Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 14.704
Filter
1.
J Vis Exp ; (209)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39141550

ABSTRACT

Microglia are tissue-resident macrophages of the central nervous system (CNS), performing numerous functions that support neuronal health and CNS homeostasis. They are a major population of immune cells associated with CNS disease activity, adopting reactive phenotypes that potentially contribute to neuronal injury during chronic neurodegenerative diseases such as multiple sclerosis (MS). The distinct mechanisms by which microglia regulate neuronal function and survival during health and disease remain limited due to challenges in resolving the complex in vivo interactions between microglia, neurons, and other CNS environmental factors. Thus, the in vitro approach of co-culturing microglia and neurons remains a valuable tool for studying microglia-neuronal interactions. Here, we present a protocol to generate and co-culture primary microglia and neurons from mice. Specifically, microglia were isolated after 9-10 days in vitro from a mixed glia culture established from brain homogenates derived from neonatal mice between post-natal days 0-2. Neuronal cells were isolated from brain cortices of mouse embryos between embryonic days 16-18. After 4-5 days in vitro, neuronal cells were seeded in 96-well plates, followed by the addition of microglia to form the co-culture. Careful timing is critical for this protocol as both cell types need to reach experimental maturity to establish the co-culture. Overall, this co-culture can be useful for studying microglia-neuron interactions and can provide multiple readouts, including immunofluorescence microscopy, live imaging, as well as RNA and protein assays.


Subject(s)
Cerebral Cortex , Coculture Techniques , Microglia , Neurons , Animals , Coculture Techniques/methods , Microglia/cytology , Mice , Neurons/cytology , Cerebral Cortex/cytology , Cytological Techniques/methods
2.
Nat Commun ; 15(1): 6979, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143079

ABSTRACT

Oligodendrocyte precursor cells (OPCs) give rise to myelinating oligodendrocytes of the brain. This process persists throughout life and is essential for recovery from neurodegeneration. To better understand the cellular checkpoints that occur during oligodendrogenesis, we determined the mitochondrial distribution and morphometrics across the oligodendrocyte lineage in mouse and human cerebral cortex. During oligodendrocyte generation, mitochondrial content expands concurrently with a change in subcellular partitioning towards the distal processes. These changes are followed by an abrupt loss of mitochondria in the oligodendrocyte processes and myelin, coinciding with sheath compaction. This reorganization and extensive expansion and depletion take 3 days. Oligodendrocyte mitochondria are stationary over days while OPC mitochondrial motility is modulated by animal arousal state within minutes. Aged OPCs also display decreased mitochondrial size, volume fraction, and motility. Thus, mitochondrial dynamics are linked to oligodendrocyte generation, dynamically modified by their local microenvironment, and altered in the aging brain.


Subject(s)
Mitochondria , Myelin Sheath , Oligodendroglia , Animals , Mitochondria/metabolism , Humans , Oligodendroglia/metabolism , Oligodendroglia/cytology , Mice , Myelin Sheath/metabolism , Oligodendrocyte Precursor Cells/metabolism , Oligodendrocyte Precursor Cells/cytology , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Mice, Inbred C57BL , Male , Mitochondrial Dynamics , Cell Differentiation , Female
3.
Nat Commun ; 15(1): 7156, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169030

ABSTRACT

Egocentric neural representations of environmental features, such as edges and vertices, are important for constructing a geometrically detailed egocentric cognitive map for goal-directed navigation and episodic memory. While egocentric neural representations of edges like egocentric boundary/border cells exist, those that selectively represent vertices egocentrically are yet unknown. Here we report that granular retrosplenial cortex (RSC) neurons in male mice generate spatial receptive fields exclusively near the vertices of environmental geometries during free exploration, termed vertex cells. Their spatial receptive fields occurred at a specific orientation and distance relative to the heading direction of mice, indicating egocentric vector coding of vertex. Removing physical boundaries defining the environmental geometry abolished the egocentric vector coding of vertex, and goal-directed navigation strengthened the egocentric vector coding at the goal-located vertex. Our findings suggest that egocentric vector coding of vertex by granular RSC neurons helps construct an egocentric cognitive map that guides goal-directed navigation.


