Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57.799
Filter
1.
Cell Mol Biol Lett ; 29(1): 108, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39127627

ABSTRACT

Palmitoylation is a type of lipid modification that plays an important role in various aspects of neuronal function. Over the past few decades, several studies have shown that the palmitoylation of synaptic proteins is involved in neurotransmission and synaptic functions. Palmitoyl acyltransferases (PATs), which belong to the DHHC family, are major players in the regulation of palmitoylation. Dysregulated palmitoylation of synaptic proteins and mutated/dysregulated DHHC proteins are associated with several neurodegenerative diseases, such as Alzheimer's disease (AD), Huntington's disease (HD), and Parkinson's disease (PD). In this review, we summarize the recent discoveries on the subcellular distribution of DHHC proteins and analyze their expression patterns in different brain cells. In particular, this review discusses how palmitoylation of synaptic proteins regulates synaptic vesicle exocytotic fusion and the localization, clustering, and transport of several postsynaptic receptors, as well as the role of palmitoylation of other proteins in regulating synaptic proteins. Additionally, some of the specific known associations of these factors with neurodegenerative disorders are explored, with a few suggestions for the development of therapeutic strategies. Finally, this review provides possible directions for future research to reveal detailed and specific mechanisms underlying the roles of synaptic protein palmitoylation.


Subject(s)
Lipoylation , Neurodegenerative Diseases , Synapses , Humans , Neurodegenerative Diseases/metabolism , Animals , Synapses/metabolism , Acyltransferases/metabolism , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Synaptic Transmission
2.
Int J Mol Sci ; 25(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39125997

ABSTRACT

The transmembrane protein ß-amyloid precursor protein (APP) is central to the pathophysiology of Alzheimer's disease (AD). The ß-amyloid hypothesis posits that aberrant processing of APP forms neurotoxic ß-amyloid aggregates, which lead to the cognitive impairments observed in AD. Although numerous additional factors contribute to AD, there is a need to better understand the synaptic function of APP. We have found that Drosophila APP-like (APPL) has both shared and non-shared roles at the synapse with Kismet (Kis), a chromatin helicase binding domain (CHD) protein. Kis is the homolog of CHD7 and CHD8, both of which are implicated in neurodevelopmental disorders including CHARGE Syndrome and autism spectrum disorders, respectively. Loss of function mutations in kis and animals expressing human APP and BACE in their central nervous system show reductions in the glutamate receptor subunit, GluRIIC, the GTPase Rab11, and the bone morphogenetic protein (BMP), pMad, at the Drosophila larval neuromuscular junction (NMJ). Similarly, processes like endocytosis, larval locomotion, and neurotransmission are deficient in these animals. Our pharmacological and epistasis experiments indicate that there is a functional relationship between Kis and APPL, but Kis does not regulate appl expression at the larval NMJ. Instead, Kis likely influences the synaptic localization of APPL, possibly by promoting rab11 transcription. These data identify a potential mechanistic connection between chromatin remodeling proteins and aberrant synaptic function in AD.


Subject(s)
Amyloid beta-Protein Precursor , Drosophila Proteins , Neuromuscular Junction , rab GTP-Binding Proteins , Animals , Neuromuscular Junction/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/genetics , Synaptic Transmission , Synapses/metabolism , Receptors, Glutamate/metabolism , Receptors, Glutamate/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Humans , DNA Helicases/metabolism , DNA Helicases/genetics , Membrane Proteins , Nerve Tissue Proteins , Homeodomain Proteins , Receptors, Ionotropic Glutamate
3.
Neurol Neuroimmunol Neuroinflamm ; 11(5): e200284, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39141878

