Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 940
Filter
1.
Cereb Cortex ; 34(5)2024 May 02.
Article in English | MEDLINE | ID: mdl-38745556

ABSTRACT

The basic building block of the cerebral cortex, the pyramidal cell, has been shown to be characterized by a markedly different dendritic structure among layers, cortical areas, and species. Functionally, differences in the structure of their dendrites and axons are critical in determining how neurons integrate information. However, within the human cortex, these neurons have not been quantified in detail. In the present work, we performed intracellular injections of Lucifer Yellow and 3D reconstructed over 200 pyramidal neurons, including apical and basal dendritic and local axonal arbors and dendritic spines, from human occipital primary visual area and associative temporal cortex. We found that human pyramidal neurons from temporal cortex were larger, displayed more complex apical and basal structural organization, and had more spines compared to those in primary sensory cortex. Moreover, these human neocortical neurons displayed specific shared and distinct characteristics in comparison to previously published human hippocampal pyramidal neurons. Additionally, we identified distinct morphological features in human neurons that set them apart from mouse neurons. Lastly, we observed certain consistent organizational patterns shared across species. This study emphasizes the existing diversity within pyramidal cell structures across different cortical areas and species, suggesting substantial species-specific variations in their computational properties.


Subject(s)
Pyramidal Cells , Humans , Pyramidal Cells/physiology , Animals , Male , Female , Mice , Adult , Dendritic Spines/physiology , Dendritic Spines/ultrastructure , Temporal Lobe/cytology , Dendrites/physiology , Middle Aged , Axons/physiology , Species Specificity
2.
Adv Sci (Weinh) ; 11(17): e2306630, 2024 May.
Article in English | MEDLINE | ID: mdl-38493494

ABSTRACT

The modification of synaptic and neural connections in adults, including the formation and removal of synapses, depends on activity-dependent synaptic and structural plasticity. MicroRNAs (miRNAs) play crucial roles in regulating these changes by targeting specific genes and regulating their expression. The fact that somatic and dendritic activity in neurons often occurs asynchronously highlights the need for spatial and dynamic regulation of protein synthesis in specific milieu and cellular loci. MicroRNAs, which can show distinct patterns of enrichment, help to establish the localized distribution of plasticity-related proteins. The recent study using atomic force microscopy (AFM)-based nanoscale imaging reveals that the abundance of miRNA(miR)-134 is inversely correlated with the functional activity of dendritic spine structures. However, the miRNAs that are selectively upregulated in potentiated synapses, and which can thereby support prospective changes in synaptic efficacy, remain largely unknown. Using AFM force imaging, significant increases in miR-132 in the dendritic regions abutting functionally-active spines is discovered. This study provides evidence for miR-132 as a novel positive miRNA regulator residing in dendritic shafts, and also suggests that activity-dependent miRNAs localized in distinct sub-compartments of neurons play bi-directional roles in controlling synaptic transmission and synaptic plasticity.


Subject(s)
MicroRNAs , Microscopy, Atomic Force , Neuronal Plasticity , Synapses , Animals , Mice , Dendritic Spines/metabolism , Dendritic Spines/genetics , Dendritic Spines/ultrastructure , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Microscopy, Atomic Force/methods , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Neurons/metabolism , Synapses/metabolism , Synapses/genetics
3.
Ageing Res Rev ; 87: 101933, 2023 06.
Article in English | MEDLINE | ID: mdl-37061201

ABSTRACT

Neural circuits, such as synaptic plasticity and neural activity, are critical components of healthy brain function. The consequent dynamic remodeling of neural circuits is an ongoing procedure affecting neuronal activities. Disruption of this essential process results in diseases. Advanced microscopic applications such as two-photon laser scanning microscopy have recently been applied to understand neural circuit changes during disease since it can visualize fine structural and functional cellular activation in living animals. In this review, we have summarized the latest work assessing the dynamic rewiring of postsynaptic dendritic spines and modulation of calcium transients in neurons of the intact living brain, focusing on their potential roles in neurological disorders (e.g. Alzheimer's disease, stroke, and epilepsy). Understanding the fine changes that occurred in the brain during disease is crucial for future clinical intervention developments.