Subject(s)
Neurons , Animals , Male , Neurons/physiology , Mice , Mice, Inbred C57BL , Space Perception/physiology , Cerebral Cortex/physiology , Cerebral Cortex/cytology , Gyrus Cinguli/physiology , Gyrus Cinguli/cytology , Orientation/physiology , Spatial Navigation/physiology
4.
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
5.
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
6.
Methods Mol Biol ; 2831: 1-9, 2024.
Article in English | MEDLINE | ID: mdl-39134839

ABSTRACT

Primary neuronal cultures allow for in vitro analysis of early developmental processes such as axon pathfinding and growth dynamics. When coupled with methods to visualize and measure microtubule dynamics, this methodology enables an inside look at how the cytoskeleton changes in response to extracellular signaling cues. Here, we describe the culturing conditions and tools required to extract primary cortical neurons from postnatal mouse brains and visualize cytoskeletal components.


Subject(s)
Cerebral Cortex , Neurons , Animals , Mice , Neurons/cytology , Neurons/metabolism , Cerebral Cortex/cytology , Cells, Cultured , Microtubules/metabolism , Primary Cell Culture/methods , Cell Culture Techniques/methods , Cytoskeleton/metabolism
7.
Methods Mol Biol ; 2831: 81-95, 2024.
Article in English | MEDLINE | ID: mdl-39134845

ABSTRACT

During the development of mammalian brains, pyramidal neurons in the cerebral cortex form highly organized six layers with different functions. These neurons undergo developmental processes such as axon extension, dendrite outgrowth, and synapse formation. A proper integration of the neuronal connectivity through dynamic changes of dendritic branches and spines is required for learning and memory. Disruption of these crucial developmental processes is associated with many neurodevelopmental and neurodegenerative disorders. To investigate the complex dendritic architecture, several useful staining tools and genetic methods to label neurons have been well established. Monitoring the dynamics of dendritic spine in a single neuron is still a challenging task. Here, we provide a methodology that combines in vivo two-photon brain imaging and in utero electroporation, which sparsely labels cortical neurons with fluorescent proteins. This protocol may help elucidate the dynamics of microstructure and neural complexity in living rodents under normal and disease conditions.


Subject(s)
Neurons , Animals , Mice , Neurons/cytology , Neurons/metabolism , Electroporation/methods , Microscopy, Fluorescence, Multiphoton/methods , Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , Pyramidal Cells/metabolism , Pyramidal Cells/cytology , Female , Cerebral Cortex/cytology , Dendrites/metabolism
8.
Methods Mol Biol ; 2831: 113-132, 2024.
Article in English | MEDLINE | ID: mdl-39134847

ABSTRACT

Neuronal development is characterized by the unidirectional flow of signal from the axon to the dendrites via synapses. Neuronal polarization is a critical step during development that allows the specification of the different neuronal processes as a single axon and multiple dendrites both structurally and functionally, allowing the unidirectional flow of information. Along with extrinsic and intrinsic signaling, a whole network of molecular complexes involved in positive and negative feedback loops play a major role in this critical distinction of neuronal processes. As a result, neuronal morphology is drastically altered during establishment of polarity. In this chapter, we discuss how we can analyze the morphological alterations of neurons in vitro in culture to assess the development and polarity status of the neuron. We also discuss how these studies can be conducted in vivo, where polarity studies pose a greater challenge with promising results for addressing multiple pathological conditions. Our experimental model is limited to rodent hippocampal/cortical neurons in culture and cortical neurons in brain tissues, which are well-characterized model systems for understanding neuronal polarization.


Subject(s)
Cell Polarity , Hippocampus , Neurons , Animals , Neurons/cytology , Neurons/physiology , Neurons/metabolism , Mice , Hippocampus/cytology , Cells, Cultured , Rats , Axons/physiology , Axons/metabolism , Dendrites/physiology , Dendrites/metabolism , Cerebral Cortex/cytology
9.
Cereb Cortex ; 34(7)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38960704

ABSTRACT

The Polycomb Repressive Complex 2 (PRC2) regulates corticogenesis, yet the consequences of mutations to this epigenetic modifier in the mature brain are poorly defined. Importantly, PRC2 core genes are haploinsufficient and causative of several human neurodevelopmental disorders. To address the role of PRC2 in mature cortical structure and function, we conditionally deleted the PRC2 gene Eed from the developing mouse dorsal telencephalon. Adult homozygotes displayed smaller forebrain structures. Single-nucleus transcriptomics revealed that glutamatergic neurons were particularly affected, exhibiting dysregulated gene expression profiles, accompanied by aberrations in neuronal morphology and connectivity. Remarkably, homozygous mice performed well on challenging cognitive tasks. In contrast, while heterozygous mice did not exhibit clear anatomical or behavioral differences, they displayed dysregulation of neuronal genes and altered neuronal morphology that was strikingly different from homozygous phenotypes. Collectively, these data reveal how alterations to PRC2 function shape the mature brain and reveal a dose-specific role for PRC2 in determining glutamatergic neuron identity.


Subject(s)
Glutamic Acid , Neurogenesis , Neurons , Polycomb Repressive Complex 2 , Animals , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Neurons/metabolism , Neurons/physiology , Mice , Neurogenesis/physiology , Glutamic Acid/metabolism , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Male , Mice, Inbred C57BL , Female , Mice, Transgenic
10.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-39000184

ABSTRACT

Microglia migrate to the cerebral cortex during early embryonic stages. However, the precise mechanisms underlying microglia migration remain incompletely understood. As an extracellular matrix protein, Netrin-1 is involved in modulating the motility of diverse cells. In this paper, we found that Netrin-1 promoted microglial BV2 cell migration in vitro. Mechanism studies indicated that the activation of GSK3ß activity contributed to Netrin-1-mediated microglia migration. Furthermore, Integrin α6/ß1 might be the relevant receptor. Single-cell data analysis revealed the higher expression of Integrin α6 subunit and ß1 subunit in microglia in comparison with classical receptors, including Dcc, Neo1, Unc5a, Unc5b, Unc5c, Unc5d, and Dscam. Microscale thermophoresis (MST) measurement confirmed the high binding affinity between Integrin α6/ß1 and Netrin-1. Importantly, activation of Integrin α6/ß1 with IKVAV peptides mirrored the microglia migration and GSK3 activation induced by Netrin-1. Finally, conditional knockout (CKO) of Netrin-1 in radial glial cells and their progeny led to a reduction in microglia population in the cerebral cortex at early developmental stages. Together, our findings highlight the role of Netrin-1 in microglia migration and underscore its therapeutic potential in microglia-related brain diseases.


Subject(s)
Cell Movement , Microglia , Netrin-1 , Netrin-1/metabolism , Netrin-1/genetics , Microglia/metabolism , Animals , Mice , Mice, Knockout , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Cell Line , Integrin beta1/metabolism , Integrin beta1/genetics
11.
J Neurophysiol ; 132(3): 757-764, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39015071

ABSTRACT

To support complex cognition, neuronal circuits must integrate information across multiple temporal scales, ranging from milliseconds to decades. Neuronal timescales describe the duration over which activity within a network persists, posing a putative explanatory mechanism for how information might be integrated over multiple temporal scales. Little is known about how timescales develop in human neural circuits or other model systems, limiting insight into how the functional dynamics necessary for cognition emerge. In our work, we show that neuronal timescales develop in a nonlinear fashion in human cortical organoids, which is partially replicated in dissociated rat hippocampus cultures. We use spectral parameterization of spiking activity to extract an estimate of neuronal timescale that is unbiased by coevolving oscillations. Cortical organoid timescales begin to increase around month 6 postdifferentiation. In rodent hippocampal dissociated cultures, we see that timescales decrease from in vitro days 13-23 before stabilizing. We speculate that cortical organoid development over the duration studied here reflects an earlier stage of a generalized developmental timeline in contrast to the rodent hippocampal cultures, potentially accounting for differences in timescale developmental trajectories. The fluctuation of timescales might be an important developmental feature that reflects the changing complexity and information capacity in developing neuronal circuits.NEW & NOTEWORTHY Neuronal timescales describe the persistence of activity within a network of neurons. Timescales were found to fluctuate with development in two model systems. In cortical organoids timescales increased, peaked, and then decreased throughout development; in rat hippocampal dissociated cultures timescales decreased over development. These distinct developmental models overlap to highlight a critical window in which timescales lengthen and contract, potentially indexing changes in the information capacity of neuronal systems.


Subject(s)
Hippocampus , Neurons , Organoids , Animals , Organoids/physiology , Organoids/cytology , Hippocampus/physiology , Hippocampus/cytology , Rats , Humans , Neurons/physiology , Cerebral Cortex/physiology , Cerebral Cortex/cytology , Cells, Cultured , Action Potentials/physiology , Time Factors
12.
Cereb Cortex ; 34(7)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-39024157

ABSTRACT

The centrosome is the main microtubule organizing center in stem cells, and its mother centriole, anchored to the cell membrane, serves as the basal body of the primary cilium. Prolonged anchorage of centrosomes and primary cilia to the apical segment of the membrane of apical neural progenitor cells is considered vital for interkinetic nuclear translocation and repetitive cycling in the ventricular zone. In contrast, the basolateral anchorage of primary cilia has been regarded as the first step in delamination and conversion of apical to basal neural progenitor cells or neurons. Using electron microscopy analysis of serial sections, we show that centrosomes, in a fraction of cells, anchor to the basolateral cell membrane immediately after cell division and before development of cilia. In other cells, centrosomes situate freely in the cytoplasm, increasing their probability of subsequent apical anchorage. In mice, anchored centrosomes in the cells shortly after mitosis predominate during the entire cerebral neurogenesis, whereas in macaque monkeys, cytoplasmic centrosomes are more numerous. Species-specific differences in the ratio of anchored and free cytoplasmic centrosomes appear to be related to prolonged neurogenesis in the ventricular zone that is essential for lateral expansion of the cerebral cortex in primates.


Subject(s)
Centrosome , Cerebral Cortex , Neural Stem Cells , Neurogenesis , Animals , Centrosome/metabolism , Cerebral Cortex/cytology , Neural Stem Cells/physiology , Mice , Neurogenesis/physiology
13.
Lab Chip ; 24(16): 3945-3957, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-38994783

ABSTRACT

Electrical stimulation (ES) techniques, such as deep brain and transcranial electrical stimulation, have shown promise in alleviating the symptoms of depression and other neurological disorders in vivo. A new noninvasive ES method called temporal interference stimulation (TIS), possesses great potential as it can be used to steer the stimulation and possibly selectively modulate different brain regions. To study TIS in a controlled environment, we successfully established an in vitro 'TIS on a chip' setup using rat cortical neurons on microelectrode arrays (MEAs) in combination with a current stimulator. We validated the developed TIS system and demonstrated the spatial steerability of the stimulation by direct electric field measurements in the chip setup. We stimulated cultures of rat cortical neurons at 28 days in vitro (DIV) by two-channel stimulation delivering 1) TIS at 653 Hz and 643 Hz, resulting in a 10 Hz frequency envelope, 2) low-frequency stimulation (LFS) at 10 Hz and 3) high-frequency stimulation (HFS) at 653 Hz. Unstimulated cultures were used as control/sham. We observed the differences in the electric field strengths during TIS, HFS, and LFS. Moreover, HFS and LFS had the smallest effects on neuronal activity. Instead, TIS elicited neuronal electrophysiological responses, especially 24 hours after stimulation. Our 'TIS on a chip' approach eludicates the applicability of TIS as a method to modulate neuronal electrophysiological activity. The TIS on a chip approach provides spatially steerable stimuli while mitigating the effects of high stimulus fields near the stimulation electrodes. Thus, the approach opens new avenues for stimulation on a chip applications, allowing the study of neuronal responses to gain insights into the potential clinical applications of TIS in treating various brain disorders.


Subject(s)
Electric Stimulation , Microelectrodes , Neurons , Animals , Neurons/physiology , Rats , Cells, Cultured , Rats, Sprague-Dawley , Cerebral Cortex/cytology , Cerebral Cortex/physiology
14.
J Neurophysiol ; 132(3): 653-665, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38988287

ABSTRACT

Generation of human induced pluripotent stem cells (iPSCs) through reprogramming was a transformational change in the field of regenerative medicine that led to new possibilities for drug discovery and cell replacement therapy. Several protocols have been established to differentiate hiPSCs into neuronal lineages. However, low differentiation efficiency is one of the major drawbacks of these approaches. Here, we compared the efficiency of two methods of neuronal differentiation from iPSCs cultured in two different culture media, StemFlex Medium (SFM) and Essential 8 Medium (E8M). The results indicated that iPSCs cultured in E8M efficiently generated different types of neurons in a shorter time and without the growth of undifferentiated nonneuronal cells in the culture as compared with those generated from iPSCs in SFM. Furthermore, these neurons were validated as functional units immunocytochemically by confirming the expression of mature neuronal markers (i.e., NeuN, ß tubulin, and Synapsin I) and whole cell patch-clamp recordings. Long-read single-cell RNA sequencing confirms the presence of upper and deep layer cortical layer excitatory and inhibitory neuronal subtypes in addition to small populations of GABAergic neurons in day 30 neuronal cultures. Pathway analysis indicated that our protocol triggers the signaling transcriptional networks important for the process of neuronal differentiation in vivo.NEW & NOTEWORTHY Low differentiation efficiency is one of the major drawbacks of the existing protocols to differentiate iPSCs into neuronal lineages. Here, we present time-efficient and robust approach of neuronal differentiation leading to the generation of functional brain units, cortical layer neurons. We found iPSCs cultured in Essential 8 media (E8M) resulted in neuronal differentiation without the signs of growth of spontaneously differentiated cells in culture at any point in 35 days compared with Stemflex media (SFM).


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells , Neurons , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/physiology , Humans , Neurons/physiology , Neurons/cytology , Cell Differentiation/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Neurogenesis/physiology , Protein Isoforms/metabolism , Culture Media
15.
Nat Commun ; 15(1): 5512, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951525

ABSTRACT

Microglia are important players in surveillance and repair of the brain. Implanting an electrode into the cortex activates microglia, produces an inflammatory cascade, triggers the foreign body response, and opens the blood-brain barrier. These changes can impede intracortical brain-computer interfaces performance. Using two-photon imaging of implanted microelectrodes, we test the hypothesis that low-intensity pulsed ultrasound stimulation can reduce microglia-mediated neuroinflammation following the implantation of microelectrodes. In the first week of treatment, we found that low-intensity pulsed ultrasound stimulation increased microglia migration speed by 128%, enhanced microglia expansion area by 109%, and a reduction in microglial activation by 17%, indicating improved tissue healing and surveillance. Microglial coverage of the microelectrode was reduced by 50% and astrocytic scarring by 36% resulting in an increase in recording performance at chronic time. The data indicate that low-intensity pulsed ultrasound stimulation helps reduce the foreign body response around chronic intracortical microelectrodes.


Subject(s)
Electrodes, Implanted , Microelectrodes , Microglia , Ultrasonic Waves , Microglia/radiation effects , Microglia/metabolism , Animals , Male , Foreign-Body Reaction/prevention & control , Foreign-Body Reaction/etiology , Mice , Cerebral Cortex/radiation effects , Cerebral Cortex/cytology , Brain-Computer Interfaces , Cell Movement/radiation effects , Rats
16.
Cell Rep ; 43(8): 114503, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39018245

ABSTRACT

Synaptic plasticities, such as long-term potentiation (LTP) and depression (LTD), tune synaptic efficacy and are essential for learning and memory. Current studies of synaptic plasticity in humans are limited by a lack of adequate human models. Here, we modeled the thalamocortical system by fusing human induced pluripotent stem cell-derived thalamic and cortical organoids. Single-nucleus RNA sequencing revealed that >80% of cells in thalamic organoids were glutamatergic neurons. When fused to form thalamocortical assembloids, thalamic and cortical organoids formed reciprocal long-range axonal projections and reciprocal synapses detectable by light and electron microscopy, respectively. Using whole-cell patch-clamp electrophysiology and two-photon imaging, we characterized glutamatergic synaptic transmission. Thalamocortical and corticothalamic synapses displayed short-term plasticity analogous to that in animal models. LTP and LTD were reliably induced at both synapses; however, their mechanisms differed from those previously described in rodents. Thus, thalamocortical assembloids provide a model system for exploring synaptic plasticity in human circuits.


Subject(s)
Neuronal Plasticity , Thalamus , Humans , Thalamus/physiology , Thalamus/cytology , Neuronal Plasticity/physiology , Synapses/physiology , Synapses/metabolism , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Cerebral Cortex/physiology , Cerebral Cortex/cytology , Organoids/metabolism , Long-Term Potentiation/physiology , Neurons/physiology , Neurons/metabolism
17.
Eur J Neurosci ; 60(5): 4893-4906, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39073048

ABSTRACT

We investigated the impact of the human-specific gene CHRFAM7A on the function of α7 nicotinic acetylcholine receptors (α7 nAChRs) in two different types of neurons: human-induced pluripotent stem cell (hiPSC)-derived cortical neurons, and superior cervical ganglion (SCG) neurons, taken from transgenic mice expressing CHRFAM7A. dupα7, the gene product of CHRFAM7A, which lacks a major part of the extracellular N-terminal ligand-binding domain, co-assembles with α7, the gene product of CHRNA7. We assessed the receptor function in hiPSC-derived cortical and SCG neurons with Fura-2 calcium imaging and three different α7-specific ligands: PNU282987, choline, and 4BP-TQS. Given the short-lived open state of α7 receptors, we combined the two orthosteric agonists PNU282987 and choline with the type-2 positive allosteric modulator (PAM II) PNU120596. In line with different cellular models used previously, we demonstrate that CHRFAM7A has a major impact on nicotinic α7 nAChRs by reducing calcium transients in response to all three agonists.


Subject(s)
Induced Pluripotent Stem Cells , Mice, Transgenic , Neurons , alpha7 Nicotinic Acetylcholine Receptor , alpha7 Nicotinic Acetylcholine Receptor/metabolism , alpha7 Nicotinic Acetylcholine Receptor/genetics , Animals , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Humans , Neurons/metabolism , Neurons/drug effects , Mice , Choline/pharmacology , Choline/metabolism , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/metabolism , Bridged Bicyclo Compounds/pharmacology , Nicotinic Agonists/pharmacology , Benzamides/pharmacology , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Cerebral Cortex/drug effects , Calcium/metabolism , Isoxazoles , Phenylurea Compounds
18.
J Comp Neurol ; 532(6): e25626, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39031698

ABSTRACT

We have examined the number and distribution of NeuN-immunoreactive cortical white matter interstitial cells (WMICs) and compared them to the neurons in layers 1-6 across the overlying cortex in coronal sections from postnatal macaques. The data have been gathered from over 300 selected regions at gyral crowns, at sulci, and at linear regions of the cortex where we also determined cortical layer thicknesses: standard thicknesses and tangential thicknesses. Cortical thicknesses and cell numbers showed variability according to gyral, linear, or sulcal regions. In spite of these variations, our standardized cell numbers in layers 1 to 6b and interstitial cells underlying layer 6b-white matter boundary have shown a consistent correlation between the number of WMICs and the number of layer 5 and 6a cortical neurons on all cortical regions studied: for each WMIC, there are on the order of five cortical neurons in layer 5 and approximately three cortical neurons in layer 6a, irrespective of the origins of the selected cortical area or whether they are from gyral, linear, or sulcal regions. We propose that the number of interstitial neurons in the postnatal macaque cortex is correlated to the density of neurons within layers 5 and 6a and, from a clinical perspective, the change in density or distribution of interstitial neurons in schizophrenia or epilepsy may in fact be linked to the number of layers 5 and 6a neurons.


Subject(s)
Cerebral Cortex , Neurons , White Matter , Animals , Neurons/cytology , Cerebral Cortex/cytology , White Matter/cytology , White Matter/anatomy & histology , Cell Count , Animals, Newborn , Macaca mulatta , Male , Female
19.
J Mol Neurosci ; 74(3): 60, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38904846

ABSTRACT

Our former studies have identified the alleviating effect of Calycosin (CA) on spinal cord injury (SCI). In this study, our purpose is to explore the influence of CA on SCI from the perspective of promoting axon growth. The SCI animal model was constructed by spinal cord compression, wherein rat primary cortex neuronal isolation was performed, and the axonal growth restriction cell model was established via chondroitin sulfate proteoglycan (CSPG) treatment. The expressions of axon regeneration markers were measured via immunofluorescent staining and western blot, and the direct target of CA was examined using silver staining. Finally, the expression of the protein tyrosine phosphatase receptor type S (PTPRS) was assessed using western blot. CA treatment increased neuronal process outgrowth and the expressions of axon regeneration markers, such as neurofilament H (NF-H), vesicular glutamate transporter 1 (vGlut1), and synaptophysin (Syn) in both SCI model rats and CSPG-treated primary cortical neurons, and PTPRS levels were elevated after SCI induction. In addition, PTPRS was the direct target of CA, and according to in vivo findings, exposure to CA reduced the PTPRS content. Furthermore, PTPRS overexpression inhibited CA's enhancement of axon regeneration marker content and neuronal axon lengths. CA improves SCI by increasing axon development through regulating PTPRS expression.


Subject(s)
Axons , Isoflavones , Rats, Sprague-Dawley , Spinal Cord Injuries , Synaptophysin , Animals , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/drug therapy , Rats , Isoflavones/pharmacology , Isoflavones/therapeutic use , Axons/drug effects , Axons/metabolism , Cells, Cultured , Synaptophysin/metabolism , Synaptophysin/genetics , Neurofilament Proteins/metabolism , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Glutamate Transport Protein 1/genetics , Neurons/metabolism , Neurons/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/cytology , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Male , Chondroitin Sulfate Proteoglycans/metabolism , Neuronal Outgrowth/drug effects , Female , Vesicular Glutamate Transport Protein 2
20.
Glia ; 72(9): 1693-1706, 2024 09.
Article in English | MEDLINE | ID: mdl-38852127

ABSTRACT

Astrocytes that reside in superficial (SL) and deep cortical layers have distinct molecular profiles and morphologies, which may underlie specific functions. Here, we demonstrate that the production of SL and deep layer (DL) astrocyte populations from neural progenitor cells in the mouse is temporally regulated. Lineage tracking following in utero and postnatal electroporation with PiggyBac (PB) EGFP and birth dating with EdU and FlashTag, showed that apical progenitors produce astrocytes during late embryogenesis (E16.5) that are biased to the SL, while postnatally labeled (P0) astrocytes are biased to the DL. In contrast, astrocytes born during the predominantly neurogenic window (E14.5) showed a random distribution in the SL and DL. Of interest, E13.5 astrocytes birth dated at E13.5 with EdU showed a lower layer bias, while FT labeling of apical progenitors showed no bias. Finally, examination of the morphologies of "biased" E16.5- and P0-labeled astrocytes demonstrated that E16.5-labeled astrocytes exhibit different morphologies in different layers, while P0-labeled astrocytes do not. Differences based on time of birth are also observed in the molecular profiles of E16.5 versus P0-labeled astrocytes. Altogether, these results suggest that the morphological, molecular, and positional diversity of cortical astrocytes is related to their time of birth from ventricular/subventricular zone progenitors.


Subject(s)
Astrocytes , Cerebral Cortex , Neural Stem Cells , Animals , Astrocytes/metabolism , Astrocytes/cytology , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Mice , Mice, Transgenic , Female , Animals, Newborn , Gene Expression Regulation, Developmental , Transcriptome , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Cerebral Ventricles/cytology , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL