (L-NAME, 100 µmol/L) or 7-nitroindazole (7-NI, 100 µmol/L), also did not alter the effect of GLY on dopamine release. Finally, pretreatment of the animals with mecamylamine, an antagonist of nicotinic receptors, decreased the effect of GLY on dopamine release by 49%, whereas atropine, a muscarinic antagonist, had no significant effect. These results indicate that GLY-induced dopamine release largely depends on the activation of NMDA and nicotinic receptors in rat dorsal striatum. Future research is needed to determine the effects of this pesticide at environmentally relevant concentrations.

13.
Mol Cell Neurosci ; 124: 103819, 2023 03.
Article in English | MEDLINE | ID: mdl-36720293

ABSTRACT

The postsynaptic density (PSD) of excitatory synapses is built from a wide variety of scaffolding proteins, receptors, and signaling molecules that collectively orchestrate synaptic transmission. Seminal work over the past decades has led to the identification and functional characterization of many PSD components. In contrast, we know far less about how these constituents are assembled within synapses, and how this organization contributes to synapse function. Notably, recent evidence from high-resolution microscopy studies and in silico models, highlights the importance of the precise subsynaptic structure of the PSD for controlling the strength of synaptic transmission. Even further, activity-driven changes in the distribution of glutamate receptors are acknowledged to contribute to long-term changes in synaptic efficacy. Thus, defining the mechanisms that drive structural changes within the PSD are important for a molecular understanding of synaptic transmission and plasticity. Here, we review the current literature on how the PSD is organized to mediate basal synaptic transmission and how synaptic activity alters the nanoscale organization of synapses to sustain changes in synaptic strength.


Subject(s)
Nanostructures , Synapses , Synapses/metabolism , Synaptic Transmission/physiology , Receptors, Glutamate/metabolism , Post-Synaptic Density/metabolism , Neuronal Plasticity/physiology
14.
J Labelled Comp Radiopharm ; 67(4): 120-130, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38332677

ABSTRACT

Kainate receptors play a crucial role in mediating synaptic transmission within the central nervous system. However, the lack of selective pharmacological tool compounds for the GluK3 subunit represents a significant challenge in studying these receptors. Recently presented compound 1 stands out as a potent antagonist of GluK3 receptors, exhibiting nanomolar affinity at GluK3 receptors and strongly inhibiting glutamate-induced currents at homomeric GluK1 and GluK3 receptors in HEK293 cells with Kb values of 65 and 39 nM, respectively. This study presents the synthesis of two potent GluK3-preferring iodine derivatives of compound 1, serving as precursors for radiolabelling. Furthermore, we demonstrate the optimisation of dehalogenation conditions using hydrogen and deuterium, resulting in [2H]-1, and demonstrate the efficient synthesis of the radioligand [3H]-1 with a specific activity of 1.48 TBq/mmol (40.1 Ci/mmol). Radioligand binding studies conducted with [3H]-1 as a radiotracer at GluK1, GluK2, and GluK3 receptors expressed in Sf9 and rat P2 membranes demonstrated its potential applicability for selectively studying native GluK3 receptors in the presence of GluK1 and 2-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor-blocking ligands.


Subject(s)
Glutamic Acid , Receptors, Kainic Acid , Rats , Animals , Humans , Tritium , Deuterium , HEK293 Cells , Receptors, Kainic Acid/chemistry , Receptors, Kainic Acid/metabolism , Receptors, AMPA/chemistry , Receptors, AMPA/metabolism
15.
Int J Mol Sci ; 25(2)2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38255760

