Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Exp Neurol ; 379: 114874, 2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38914275

ABSTRACT

The mechanistic target of rapamycin complex 1 (mTORC1) signaling pathway is a ubiquitous cellular pathway. mTORopathies, a group of disorders characterized by hyperactivity of the mTORC1 pathway, illustrate the prominent role of the mTOR pathway in disease pathology, often profoundly affecting the central nervous system. One of the most debilitating symptoms of mTORopathies is drug-resistant epilepsy, emphasizing the urgent need for a deeper understanding of disease mechanisms to develop novel anti-epileptic drugs. In this study, we explored the multiwell Multi-electrode array (MEA) system as a tool to identify robust network activity parameters in an approach to model mTORopathy-related epilepsy in vitro. To this extent, we cultured mouse primary hippocampal neurons on the multiwell MEA to identify robust network activity phenotypes in mTORC1-hyperactive neuronal networks. mTOR-hyperactivity was induced either through deletion of Tsc1 or overexpression of a constitutively active RHEB variant identified in patients, RHEBp.P37L. mTORC1 dependency of the phenotypes was assessed using rapamycin, and vigabatrin was applied to treat epilepsy-like phenotypes. We show that hyperactivity of the mTORC1 pathway leads to aberrant network activity. In both the Tsc1-KO and RHEB-p.P37L models, we identified changes in network synchronicity, rhythmicity, and burst characteristics. The presence of these phenotypes is prevented upon early treatment with the mTORC1-inhibitor rapamycin. Application of rapamycin in mature neuronal cultures could only partially rescue the network activity phenotypes. Additionally, treatment with the anti-epileptic drug vigabatrin reduced network activity and restored burst characteristics. Taken together, we showed that mTORC1-hyperactive neuronal cultures on the multiwell MEA system present reliable network activity phenotypes that can be used as an assay to explore the potency of new drug treatments targeting epilepsy in mTORopathy patients and may give more insights into the pathophysiological mechanisms underlying epilepsy in these patients.

2.
Biosensors (Basel) ; 14(6)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38920600

ABSTRACT

Development and optimisation of bioelectronic monitoring techniques like microelectrode array-based field potential measurement and impedance spectroscopy for the functional, label-free and non-invasive monitoring of in vitro neuronal networks is widely investigated in the field of biosensors. Thus, these techniques were individually used to demonstrate the capabilities of, e.g., detecting compound-induced toxicity in neuronal culture models. In contrast, extended application for investigating the effects of central nervous system infecting viruses are rarely described. In this context, we wanted to analyse the effect of herpesviruses on functional neuronal networks. Therefore, we developed a unique hybrid bioelectronic monitoring platform that allows for performing field potential monitoring and impedance spectroscopy on the same microelectrode. In the first step, a neuronal culture model based on primary hippocampal cells from neonatal rats was established with reproducible and stable synchronised electrophysiological network activity after 21 days of cultivation on microelectrode arrays. For a proof of concept, the pseudorabies model virus PrV Kaplan-ΔgG-GFP was applied and the effect on the neuronal networks was monitored by impedance spectroscopy and field potential measurement for 72 h in a multiparametric mode. Analysis of several bioelectronic parameters revealed a virus concentration-dependent degeneration of the neuronal network within 24-48 h, with a significant early change in electrophysiological activity, subsequently leading to a loss of activity and network synchronicity. In conclusion, we successfully developed a microelectrode array-based hybrid bioelectronic measurement platform for quantitative monitoring of pathologic effects of a herpesvirus on electrophysiological active neuronal networks.


Subject(s)
Biosensing Techniques , Dielectric Spectroscopy , Neurons , Animals , Rats , Neurons/virology , Nerve Net , Microelectrodes , Hippocampus/virology , Herpesvirus 1, Suid , Cells, Cultured , Pseudorabies/virology
3.
Methods Mol Biol ; 2799: 29-46, 2024.
Article in English | MEDLINE | ID: mdl-38727901

ABSTRACT

The expression and activity of ionotropic glutamate receptors control signal transduction at the excitatory synapses in the CNS. The NMDAR comprises two obligatory GluN1 subunits and two GluN2 or GluN3 subunits in different combinations. Each GluN subunit consists of four domains: the extracellular amino-terminal and agonist-binding domains, the transmembrane domain, and the intracellular C-terminal domain (CTD). The CTD interaction with various classes of intracellular proteins is critical for trafficking and synaptic localization of NMDARs. Amino acid mutations or the inclusion of premature stop codons in the CTD could contribute to the emergence of neurodevelopmental and neuropsychiatric disorders. Here, we describe the method of preparing primary hippocampal neurons and lentiviral particles expressing GluN subunits that can be used as a model to study cell surface expression and synaptic localization of NMDARs. We also show a simple method of fluorescence immunostaining of eGFP-tagged GluN2 subunits and subsequent microscopy technique and image analysis to study the effects of disease-associated mutations in the CTDs of GluN2A and GluN2B subunits.


Subject(s)
Hippocampus , Neurons , Receptors, N-Methyl-D-Aspartate , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Hippocampus/metabolism , Hippocampus/cytology , Neurons/metabolism , Animals , Protein Subunits/metabolism , Protein Subunits/genetics , Cells, Cultured , Rats , Humans , Lentivirus/genetics , Primary Cell Culture/methods , Gene Expression
4.
Dev Cell ; 59(12): 1571-1592.e9, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38626765

ABSTRACT

Neuronal endosomal and lysosomal abnormalities are among the early changes observed in Alzheimer's disease (AD) before plaques appear. However, it is unclear whether distinct endolysosomal defects are temporally organized and how altered γ-secretase function or amyloid precursor protein (APP) metabolism contribute to these changes. Inhibiting γ-secretase chronically, in mouse embryonic fibroblast and hippocampal neurons, led to a gradual endolysosomal collapse initiated by decreased lysosomal calcium and increased cholesterol, causing downstream defects in endosomal recycling and maturation. This endolysosomal demise is γ-secretase dependent, requires membrane-tethered APP cytoplasmic domains, and is rescued by APP depletion. APP C-terminal fragments (CTFs) localized to late endosome/lysosome-endoplasmic reticulum contacts; an excess of APP-CTFs herein reduced lysosomal Ca2+ refilling from the endoplasmic reticulum, promoting cholesterol accretion. Tonic regulation by APP-CTFs provides a mechanistic explanation for their cellular toxicity: failure to timely degrade APP-CTFs sustains downstream signaling, instigating lysosomal dyshomeostasis, as observed in prodromal AD. This is the opposite of substrates such as Notch, which require intramembrane proteolysis to initiate signaling.


Subject(s)
Alzheimer Disease , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor , Endoplasmic Reticulum , Endosomes , Lysosomes , Neurons , Lysosomes/metabolism , Animals , Endosomes/metabolism , Amyloid beta-Protein Precursor/metabolism , Mice , Endoplasmic Reticulum/metabolism , Amyloid Precursor Protein Secretases/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Neurons/metabolism , Cholesterol/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Calcium/metabolism , Humans , Fibroblasts/metabolism , Signal Transduction , Proteolysis
5.
Mar Drugs ; 21(7)2023 Jul 19.
Article in English | MEDLINE | ID: mdl-37504941

ABSTRACT

Nerve damage caused by accumulated oxidative stress is one of the characteristics and main mechanisms of Alzheimer's disease (AD). Previous studies have shown that phosphatidylserine (PS) rich in eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) plays a significant role in preventing and mitigating the progression of AD. However, whether DHA-PS and EPA-PS can directly protect primary hippocampal neurons against oxidative damage has not been studied. Here, the neuroprotective functions of DHA-PS and EPA-PS against H2O2/t-BHP-induced oxidative damage and the possible mechanisms were evaluated in primary hippocampal neurons. It was found that DHA-PS and EPA-PS could significantly improve cell morphology and promote the restoration of neural network structure. Further studies showed that both of them significantly alleviated oxidative stress-mediated mitochondrial dysfunction. EPA-PS significantly inhibited the phosphorylation of ERK, thus playing an anti-apoptotic role, and EPA-PS significantly increased the protein expressions of p-TrkB and p-CREB, thus playing a neuroprotective role. In addition, EPA-PS, rather than DHA-PS could enhance synaptic plasticity by increasing the expression of SYN, and both could significantly reduce the expression levels of p-GSK3ß and p-Tau. These results provide a scientific basis for the use of DHA/EPA-enriched phospholipids in the treatment of neurodegenerative diseases, and also provide a reference for the development of related functional foods.