Subject(s)
Alzheimer Disease , Stroke , Animals , Humans , Neurons , Neuronal Plasticity/physiology , Brain/diagnostic imaging , Brain/physiology , Alzheimer Disease/diagnostic imaging , Dendritic Spines/physiology , Dendritic Spines/ultrastructure
4.
Behav Brain Res ; 418: 113621, 2022 02 10.
Article in English | MEDLINE | ID: mdl-34624424

ABSTRACT

Sleep is essential for important physiological functions. Impairment of learning and memory function caused by lack of sleep is a common physiological phenomenon of which underlying changes in synaptic plasticity in the hippocampus are not well understood. The possible different effects of sleep deprivation (SD) lasting for various durations on learning and memory function and hippocampal synaptic plasticity are still not completely clear. In this study, we used a modified multiple platform method (MMPM) to induce rapid eye movement SD (REM SD), lasting for 24 h, 48 h, and 72 h, separately. The novel place recognition (NPR) and novel object recognition (NOR) tasks were used to test the novelty-related object recognition memory (ORM) and object location memory (OLM) of mice. Then, hippocampal synaptic plasticity was evaluated after all behavioural experiments. The results showed that REM SD played a key role in OLM but not in ORM. Specifically, 24 h REM SD improved novelty-related OLM, accompanied by a significantly increased hippocampal synaptic plasticity, including gain of dendritic spines, increased expression of hippocampal GluA1, and enhanced long-term potentiation (LTP), whereas 48 h REM SD showed no effect on OLM or the hippocampal synaptic plasticity mentioned above; however, 72 h REM SD impaired novelty-related OLM and weakened hippocampal synaptic plasticity, including serious loss of dendritic spines, decreased expression of hippocampal GluA1, and significantly attenuated LTP. Our results suggest that REM SD of various durations has different effects on both novelty-related OLM and hippocampal synaptic plasticity.


Subject(s)
Learning/physiology , Memory/physiology , Neuronal Plasticity/physiology , Recognition, Psychology/physiology , Sleep Deprivation/physiopathology , Animals , Dendritic Spines/ultrastructure , Hippocampus/metabolism , Long-Term Potentiation/physiology , Male , Mice , Mice, Inbred C57BL , Sleep, REM , Time Factors , Visual Perception/physiology
5.
PLoS Biol ; 19(8): e3001375, 2021 08.
Article in English | MEDLINE | ID: mdl-34428203

ABSTRACT

Pyramidal neurons (PNs) are covered by thousands of dendritic spines receiving excitatory synaptic inputs. The ultrastructure of dendritic spines shapes signal compartmentalization, but ultrastructural diversity is rarely taken into account in computational models of synaptic integration. Here, we developed a 3D correlative light-electron microscopy (3D-CLEM) approach allowing the analysis of specific populations of synapses in genetically defined neuronal types in intact brain circuits. We used it to reconstruct segments of basal dendrites of layer 2/3 PNs of adult mouse somatosensory cortex and quantify spine ultrastructural diversity. We found that 10% of spines were dually innervated and 38% of inhibitory synapses localized to spines. Using our morphometric data to constrain a model of synaptic signal compartmentalization, we assessed the impact of spinous versus dendritic shaft inhibition. Our results indicate that spinous inhibition is locally more efficient than shaft inhibition and that it can decouple voltage and calcium signaling, potentially impacting synaptic plasticity.


Subject(s)
Dendritic Spines/ultrastructure , Excitatory Postsynaptic Potentials , Inhibitory Postsynaptic Potentials , Models, Neurological , Pyramidal Cells/ultrastructure , Animals , Calcium Signaling , Dendritic Spines/physiology , Female , Mice , Microscopy, Electron, Scanning/methods , Neuronal Plasticity , Pregnancy , Somatosensory Cortex/physiology , Somatosensory Cortex/ultrastructure
6.
Mol Brain ; 14(1): 129, 2021 08 21.
Article in English | MEDLINE | ID: mdl-34419133

ABSTRACT

Hypobaric hypoxia (HH) is a typical characteristic of high altitude environment and causes a spectrum of pathophysiological effects, including headaches, gliovascular dysfunction and cognitive retardation. Here, we sought to understand the mechanisms underlying cognitive deficits under HH exposure. Our results showed that hypobaric hypoxia exposure impaired cognitive function and suppressed dendritic spine density accompanied with increased neck length in both basal and apical hippocampal CA1 region neurons in mice. The expression of PSD95, a vital synaptic scaffolding molecule, is down-regulated by hypobaric hypoxia exposure and post-transcriptionally regulated by cold-inducible RNA-binding protein (Cirbp) through 3'-UTR region binding. PSD95 expressing alleviates hypoxia-induced dendritic spine morphology changes of hippocampal neurons and memory deterioration. Moreover, overexpressed Cirbp in hippocampus rescues HH-induced abnormal expression of PSD95 and attenuates hypoxia-induced dendritic spine injury and cognitive retardation. Thus, our findings reveal a novel mechanism that Cirbp-PSD-95 axis appears to play an essential role in HH-induced cognitive dysfunction in mice.