ABSTRACT

Noribogaine (noribo) is the primary metabolite from ibogaine, an atypical psychedelic alkaloid isolated from the root bark of the African shrub Tabernanthe iboga. The main objective of this study was to test the hypothesis that molecular, electrophysiological, and behavioral responses of noribo are mediated by the 5-HT2A receptor (5-HT2AR) in mice. In that regard, we used male and female, 5-HT2AR knockout (KO) and wild type (WT) mice injected with a single noribo dose (10 or 40 mg/kg; i.p.). After 30 min., locomotor activity was recorded followed by mRNA measurements by qPCR (immediate early genes; IEG, glutamate receptors, and 5-HT2AR levels) and electrophysiology recordings of layer V pyramidal neurons from the medial prefrontal cortex. Noribo 40 decreased locomotion in male, but not female WT. Sex and genotype differences were observed for IEG and glutamate receptor expression. Expression of 5-HT2AR mRNA increased in the mPFC of WT mice following Noribo 10 (males) or Noribo 40 (females). Patch-clamp recordings showed that Noribo 40 reduced the NMDA-mediated postsynaptic current density in mPFC pyramidal neurons only in male WT mice, but no effects were found for either KO males or females. Our results highlight that noribo produces sexually dimorphic effects while the genetic removal of 5HT2AR blunted noribo-mediated responses to NMDA synaptic transmission.


Subject(s)
Ibogaine , Female , Male , Animals , Mice , Mice, Knockout , Ibogaine/pharmacology , Receptor, Serotonin, 5-HT2A/genetics , N-Methylaspartate , Serotonin , Glutamic Acid , RNA, Messenger
16.
Molecules ; 29(2)2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38257270

ABSTRACT

Brain-derived neurotrophic factor (BDNF) plays an important role in neurogenesis, synaptic plasticity, and cognition. BDNF is a neurotrophin that binds to tropomyosin receptor kinase B (TrkB), a specific receptor on target cell surfaces; it acts on neuronal formation, development, growth, and repair via transcription factors, such as cAMP response element-binding protein (CREB), and it is involved in learning and memory. BDNF expression is decreased in patients with Alzheimer's disease (AD). Exercise and the intake of several different foods or ingredients can increase BDNF expression, as confirmed with lutein, xanthophylls (polar carotenoids), and ethanolamine plasmalogen (PlsEtn), which are present at high levels in the brain. This study examined the effects of combining lutein and PlsEtn using lutein-rich Chlorella and ascidian extracts containing high levels of PlsEtn bearing docosahexaenoic acid, which is abundant in the human brain, on the activation of the BDNF-TrkB-CREB signaling pathway in the hippocampus of Sprague-Dawley rats. Although activation of the BDNF-TrkB-CREB signaling pathway in the hippocampus was not observed in Chlorella or ascidian PlsEtn monotherapy, activation was observed with combination therapy at an equal dose. The results of this study suggest that the combination of Chlorella and ascidian PlsEtn may have a preventive effect against dementia, including AD.


Subject(s)
Alzheimer Disease , Chlorella , Plasmalogens , Humans , Rats , Animals , Brain-Derived Neurotrophic Factor , Lutein , Rats, Sprague-Dawley , Signal Transduction , Brain , Alzheimer Disease/drug therapy
17.
J Neurosci ; 42(29): 5672-5680, 2022 07 20.
Article in English | MEDLINE | ID: mdl-35705487

ABSTRACT

NMDARs are ionotropic glutamate receptors widely expressed in the CNS, where they mediate phenomena as diverse as neurotransmission, information processing, synaptogenesis, and cellular toxicity. They function as glutamate-gated Ca2+-permeable channels, which require glycine as coagonist, and can be modulated by many diffusible ligands and cellular cues, including mechanical stimuli. Previously, we found that, in cultured astrocytes, shear stress initiates NMDAR-mediated Ca2+ entry in the absence of added agonists, suggesting that more than being mechanosensitive, NMDARs may be mechanically activated. Here, we used controlled expression of rat recombinant receptors and noninvasive on-cell single-channel current recordings to show that mild membrane stretch can substitute for the neurotransmitter glutamate in gating NMDAR currents. Notably, stretch-activated currents maintained the hallmark features of the glutamate-gated currents, including glycine-requirement, large unitary conductance, high Ca2+ permeability, and voltage-dependent Mg2+ blockade. Further, we found that the stretch-gated current required the receptor's intracellular domain. Our results are consistent with the hypothesis that mechanical forces can gate endogenous NMDAR currents even in the absence of synaptic glutamate release, which has important implications for understanding mechanotransduction and the physiological and pathologic effects of mechanical forces on cells of the CNS.SIGNIFICANCE STATEMENT We show that, in addition to enhancing currents elicited with low agonist concentrations, membrane stretch can gate NMDARs in the absence of the neurotransmitter glutamate. Stretch-gated currents have the principal hallmarks of the glutamate-gated currents, including requirement for glycine, large Na+ conductance, high Ca2+ permeability, and voltage-dependent Mg2+ block. Therefore, results suggest that mechanical forces can initiate cellular processes presently attributed to glutamatergic neurotransmission, such as synaptic plasticity and cytotoxicity. Given the ubiquitous presence of mechanical forces in the CNS, this discovery identifies NMDARs as possibly important mechanotransducers during development and across the lifespan, and during pathologic processes, such as those associated with traumatic brain injuries, shaken infant syndrome, and chronic traumatic encephalopathy.