Subject(s)
Alzheimer Disease , Neuroprotective Agents , Humans , Eicosapentaenoic Acid/pharmacology , Eicosapentaenoic Acid/metabolism , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/metabolism , Phosphatidylserines/pharmacology , Phosphatidylserines/chemistry , Hydrogen Peroxide/toxicity , Hydrogen Peroxide/metabolism , Oxidative Stress , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Neurons , Hippocampus
6.
Neurosci Lett ; 794: 137009, 2023 01 18.
Article in English | MEDLINE | ID: mdl-36493898

ABSTRACT

Thyroid hormones (THs) have an essential role in normal brain development and function. Methamphetamine (MA) is a widely abused psychostimulant that induces irreversible damages to neuronal cells. In the current study, we used rat primary hippocampal neurons (PHNs) to investigate the neuroprotective effect of THs against MA neurotoxicity. PHNs were prepared from 18-day rat embryos and cell viability was assessed using MTT assay, following treatment with various concentrations of MA, T3, T4 or tetrac, an integrin αvß3 cell surface receptor antagonist. Our results showed that 7 mM MA induced an approximately 50 % reduction in the PHNs viability. Treatment with 800 nM T3 or 8 µM T4 protected PHNs against MA toxicity, an effect which was blocked in the presence of tetrac. These findings suggest that THs protect PHNs against MA-induced cell death by the activation of integrin αvß3 cell surface receptors. So, targeting integrin αvß3 receptors or using THs can be considered as promising therapeutic strategies to overcome MA neurotoxicity.


Subject(s)
Methamphetamine , Neuroprotective Agents , Rats , Animals , Neuroprotective Agents/pharmacology , Triiodothyronine , Methamphetamine/toxicity , Integrin alphaVbeta3/metabolism , Thyroid Hormones/metabolism , Thyroxine/pharmacology , Thyroxine/metabolism
7.
Chinese Pharmacological Bulletin ; (12): 1189-1194, 2023.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1013795

ABSTRACT

Aim To explore the protective effect of Zishen Huoxue Prescription on OGD/R-induced primary hippocampal neuron damage in rats and the possible mechanism. Methods After the isolated primary hippocampal neurons were identified by immunofluorescence, OGD/R induced neuronal damage, and the changes of autophagic flux at different re-oxygenation time were observed by confocal laser scanning microscopy. After OGD/R-induced primary hippocampal neurons were intervened with serum containing Zishen Huoxue Prescription, cell viability was detected by CCK-8, cell apoptosis was detected by flow cytometry, autophagosomes were detected by transmission electron microscopy, and autophagy-related protein expressions were detected by Western blot. Results 10% Zishen Huoxue Prescription-containing serum could significantly improve cell viability and reduce the proportion of cell apoptosis, increase the number of autophagosomes in neurons, and up-regulate the expression of autophagy-related protein PINK1, Parkin, and pATG16L1. Conclusions Zishen Huoxue Prescription can effectively resist OGD/R-induced apoptosis of primary hippocampal neurons in rats, and its effect may be related to the regulation of PINK1-Parkin pathway to promote mitophagy.

9.
Front Psychiatry ; 12: 627056, 2021.
Article in English | MEDLINE | ID: mdl-34122166

ABSTRACT

Objective: Corticosterone causes significant neurotoxicity in primary hippocampal neurons which is associated with depression. Dysfunctional autophagy is implicated in cognitive impairment and depressive-like behavior. The traditional Chinese medicine Sinisan (SNS) is highly effective in clinical treatment of depression. However, the molecular mechanisms underlying therapeutic effects of SNS are unknown. Purpose: The aim of this study was to elucidate the protective effect of SNS and the underlying mechanisms against corticosterone-induced neuronal damage. Study Design: The effects of serum derived from rats containing SNS (or untreated controls) on the expression of autophagy-related molecules in primary rat hippocampal neurons exposed to different concentrations of corticosterone for different intervals were explored. Methods: CCK-8 assay, LDH assay were used to analyze cell viability and LDH activity. Western blot, qRT-PCR, and immunofluorescence assays were used to determine protein and mRNA expression levels of molecules such as LC3, p62, Beclin1, ULK1, PI3K, p-PI3K, Akt p-Akt, mTOR, p-mTOR, p70S6, p-p70S6, 4ebp1 and p-4ebp1. Results: Corticosterone induced a dose- and time-dependent reduction in cellular viability. Moreover, corticosterone (100-400 µM) treatment for 24 h increased LC3-II/LC3-I protein ratio, increased Beclin1 and ULK1 protein expression levels, and decreased p62, PI3K, p-PI3K, p-Akt, p-mTOR, p-p70S6, and p-4ebp1 protein expression levels. Notably, SNS-containing serum reversed corticosterone-induced reduction of neuronal viability, and increased p62, PI3K, p-Akt, p-mTOR, p-p70S6, and p-4ebp1 protein and mRNA expression levels. In addition, SNS-containing serum decreased LC3-II/LC3-I protein ratio, and downregulated Beclin1, and ULK1 protein and mRNA expression in primary hippocampal neurons. Conclusion: SNS protects primary hippocampal neurons against corticosterone-induced neurotoxicity by preventing excessive autophagy through activation of PI3K/AKT/mTOR pathway.

10.
Folia Histochem Cytobiol ; 59(1): 57-65, 2021.
Article in English | MEDLINE | ID: mdl-33651374

ABSTRACT

INTRODUCTION: The pathological mechanism of cerebral ischemia/reperfusion (CIR) injury is complicated and unclear. Apart from the involvement of many low-molecular factors it was found that several miRNAs were dysregulated during and after CIR injury in cell models. This study aimed to explore the effects of miR-378a-5p on in vitro model of (CIR) injury-induced neuronal apoptosis and provide a new mechanism of CIR injury. MATERIAL AND METHODS: Primary hippocampal neurons were isolated from newborn Sprague-Dawley rats. Oxygen- glucose deprivation/reoxygenation (OGDR) for 24 h and 48 h was used as an in vitro model of CIR. Cell viability was measured using MTT assay and apoptosis was determined by flow cytometry. Quantitative real time PCR (qRT-PCR) assay and Western blotting were used to examine mRNA and protein expressions, respectively. TargetScan was used to predict the direct target of miR-378a-5p and luciferase assay was used to validate that calmodulin-dependent protein kinase kinase-2 (CAMKK2) was the direct target of miR-378a-5p. RESULTS: miR-378a-5p expression was significantly increased after OGDR at 24 h and 48 h. After OGDR, cell viability was reduced, which was reversed by miR-378a-5p and enhanced by shCAMKK2 plasmid. Cell apoptosis was increased after OGDR, which was prevented by miR-378a-5p and enhanced by shCAMKK2 plasmid. Results of TargetScan and luciferase assay demonstrated that miR-378a-5p could directly bind to 3'-untranslated region (3'-UTR) of CAMKK2. Both mRNA and protein expression of CAMKK2 were downregulated by miR-378a-5p mimics and upregulated by miR-378a-5p inhibitors. Phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) was positively associated with expression of CAMKK2. CONCLUSIONS: Data of this study indicated that miR-378a-5p was significantly overexpressed after OGDR. miR-378a-5p could bind to 3'-UTR of CAMKK2 to inhibit cell proliferation through regulation of CAMKK2/AMPK pathway providing a new mechanism and biomarker for the diagnosis and potential treatment of CIR injury.


Subject(s)
Apoptosis/physiology , MicroRNAs/metabolism , Neurons/metabolism , Reperfusion Injury/physiopathology , Signal Transduction/physiology , AMP-Activated Protein Kinases/metabolism , Animals , Animals, Newborn , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Cell Proliferation/physiology , Cell Survival/physiology , HEK293 Cells , Hippocampus/pathology , Humans , Rats, Sprague-Dawley
11.
Cannabis Cannabinoid Res ; 6(1): 40-47, 2021.
Article in English | MEDLINE | ID: mdl-33614951

ABSTRACT

Introduction: Reports on the neurotoxic and neuroprotective effects of cannabidiol (CBD) have not been in complete accord, showing different and somewhat contradictory results depending upon the brain cell types and experimental conditions employed. This work systematically examines the neuroprotective capability of CBD against oxidative stress (i.e., hydrogen peroxide [H2O2]) as well as its toxicity profile in the in vitro culture platform of primary hippocampal neurons. Materials and Methods: The low cell-density (100 neurons per mm2) culture was used for analyzing the viability and morphology of neurons at a single-cell level with a confocal laser-scanning microscope (CLSM). Primary neurons were obtained from the hippocampal tissues of embryonic day-18 (E18) Sprague-Dawley rat pups and treated with CBD (0.1-100 µM) and/or H2O2 (0.1-50 µM) at 1 DIV (days in vitro). Results: The lethal concentration 50 (LC50) value (the concentration causing 50% cell death) of CBD was calculated to be 9.85 µM after 24 h of incubation, and that of H2O2 was 2.46 µM under the same conditions. The neuroprotection ratio of CBD against H2O2 ([H2O2]=10 µM) was 2.40 with 5 µM of CBD, increasing the cell viability to 57% from 24%. The CLSM analysis suggested that the cell-death mechanisms were different for CBD and H2O2, and CBD did not completely rescue the morphological alterations of primary hippocampal neurons caused by H2O2, such as neurite degeneration, at least in the in vitro neuron culture. Conclusion: Although CBD showed both neurotoxic and neuroprotective effects on hippocampal neurons in the in vitro setting, the use of low-concentrated (i.e., 5 µM) CBD, not causing toxic effects on the neurons, significantly rescued the neurons from the oxidative stress (H2O2), confirming its neuroprotection capability.


Subject(s)
Cannabidiol/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Cell Adhesion/drug effects , Cell Death/drug effects , Cell Survival/drug effects , Hydrogen Peroxide/toxicity , Microscopy, Confocal , Oxidative Stress/drug effects , Primary Cell Culture , Rats, Sprague-Dawley
12.
Chemosphere ; 264(Pt 1): 128450, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33007573

ABSTRACT

BPA analogs, including bisphenol S (BPS) and bisphenol B (BPB), have been used to replace BPA since it was banned to be added. To investigate whether BPA and its analogs cause oxidative damage effects on primary hippocampal neurons of rats, reactive oxygen species (ROS), malondialdehyde (MDA), superoxide dismutase (SOD), mitochondrial membrane potential (MMP), apoptosis and cell viability assays were conducted after hippocampal neurons exposure to different concentrations of BPA, BPS, and BPB (1, 10, 100 nM and 1, 10, 100 µM). Moreover, the effects of EGCG (5 and 6 µM for male and female, respectively) added on neurons exposed to BPA were assessed. Results showed that 24 h exposure to these bisphenols (BPs) could increase the levels of ROS and contents of MDA, but reduce the activity of SOD significantly. A decline of cell viabilities accompanied with the increasing of apoptosis rates was observed after 7 d exposure to BPs and the reduction of MMP was also observed after 7 d exposure to BPA. Interestingly, BPS has the lower toxicity to hippocampal neurons compared with BPA and BPB. Non-monotonic dose-effect relationships between the concentrations of BPs and the cytotoxic effects were observed, and the effects of BPs on male hippocampal neurons are greater than those of female ones in general. While EGCG can protect neurons free of oxidative damages. In conclusion, the results suggest that BPs may induce sex-specific neurotoxic effects involving oxidative stress, which can be attenuated by EGCG, and males are more sensitive to BPs than females.


Subject(s)
Benzhydryl Compounds , Oxidative Stress , Animals , Benzhydryl Compounds/toxicity , Female , Hippocampus , Male , Membrane Potential, Mitochondrial , Neurons , Rats , Reactive Oxygen Species
13.
Front Cell Neurosci ; 14: 573208, 2020.
Article in English | MEDLINE | ID: mdl-33192317

ABSTRACT

In the immune system, Major Histocompatibility Complex class I (MHC-I) molecules are located on the surface of most nucleated cells in vertebrates where they mediate immune responses. Accumulating evidence indicates that MHC-I molecules are also expressed in the central nervous system (CNS) where they play important roles that are significantly different from their immune functions. Classical MHC-I molecules are temporally and spatially expressed in the developing and adult CNS, where they participate in the synaptic formation, remodeling and plasticity. Therefore, clarifying the regulation of MHC-I expression is necessary to develop an accurate understanding of its function in the CNS. Here, we show that microRNA 34a (miR34a), a brain enriched noncoding RNA, is temporally expressed in developing hippocampal neurons, and its expression is significantly increased after MHC-I protein abundance is decreased in the hippocampus. Computational algorithms identify putative miR34a target sites in the 3'UTR of MHC-I mRNA, and here we demonstrate direct targeting of miR34a to MHC-I mRNA using a dual-luciferase reporter assay system. MiR34a targeting can decrease constitutive MHC-I expression in both Neuro-2a neuroblastoma cells and primary hippocampal neurons. Finally, miR34a mediated reduction of MHC-I results in increased dendritic growth and branching in cultured hippocampal neurons. Taken together, our findings identify miR34a as a novel regulator of MHC-I for shaping neural morphology in developing hippocampal neurons.

14.
Neurotox Res ; 38(2): 370-384, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32378057

ABSTRACT

Major depression disorder is one of the most common psychiatric disorders that greatly threaten the mental health of a large population worldwide. Previous studies have shown that endoplasmic reticulum (ER) stress plays an important role in the pathophysiology of depression, and current research suggests that brain-derived neurotrophic factor precursor (proBDNF) is involved in the development of depression. However, the relationship between ER and proBDNF in the pathophysiology of depression is not well elucidated. Here, we treated primary hippocampal neurons of mice with corticosterone (CORT) and evaluated the relationship between proBDNF and ERS. Our results showed that CORT induced ERS and upregulated the expression of proBDNF and its receptor, Follistatin-like protein 4 (FSTL4), which contributed to significantly decreased neuronal viability and expression of synaptic-related proteins including NR2A, PSD95, and SYN. Anti-proBDNF neutralization and ISRIB (an inhibitor of the ERS) treatment, respective ly, protected neuronal viabilities and increased the expression of synaptic-related proteins in corticosterone-exposed neurons. ISRIB treatment reduced the expression of proBDNF and FSTL4, whereas anti-proBDNF treatment did not affect ERS markers (Grp78, p-PERK, ATF4) expression. Our study presented evidence that CORT-induced ERS negatively regulated the neuronal viability and the level of synaptic-related protein of primary neurons via the proBDNF/FSTL4 pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Brain-Derived Neurotrophic Factor/drug effects , Corticosterone/pharmacology , Endoplasmic Reticulum Stress/drug effects , Hippocampus/cytology , Neurons/drug effects , Protein Precursors/drug effects , Animals , Brain-Derived Neurotrophic Factor/metabolism , Depressive Disorder, Major , Disks Large Homolog 4 Protein/drug effects , Disks Large Homolog 4 Protein/metabolism , Endoplasmic Reticulum Chaperone BiP , Follistatin-Related Proteins/drug effects , Follistatin-Related Proteins/metabolism , Mice , Neurons/metabolism , Primary Cell Culture , Protein Precursors/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptophysin/metabolism
15.
J Neurochem ; 154(6): 647-661, 2020 09.
Article in English | MEDLINE | ID: mdl-32233089

ABSTRACT

SUMOylation is a dynamic post-translational protein modification that primarily takes place in cell nuclei, where it plays a key role in multiple DNA-related processes. In neurons, the SUMOylation-dependent control of a subset of neuronal transcription factors is known to regulate various aspects of nerve cell differentiation, development, and function. In an unbiased screen for endogenous SUMOylation targets in the developing mouse brain, based on a His6 -HA-SUMO1 knock-in mouse line, we previously identified the transcription factor Zinc finger and BTB domain-containing 20 (Zbtb20) as a new SUMO1-conjugate. We show here that the three key SUMO paralogues SUMO1, SUMO2, and SUMO3 can all be conjugated to Zbtb20 in vitro in HEK293FT cells, and we confirm the SUMOylation of Zbtb20 in vivo in mouse brain. Using primary hippocampal neurons from wild-type and Zbtb20 knock-out (KO) mice as a model system, we then demonstrate that the expression of Zbtb20 is required for proper nerve cell development and neurite growth and branching. Furthermore, we show that the SUMOylation of Zbtb20 is essential for its function in this context, and provide evidence indicating that SUMOylation affects the Zbtb20-dependent transcriptional profile of neurons. Our data highlight the role of SUMOylation in the regulation of neuronal transcription factors that determine nerve cell development, and they demonstrate that key functions of the transcription factor Zbtb20 in neuronal development and neurite growth are under obligatory SUMOylation control.


Subject(s)
Nervous System/growth & development , Sumoylation/physiology , Transcription Factors/genetics , Transcription Factors/physiology , Animals , Cell Survival , Gene Expression Profiling , Gene Knock-In Techniques , HEK293 Cells , Hippocampus/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurites/physiology , Neurons/metabolism , Primary Cell Culture , RNA/biosynthesis , RNA/genetics
16.
Adv Healthc Mater ; 9(5): e1901072, 2020 03.
Article in English | MEDLINE | ID: mdl-31957248

ABSTRACT

Astrocytes, the most representative glial cells in the brain, play a multitude of crucial functions for proper neuronal development and synaptic-network formation, including neuroprotection as well as physical and chemical support. However, little attention has been paid, in the neuroregenerative medicine and related fields, to the cytoprotective incorporation of astrocytes into neuron-culture scaffolds and full-fledged functional utilization of encapsulated astrocytes for controlled neuronal development. In this article, a 3D neurosupportive culture system for enhanced induction of neuronal circuit generation is reported, where astrocytes are confined in hydrogel microfibers and protected from the outside. The astrocyte-encapsulated microfibers significantly accelerate the neurite outgrowth and guide its directionality, and enhance the synaptic formation, without any physical contact with the neurons. This astrocyte-laden system provides a pivotal culture scaffold for advanced development of cell-based therapeutics for neural injuries, such as spinal cord injury.


Subject(s)
Astrocytes , Hydrogels , Cells, Cultured , Coculture Techniques , Neurogenesis , Neurons
17.
Methods Mol Biol ; 2101: 327-351, 2020.
Article in English | MEDLINE | ID: mdl-31879912

ABSTRACT

Microtubules, as integral part of the eukaryotic cytoskeleton, exert numerous essential functions in cells. A mechanism to control these diverse functions are the posttranslational modifications of tubulin. Despite being known for decades, relatively little insight into the cellular functions of these modifications has been gained so far. The discovery of tubulin-modifying enzymes and a growing number of available knockout mice now allow working with primary cells from those mouse models to address biological functions and molecular mechanisms behind those modifications. However, a number of those mouse models show either lethality or sterility, making it difficult to impossible to obtain a sufficient number of animals for a systematic study with primary cells. Moreover, many of those modifications are controlled by several redundant enzymes, and it is often necessary to knock out several enzymes in parallel to obtain a significant change in a given tubulin modification. Here we describe a method to generate primary cells with combinatorial knockout genotypes using conditional knockout mice. The conditional alleles are converted into knockout in the cultured primary cells by transduction with a lentivirus encoding cre-recombinase. This approach has allowed us to knock out the two main brain deglutamylases in mouse primary neurons, which leads to strongly increased polyglutamylation in these cells. Our method can be applied to measure different cellular processes, such as axonal transport, for which it can be combined with the expression of different fluorescent reporters to label intracellular proteins. Using a panel of conditional knockout mice, our method can further be applied to study the functions of a variety of tubulin modifications that require simultaneous knockout of multiple genes.


Subject(s)
Gene Knockdown Techniques , Neurons/metabolism , Protein Processing, Post-Translational , Tubulin/genetics , Tubulin/metabolism , Animals , Cell Culture Techniques , Cells, Cultured , Cloning, Molecular , Cytoskeleton/genetics , Cytoskeleton/metabolism , Electrophoresis, Polyacrylamide Gel , Gene Expression , Genes, Reporter , Genetic Vectors/genetics , Humans , Mice, Knockout , Pyramidal Cells/metabolism , Transduction, Genetic
18.
ACS Appl Mater Interfaces ; 11(41): 38190-38204, 2019 Oct 16.
Article in English | MEDLINE | ID: mdl-31550131

ABSTRACT

Despite broad application of nanotechnology in neuroscience, the nanoneurotoxicity of magnetic nanoparticles in primary hippocampal neurons remains poorly characterized. In particular, understanding how magnetic nanoparticles perturb neuronal calcium homeostasis is critical when considering magnetic nanoparticles as a nonviral vector for effective gene therapy in neuronal diseases. Here, we address the pressing need to systematically investigate the neurotoxicity of magnetic nanoparticles with different surface charges in primary hippocampal neurons. We found that unlike negative and neutral nanoparticles, positively charged magnetic nanoparticles (magnetic poly(lactic-co-glycolic acid) (PLGA)-polyethylenimine (PEI) nanoparticles, MNP-PLGA-PEI NPs) rapidly elevated cytoplasmic calcium levels in primary hippocampal neurons, mainly via extracellular calcium influx regulated by voltage-gated calcium channels. We went on to show that this perturbation of intracellular calcium homeostasis elicited serious cytotoxicity in primary hippocampal neurons. However, our next experiment demonstrated that PEGylation on the surface of MNP-PLGA-PEI NPs shielded the surface charge, thereby preventing the perturbation of intracellular calcium homeostasis. That is, PEGylated MNP-PLGA-PEI NPs reduced nanoneurotoxicity. Importantly, biocompatible PEGylated MNP-PLGA-PEI NPs under an external magnetic field enhanced transfection efficiency (>7%) of plasmid DNA encoding GFP in primary hippocampal neurons compared to NPs without external magnetic field mediation. Moreover, under an external magnetic field, this system achieved gene transfection in the hippocampus of the C57 mouse. Overall, this study is the first to successfully employ biocompatible PEGylated MNP-PLGA-PEI NPs for transfection using a magnetofection strategy in primary hippocampal neurons, thereby providing a nanoplatform as a new perspective for treating neuronal diseases or modulating neuron activities.


Subject(s)
Green Fluorescent Proteins , Hippocampus/metabolism , Nanoparticles/chemistry , Neurons/metabolism , Plasmids , Transfection , Animals , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , HEK293 Cells , Hippocampus/cytology , Humans , Mice , Neurons/cytology , Plasmids/chemistry , Plasmids/genetics , Plasmids/pharmacology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Polyethyleneimine/chemistry , Polyethyleneimine/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology , Rats , Rats, Sprague-Dawley
19.
Neurosci Lett ; 701: 38-47, 2019 05 14.
Article in English | MEDLINE | ID: mdl-30776494

ABSTRACT

BACKGROUND: Alpha-synuclein (αSN) is an abundant presynaptic brain protein that its aggregated species believed to play pivotal roles in the development of neurodegenerative diseases, especially Parkinson's disease (PD). In this study, we compared the response of primary neuronal cells with a well-known cell line model, PC12, against the toxic aggregates of αSN. METHODS: Primary hippocampal neurons (PHNs) were isolated from 17 to 18 days old rat embryos. Fibrillization was induced in recombinant αSN and monitored by standard methods. The toxicity of different aggregates of αSN on the treated cells was then studied. Furthermore, changes in the intracellular reactive oxygen species (ROS) and Ca2+ levels were also compared in two kinds of treated cells. We also studied the gene expression profile of certain Ca2+ channels and carriers using the GEO2 database. RESULTS: The viability rate was significantly lower in PC12 versus PHNs, in response to αSN. This is while the intracellular ROS and Ca2+ levels were significantly increased in both cell types. Analysis of microarray data indicated that some factors involved in Ca2+ hemostasis may face significant changes in the PD condition. CONCLUSION: By putting these data together, it is clear that PHN is more resistant than PC12 toward αSN cytotoxicity even in the presence of rising cytoplasmic ROS and Ca2+ levels. Exploring the supporting mechanisms which PHN uses to be more resistant to αSN cytotoxicity can help to open a roadmap toward therapeutic plans in PD and other synucleinopathy disorders.


Subject(s)
Hippocampus/drug effects , Neurons/drug effects , PC12 Cells/drug effects , alpha-Synuclein/toxicity , Animals , Calcium/metabolism , Cell Survival/drug effects , Parkinson Disease , Primary Cell Culture , Rats , Reactive Oxygen Species/metabolism
20.
Cell Physiol Biochem ; 51(1): 97-112, 2018.
Article in English | MEDLINE | ID: mdl-30439717

ABSTRACT

BACKGROUND/AIMS: The N-methyl-D-aspartic acid receptor (NMDAR) has been extensively studied for its important roles in synaptic plasticity and learning and memory. However, the effects of microwave radiation on the subunit composition and activity of NMDARs and the relationship between NMDARs and microwave-induced synaptic plasticity have not been thoroughly elucidated to date. MATERIALS: In our study, primary hippocampal neurons were used to evaluate the effects of microwave radiation on synaptic plasticity. Structural changes were observed by diolistic (Dil) labeling and scanning electron microscopy (SEM) observation. Functional synaptic plasticity was reflected by the NMDAR currents, which were detected by whole cell patch clamp. We also detected the expression of NMDAR subunits by real-time PCR and Western blot analysis. To clarify the effects of microwave radiation on NMDAR-induced synaptic plasticity, suitable agonists or inhibitors were added to confirm the role of NMDARs on microwave-induced synaptic plasticity. Dil labeling, SEM observation, whole cell patch clamp, real-time PCR and Western blot analysis were used to evaluate changes in synaptic plasticity after treatment with agonists or inhibitors. RESULTS: Our results found that microwave exposure impaired neurite development and decreased mRNA and protein levels and the current density of NMDARs. Due to the decreased expression of NMDAR subunits after microwave exposure, the selective agonist NMDA was added to identify the role of NMDARs on microwave-induced synaptic plasticity injuries. After adding the agonist, the expression of NMDAR subunits recovered to the normal levels. In addition, the microwave-induced structural and functional synaptic plasticity injuries recovered, including the number and length of neurites, the connections between neurons, and the NMDAR current. CONCLUSION: Microwave radiation caused neuronal synaptic plasticity injuries in primary hippocampal neurons, and NMDARs played protective roles on the damage process.


Subject(s)
Microwaves , Neuronal Plasticity/radiation effects , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Microscopy, Confocal , N-Methylaspartate/pharmacology , Neurites/physiology , Neurites/radiation effects , Neuronal Plasticity/drug effects , Neurons/cytology , Neurons/metabolism , Patch-Clamp Techniques , Phosphorylation/drug effects , Protein Subunits/agonists , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...