ABSTRACT

BACKGROUND AND OBJECTIVES: Autoantibodies against the protein leucine-rich glioma inactivated 1 (LGI1) cause the most common subtype of autoimmune encephalitis with predominant involvement of the limbic system, associated with seizures and memory deficits. LGI1 and its receptor ADAM22 are part of a transsynaptic protein complex that includes several proteins involved in presynaptic neurotransmitter release and postsynaptic glutamate sensing. Autoantibodies against LGI1 increase excitatory synaptic strength, but studies that genetically disrupt the LGI1-ADAM22 complex report a reduction in postsynaptic glutamate receptor-mediated responses. Thus, the mechanisms underlying the increased synaptic strength induced by LGI1 autoantibodies remain elusive, and the contributions of presynaptic molecules to the LGI1-transsynaptic complex remain unclear. We therefore investigated the presynaptic mechanisms that mediate autoantibody-induced synaptic strengthening. METHODS: We studied the effects of patient-derived purified polyclonal LGI1 autoantibodies on synaptic structure and function by combining direct patch-clamp recordings from presynaptic boutons and somata of hippocampal neurons with super-resolution light and electron microscopy of hippocampal cultures and brain slices. We also identified the protein domain mediating the presynaptic effect using domain-specific patient-derived monoclonal antibodies. RESULTS: LGI1 autoantibodies dose-dependently increased short-term depression during high-frequency transmission, consistent with increased release probability. The increased neurotransmission was not related to presynaptic calcium channels because presynaptic Cav2.1 channel density, calcium current amplitude, and calcium channel gating were unaffected by LGI1 autoantibodies. By contrast, application of LGI1 autoantibodies homogeneously reduced Kv1.1 and Kv1.2 channel density on the surface of presynaptic boutons. Direct presynaptic patch-clamp recordings revealed that LGI1 autoantibodies cause a pronounced broadening of the presynaptic action potential. Domain-specific effects of LGI1 autoantibodies were analyzed at the neuronal soma. Somatic action potential broadening was induced by polyclonal LGI1 autoantibodies and patient-derived monoclonal autoantibodies targeting the epitempin domain, but not the leucin-rich repeat domain. DISCUSSION: Our results indicate that LGI1 autoantibodies reduce the density of both Kv1.1 and Kv1.2 on presynaptic boutons, without actions on calcium channel density or function, thereby broadening the presynaptic action potential and increasing neurotransmitter release. This study provides a molecular explanation for the neuronal hyperactivity observed in patients with LGI1 autoantibodies.


Subject(s)
Action Potentials , Autoantibodies , Intracellular Signaling Peptides and Proteins , Presynaptic Terminals , Synaptic Transmission , Autoantibodies/immunology , Autoantibodies/pharmacology , Humans , Animals , Synaptic Transmission/physiology , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Presynaptic Terminals/metabolism , Action Potentials/physiology , Action Potentials/drug effects , Hippocampus/metabolism , Rats , Kv1.1 Potassium Channel/immunology , Proteins/immunology , Proteins/metabolism , Male , Cells, Cultured
4.
Int J Mol Sci ; 25(15)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39125587

ABSTRACT

Age-related conditions, such as sarcopenia, cause physical disabilities for an increasing section of society. At the neuromuscular junction, the postsynaptic-derived neurotrophic factors brain-derived neurotrophic factor (BDNF) and neurotrophin 4 (NT-4) have neuroprotective functions and contribute to the correct regulation of the exocytotic machinery. Similarly, presynaptic muscarinic signalling plays a fundamental modulatory function in this synapse. However, whether or not these signalling pathways are compromised in ageing neuromuscular system has not yet been analysed. The present study analyses, through Western blotting, the differences in expression and activation of the main key proteins of the BDNF/NT-4 and muscarinic pathways related to neurotransmission in young versus ageing Extensor digitorum longus (EDL) rat muscles. The main results show an imbalance in several sections of these pathways: (i) a change in the stoichiometry of BDNF/NT-4, (ii) an imbalance of Tropomyosin-related kinase B receptor (TrkB)-FL/TrkB-T1 and neurotrophic receptor p 75 (p75NTR), (iii) no changes in the cytosol/membrane distribution of phosphorylated downstream protein kinase C (PKC)ßI and PKCε, (iv) a reduction in the M2-subtype muscarinic receptor and P/Q-subtype voltage-gated calcium channel, (v) an imbalance of phosphorylated mammalian uncoordinated-18-1 (Munc18-1) (S313) and synaptosomal-associated protein 25 (SNAP-25) (S187), and (vi) normal levels of molecules related to the management of acetylcholine (Ach). Based on this descriptive analysis, we hypothesise that these pathways can be adjusted to ensure neurotransmission rather than undergoing negative alterations caused by ageing. However, further studies are needed to assess this hypothetical suggestion. Our results contribute to the understanding of some previously described neuromuscular functional age-related impairments. Strategies to promote these signalling pathways could improve the neuromuscular physiology and quality of life of older people.


Subject(s)
Aging , Brain-Derived Neurotrophic Factor , Neuromuscular Junction , Receptor, trkB , Signal Transduction , Brain-Derived Neurotrophic Factor/metabolism , Animals , Neuromuscular Junction/metabolism , Aging/metabolism , Rats , Receptor, trkB/metabolism , Nerve Growth Factors/metabolism , Male , Receptors, Muscarinic/metabolism , Synaptic Transmission , Receptors, Nerve Growth Factor/metabolism , Rats, Wistar
5.
Nat Commun ; 15(1): 6594, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39097618

ABSTRACT

Neurons coordinate inter-tissue protein homeostasis to systemically manage cytotoxic stress. In response to neuronal mitochondrial stress, specific neuronal signals coordinate the systemic mitochondrial unfolded protein response (UPRmt) to promote organismal survival. Yet, whether chemical neurotransmitters are sufficient to control the UPRmt in physiological conditions is not well understood. Here, we show that gamma-aminobutyric acid (GABA) inhibits, and acetylcholine (ACh) promotes the UPRmt in the Caenorhabditis elegans intestine. GABA controls the UPRmt by regulating extra-synaptic ACh release through metabotropic GABAB receptors GBB-1/2. We find that elevated ACh levels in animals that are GABA-deficient or lack ACh-degradative enzymes induce the UPRmt through ACR-11, an intestinal nicotinic α7 receptor. This neuro-intestinal circuit is critical for non-autonomously regulating organismal survival of oxidative stress. These findings establish chemical neurotransmission as a crucial regulatory layer for nervous system control of systemic protein homeostasis and stress responses.