Subject(s)
Mechanotransduction, Cellular , Receptors, N-Methyl-D-Aspartate , Animals , Glutamic Acid/metabolism , Glycine/metabolism , Glycine/pharmacology , Humans , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission
18.
J Neurosci ; 42(13): 2662-2677, 2022 03 30.
Article in English | MEDLINE | ID: mdl-35165175

ABSTRACT

Palmitoylation may be relevant to the processes of learning and memory, and even disorders, such as post-traumatic stress disorder and aging-related cognitive decline. However, underlying mechanisms of palmitoylation in these processes remain unclear. Herein, we used acyl-biotin exchange, coimmunoprecipitation and biotinylation assays, and behavioral and electrophysiological methods, to explore whether palmitoylation is required for hippocampal synaptic transmission and fear memory formation, and involved in functional modification of synaptic proteins, such as postsynapse density-95 (PSD-95) and glutamate receptors, and detected if depalmitoylation by specific enzymes has influence on glutamatergic synaptic plasticity. Our results showed that global palmitoylation level, palmitoylation of PSD-95 and glutamate receptors, postsynapse density localization of PSD-95, surface expression of AMPARs, and synaptic strength of cultured hippocampal neurons were all enhanced by TTX pretreatment, and these can be reversed by inhibition of palmitoylation with palmitoyl acyl transferases inhibitors, 2-bromopalmitate and N-(tert-butyl) hydroxylamine hydrochloride. Importantly, we also found that acyl-protein thioesterase 1 (APT1)-mediated depalmitoylation is involved in palmitoylation of PSD-95 and glutamatergic synaptic transmission. Knockdown of APT1, not protein palmitoyl thioesterase 1, with shRNA, or selective inhibition, significantly increased AMPAR-mediated synaptic strength, palmitoylation levels, and synaptic or surface expression of PSD-95 and AMPARs. Results from hippocampal tissues and fear-conditioned rats showed that palmitoylation is required for synaptic strengthening and fear memory formation. These results suggest that palmitoylation and APT1-mediated depalmitoylation have critical effects on the regulation of glutamatergic synaptic plasticity, and it may serve as a potential target for learning and memory-associated disorders.SIGNIFICANCE STATEMENT Fear-related anxiety disorders, including post-traumatic stress disorder, are prevalent psychiatric conditions, and fear memory is associated with hyperexcitability in the hippocampal CA1 region. Palmitoylation is involved in learning and memory, but mechanisms coupling palmitoylation with fear memory acquisition remain poorly understood. This study demonstrated that palmitoylation is essential for postsynapse density-95 clustering and hippocampal glutamatergic synaptic transmission, and APT1-mediated depalmitoylation plays critical roles in the regulation of synaptic plasticity. Our study revealed that molecular mechanism about downregulation of APT1 leads to enhancement of AMPAR-mediated synaptic transmission, and that palmitoylation cycling is implicated in fear conditioning-induced synaptic strengthening and fear memory formation.


