Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 14.728
Filter
1.
J Neurosci ; 44(40)2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39358028

ABSTRACT

The brain is a highly adaptable organ that is molded by experience throughout life. Although the field of neuroscience has historically focused on intrinsic neuronal mechanisms of plasticity, there is growing evidence that multiple glial populations regulate the timing and extent of neuronal plasticity, particularly over the course of development. This review highlights recent discoveries on the role of glial cells in the establishment of cortical circuits and the regulation of experience-dependent neuronal plasticity during critical periods of neurodevelopment. These studies provide strong evidence that neuronal circuit maturation and plasticity are non-cell autonomous processes that require both glial-neuronal and glial-glial cross talk to proceed. We conclude by discussing open questions that will continue to guide research in this nascent field.


Subject(s)
Cerebral Cortex , Neuroglia , Neuronal Plasticity , Neurons , Neuronal Plasticity/physiology , Animals , Neuroglia/physiology , Humans , Cerebral Cortex/physiology , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Neurons/physiology , Nerve Net/physiology , Nerve Net/growth & development , Neurogenesis/physiology
2.
Commun Biol ; 7(1): 1286, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39384971

ABSTRACT

Bidirectional communication between neurons and glial cells is crucial to establishing and maintaining normal brain function. Some of these interactions are activity-dependent, yet it remains largely unexplored how acute changes in neuronal activity affect glial-to-neuron and neuron-to-glial dynamics. Here, we use excitatory and inhibitory designer receptors exclusively activated by designer drugs (DREADD) to study the effects of acute chemogenetic manipulations of a subpopulation of layer 5 cortical projection and dentate gyrus neurons in adult (Rbp4Cre) mouse brains. We show that acute chemogenetic neuronal activation reduces synaptic density, and increases microglia and astrocyte reactivity, but does not affect parvalbumin (PV+) neurons, only perineuronal nets (PNN). Conversely, acute silencing increases synaptic density and decreases glial reactivity. We show fast glial response upon clozapine-N-oxide (CNO) administration in cortical and subcortical regions. Together, our work provides evidence of fast, activity-dependent, bidirectional interactions between neurons and glial cells.


Subject(s)
Clozapine , Neuroglia , Neurons , Animals , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Neuroglia/metabolism , Neuroglia/drug effects , Neuroglia/physiology , Mice , Clozapine/pharmacology , Clozapine/analogs & derivatives , Male , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Parvalbumins/metabolism , Mice, Transgenic
3.
Elife ; 132024 Sep 11.
Article in English | MEDLINE | ID: mdl-39259197

ABSTRACT

The emergence of myelinating oligodendrocytes represents a pivotal developmental milestone in vertebrates, given their capacity to ensheath axons and facilitate the swift conduction of action potentials. It is widely accepted that cortical oligodendrocyte progenitor cells (OPCs) arise from medial ganglionic eminence (MGE), lateral/caudal ganglionic eminence (LGE/CGE), and cortical radial glial cells (RGCs). Here, we used two different fate mapping strategies to challenge the established notion that the LGE generates cortical OPCs. Furthermore, we used a Cre/loxP-dependent exclusion strategy to reveal that the LGE/CGE does not give rise to cortical OPCs. Additionally, we showed that specifically eliminating MGE-derived OPCs leads to a significant reduction of cortical OPCs. Together, our findings indicate that the LGE does not generate cortical OPCs, contrary to previous beliefs. These findings provide a new view of the developmental origins of cortical OPCs and a valuable foundation for future research on both normal development and oligodendrocyte-related disease.