Subject(s)
Altitude Sickness/physiopathology , CA1 Region, Hippocampal/pathology , Cognition Disorders/prevention & control , Dendritic Spines/ultrastructure , Disks Large Homolog 4 Protein/physiology , RNA-Binding Proteins/physiology , 3' Untranslated Regions , Animals , Avoidance Learning , Base Sequence , Cells, Cultured , Cognition Disorders/etiology , Disks Large Homolog 4 Protein/biosynthesis , Disks Large Homolog 4 Protein/genetics , Gene Expression Regulation , Genes, Reporter , Genetic Vectors/administration & dosage , Memory Disorders/etiology , Memory Disorders/prevention & control , Mice , Mice, Inbred C57BL , Morris Water Maze Test , Neurons/physiology , Neurons/ultrastructure , Open Field Test , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/genetics , Random Allocation , Recombinant Fusion Proteins/metabolism
7.
J Neurosci ; 41(33): 7003-7014, 2021 08 18.
Article in English | MEDLINE | ID: mdl-34266899

ABSTRACT

The structural plasticity of dendritic spines is considered to be an important basis of synaptic plasticity, learning, and memory. Here, we induced input-specific structural LTP (sLTP) in single dendritic spines in organotypic hippocampal slices from mice of either sex and performed ultrastructural analyses of the spines using efficient correlative light and electron microscopy. We observed reorganization of the PSD nanostructure, such as perforation and segmentation, at 2-3, 20, and 120 min after sLTP induction. In addition, PSD and nonsynaptic axon-spine interface (nsASI) membrane expanded unevenly during sLTP. Specifically, the PSD area showed a transient increase at 2-3 min after sLTP induction. The PSD growth was to a degree less than spine volume growth at 2-3 min and 20 min after sLTP induction but became similar at 120 min. On the other hand, the nsASI area showed a profound and lasting expansion, to a degree similar to spine volume growth throughout the process. These rapid ultrastructural changes in PSD and surrounding membrane may contribute to rapid electrophysiological plasticity during sLTP.SIGNIFICANCE STATEMENT To understand the ultrastructural changes during synaptic plasticity, it is desired to efficiently image single dendritic spines that underwent structural plasticity in electron microscopy. We induced structural long-term potentiation (sLTP) in single dendritic spines by two-photon glutamate uncaging. We then identified the same spines at different phases of sLTP and performed ultrastructural analysis by using an efficient correlative light and electron microscopy method. We found that postsynaptic density undergoes dramatic modification in its structural complexity immediately after sLTP induction. Meanwhile, the nonsynaptic axon-spine interface area shows a rapid and sustained increase throughout sLTP. Our results indicate that the uneven modification of synaptic and nonsynaptic postsynaptic membrane might contribute to rapid electrophysiological plasticity during sLTP.


Subject(s)
Dendritic Spines/ultrastructure , Hippocampus/ultrastructure , Long-Term Potentiation , Post-Synaptic Density/ultrastructure , Animals , Axons/ultrastructure , Biolistics , Cell Membrane/ultrastructure , Dendritic Spines/physiology , Female , Glutamates/radiation effects , Image Processing, Computer-Assisted , Indoles/radiation effects , Male , Mice , Microscopy, Electron, Scanning , Photochemistry
8.
Brain Res Bull ; 174: 92-102, 2021 09.
Article in English | MEDLINE | ID: mdl-34098041

ABSTRACT

Treatment of neuropathic pain (NP) resulting from nerve injury is one of the most complicated and challenging in modern practice. Pharmacological treatments for NP are not fully effectively and novel approaches are requisite. Recently, transplantation of bone mesenchymal stem cells (BMSCs) has represented a promising approach for pain relief and neural repair, but how it produces beneficial effects on resiniferatoxin (RTX) induced nerve injury is still unclear. Here, we identified the BMSCs' analgesic effects and their potential mechanisms of microglial cells activation on RTX induced neuralgia. Immunostaining, biochemical studies demonstrated that microglia rather than astrocyte cells activation involved in RTX induced mechanical hyperalgesia, whereas the GFP-labeled BMSCs alleviated this mechanical hyperalgesia. Moreover, pain-related TRPA1, PKCδ, CaMKIIɑ (Calcium/calmodulin dependent protein kinase II), P38/MAPK (mitogen-activated protein kinase), p-P65 activation and expression in the spinal cord were significantly inhibited after BMSC administration. In addition, BMSCs treated RTX mice displayed a lower density of mushroom dendritic spines. Our research suggested that activation of PKCδ-CaMKIIɑ-P38/MAPK-p-P65 pathway and mushroom dendritic spines abnormal increase in the spinal cord is the main mechanism of RTX induced neuropathic pain, and transplant of BMSCs to the damaged nerve may offer promising approach for neuropathic pain.


Subject(s)
Bone Marrow Transplantation/methods , Diterpenes/toxicity , MAP Kinase Signaling System/drug effects , Mesenchymal Stem Cell Transplantation/methods , Neuralgia/chemically induced , Neuralgia/prevention & control , Protein Kinase C-delta/antagonists & inhibitors , TRPA1 Cation Channel/antagonists & inhibitors , Transcription Factor RelA/drug effects , Animals , Behavior, Animal , Dendritic Spines/pathology , Dendritic Spines/ultrastructure , Hyperalgesia/prevention & control , Macrophage Activation , Male , Mice , Mice, Inbred C57BL , Microglia , Neuralgia/psychology , Spinal Cord/metabolism , Spinal Cord/pathology
9.
Nat Neurosci ; 24(8): 1151-1162, 2021 08.
Article in English | MEDLINE | ID: mdl-34168338

ABSTRACT

Dendritic spines, the postsynaptic compartments of excitatory neurotransmission, have different shapes classified from 'stubby' to 'mushroom-like'. Whereas mushroom spines are essential for adult brain function, stubby spines disappear during brain maturation. It is still unclear whether and how they differ in protein composition. To address this, we combined electron microscopy and quantitative biochemistry with super-resolution microscopy to annotate more than 47,000 spines for more than 100 synaptic targets. Surprisingly, mushroom and stubby spines have similar average protein copy numbers and topologies. However, an analysis of the correlation of each protein to the postsynaptic density mass, used as a marker of synaptic strength, showed substantially more significant results for the mushroom spines. Secretion and trafficking proteins correlated particularly poorly to the strength of stubby spines. This suggests that stubby spines are less likely to adequately respond to dynamic changes in synaptic transmission than mushroom spines, which possibly explains their loss during brain maturation.


Subject(s)
Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , Post-Synaptic Density/metabolism , Post-Synaptic Density/ultrastructure , Animals , Brain/metabolism , Brain/ultrastructure , Microscopy, Electron, Transmission , Proteome , Rats , Rats, Wistar , Synaptic Transmission/physiology
10.
Neurobiol Dis ; 156: 105420, 2021 08.
Article in English | MEDLINE | ID: mdl-34102277

ABSTRACT

STED microscopy is one of several fluorescence microscopy techniques that permit imaging at higher spatial resolution than what the diffraction-limit of light dictates. STED imaging is unique among these super-resolution modalities in being a beam-scanning microscopy technique based on confocal or 2-photon imaging, which provides the advantage of superior optical sectioning in thick samples. Compared to the other super-resolution techniques that are based on widefield microscopy, this makes STED particularly suited for imaging inside live brain tissue, such as in slices or in vivo. Notably, the 50 nm resolution provided by STED microscopy enables analysis of neural morphologies that conventional confocal and 2-photon microscopy approaches cannot resolve, including all-important synaptic structures. Over the course of the last 20 years, STED microscopy has undergone extensive developments towards ever more versatile use, and has facilitated remarkable neurophysiological discoveries. The technique is still not widely adopted for live tissue imaging, even though one of its particular strengths is exactly in resolving the nanoscale dynamics of synaptic structures in brain tissue, as well as in addressing the complex morphologies of glial cells, and revealing the intricate structure of the brain extracellular space. Not least, live tissue STED microscopy has so far hardly been applied in settings of pathophysiology, though also here it shows great promise for providing new insights. This review outlines the technical advantages of STED microscopy for imaging in live brain tissue, and highlights key neurobiological findings brought about by the technique.


Subject(s)
Brain/metabolism , Dendritic Spines/metabolism , Fluorescent Dyes/metabolism , Microscopy, Fluorescence/methods , Synapses/metabolism , Animals , Brain/cytology , Brain/ultrastructure , Dendritic Spines/ultrastructure , Humans , Microscopy, Fluorescence/trends , Neurons/metabolism , Neurons/ultrastructure , Synapses/ultrastructure
11.
Cereb Cortex ; 31(10): 4742-4764, 2021 08 26.
Article in English | MEDLINE | ID: mdl-33999122

ABSTRACT

In the present study, we have used focused ion beam/scanning electron microscopy (FIB/SEM) to perform a study of the synaptic organization of layer III of Brodmann's area 21 in human tissue samples obtained from autopsies and biopsies. We analyzed the synaptic density, 3D spatial distribution, and type (asymmetric/symmetric), as well as the size and shape of each synaptic junction of 4945 synapses that were fully reconstructed in 3D. Significant differences in the mean synaptic density between autopsy and biopsy samples were found (0.49 and 0.66 synapses/µm3, respectively). However, in both types of samples (autopsy and biopsy), the asymmetric:symmetric ratio was similar (93:7) and most asymmetric synapses were established on dendritic spines (75%), while most symmetric synapses were established on dendritic shafts (85%). We also compared several electron microscopy methods and analysis tools to estimate the synaptic density in the same brain tissue. We have shown that FIB/SEM is much more reliable and robust than the majority of the other commonly used EM techniques. The present work constitutes a detailed description of the synaptic organization of cortical layer III. Further studies on the rest of the cortical layers are necessary to better understand the functional organization of this temporal cortical region.


Subject(s)
Neocortex/cytology , Synapses/ultrastructure , Temporal Lobe/cytology , Adult , Autopsy , Biopsy , Cell Count , Dendritic Spines/physiology , Dendritic Spines/ultrastructure , Female , Humans , Imaging, Three-Dimensional , Male , Microscopy, Electron, Scanning , Middle Aged , Neocortex/ultrastructure , Neuroimaging , Temporal Lobe/ultrastructure , Young Adult
12.
Nat Rev Neurosci ; 22(7): 407-422, 2021 07.
Article in English | MEDLINE | ID: mdl-34050339

ABSTRACT

In the brain, most synapses are formed on minute protrusions known as dendritic spines. Unlike their artificial intelligence counterparts, spines are not merely tuneable memory elements: they also embody algorithms that implement the brain's ability to learn from experience and cope with new challenges. Importantly, they exhibit structural dynamics that depend on activity, excitatory input and inhibitory input (synaptic plasticity or 'extrinsic' dynamics) and dynamics independent of activity ('intrinsic' dynamics), both of which are subject to neuromodulatory influences and reinforcers such as dopamine. Here we succinctly review extrinsic and intrinsic dynamics, compare these with parallels in machine learning where they exist, describe the importance of intrinsic dynamics for memory management and adaptation, and speculate on how disruption of extrinsic and intrinsic dynamics may give rise to mental disorders. Throughout, we also highlight algorithmic features of spine dynamics that may be relevant to future artificial intelligence developments.


Subject(s)
Brain/physiology , Dendritic Spines/physiology , Mental Disorders/physiopathology , Models, Neurological , Neural Networks, Computer , Algorithms , Animals , Artificial Intelligence , Brain/cytology , Dendritic Spines/ultrastructure , Dopamine/physiology , Humans , Machine Learning , Memory, Short-Term/physiology , Mental Processes/physiology , Neuronal Plasticity , Neurotransmitter Agents/physiology , Optogenetics , Receptors, Dopamine/physiology , Reward , Species Specificity , Synapses/physiology
13.
Brain Res Bull ; 173: 28-36, 2021 08.
Article in English | MEDLINE | ID: mdl-33984429

ABSTRACT

Plasticity of glutamatergic synapses in the hippocampus is believed to underlie learning and memory processes. Surprisingly, very few studies report long-lasting structural changes of synapses induced by behavioral training. It remains, therefore, unclear which synaptic changes in the hippocampus contribute to memory storage. Here, we systematically compare how long-term potentiation of synaptic transmission (LTP) (a primary form of synaptic plasticity and cellular model of memory) and behavioral training affect hippocampal glutamatergic synapses at the ultrastructural level enabled by electron microscopy. The review of the literature indicates that while LTP induces growth of dendritic spines and post-synaptic densities (PSD), that represent postsynaptic part of a glutamatergic synapse, after behavioral training there is transient (< 6 h) synaptogenesis and long-lasting (> 24 h) increase in PSD volume (without a significant change of dendritic spine volume), indicating that training-induced PSD growth may reflect long-term enhancement of synaptic functions. Additionally, formation of multi-innervated spines (MIS), is associated with long-term memory in aged mice and LTP-deficient mutant mice. Since volume of PSD, as well as atypical synapses, can be reliably observed only with electron microscopy, we argue that the ultrastructural level of analysis is required to reveal synaptic changes that are associated with long-term storage of information in the brain.


Subject(s)
Dendritic Spines/ultrastructure , Hippocampus/ultrastructure , Long-Term Potentiation/physiology , Memory/physiology , Neurons/ultrastructure , Synapses/ultrastructure , Animals , Microscopy, Electron
14.
J Comp Neurol ; 529(11): 3112-3126, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33864263

ABSTRACT

Local translation can provide a rapid, spatially targeted supply of new proteins in distal dendrites to support synaptic changes that underlie learning. Learning and memory are especially sensitive to manipulations of translational control mechanisms, particularly those that target the initiation step, and translation initiation at synapses could be a means of maintaining synapse specificity during plasticity. Initiation predominantly occurs via recruitment of ribosomes to the 5' mRNA cap by complexes of eukaryotic initiation factors (eIFs), and the interaction between eIF4E and eIF4G1 is a particularly important target of translational control pathways. Pharmacological inhibition of eIF4E-eIF4G1 binding impairs formation of memory for aversive Pavlovian conditioning as well as the accompanying increase in polyribosomes in the heads of dendritic spines in the lateral amygdala (LA). This is consistent with a role for initiation at synapses in memory formation, but whether eIFs are even present near synapses is unknown. To determine whether dendritic spines contain eIFs and whether eIF distribution is affected by learning, we combined immunolabeling with serial section transmission electron microscopy (ssTEM) volume reconstructions of LA dendrites after Pavlovian conditioning. Labeling for eIF4E, eIF4G1, and eIF2α-another key target of regulation-occurred in roughly half of dendritic spines, but learning effects were only found for eIF4E, which was upregulated in the heads of dendritic spines. Our results support the possibility of regulated translation initiation as a means of synapse-specific protein targeting during learning and are consistent with the model of eIF4E availability as a central point of control.


Subject(s)
Dendritic Spines/metabolism , Eukaryotic Initiation Factor-4E/biosynthesis , Memory/physiology , Protein Biosynthesis/physiology , Up-Regulation/physiology , Animals , Dendritic Spines/ultrastructure , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factors/biosynthesis , Eukaryotic Initiation Factors/genetics , Male , Rats , Rats, Sprague-Dawley
15.
Neurosci Lett ; 751: 135807, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33705934

ABSTRACT

Reduced cerebellar volume and motor dysfunction have previously been observed in the GFAP-IL6 murine model of chronic neuroinflammation. This study aims to extend these findings by investigating the effect of microglial activation and ageing on the total number of Purkinje cells and the morphology of their dendritic arborization. Through comparison of transgenic GFAP-IL6 mice and their wild-type counterparts at the ages of 12 and 24-months, we were able to investigate the effects of ageing and chronic microglial activation on Purkinje cells. Unbiased stereology was used to estimate the number of microglia in Iba1+ stained tissue and Purkinje cells in calbindin stained tissue. Morphological analyses were made using 3D reconstructions of images acquired from the Golgi-stained cerebellar tissue. We found that the total number of microglia increased by approximately 5 times in the cerebellum of GFAP-IL6 mice compared to their WT littermates. The number of Purkinje cells decreased by as much as 50 % in aged wild type mice and 83 % in aged GFAP-IL6 mice. The remaining Purkinje cells in these cohorts were found to have significant reductions in their total dendritic length and number of branching points, indicating how the complexity of the Purkinje cell dendritic arbor reduces through age and inflammation. GFAP-IL6 mice, when compared to WT mice, had higher levels of microglial activation and more profound neurodegenerative changes in the cerebellum. The presence of constitutive IL6 production, driving chronic neuroinflammation, may account for these neurodegenerative changes in GFAP-IL6 mice.


Subject(s)
Aging/pathology , Glial Fibrillary Acidic Protein/metabolism , Interleukin-6/metabolism , Microglia/metabolism , Purkinje Cells/cytology , Aging/metabolism , Animals , Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , Glial Fibrillary Acidic Protein/genetics , Inflammation/metabolism , Interleukin-6/genetics , Mice , Microglia/cytology , Purkinje Cells/metabolism , Purkinje Cells/pathology
16.
Cereb Cortex ; 31(8): 3592-3609, 2021 07 05.
Article in English | MEDLINE | ID: mdl-33723567

ABSTRACT

Pyramidal neurons are the most abundant and characteristic neuronal type in the cerebral cortex and their dendritic spines are the main postsynaptic elements of cortical excitatory synapses. Previous studies have shown that pyramidal cell structure differs across layers, cortical areas, and species. However, within the human cortex, the pyramidal dendritic morphology has been quantified in detail in relatively few cortical areas. In the present work, we performed intracellular injections of Lucifer Yellow at several distances from the temporal pole. We found regional differences in pyramidal cell morphology, which showed large inter-individual variability in most of the morphological variables measured. However, some values remained similar in all cases. The smallest and least complex cells in the most posterior temporal region showed the greatest dendritic spine density. Neurons in the temporal pole showed the greatest sizes with the highest number of spines. Layer V cells were larger, more complex, and had a greater number of dendritic spines than those in layer III. The present results suggest that, while some aspects of pyramidal structure are conserved, there are specific variations across cortical regions, and species.


Subject(s)
Pyramidal Cells/ultrastructure , Temporal Lobe/ultrastructure , Adult , Dendrites , Dendritic Spines/ultrastructure , Epilepsy/pathology , Epilepsy/surgery , Female , Humans , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Individuality , Male , Middle Aged , Neuroimaging , Neurons/ultrastructure , Temporal Lobe/cytology
17.
Acta Neuropathol Commun ; 9(1): 34, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33648591

ABSTRACT

Cognitive dysfunction occurs in greater than 50% of individuals with multiple sclerosis (MS). Hippocampal demyelination is a prominent feature of postmortem MS brains and hippocampal atrophy correlates with cognitive decline in MS patients. Cellular and molecular mechanisms responsible for neuronal dysfunction in demyelinated hippocampi are not fully understood. Here we investigate a mouse model of hippocampal demyelination where twelve weeks of treatment with the oligodendrocyte toxin, cuprizone, demyelinates over 90% of the hippocampus and causes decreased memory/learning. Long-term potentiation (LTP) of hippocampal CA1 pyramidal neurons is considered to be a major cellular readout of learning and memory in the mammalian brain. In acute slices, we establish that hippocampal demyelination abolishes LTP and excitatory post-synaptic potentials of CA1 neurons, while pre-synaptic function of Schaeffer collateral fibers is preserved. Demyelination also reduced Ca2+-mediated firing of hippocampal neurons in vivo. Using three-dimensional electron microscopy, we investigated the number, shape (mushroom, stubby, thin), and post-synaptic densities (PSDs) of dendritic spines that facilitate LTP. Hippocampal demyelination did not alter the number of dendritic spines. Surprisingly, dendritic spines appeared to be more mature in demyelinated hippocampi, with a significant increase in mushroom-shaped spines, more perforated PSDs, and more astrocyte participation in the tripartite synapse. RNA sequencing experiments identified 400 altered transcripts in demyelinated hippocampi. Gene transcripts that regulate myelination, synaptic signaling, astrocyte function, and innate immunity were altered in demyelinated hippocampi. Hippocampal remyelination rescued synaptic transmission, LTP, and the majority of gene transcript changes. We establish that CA1 neurons projecting demyelinated axons silence their dendritic spines and hibernate in a state that may protect the demyelinated axon and facilitates functional recovery following remyelination.


Subject(s)
Cognitive Dysfunction/physiopathology , Demyelinating Diseases/physiopathology , Dendritic Spines/ultrastructure , Hippocampus/pathology , Hippocampus/physiopathology , Multiple Sclerosis/physiopathology , Neurons/metabolism , Neurons/pathology , Animals , Astrocytes/metabolism , Cognitive Dysfunction/etiology , Cuprizone/administration & dosage , Cuprizone/toxicity , Demyelinating Diseases/diagnostic imaging , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Disease Models, Animal , Long-Term Potentiation , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron , Multiple Sclerosis/diagnostic imaging , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Post-Synaptic Density/metabolism , Sequence Analysis, RNA
18.
J Neurosci ; 41(19): 4172-4186, 2021 05 12.
Article in English | MEDLINE | ID: mdl-33785644

ABSTRACT

Microglia, the resident immune cells of the CNS, have emerged as key regulators of neural precursor cell activity in the adult brain. However, the microglia-derived factors that mediate these effects remain largely unknown. In the present study, we investigated a role for microglial brain-derived neurotrophic factor (BDNF), a neurotrophic factor with well known effects on neuronal survival and plasticity. Surprisingly, we found that selective genetic ablation of BDNF from microglia increased the production of newborn neurons under both physiological and inflammatory conditions (e.g., LPS-induced infection and traumatic brain injury). Genetic ablation of BDNF from microglia otherwise also interfered with self-renewal/proliferation, reducing their overall density. In conclusion, we identify microglial BDNF as an important factor regulating microglia population dynamics and states, which in turn influences neurogenesis under both homeostatic and pathologic conditions.SIGNIFICANCE STATEMENT (1) Microglial BDNF contributes to self-renewal and density of microglia in the brain. (2) Selective ablation of BDNF in microglia stimulates neural precursor proliferation. (3) Loss of microglial BDNF augments working memory following traumatic brain injury. (4) Benefits of repopulating microglia on brain injury are not mediated via microglial BDNF.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Hippocampus/physiology , Microglia/metabolism , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Neurogenesis/genetics , Neurogenesis/physiology , Animals , Cell Proliferation , Cell Survival/genetics , Dendrites/ultrastructure , Dendritic Spines/ultrastructure , Encephalitis/chemically induced , Encephalitis/pathology , Learning/physiology , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/physiology , Neural Stem Cells/ultrastructure
19.
Int J Mol Sci ; 22(3)2021 Jan 26.
Article in English | MEDLINE | ID: mdl-33530380

ABSTRACT

Three-dimensional (3D) reconstruction from electron microscopy (EM) datasets is a widely used tool that has improved our knowledge of synapse ultrastructure and organization in the brain. Rearrangements of synapse structure following maturation and in synaptic plasticity have been broadly described and, in many cases, the defective architecture of the synapse has been associated to functional impairments. It is therefore important, when studying brain connectivity, to map these rearrangements with the highest accuracy possible, considering the affordability of the different EM approaches to provide solid and reliable data about the structure of such a small complex. The aim of this work is to compare quantitative data from two dimensional (2D) and 3D EM of mouse hippocampal CA1 (apical dendrites), to define whether the results from the two approaches are consistent. We examined asymmetric excitatory synapses focusing on post synaptic density and dendritic spine area and volume as well as spine density, and we compared the results obtained with the two methods. The consistency between the 2D and 3D results questions the need-for many applications-of using volumetric datasets (costly and time consuming in terms of both acquisition and analysis), with respect to the more accessible measurements from 2D EM projections.


Subject(s)
CA1 Region, Hippocampal/ultrastructure , Dendritic Spines/ultrastructure , Pyramidal Cells/ultrastructure , Animals , Imaging, Three-Dimensional , Mice , Microscopy, Electron , Synapses/ultrastructure
20.
Sci Rep ; 11(1): 1375, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33446758

ABSTRACT

Dendritic spines are the primary sites of excitatory transmission in the mammalian brain. Spines of cerebellar Purkinje Cells (PCs) are plastic, but they differ from forebrain spines in a number of important respects, and the mechanisms of spine plasticity differ between forebrain and cerebellum. Our previous studies indicate that in hippocampal spines cortactin-a protein that stabilizes actin branch points-resides in the spine core, avoiding the spine shell. To see whether the distribution of cortactin differs in PC spines, we examined its subcellular organization using quantitative preembedding immunoelectron microscopy. We found that cortactin was enriched in the spine shell, associated with the non-synaptic membrane, and was also situated within the postsynaptic density (PSD). This previously unrecognized distribution of cortactin within PC spines may underlie structural and functional differences in excitatory spine synapses between forebrain, and cerebellum.


Subject(s)
Cortactin/metabolism , Dendritic Spines/metabolism , Post-Synaptic Density/metabolism , Purkinje Cells/metabolism , Animals , Dendritic Spines/ultrastructure , Male , Post-Synaptic Density/ultrastructure , Purkinje Cells/ultrastructure , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...