Subject(s)
Hippocampus , Synapses , Animals , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Neuronal Plasticity , Rats , Synapses/metabolism , Synaptic Transmission/physiology
19.
J Neurosci ; 42(5): 731-748, 2022 02 02.
Article in English | MEDLINE | ID: mdl-34907024

ABSTRACT

Group I mGluRs have diverse functions in some fundamental neuronal processes, including modulation of synaptic plasticity; and dysregulation of these receptors could lead to various neuropsychiatric disorders. Trafficking of Group I mGluRs plays critical roles in controlling the precise spatiotemporal localization and activity of these receptors, both of which contribute to proper downstream signaling. Using "molecular replacement" approach in hippocampal neurons derived from mice of both sexes, we demonstrate a critical role for the postsynaptic density protein Norbin in regulating the ligand-induced internalization of Group I mGluRs. We show that Norbin associates with protein kinase A (PKA) through its N-terminus and anchors mGluR5 through its C-terminus, both of which are necessary for the ligand-mediated endocytosis of mGluR5, a member of the Group I mGluR family. A point mutation (A687G) at the C-terminus of Norbin inhibits the binding of Norbin to mGluR5 and blocks mGluR5 endocytosis. Finally, we demonstrate an important mechanism by which Norbin regulates mGluR-mediated AMPAR endocytosis in hippocampal neurons, a cellular correlate for mGluR-dependent synaptic plasticity. Norbin, through its PKA-binding regions, recruits PKA to AMPARs on activation of mGluRs; and deletion of the PKA-binding regions of Norbin inhibits mGluR-triggered AMPAR endocytosis. We further report that Norbin is important specifically for the mGluR-mediated AMPAR endocytosis, but not for NMDAR-dependent AMPAR endocytosis. Thus, this study unravels a novel role for Norbin in the internalization of mGluRs and mGluR-mediated AMPAR endocytosis that can have clinical relevance to the function of Group I mGluRs in pathologic processes.SIGNIFICANCE STATEMENT The postsynaptic protein Norbin interacts with mGluR5, and both of them have been implicated in disorders, such as schizophrenia. However, the mechanistic basis underlying the regulation of mGluRs by Norbin remains elusive. We have identified Norbin as an essential mediator of ligand-mediated endocytosis of Group I mGluRs. Mechanistically, Norbin N-terminus associates with protein kinase-A (PKA) and C-terminus binds to mGluR5 to coordinate receptor internalization. A point mutation NorA687G inhibits endocytosis by disrupting this interaction. Additionally, Norbin is critical for the recruitment of PKA to AMPARs on activation of Group I mGluRs that assists in mGluR-mediated AMPAR endocytosis. Thus, Norbin has a dual function in the hippocampus: regulation of mGluR internalization and PKA-dependent modulation of mGluR-mediated AMPAR endocytosis, a prerequisite for mGluR-mediated synaptic plasticity.


Subject(s)
Endocytosis/physiology , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/physiology , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, AMPA/metabolism , Synapses/metabolism , Animals , Animals, Newborn , Cells, Cultured , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Receptor, Metabotropic Glutamate 5/genetics , Receptors, AMPA/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/genetics
20.
Glia ; 71(4): 819-847, 2023 04.
Article in English | MEDLINE | ID: mdl-36453615

ABSTRACT

Receptors for α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPARs) are ligand-gated ionotropic receptors for glutamate that is a major excitatory neurotransmitter in the central nervous system. AMPARs are located at postsynaptic sites of neuronal synapses where they mediate fast synaptic signaling and synaptic plasticity. Remarkably, AMPARs are also expressed by glial cells. Their expression by the oligodendrocyte (OL) lineage cells is of special interest because AMPARs mediate fast synaptic communication between neurons and oligodendrocyte progenitor cells (OPCs), modulate proliferation and differentiation of OPCs, and may also be involved in regulation of myelination. On the other hand, during pathological conditions, AMPARs may mediate damage of the OL lineage cells. In the present review, we focus on the technical approaches that have been used to study AMPARs in the OL lineage cells, and discuss future perspectives of AMPAR research in these glial cells.


Subject(s)
Neurons , Receptors, AMPA , Receptors, AMPA/metabolism , Cell Lineage , Neurons/metabolism , Neuroglia/metabolism , Oligodendroglia/metabolism , Synapses/metabolism , Synaptic Transmission
SELECTION OF CITATIONS
SEARCH DETAIL