Subject(s)
Cerebral Cortex , Oligodendroglia , Animals , Oligodendroglia/physiology , Oligodendroglia/cytology , Mice , Cerebral Cortex/embryology , Cerebral Cortex/physiology , Cerebral Cortex/cytology , Oligodendrocyte Precursor Cells/physiology , Oligodendrocyte Precursor Cells/cytology , Cell Differentiation , Ganglionic Eminence
4.
Nat Commun ; 15(1): 7958, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261492

ABSTRACT

Cortical neurons exhibit temporally irregular spiking patterns and heterogeneous firing rates. These features arise in model circuits operating in a 'fluctuation-driven regime', in which fluctuations in membrane potentials emerge from the network dynamics. However, it is still debated whether the cortex operates in such a regime. We evaluated the fluctuation-driven hypothesis by analyzing spiking and sub-threshold membrane potentials of neurons in the frontal cortex of mice performing a decision-making task. We showed that while standard fluctuation-driven models successfully account for spiking statistics, they fall short in capturing the heterogeneity in sub-threshold activity. This limitation is an inevitable outcome of bombarding single-compartment neurons with a large number of pre-synaptic inputs, thereby clamping the voltage of all neurons to more or less the same average voltage. To address this, we effectively incorporated dendritic morphology into the standard models. Inclusion of dendritic morphology in the neuronal models increased neuronal selectivity and reduced error trials, suggesting a functional role for dendrites during decision-making. Our work suggests that, during decision-making, cortical neurons in high-order cortical areas operate in a fluctuation-driven regime.


Subject(s)
Action Potentials , Models, Neurological , Neurons , Animals , Neurons/physiology , Mice , Action Potentials/physiology , Cerebral Cortex/physiology , Cerebral Cortex/cytology , Decision Making/physiology , Membrane Potentials/physiology , Dendrites/physiology , Male , Mice, Inbred C57BL , Frontal Lobe/physiology , Frontal Lobe/cytology
5.
PLoS Biol ; 22(9): e3002760, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39226322

ABSTRACT

53BP1 is a well-established DNA damage repair factor that has recently emerged to critically regulate gene expression for tumor suppression and neural development. However, its precise function and regulatory mechanisms remain unclear. Here, we showed that phosphorylation of 53BP1 at serine 25 by ATM is required for neural progenitor cell proliferation and neuronal differentiation in cortical brain organoids. Dynamic phosphorylation of 53BP1-serine 25 controls 53BP1 target genes governing neuronal differentiation and function, cellular response to stress, and apoptosis. Mechanistically, ATM and RNF168 govern 53BP1's binding to gene loci to directly affect gene regulation, especially at genes for neuronal differentiation and maturation. 53BP1 serine 25 phosphorylation effectively impedes its binding to bivalent or H3K27me3-occupied promoters, especially at genes regulating H3K4 methylation, neuronal functions, and cell proliferation. Beyond 53BP1, ATM-dependent phosphorylation displays wide-ranging effects, regulating factors in neuronal differentiation, cytoskeleton, p53 regulation, as well as key signaling pathways such as ATM, BDNF, and WNT during cortical organoid differentiation. Together, our data suggest that the interplay between 53BP1 and ATM orchestrates essential genetic programs for cell morphogenesis, tissue organization, and developmental pathways crucial for human cortical development.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , Organoids , Tumor Suppressor p53-Binding Protein 1 , Tumor Suppressor p53-Binding Protein 1/metabolism , Tumor Suppressor p53-Binding Protein 1/genetics , Organoids/metabolism , Humans , Ataxia Telangiectasia Mutated Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Phosphorylation , DNA Damage , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Neural Stem Cells/metabolism , Cell Differentiation/genetics , Cell Proliferation , DNA Repair , Neurogenesis/genetics , Neurons/metabolism , Signal Transduction
6.
Nat Commun ; 15(1): 8025, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39271679

ABSTRACT

Complex sensory information arrives in the brain from an animal's first-person ('egocentric') perspective. However, animals can efficiently navigate as if referencing map-like ('allocentric') representations. The postrhinal (POR) and retrosplenial (RSC) cortices are thought to mediate between sensory input and internal maps, combining egocentric representations of physical cues with allocentric head direction (HD) information. Here we show that neurons in the POR and RSC of female Long-Evans rats are tuned to distinct but complementary aspects of local space. Egocentric bearing (EB) cells recorded in square and L-shaped environments reveal that RSC cells encode local geometric features, while POR cells encode a more global account of boundary geometry. Additionally, POR HD cells can incorporate egocentric information to fire in two opposite directions with two oppositely placed identical visual landmarks, while only a subset of RSC HD cells possess this property. Entorhinal grid and HD cells exhibit consistently allocentric spatial firing properties. These results reveal significant regional differences in the neural encoding of spatial reference frames.