Subject(s)
Acetylcholine , Caenorhabditis elegans , Mitochondria , Signal Transduction , Animals , Acetylcholine/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , alpha7 Nicotinic Acetylcholine Receptor/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , gamma-Aminobutyric Acid/metabolism , Intestines/physiology , Mitochondria/metabolism , Neurons/metabolism , Oxidative Stress , Receptors, GABA-B/metabolism , Receptors, GABA-B/genetics , Stress, Physiological , Synaptic Transmission/physiology , Unfolded Protein Response
6.
Cell Mol Life Sci ; 81(1): 342, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39123091

ABSTRACT

A Disintegrin And Metalloproteinase 10 (ADAM10) plays a pivotal role in shaping neuronal networks by orchestrating the activity of numerous membrane proteins through the shedding of their extracellular domains. Despite its significance in the brain, the specific cellular localization of ADAM10 remains not well understood due to a lack of appropriate tools. Here, using a specific ADAM10 antibody suitable for immunostainings, we observed that ADAM10 is localized to presynapses and especially enriched at presynaptic vesicles of mossy fiber (MF)-CA3 synapses in the hippocampus. These synapses undergo pronounced frequency facilitation of neurotransmitter release, a process that play critical roles in information transfer and neural computation. We demonstrate, that in conditional ADAM10 knockout mice the ability of MF synapses to undergo this type of synaptic plasticity is greatly reduced. The loss of facilitation depends on the cytosolic domain of ADAM10 and association with the calcium sensor synaptotagmin 7 rather than ADAM10's proteolytic activity. Our findings unveil a new role of ADAM10 in the regulation of synaptic vesicle exocytosis.


Subject(s)
ADAM10 Protein , Amyloid Precursor Protein Secretases , Membrane Proteins , Mice, Knockout , Neuronal Plasticity , Synaptic Vesicles , Animals , ADAM10 Protein/metabolism , ADAM10 Protein/genetics , Neuronal Plasticity/physiology , Amyloid Precursor Protein Secretases/metabolism , Amyloid Precursor Protein Secretases/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice , Synaptic Vesicles/metabolism , Mice, Inbred C57BL , Synapses/metabolism , Mossy Fibers, Hippocampal/metabolism , Hippocampus/metabolism , Exocytosis/physiology , Presynaptic Terminals/metabolism , Synaptic Transmission , Synaptotagmins/metabolism , Synaptotagmins/genetics
7.
Hippocampus ; 34(9): 454-463, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39150316

ABSTRACT

Estrogens are believed to modulate cognitive functions in part through the modulation of synaptic transmission in the cortex and hippocampus. Administration of 17ß-estradiol (E2) can rapidly enhance excitatory synaptic transmission in the hippocampus and facilitate excitatory synaptic transmission in rat lateral entorhinal cortex via activation of the G protein-coupled estrogen receptor-1 (GPER1). To assess the mechanisms through which GPER1 activation facilitates synaptic transmission, we assessed the effects of acute 10 nM E2 administration on pharmacologically isolated evoked excitatory and inhibitory synaptic currents in layer II/III entorhinal neurons. Female Long-Evans rats were ovariectomized between postnatal day (PD) 63 and 74 and implanted with a subdermal E2 capsule to maintain continuous low levels of E2. Electrophysiological recordings were obtained between 7 and 20 days after ovariectomy. Application of E2 for 20 min did not significantly affect AMPA or NMDA receptor-mediated excitatory synaptic currents. However, GABA receptor-mediated inhibitory synaptic currents (IPSCs) were markedly reduced by E2 and returned towards baseline levels during the 20-min washout period. The inhibition of GABA-mediated IPSCs was blocked in the presence of the GPER1 receptor antagonist G15. GPER1 can modulate protein kinase A (PKA), but blocking PKA with intracellular KT5720 did not prevent the E2-induced reduction in IPSCs. GPER1 can also stimulate extracellular signal-regulated kinase (ERK), a negative modulator of GABAA receptors, and blocking activation of ERK with PD90859 prevented the E2-induced reduction of IPSCs. E2 can therefore result in a rapid GPER1 and ERK signaling-mediated reduction in GABA-mediated IPSCs. This provides a novel mechanism through which E2 can rapidly modulate synaptic excitability in entorhinal layer II/III neurons and may also contribute to E2 and ERK-dependent alterations in synaptic transmission in other brain areas.


Subject(s)
Entorhinal Cortex , Estradiol , Extracellular Signal-Regulated MAP Kinases , Neurons , Rats, Long-Evans , Receptors, G-Protein-Coupled , Animals , Entorhinal Cortex/drug effects , Entorhinal Cortex/physiology , Receptors, G-Protein-Coupled/metabolism , Estradiol/pharmacology , Female , Neurons/drug effects , Neurons/metabolism , Rats , Extracellular Signal-Regulated MAP Kinases/metabolism , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Receptors, Estrogen/metabolism , Ovariectomy , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Patch-Clamp Techniques , Estrogens/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors
8.
Georgian Med News ; (350): 36-41, 2024 May.
Article in English | MEDLINE | ID: mdl-39089268

ABSTRACT

Methionine sulfoxide reductase A (MsrA) is an antioxidant enzyme that repairs the oxidation of methionine residues in proteins and free methionine in autism spectrum disorder (ASD). The present study aimed to assess the level of MsrA and neurotransmission enzymes in ASD individuals. Results confirmed that ASD associated with significant (P<0.05) reduction of MsrA and modulated mission enzymes. The role of MsrA as repair enzyme should be taken into account for study the activity of brain enzymes and proteins in ASD including ASMT that has a role in melatonin problems production in ASD due to higher AANAT level. The influence of MsrA also should be studied with MAT in mice to give more evidence.


Subject(s)
Autism Spectrum Disorder , Methionine Sulfoxide Reductases , Humans , Autism Spectrum Disorder/enzymology , Methionine Sulfoxide Reductases/metabolism , Methionine Sulfoxide Reductases/genetics , Male , Female , Synaptic Transmission , Child , Melatonin/metabolism , Adolescent , Child, Preschool , Adult , Case-Control Studies , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/metabolism , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/genetics
9.
PLoS One ; 19(8): e0306423, 2024.
Article in English | MEDLINE | ID: mdl-39088455

ABSTRACT

Hyperexcitability of motor neurons and spinal cord motor circuitry has been widely reported in the early stages of Amyotrophic Lateral Sclerosis (ALS). Changes in the relative amount of excitatory to inhibitory inputs onto a neuron (E:I synaptic ratio), possibly through a developmental shift in synapse formation in favour of excitatory transmission, could underlie pathological hyperexcitability. Given that astrocytes play a major role in early synaptogenesis and are implicated in ALS pathogenesis, their potential contribution to disease mechanisms involving synaptic imbalances and subsequent hyperexcitability is also of great interest. In order to assess E:I ratios in ALS, we utilised a novel primary spinal neuron / astrocyte co-culture system, derived from neonatal mice, in which synapses are formed in vitro. Using multiple ALS mouse models we found that no combination of astrocyte or neuron genotype produced alterations in E:I synaptic ratios assessed using pre- and post-synaptic anatomical markers. Similarly, we observed that ephrin-B1, a major contact-dependent astrocytic synaptogenic protein, was not differentially expressed by ALS primary astrocytes. Further to this, analysis of E:I ratios across the entire grey matter of the lumbar spinal cord in young (post-natal day 16-19) ALS mice revealed no differences versus controls. Finally, analysis in co-cultures of human iPSC-derived motor neurons and astrocytes harbouring the pathogenic C9orf72 hexanucleotide repeat expansion showed no evidence of a bias toward excitatory versus inhibitory synapse formation. We therefore conclude, utilising multiple ALS models, that we do not observe significant changes in the relative abundance of excitatory versus inhibitory synapses as would be expected if imbalances in synaptic inputs contribute to early hyperexcitability.


Subject(s)
Amyotrophic Lateral Sclerosis , Astrocytes , Coculture Techniques , Disease Models, Animal , Motor Neurons , Spinal Cord , Synapses , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Astrocytes/metabolism , Astrocytes/pathology , Mice , Synapses/metabolism , Synapses/physiology , Motor Neurons/metabolism , Motor Neurons/pathology , Motor Neurons/physiology , Spinal Cord/metabolism , Spinal Cord/pathology , Humans , Excitatory Postsynaptic Potentials , Mice, Transgenic , Cells, Cultured , Synaptic Transmission
10.
Elife ; 122024 Aug 06.
Article in English | MEDLINE | ID: mdl-39106188

ABSTRACT

Biological synaptic transmission is unreliable, and this unreliability likely degrades neural circuit performance. While there are biophysical mechanisms that can increase reliability, for instance by increasing vesicle release probability, these mechanisms cost energy. We examined four such mechanisms along with the associated scaling of the energetic costs. We then embedded these energetic costs for reliability in artificial neural networks (ANNs) with trainable stochastic synapses, and trained these networks on standard image classification tasks. The resulting networks revealed a tradeoff between circuit performance and the energetic cost of synaptic reliability. Additionally, the optimised networks exhibited two testable predictions consistent with pre-existing experimental data. Specifically, synapses with lower variability tended to have (1) higher input firing rates and (2) lower learning rates. Surprisingly, these predictions also arise when synapse statistics are inferred through Bayesian inference. Indeed, we were able to find a formal, theoretical link between the performance-reliability cost tradeoff and Bayesian inference. This connection suggests two incompatible possibilities: evolution may have chanced upon a scheme for implementing Bayesian inference by optimising energy efficiency, or alternatively, energy-efficient synapses may display signatures of Bayesian inference without actually using Bayes to reason about uncertainty.


Subject(s)
Bayes Theorem , Neural Networks, Computer , Synapses , Synapses/physiology , Models, Neurological , Synaptic Transmission/physiology , Energy Metabolism , Animals , Neurons/physiology
11.
Sci Rep ; 14(1): 15239, 2024 07 02.
Article in English | MEDLINE | ID: mdl-38956130

ABSTRACT

Dysbindin-1, a protein encoded by the schizophrenia susceptibility gene DTNBP1, is reduced in the hippocampus of schizophrenia patients. It is expressed in various cellular populations of the brain and implicated in dopaminergic and glutamatergic transmission. To investigate the impact of reduced dysbindin-1 in excitatory cells on hippocampal-associated behaviors and synaptic transmission, we developed a conditional knockout mouse model with deletion of dysbindin-1 gene in CaMKIIα expressing cells. We found that dysbindin-1 reduction in CaMKII expressing cells resulted in impaired spatial and social memories, and attenuation of the effects of glutamate N-methyl-d-asparate receptor (NMDAR) antagonist MK801 on locomotor activity and prepulse inhibition of startle (PPI). Dysbindin-1 deficiency in CaMKII expressing cells also resulted in reduced protein levels of NMDAR subunit GluN1 and GluN2B. These changes were associated with increased expression of immature dendritic spines in basiliar dendrites and abnormalities in excitatory synaptic transmission in the ventral hippocampus. These results highlight the functional relevance of dysbindin-1 in excitatory cells and its implication in schizophrenia-related pathologies.


Subject(s)
Dysbindin , Hippocampus , Mice, Knockout , Neurons , Receptors, N-Methyl-D-Aspartate , Synaptic Transmission , Animals , Dysbindin/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Hippocampus/metabolism , Mice , Neurons/metabolism , Schizophrenia/metabolism , Schizophrenia/pathology , Schizophrenia/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Male , Dizocilpine Maleate/pharmacology , Behavior, Animal , Dendritic Spines/metabolism , Nerve Tissue Proteins
12.
eNeuro ; 11(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38997143

ABSTRACT

Psychotic symptoms and delusional beliefs have been linked to dopamine transmission in both healthy and clinical samples and are assumed to result at least in part from perceiving illusory patterns in noise. However, the existing literature on the role of dopamine in detecting patterns in noise is inconclusive. To address this issue, we assessed the effect of manipulating dopaminergic neurotransmission on illusory pattern perception in healthy individuals (n = 48, n = 19 female) in a double-blind placebo-controlled within-subjects design (see preregistration at https://osf.io/a4k9j/). We predicted individuals on versus off ʟ-DOPA to be more likely to perceive illusory patterns, specifically objects in images containing only noise. Using a signal detection model, however, we found no credible evidence that ʟ-DOPA compared with placebo increased false alarm rates. Further, ʟ-DOPA did not reliably modulate measures of accuracy, discrimination sensitivity, and response bias. In all cases, Bayesian statistics revealed strong evidence in favor of the null hypothesis. The task design followed previous work on illusory pattern perception and comprised a limited number of items per condition. The results therefore need to be interpreted with caution, as power was limited. Future studies should address illusory pattern perception using more items and take into account potential dose-dependent effects and differential effects in healthy versus clinical samples.


Subject(s)
Dopamine , Illusions , Levodopa , Humans , Female , Male , Double-Blind Method , Adult , Illusions/physiology , Illusions/drug effects , Dopamine/metabolism , Young Adult , Levodopa/pharmacology , Levodopa/administration & dosage , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Dopamine Agents/pharmacology , Pattern Recognition, Visual/physiology , Pattern Recognition, Visual/drug effects , Bayes Theorem
13.
Cells ; 13(13)2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38994982

ABSTRACT

There has been a significant increase in the consumption of cannabis for both recreational and medicinal purposes in recent years, and its use can have long-term consequences on cognitive functions, including memory. Here, we review the immediate and long-term effects of cannabis and its derivatives on glutamatergic neurotransmission, with a focus on both the presynaptic and postsynaptic alterations. Several factors can influence cannabinoid-mediated changes in glutamatergic neurotransmission, including dosage, sex, age, and frequency of use. Acute exposure to cannabis typically inhibits glutamate release, whereas chronic use tends to increase glutamate release. Conversely, the postsynaptic alterations are more complicated than the presynaptic effects, as cannabis can affect the glutamate receptor expression and the downstream signaling of glutamate. All these effects ultimately influence cognitive functions, particularly memory. This review will cover the current research on glutamate-cannabis interactions, as well as the future directions of research needed to understand cannabis-related health effects and neurological and psychological aspects of cannabis use.