Subject(s)
Neurons , Rats, Long-Evans , Space Perception , Animals , Female , Neurons/physiology , Rats , Space Perception/physiology , Cues , Entorhinal Cortex/physiology , Entorhinal Cortex/cytology , Environment , Cerebral Cortex/physiology , Cerebral Cortex/cytology
7.
Mol Brain ; 17(1): 66, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39267102

ABSTRACT

TRPM4 is a non-selective cation channel activated by intracellular Ca2+ but only permeable to monovalent cations, its activation regulates membrane potential and intracellular calcium. This channel participates in the migration and adhesion of non-excitable cells and forms an integral part of the focal adhesion complex. In neurons, TRPM4 expression starts before birth and its function at this stage is not clear, but it may function in processes such as neurite development. Here we investigate the role of TRPM4 in neuritogenesis. We found that neurons at DIV 0 express TRPM4, the inhibition of TRPM4 using 9-Ph reduces neurite number and slows the progression of neurite development, keeping neurons in stage 1. The genetic suppression of TRPM4 using an shRNA at later stages (DIV2) reduces neurite length. Conversely, at DIV 0, TRPM4 inhibition augments the Cch-induced Ca2 + i increase, altering the calcium homeostasis. Together, these results show that TRPM4 participates in progression of neurite development and suggest a critical role of the calcium modulation during this stage of neuronal development.


Subject(s)
Calcium , Cerebral Cortex , Neurites , Neurogenesis , TRPM Cation Channels , TRPM Cation Channels/metabolism , TRPM Cation Channels/antagonists & inhibitors , Animals , Neurites/metabolism , Neurites/drug effects , Calcium/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Neurons/metabolism
8.
J Photochem Photobiol B ; 259: 113021, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39222549

ABSTRACT

Alzheimer's disease, a prevalent neurodegenerative condition primarily affecting older adults, remains incurable. Its principle pathological hallmark is the accelerated accumulation of amyloid ß (Aß) protein. This study investigates the potential of photobiomodulation using near infrared light to counteract Aß1-42-induced synaptic degeneration and neurotoxicity. We focused on the effect of 808 nm near-infrared laser diode (LD) on Aß1-42 cytotoxicity in primary cultured cortical neurons. We assessed cell survival using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, observing substantial benefits from LD irradiation with a power of 10 mW and a dose of 30 J. Cells exposed to Aß1-42 exhibited morphological changes indicative of synaptic damage and a significant decrease in the number of postsynaptic density protein-95 (PSD-95) contacts, which were significantly improved with near-infrared LD therapy. Furthermore, this therapy reduced Aß and phosphorylated tau (P-tau) protein accumulation. Additionally, near-infrared LD irradiation substantially lessened the Aß1-42-induced rise in glial fibrillary acid protein (GFAP) and ionized calcium-binding adaptor molecule 1 (IBA1) in astrocytes and microglia. Remarkably, near-infrared LD irradiation effectively inhibited phosphorylation of key proteins involved in Aß1-42-induced necroptosis, namely Receptor-interacting protein kinase-3 (RIP3) and Mixed Lineage Kinase domain-Like protein (MLKL). Our findings suggest that near-infrared LD treatment significantly reduces neurodegeneration by reducing glial overactivation and neuronal necroptosis triggered by Aß1-42. Thus, near-infrared LD treatment emerges as a promising approach for slowing or treating Alzheimer's disease, offering new avenues in its management.


Subject(s)
Amyloid beta-Peptides , Cell Survival , Infrared Rays , Neurons , Peptide Fragments , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Animals , Neurons/radiation effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Peptide Fragments/metabolism , Cell Survival/drug effects , Cell Survival/radiation effects , Rats , Lasers, Semiconductor , tau Proteins/metabolism , Low-Level Light Therapy , Cells, Cultured , Disks Large Homolog 4 Protein/metabolism , Glial Fibrillary Acidic Protein/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/radiation effects , Astrocytes/metabolism , Astrocytes/drug effects , Astrocytes/radiation effects
9.
Int J Mol Sci ; 25(17)2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39273571

ABSTRACT

Amyloid-beta peptide (Aß) is a neurotoxic constituent of senile plaques in the brains of Alzheimer's disease (AD) patients. The detailed mechanisms by which protein kinase C-delta (PKCδ) contributes to Aß toxicity is not yet entirely understood. Using fully differentiated primary rat cortical neurons, we found that inhibition of Aß25-35-induced PKCδ increased cell viability with restoration of neuronal morphology. Using cyclin D1, proliferating cell nuclear antigen (PCNA), and histone H3 phosphorylated at Ser-10 (p-Histone H3) as the respective markers for the G1-, S-, and G2/M-phases, PKCδ inhibition mitigated cell cycle reentry (CCR) and subsequent caspase-3 cleavage induced by both Aß25-35 and Aß1-42 in the post-mitotic cortical neurons. Upstream of PKCδ, signal transducers and activators of transcription (STAT)-3 mediated PKCδ induction, CCR, and caspase-3 cleavage upon Aß exposure. Downstream of PKCδ, aberrant neuronal CCR was triggered by overactivating cyclin-dependent kinase-5 (CDK5) via calpain2-dependent p35 cleavage into p25. Finally, PKCδ and CDK5 also contributed to Aß25-35 induction of p53-upregulated modulator of apoptosis (PUMA) in cortical neurons. Together, we demonstrated that, in the post-mitotic neurons exposed to Aßs, STAT3-dependent PKCδ expression triggers calpain2-mediated p35 cleavage into p25 to overactivate CDK5, thus leading to aberrant CCR, PUMA induction, caspase-3 cleavage, and ultimately apoptosis.


Subject(s)
Amyloid beta-Peptides , Apoptosis , Cell Cycle , Cerebral Cortex , Neurons , Protein Kinase C-delta , Amyloid beta-Peptides/metabolism , Animals , Neurons/metabolism , Neurons/drug effects , Apoptosis/drug effects , Rats , Protein Kinase C-delta/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Cell Cycle/drug effects , Cyclin-Dependent Kinase 5/metabolism , Peptide Fragments/pharmacology , Peptide Fragments/metabolism , Caspase 3/metabolism , Rats, Sprague-Dawley , Cells, Cultured , Signal Transduction/drug effects
10.
Nature ; 634(8033): 415-423, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39232166

ABSTRACT

Astrocytes are the most abundant cell type in the mammalian brain and provide structural and metabolic support to neurons, regulate synapses and become reactive after injury and disease. However, a small subset of astrocytes settles in specialized areas of the adult brain where these astrocytes instead actively generate differentiated neuronal and glial progeny and are therefore referred to as neural stem cells1-3. Common parenchymal astrocytes and quiescent neural stem cells share similar transcriptomes despite their very distinct functions4-6. Thus, how stem cell activity is molecularly encoded remains unknown. Here we examine the transcriptome, chromatin accessibility and methylome of neural stem cells and their progeny, and of astrocytes from the striatum and cortex in the healthy and ischaemic adult mouse brain. We identify distinct methylation profiles associated with either astrocyte or stem cell function. Stem cell function is mediated by methylation of astrocyte genes and demethylation of stem cell genes that are expressed later. Ischaemic injury to the brain induces gain of stemness in striatal astrocytes7. We show that this response involves reprogramming the astrocyte methylome to a stem cell methylome and is absent if the de novo methyltransferase DNMT3A is missing. Overall, we unveil DNA methylation as a promising target for regenerative medicine.


Subject(s)
Astrocytes , Brain Ischemia , DNA Methylation , DNA Methyltransferase 3A , Neural Stem Cells , Astrocytes/metabolism , Astrocytes/cytology , Animals , DNA Methylation/genetics , Mice , DNA Methyltransferase 3A/metabolism , Brain Ischemia/pathology , Brain Ischemia/metabolism , Brain Ischemia/genetics , Male , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Transcriptome , Epigenome , Female , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , Chromatin/metabolism , Chromatin/genetics , Corpus Striatum/cytology , Corpus Striatum/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Mice, Inbred C57BL , Cellular Reprogramming/genetics , Regenerative Medicine
11.
Development ; 151(17)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39250533

ABSTRACT

The cell-intrinsic mechanisms underlying the decision of a stem/progenitor cell to either proliferate or differentiate remain incompletely understood. Here, we identify the transmembrane protein Lrig1 as a physiological homeostatic regulator of FGF2-driven proliferation and self-renewal of neural progenitors at early-to-mid embryonic stages of cortical development. We show that Lrig1 is expressed in cortical progenitors (CPs), and its ablation caused expansion and increased proliferation of radial/apical progenitors and of neurogenic transit-amplifying Tbr2+ intermediate progenitors. Notably, our findings identify a previously unreported EGF-independent mechanism through which Lrig1 negatively regulates neural progenitor proliferation by modulating the FGF2-induced IL6/Jak2/Stat3 pathway, a molecular cascade that plays a pivotal role in the generation and maintenance of CPs. Consistently, Lrig1 knockout mice showed a significant increase in the density of pyramidal glutamatergic neurons placed in superficial layers 2 and 3 of the postnatal neocortex. Together, these results support a model in which Lrig1 regulates cortical neurogenesis by influencing the cycling activity of a set of progenitors that are temporally specified to produce upper layer glutamatergic neurons.


Subject(s)
Janus Kinase 2 , Membrane Glycoproteins , Mice, Knockout , Neural Stem Cells , Neurogenesis , Neurons , STAT3 Transcription Factor , Signal Transduction , Animals , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Janus Kinase 2/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Mice , Neurogenesis/genetics , Neurons/metabolism , Neurons/cytology , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Cell Proliferation , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cell Differentiation , Fibroblast Growth Factors/metabolism , Nerve Tissue Proteins
12.
Neuromolecular Med ; 26(1): 36, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39254908

ABSTRACT

The symptoms of fragile X syndrome (FXS), caused by a single gene mutation to Fmr1, have been increasingly linked to disordered astrocyte signalling within the cerebral cortex. We have recently demonstrated that the purinergic signalling pathway, which utilizes nucleoside triphosphates and their metabolites to facilitate bidirectional glial and glial-neuronal interactions, is upregulated in cortical astrocytes derived from the Fmr1 knockout (KO) mouse model of FXS. Heightened Fmr1 KO P2Y purinergic receptor levels were correlated with prolonged intracellular calcium release, elevated synaptogenic protein secretion, and hyperactivity of developing circuits. However, due to the relative lack of sensitive and reproducible quantification methods available for measuring purines and pyrimidines, determining the abundance of these factors in Fmr1 KO astrocytes was limited. We therefore developed a hydrophilic interaction liquid chromatography protocol coupled with mass spectrometry to compare the abundance of intracellular and extracellular purinergic molecules between wildtype and Fmr1 KO mouse astrocytes. Significant differences in the concentrations of UDP, ATP, AMP, and adenosine intracellular stores were found within Fmr1 KO astrocytes relative to WT. The extracellular level of adenosine was also significantly elevated in Fmr1 KO astrocyte-conditioned media in comparison to media collected from WT astrocytes. Glycosylation of the astrocyte membrane-bound CD39 ectonucleotidase, which facilitates ligand breakdown following synaptic release, was also elevated in Fmr1 KO astrocyte cultures. Together, these differences demonstrated further dysregulation of the purinergic signalling system within Fmr1 KO cortical astrocytes, potentially leading to significant alterations in FXS purinergic receptor activation and cellular pathology.


Subject(s)
Astrocytes , Cerebral Cortex , Fragile X Mental Retardation Protein , Fragile X Syndrome , Mice, Knockout , Signal Transduction , Animals , Astrocytes/metabolism , Mice , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Apyrase/genetics , Apyrase/metabolism , Cells, Cultured , Adenosine Triphosphate/metabolism , Culture Media, Conditioned , Adenosine/metabolism , Adenosine/analogs & derivatives , Receptors, Purinergic P2Y/metabolism , Receptors, Purinergic P2Y/genetics , Mice, Inbred C57BL , Antigens, CD
13.
Nat Commun ; 15(1): 6872, 2024 Aug 11.
Article in English | MEDLINE | ID: mdl-39127731

ABSTRACT

Cortical neurons store information across different timescales, from seconds to years. Although information stability is variable across regions, it can vary within a region as well. Association areas are known to multiplex behaviorally relevant variables, but the stability of their representations is not well understood. Here, we longitudinally recorded the activity of neuronal populations in the mouse retrosplenial cortex (RSC) during the performance of a context-choice association task. We found that the activity of neurons exhibits different levels of stability across days. Using linear classifiers, we quantified the stability of three task-relevant variables. We find that RSC representations of context and trial outcome display higher stability than motor choice, both at the single cell and population levels. Together, our findings show an important characteristic of association areas, where diverse streams of information are stored with varying levels of stability, which may balance representational reliability and flexibility according to behavioral demands.


Subject(s)
Neurons , Animals , Neurons/physiology , Mice , Male , Mice, Inbred C57BL , Choice Behavior/physiology , Cerebral Cortex/physiology , Cerebral Cortex/cytology , Gyrus Cinguli/physiology , Gyrus Cinguli/cytology , Behavior, Animal/physiology
14.
Drug Discov Ther ; 18(4): 255-259, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39183043

ABSTRACT

Actin rearrangement and phosphorylation-dephosphorylation in the nervous system contribute to plastic alteration of neuronal structure and function. Phosphatase and actin regulator (PHACTR) family members are actin- and protein phosphatase 1 (PP1)-binding proteins. Because some family members act as regulators of neuronal morphology, studying the regulatory mechanisms of PHACTR is valuable for understanding the basis of neuronal circuit formation. Although expression patterns of PHACTR family molecules (PHACTR1-4) vary across distinct brain areas, little is known about the extracellular ligands that influence their mRNA levels. In this study, we focused on an important neurotrophin, brain-derived neurotrophic factor (BDNF), and examined its effect on mRNA expression of PHACTR family member in cortical neurons. PHACTR1-3, but not PHACTR4, were affected by stimulation of primary cultured cortical neurons with BDNF; namely, sustained downregulation of their mRNA levels was observed. The observed downregulation was blocked by an inhibitor of the extracellular signal-regulated protein kinase/mitogen-activated protein kinase (ERK/MAPK) pathway, U0126, suggesting that ERK/MAPK plays an inhibitory role for gene induction of PHACTR1-3. These findings aid the elucidation of how BDNF regulates actin- and PP1-related neuronal functions.


Subject(s)
Brain-Derived Neurotrophic Factor , Cerebral Cortex , MAP Kinase Signaling System , Neurons , RNA, Messenger , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Animals , Neurons/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , RNA, Messenger/metabolism , RNA, Messenger/genetics , Cells, Cultured , Rats , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Butadienes/pharmacology , Down-Regulation , Nitriles/pharmacology
15.
Pharmacol Rep ; 76(5): 1174-1183, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39088104

ABSTRACT

BACKGROUND: - Alcohol-induced pro-inflammatory activation might influence cellular and synaptic pathology, thus contributing to the behavioral phenotypes associated with alcohol use disorders. In the present study, the possible anti-inflammatory properties of N-[(4-trifluoromethyl)-benzyl]4-methoxybutyramide (GET73), a promising therapeutic agent for alcohol use disorder treatment, were evaluated in primary cultures of rat cortical microglia. METHODS: - Primary cultures of cerebral cortex microglial cells were treated with 100 ng/ml lipopolysaccharide (LPS; 8 h, 37 °C) or 75 mM ethanol (EtOH; 4 days, 37 °C) alone or in the presence of GET73 (1-30 µM). At the end of the incubation period, multiparametric quantification of cytokines/chemokines was performed by using the xMAP technology and Luminex platform. Furthermore, cultured microglial cell viability following the treatment with EtOH and GET73, alone or in combination, has been measured by a colorimetric assay (i.e. MTT assay). RESULTS: - GET73 (10 and 30 µM) partially or fully prevented the LPS-induced increase of IL-6, IL-1ß, RANTES/CCL5 protein and MCP-1/CCL2 levels. On the contrary, GET73 failed to attenuate the TNF-α level increase induced by LPS. Furthermore, GET73 treatment (10-30 µM) significantly attenuated or prevented the EtOH-induced increase of TNF-α, IL-6, IL-1ß and MCP-1/CCL2 levels. Finally, at all the concentrations tested (1-30 µM), the GET73 treatment did not alter the EtOH-induced reduction of microglial cell viability. CONCLUSIONS: - The current results provide the first in vitro evidence of GET73 protective properties against EtOH-induced neuroinflammation. These data add more information on the complex and multifactorial profile of action of the compound, further supporting the significance of developing GET73 as a therapeutic tool for the treatment of individuals with alcohol use disorders.


Subject(s)
Cell Survival , Cerebral Cortex , Cytokines , Ethanol , Lipopolysaccharides , Microglia , Animals , Rats , Anti-Inflammatory Agents/pharmacology , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Chemokines/metabolism , Cytokines/metabolism , Ethanol/pharmacology , Lipopolysaccharides/pharmacology , Microglia/drug effects , Microglia/metabolism , Rats, Wistar
16.
PLoS One ; 19(8): e0306656, 2024.
Article in English | MEDLINE | ID: mdl-39213294

ABSTRACT

The invention of Light Emitting Diode (LED) revolutionized energy-efficient illumination, but concerns persist regarding the potential harm of blue light to our eyes. In this study, we scrutinized the impact of LED light characteristics on eyes using two cell types: M-1 (rich in mitochondria) and CD-1 (neuronal). Variations in color rendering index (CRI) and correlated color temperature (CCT) were investigated, alongside exposure durations ranging from 0 to 24 hours. The findings illuminated the potential benefits of high-quality LED lighting, characterized by a high CRI and low CCT, which emits a greater proportion of red light. This form of lighting was associated with enhanced cell proliferation, elevated ATP levels, and reduced oxidative stress. In contrast, LEDs with low CRI and high CCT exhibited adverse effects, diminishing cell viability and increasing oxidative stress. These results suggest that high-quality LED lighting may have neuroprotective potential as a treatment option, such as for retinal ganglion cells.


Subject(s)
Cell Survival , Light , Mitochondria , Neurons , Oxidative Stress , Animals , Mice , Mitochondria/metabolism , Mitochondria/radiation effects , Cell Survival/radiation effects , Neurons/metabolism , Neurons/radiation effects , Oxidative Stress/radiation effects , Cerebral Cortex/radiation effects , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Cell Line , Cell Proliferation/radiation effects , Adenosine Triphosphate/metabolism , Lighting
17.
Cells Tissues Organs ; 213(5): 382-389, 2024.
Article in English | MEDLINE | ID: mdl-39191219

ABSTRACT

INTRODUCTION: Neurogenesis in the adult brain may play an important role in memory and cognition; however, knowledge of neurogenic markers in the human brain remains limited. We compared the single-nucleus transcriptome of the hippocampus with that of other cortical regions to identify hippocampus-specific neurogenic markers. METHODS: We analyzed 26,189 nuclei from four human brains collected within 16 h of death. Clustering and annotation were performed to examine differential expression, gene ontology, and intercellular communication. DCX expression was validated by ddPCR. RESULTS: Immature markers such as DCX, CALB2, NES, SOX2, PAX6, DPYSL3, and TUBB3 were expressed in both hippocampus and prefrontal cortex, with higher levels in the prefrontal cortex. ddPCR confirmed higher expression of DCX in the prefrontal cortex. DCX was involved in both neurogenesis and neuroprotection pathways. CONCLUSION: Neurogenic markers are not definitive indicators of adult neurogenesis as their roles are more complex than previously understood.


Subject(s)
Doublecortin Protein , Hippocampus , Neurogenesis , Humans , Hippocampus/metabolism , Doublecortin Domain Proteins , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Neuropeptides/metabolism , Neuropeptides/genetics , Transcriptome , Male , Adult , Female , Cerebral Cortex/metabolism , Cerebral Cortex/cytology
18.
Neuron ; 112(18): 3058-3068.e8, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39111306

ABSTRACT

Human brain ontogeny is characterized by a considerably prolonged neotenic development of cortical neurons and circuits. Neoteny is thought to be essential for the acquisition of advanced cognitive functions, which are typically altered in intellectual disability (ID) and autism spectrum disorders (ASDs). Human neuronal neoteny could be disrupted in some forms of ID and/or ASDs, but this has never been tested. Here, we use xenotransplantation of human cortical neurons into the mouse brain to model SYNGAP1 haploinsufficiency, one of the most prevalent genetic causes of ID/ASDs. We find that SYNGAP1-deficient human neurons display strong acceleration of morphological and functional synaptic formation and maturation alongside disrupted synaptic plasticity. At the circuit level, SYNGAP1-haploinsufficient neurons display precocious acquisition of responsiveness to visual stimulation months ahead of time. Our findings indicate that SYNGAP1 is required cell autonomously for human neuronal neoteny, providing novel links between human-specific developmental mechanisms and ID/ASDs.


Subject(s)
Cerebral Cortex , Neurons , ras GTPase-Activating Proteins , Animals , Humans , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/deficiency , ras GTPase-Activating Proteins/metabolism , Neurons/metabolism , Mice , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Haploinsufficiency , Neuronal Plasticity/physiology , Synapses/metabolism , Synapses/physiology , Intellectual Disability/genetics , Male , Female
19.
STAR Protoc ; 5(3): 103280, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39213150

ABSTRACT

The generation of human pluripotent stem cell (hPSC)-derived brain organoids is continuously refined, enhancing their reproducibility and complexity. Here, we present a guided differentiation protocol for generating cortical forebrain organoids and cortico-pericyte (CP)assembloids composed of a robust outer radial glia (oRG) population and an expanded outer subventricular zone (oSVZ). We describe the steps to generate hPSC-derived cortical organoids (COs), cortical pericytes, and CP assembloids. Moreover, we outline the procedures to characterize the organoids by immunostaining and to perform single-cell dissociation. For complete details on the use and execution of this protocol, please refer to Walsh et al.1.


Subject(s)
Cell Differentiation , Ependymoglial Cells , Organoids , Pluripotent Stem Cells , Humans , Organoids/cytology , Organoids/metabolism , Cell Differentiation/physiology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Ependymoglial Cells/cytology , Ependymoglial Cells/metabolism , Cell Culture Techniques/methods , Cerebral Cortex/cytology , Neuroglia/cytology , Pericytes/cytology , Pericytes/metabolism
20.
Trends Neurosci ; 47(9): 667-668, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39142912

ABSTRACT

The maturation of cerebral cortical networks during early life involves a major reorganization of long-range axonal connections. In a recent study, Bragg-Gonzalo, Aguilera, et al. discovered that in mice, the interhemispheric connections sent by S1L4 callosal projection neurons are pruned via the tight control of their ipsilateral synaptic integration, which relies on the early activity of specific interneurons.


Subject(s)
Cerebral Cortex , Neural Inhibition , Animals , Mice , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Corpus Callosum/cytology , Corpus Callosum/physiology , Interneurons/physiology , Nerve Net/cytology , Nerve Net/physiology , Neural Inhibition/physiology , Neural Pathways/cytology , Neural Pathways/physiology , Neurons/physiology
SELECTION OF CITATIONS
SEARCH DETAIL