Subject(s)
Cannabinoids , Cannabis , Glutamic Acid , Synaptic Transmission , Humans , Synaptic Transmission/drug effects , Cannabinoids/pharmacology , Cannabinoids/metabolism , Glutamic Acid/metabolism , Cannabis/metabolism , Animals
14.
PLoS One ; 19(7): e0306605, 2024.
Article in English | MEDLINE | ID: mdl-38968286

ABSTRACT

Delays in nerve transmission are an important topic in the field of neuroscience. Spike signals fired or received by the dendrites of a neuron travel from the axon to a presynaptic cell. The spike signal then triggers a chemical reaction at the synapse, wherein a presynaptic cell transfers neurotransmitters to the postsynaptic cell, regenerates electrical signals via a chemical reaction through ion channels, and transmits them to neighboring neurons. In the context of describing the complex physiological reaction process as a stochastic process, this study aimed to show that the distribution of the maximum time interval of spike signals follows extreme-order statistics. By considering the statistical variance in the time constant of the leaky Integrate-and-Fire model, a deterministic time evolution model for spike signals, we enabled randomness in the time interval of the spike signals. When the time constant follows an exponential distribution function, the time interval of the spike signal also follows an exponential distribution. In this case, our theory and simulations confirmed that the histogram of the maximum time interval follows the Gumbel distribution, one of the three forms of extreme-value statistics. We further confirmed that the histogram of the maximum time interval followed a Fréchet distribution when the time interval of the spike signal followed a Pareto distribution. These findings confirm that nerve transmission delay can be described using extreme value statistics and can therefore be used as a new indicator of transmission delay.


Subject(s)
Models, Neurological , Synaptic Transmission , Synaptic Transmission/physiology , Action Potentials/physiology , Neurons/physiology , Humans , Time Factors , Stochastic Processes , Computer Simulation
15.
J Biomed Sci ; 31(1): 69, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992696

ABSTRACT

BACKGROUND: Local translation at synapses is important for rapidly remodeling the synaptic proteome to sustain long-term plasticity and memory. While the regulatory mechanisms underlying memory-associated local translation have been widely elucidated in the postsynaptic/dendritic region, there is no direct evidence for which RNA-binding protein (RBP) in axons controls target-specific mRNA translation to promote long-term potentiation (LTP) and memory. We previously reported that translation controlled by cytoplasmic polyadenylation element binding protein 2 (CPEB2) is important for postsynaptic plasticity and memory. Here, we investigated whether CPEB2 regulates axonal translation to support presynaptic plasticity. METHODS: Behavioral and electrophysiological assessments were conducted in mice with pan neuron/glia- or glutamatergic neuron-specific knockout of CPEB2. Hippocampal Schaffer collateral (SC)-CA1 and temporoammonic (TA)-CA1 pathways were electro-recorded to monitor synaptic transmission and LTP evoked by 4 trains of high-frequency stimulation. RNA immunoprecipitation, coupled with bioinformatics analysis, were used to unveil CPEB2-binding axonal RNA candidates associated with learning, which were further validated by Western blotting and luciferase reporter assays. Adeno-associated viruses expressing Cre recombinase were stereotaxically delivered to the pre- or post-synaptic region of the TA circuit to ablate Cpeb2 for further electrophysiological investigation. Biochemically isolated synaptosomes and axotomized neurons cultured on a microfluidic platform were applied to measure axonal protein synthesis and FM4-64FX-loaded synaptic vesicles. RESULTS: Electrophysiological analysis of hippocampal CA1 neurons detected abnormal excitability and vesicle release probability in CPEB2-depleted SC and TA afferents, so we cross-compared the CPEB2-immunoprecipitated transcriptome with a learning-induced axonal translatome in the adult cortex to identify axonal targets possibly regulated by CPEB2. We validated that Slc17a6, encoding vesicular glutamate transporter 2 (VGLUT2), is translationally upregulated by CPEB2. Conditional knockout of CPEB2 in VGLUT2-expressing glutamatergic neurons impaired consolidation of hippocampus-dependent memory in mice. Presynaptic-specific ablation of Cpeb2 in VGLUT2-dominated TA afferents was sufficient to attenuate protein synthesis-dependent LTP. Moreover, blocking activity-induced axonal Slc17a6 translation by CPEB2 deficiency or cycloheximide diminished the releasable pool of VGLUT2-containing synaptic vesicles. CONCLUSIONS: We identified 272 CPEB2-binding transcripts with altered axonal translation post-learning and established a causal link between CPEB2-driven axonal synthesis of VGLUT2 and presynaptic translation-dependent LTP. These findings extend our understanding of memory-related translational control mechanisms in the presynaptic compartment.


Subject(s)
Neuronal Plasticity , RNA-Binding Proteins , Synaptic Transmission , Vesicular Glutamate Transport Protein 2 , Animals , Mice , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Neuronal Plasticity/physiology , Synaptic Transmission/physiology , Vesicular Glutamate Transport Protein 2/metabolism , Vesicular Glutamate Transport Protein 2/genetics , Mice, Knockout , Axons/metabolism , Axons/physiology , RNA, Messenger/metabolism , RNA, Messenger/genetics , Male , Protein Biosynthesis
16.
Int J Mol Sci ; 25(13)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38999947

ABSTRACT

Alcohol tolerance is a neuroadaptive response that leads to a reduction in the effects of alcohol caused by previous exposure. Tolerance plays a critical role in the development of alcohol use disorder (AUD) because it leads to the escalation of drinking and dependence. Understanding the molecular mechanisms underlying alcohol tolerance is therefore important for the development of effective therapeutics and for understanding addiction in general. This review explores the molecular basis of alcohol tolerance in invertebrate models, Drosophila and C. elegans, focusing on synaptic transmission. Both organisms exhibit biphasic responses to ethanol and develop tolerance similar to that of mammals. Furthermore, the availability of several genetic tools makes them a great candidate to study the molecular basis of ethanol response. Studies in invertebrate models show that tolerance involves conserved changes in the neurotransmitter systems, ion channels, and synaptic proteins. These neuroadaptive changes lead to a change in neuronal excitability, most likely to compensate for the enhanced inhibition by ethanol.


Subject(s)
Caenorhabditis elegans , Ethanol , Neuronal Plasticity , Synaptic Transmission , Animals , Neuronal Plasticity/drug effects , Ethanol/pharmacology , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/physiology , Caenorhabditis elegans/metabolism , Synaptic Transmission/drug effects , Drug Tolerance , Synapses/metabolism , Synapses/drug effects , Synapses/physiology , Alcoholism/metabolism , Drosophila/physiology , Humans , Invertebrates/physiology
17.
Cell Rep ; 43(7): 114461, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38990719

ABSTRACT

The quantal content of an evoked postsynaptic response is typically determined by dividing it by the average spontaneous miniature response. However, this approach is challenged by the notion that different synaptic vesicle pools might drive spontaneous and evoked release. Here, we "silence" synaptic vesicles through pharmacological alkalinization and subsequently rescue them by optogenetic acidification. We find that such silenced synaptic vesicles, retrieved during evoked or spontaneous activity, cross-deplete the complementary release mode in a fully reversible manner. A fluorescently tagged version of the endosomal SNARE protein Vti1a, which has been suggested to identify a separate pool of spontaneously recycling synaptic vesicles, is trafficked to synaptic vesicles significantly only upon overexpression but not when endogenously tagged by CRISPR-Cas9. Thus, both release modes draw synaptic vesicles from the same readily releasable pool.


Subject(s)
Synaptic Vesicles , Synaptic Vesicles/metabolism , Animals , Synaptic Transmission , Rats , Optogenetics
18.
Biochemistry (Mosc) ; 89(6): 1031-1044, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38981699

ABSTRACT

Alzheimer's disease (AD) is a severe neurodegenerative condition affecting millions worldwide. Prevalence of AD correlates with increased life expectancy and aging population in the developed countries. Considering that AD is a multifactorial disease involving various pathological processes such as synaptic dysfunction, neuroinflammation, oxidative stress, and improper protein folding, a comprehensive approach targeting multiple pathways may prove effective in slowing the disease progression. Cellular therapy and its further development in the form of cell vesicle and particularly mitochondrial transplantation represent promising approaches for treating neurodegeneration. The use of synaptosomes, due to uniqueness of their contents, could mark a new stage in the development of comprehensive therapies for neurodegenerative diseases, particularly AD. Synaptosomes contain unique memory mitochondria, which differ not only in size but also in functionality compared to the mitochondria in the neuronal soma. These synaptosomal mitochondria actively participate in cellular communication and signal transmission within synapses. Synaptosomes also contain other elements such as their own protein synthesis machinery, synaptic vesicles with neurotransmitters, synaptic adhesion molecules, and microRNAs - all crucial for synaptic transmission and, consequently, cognitive processes. Complex molecular ensemble ensures maintenance of the synaptic autonomy of mitochondria. Additionally, synaptosomes, with their affinity for neurons, can serve as an optimal platform for targeted drug delivery to nerve cells. This review discusses unique composition of synaptosomes, their capabilities and advantages, as well as limitations of their suggested use as therapeutic agents for treating neurodegenerative pathologies, particularly AD.


Subject(s)
Alzheimer Disease , Synaptosomes , Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Alzheimer Disease/pathology , Humans , Synaptosomes/metabolism , Animals , Mitochondria/metabolism , Synaptic Transmission , Neurons/metabolism , Synapses/metabolism
19.
Transl Psychiatry ; 14(1): 272, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961057

ABSTRACT

Valproic acid (VPA) is one of the most effective antiepileptic drugs, and exposing animals to VPA during gestation has been used as a model for autism spectrum disorder (ASD). Numerous studies have shown that impaired synaptic transmission in the cerebellar cortical circuits is one of the reasons for the social deficits and repetitive behavior seen in ASD. In this study, we investigated the effect of VPA exposure during pregnancy on tactile stimulation-evoked cerebellar mossy fiber-granule cell (MF-GC) synaptic transmission in mice anesthetized with urethane. Three-chamber testing showed that mice exposed to VPA mice exhibited a significant reduction in social interaction compared with the control group. In vivo electrophysiological recordings revealed that a pair of air-puff stimulation on ipsilateral whisker pad evoked MF-GC synaptic transmission, N1, and N2. The evoked MF-GC synaptic responses in VPA-exposed mice exhibited a significant increase in the area under the curve (AUC) of N1 and the amplitude and AUC of N2 compared with untreated mice. Cerebellar surface application of the selective N-methyl-D-aspartate (NMDA) receptor blocker D-APV significantly inhibited facial stimulation-evoked MF-GC synaptic transmission. In the presence of D-APV, there were no significant differences between the AUC of N1 and the amplitude and AUC of N2 in the VPA-exposed mice and those of the untreated mice. Notably, blockade of the GluN2A subunit-containing, but not the GluN2B subunit-containing, NMDA receptor, significantly inhibited MF-GC synaptic transmission and decreased the AUC of N1 and the amplitude and AUC of N2 in VPA-exposed mice to levels similar to those seen in untreated mice. In addition, the GluN2A subunit-containing NMDA receptor was expressed at higher levels in the GC layer of VPA-treated mice than in control mice. These results indicate that gestational VPA exposure in mice produces ASD-like behaviors, accompanied by increased cerebellar MF-GC synaptic transmission and an increase in GluN2A subunit-containing NMDA receptor expression in the offspring.


Subject(s)
Autism Spectrum Disorder , Disease Models, Animal , Prenatal Exposure Delayed Effects , Receptors, N-Methyl-D-Aspartate , Synaptic Transmission , Valproic Acid , Animals , Receptors, N-Methyl-D-Aspartate/metabolism , Valproic Acid/pharmacology , Pregnancy , Female , Mice , Prenatal Exposure Delayed Effects/physiopathology , Synaptic Transmission/drug effects , Autism Spectrum Disorder/chemically induced , Male , Cerebellum/drug effects , Cerebellum/metabolism , Anticonvulsants/pharmacology
20.
Proc Natl Acad Sci U S A ; 121(31): e2315599121, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39058581

ABSTRACT

Ribbon synapses between inner hair cells (IHCs) and type I spiral ganglion neurons (SGNs) in the inner ear are damaged by noise trauma and with aging, causing "synaptopathy" and hearing loss. Cocultures of neonatal denervated organs of Corti and newly introduced SGNs have been developed to find strategies for improving IHC synapse regeneration, but evidence of the physiological normality of regenerated synapses is missing. This study utilizes IHC optogenetic stimulation and SGN recordings, showing that, when P3-5 denervated organs of Corti are cocultured with SGNs, newly formed IHC/SGN synapses are indeed functional, exhibiting glutamatergic excitatory postsynaptic currents. When using older organs of Corti at P10-11, synaptic activity probed by deconvolution showed more mature release properties, closer to the specialized mode of IHC synaptic transmission crucial for coding the sound signal. This functional assessment of newly formed IHC synapses developed here, provides a powerful tool for testing approaches to improve synapse regeneration.


Subject(s)
Spiral Ganglion , Synapses , Animals , Spiral Ganglion/cytology , Spiral Ganglion/physiology , Synapses/physiology , Mice , Hair Cells, Auditory, Inner/physiology , Hair Cells, Auditory, Inner/metabolism , Synaptic Transmission/physiology , Neurons/physiology , Neurons/metabolism , Regeneration/physiology , Hair Cells, Auditory/physiology , Coculture Techniques/methods , Optogenetics/methods , Nerve Regeneration/physiology , Excitatory Postsynaptic Potentials/physiology , Organ of Corti/physiology , Organ of Corti/cytology , Organ of Corti/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL