Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.870
Filter
Add more filters

Publication year range
1.
Cell ; 187(4): 962-980.e19, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38309258

ABSTRACT

Microglia (MG), the brain-resident macrophages, play major roles in health and disease via a diversity of cellular states. While embryonic MG display a large heterogeneity of cellular distribution and transcriptomic states, their functions remain poorly characterized. Here, we uncovered a role for MG in the maintenance of structural integrity at two fetal cortical boundaries. At these boundaries between structures that grow in distinct directions, embryonic MG accumulate, display a state resembling post-natal axon-tract-associated microglia (ATM) and prevent the progression of microcavities into large cavitary lesions, in part via a mechanism involving the ATM-factor Spp1. MG and Spp1 furthermore contribute to the rapid repair of lesions, collectively highlighting protective functions that preserve the fetal brain from physiological morphogenetic stress and injury. Our study thus highlights key major roles for embryonic MG and Spp1 in maintaining structural integrity during morphogenesis, with major implications for our understanding of MG functions and brain development.


Subject(s)
Brain , Microglia , Axons , Brain/cytology , Brain/growth & development , Macrophages/physiology , Microglia/pathology , Morphogenesis
2.
Cell ; 186(26): 5766-5783.e25, 2023 12 21.
Article in English | MEDLINE | ID: mdl-38134874

ABSTRACT

The enhanced cognitive abilities characterizing the human species result from specialized features of neurons and circuits. Here, we report that the hominid-specific gene LRRC37B encodes a receptor expressed in human cortical pyramidal neurons (CPNs) and selectively localized to the axon initial segment (AIS), the subcellular compartment triggering action potentials. Ectopic expression of LRRC37B in mouse CPNs in vivo leads to reduced intrinsic excitability, a distinctive feature of some classes of human CPNs. Molecularly, LRRC37B binds to the secreted ligand FGF13A and to the voltage-gated sodium channel (Nav) ß-subunit SCN1B. LRRC37B concentrates inhibitory effects of FGF13A on Nav channel function, thereby reducing excitability, specifically at the AIS level. Electrophysiological recordings in adult human cortical slices reveal lower neuronal excitability in human CPNs expressing LRRC37B. LRRC37B thus acts as a species-specific modifier of human neuron excitability, linking human genome and cell evolution, with important implications for human brain function and diseases.


Subject(s)
Neurons , Pyramidal Cells , Voltage-Gated Sodium Channels , Animals , Humans , Mice , Action Potentials/physiology , Axons/metabolism , Neurons/metabolism , Voltage-Gated Sodium Channels/genetics , Voltage-Gated Sodium Channels/metabolism
3.
Cell ; 184(15): 4048-4063.e32, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34233165

ABSTRACT

Microglia, the resident immune cells of the brain, have emerged as crucial regulators of synaptic refinement and brain wiring. However, whether the remodeling of distinct synapse types during development is mediated by specialized microglia is unknown. Here, we show that GABA-receptive microglia selectively interact with inhibitory cortical synapses during a critical window of mouse postnatal development. GABA initiates a transcriptional synapse remodeling program within these specialized microglia, which in turn sculpt inhibitory connectivity without impacting excitatory synapses. Ablation of GABAB receptors within microglia impairs this process and leads to behavioral abnormalities. These findings demonstrate that brain wiring relies on the selective communication between matched neuronal and glial cell types.


Subject(s)
Microglia/metabolism , Neural Inhibition/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Animals, Newborn , Behavior, Animal , Gene Expression Regulation , HEK293 Cells , Humans , Mice , Parvalbumins/metabolism , Phenotype , Receptors, GABA-B/metabolism , Synapses/physiology , Transcription, Genetic
4.
Cell ; 184(19): 5053-5069.e23, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34390642

ABSTRACT

Genetic perturbations of cortical development can lead to neurodevelopmental disease, including autism spectrum disorder (ASD). To identify genomic regions crucial to corticogenesis, we mapped the activity of gene-regulatory elements generating a single-cell atlas of gene expression and chromatin accessibility both independently and jointly. This revealed waves of gene regulation by key transcription factors (TFs) across a nearly continuous differentiation trajectory, distinguished the expression programs of glial lineages, and identified lineage-determining TFs that exhibited strong correlation between linked gene-regulatory elements and expression levels. These highly connected genes adopted an active chromatin state in early differentiating cells, consistent with lineage commitment. Base-pair-resolution neural network models identified strong cell-type-specific enrichment of noncoding mutations predicted to be disruptive in a cohort of ASD individuals and identified frequently disrupted TF binding sites. This approach illustrates how cell-type-specific mapping can provide insights into the programs governing human development and disease.


Subject(s)
Cerebral Cortex/embryology , Chromatin/metabolism , Gene Expression Regulation, Developmental , Single-Cell Analysis , Astrocytes/cytology , Cell Differentiation , Cell Lineage/genetics , Cluster Analysis , Deep Learning , Epigenesis, Genetic , Fuzzy Logic , Glutamates/metabolism , Humans , Mutation/genetics , Neurons/metabolism , Regulatory Sequences, Nucleic Acid/genetics
5.
Cell ; 183(7): 1913-1929.e26, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33333020

ABSTRACT

Neurons in the cerebral cortex connect through descending pathways to hindbrain and spinal cord to activate muscle and generate movement. Although components of this pathway have been previously generated and studied in vitro, the assembly of this multi-synaptic circuit has not yet been achieved with human cells. Here, we derive organoids resembling the cerebral cortex or the hindbrain/spinal cord and assemble them with human skeletal muscle spheroids to generate 3D cortico-motor assembloids. Using rabies tracing, calcium imaging, and patch-clamp recordings, we show that corticofugal neurons project and connect with spinal spheroids, while spinal-derived motor neurons connect with muscle. Glutamate uncaging or optogenetic stimulation of cortical spheroids triggers robust contraction of 3D muscle, and assembloids are morphologically and functionally intact for up to 10 weeks post-fusion. Together, this system highlights the remarkable self-assembly capacity of 3D cultures to form functional circuits that could be used to understand development and disease.


Subject(s)
Cerebral Cortex/physiology , Motor Cortex/physiology , Organoids/physiology , Animals , Calcium/metabolism , Cell Differentiation , Cells, Cultured , Cervical Vertebrae , Gene Expression Regulation , Glutamates/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Muscles/physiology , Myoblasts/metabolism , Nerve Net/physiology , Optogenetics , Organoids/ultrastructure , Rhombencephalon/physiology , Spheroids, Cellular/cytology , Spinal Cord/cytology
6.
Annu Rev Cell Dev Biol ; 37: 495-517, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34416113

ABSTRACT

With the discovery of the incredible diversity of neurons, Cajal and coworkers laid the foundation of modern neuroscience. Neuron types are not only structural units of nervous systems but also evolutionary units, because their identities are encoded in the genome. With the advent of high-throughput cellular transcriptomics, neuronal identities can be characterized and compared systematically across species. The comparison of neurons in mammals, reptiles, and birds indicates that the mammalian cerebral cortex is a mosaic of deeply conserved and recently evolved neuron types. Using the cerebral cortex as a case study, this review illustrates how comparing neuron types across species is key to reconciling observations on neural development, neuroanatomy, circuit wiring, and physiology for an integrated understanding of brain evolution.


Subject(s)
Biological Evolution , Cerebral Cortex , Animals , Brain/physiology , Cerebral Cortex/anatomy & histology , Mammals , Neurogenesis , Neurons/metabolism
7.
Cell ; 173(6): 1370-1384.e16, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29856955

ABSTRACT

The cerebral cortex underwent rapid expansion and increased complexity during recent hominid evolution. Gene duplications constitute a major evolutionary force, but their impact on human brain development remains unclear. Using tailored RNA sequencing (RNA-seq), we profiled the spatial and temporal expression of hominid-specific duplicated (HS) genes in the human fetal cortex and identified a repertoire of 35 HS genes displaying robust and dynamic patterns during cortical neurogenesis. Among them NOTCH2NL, human-specific paralogs of the NOTCH2 receptor, stood out for their ability to promote cortical progenitor maintenance. NOTCH2NL promote the clonal expansion of human cortical progenitors, ultimately leading to higher neuronal output. At the molecular level, NOTCH2NL function by activating the Notch pathway through inhibition of cis Delta/Notch interactions. Our study uncovers a large repertoire of recently evolved genes active during human corticogenesis and reveals how human-specific NOTCH paralogs may have contributed to the expansion of the human cortex.


Subject(s)
Cerebral Cortex/metabolism , Gene Expression Regulation , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neurogenesis , Neurons/metabolism , Receptor, Notch2/genetics , Amino Acid Sequence , Calcium-Binding Proteins , Cell Differentiation/genetics , Cluster Analysis , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , In Situ Hybridization , Neural Stem Cells/metabolism , Signal Transduction
8.
Annu Rev Neurosci ; 46: 301-320, 2023 07 10.
Article in English | MEDLINE | ID: mdl-37428601

ABSTRACT

Despite increasing evidence of its involvement in several key functions of the cerebral cortex, the vestibular sense rarely enters our consciousness. Indeed, the extent to which these internal signals are incorporated within cortical sensory representation and how they might be relied upon for sensory-driven decision-making, during, for example, spatial navigation, is yet to be understood. Recent novel experimental approaches in rodents have probed both the physiological and behavioral significance of vestibular signals and indicate that their widespread integration with vision improves both the cortical representation and perceptual accuracy of self-motion and orientation. Here, we summarize these recent findings with a focus on cortical circuits involved in visual perception and spatial navigation and highlight the major remaining knowledge gaps. We suggest that vestibulo-visual integration reflects a process of constant updating regarding the status of self-motion, and access to such information by the cortex is used for sensory perception and predictions that may be implemented for rapid, navigation-related decision-making.


Subject(s)
Motion Perception , Vestibule, Labyrinth , Motion Perception/physiology , Cues , Visual Perception/physiology , Vestibule, Labyrinth/physiology , Cerebral Cortex/physiology
9.
Annu Rev Genet ; 55: 555-581, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34535062

ABSTRACT

The cerebral cortex is at the core of brain functions that are thought to be particularly developed in the human species. Human cortex specificities stem from divergent features of corticogenesis, leading to increased cortical size and complexity. Underlying cellular mechanisms include prolonged patterns of neuronal generation and maturation, as well as the amplification of specific types of stem/progenitor cells. While the gene regulatory networks of corticogenesis appear to be largely conserved among all mammals including humans, they have evolved in primates, particularly in the human species, through the emergence of rapidly divergent transcriptional regulatory elements, as well as recently duplicated novel genes. These human-specific molecular features together control key cellular milestones of human corticogenesis and are often affected in neurodevelopmental disorders, thus linking human neural development, evolution, and diseases.


Subject(s)
Cerebral Cortex , Neurogenesis , Animals , Cerebral Cortex/physiology , Gene Regulatory Networks/genetics , Humans , Mammals , Neurogenesis/genetics
10.
Annu Rev Neurosci ; 43: 391-415, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32250724

ABSTRACT

Neural activity and behavior are both notoriously variable, with responses differing widely between repeated presentation of identical stimuli or trials. Recent results in humans and animals reveal that these variations are not random in their nature, but may in fact be due in large part to rapid shifts in neural, cognitive, and behavioral states. Here we review recent advances in the understanding of rapid variations in the waking state, how variations are generated, and how they modulate neural and behavioral responses in both mice and humans. We propose that the brain has an identifiable set of states through which it wanders continuously in a nonrandom fashion, owing to the activity of both ascending modulatory and fast-acting corticocortical and subcortical-cortical neural pathways. These state variations provide the backdrop upon which the brain operates, and understanding them is critical to making progress in revealing the neural mechanisms underlying cognition and behavior.


Subject(s)
Behavior/physiology , Brain/physiology , Nerve Net/physiology , Neural Pathways/physiology , Animals , Cerebral Cortex/physiology , Humans , Neurons/physiology
11.
Mol Cell ; 77(2): 279-293.e8, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31784360

ABSTRACT

The genomes of mammalian neurons contain uniquely high levels of non-CG DNA methylation that can be bound by the Rett syndrome protein, MeCP2, to regulate gene expression. How patterns of non-CG methylation are established in neurons and the mechanism by which this methylation works with MeCP2 to control gene expression is unclear. Here, we find that genes repressed by MeCP2 are often located within megabase-scale regions of high non-CG methylation that correspond with topologically associating domains of chromatin folding. MeCP2 represses enhancers found in these domains that are enriched for non-CG and CG methylation, with the strongest repression occurring for enhancers located within MeCP2-repressed genes. These alterations in enhancer activity provide a mechanism for how MeCP2 disruption in disease can lead to widespread changes in gene expression. Hence, we find that DNA topology can shape non-CG DNA methylation across the genome to dictate MeCP2-mediated enhancer regulation in the brain.


Subject(s)
Chromosomes/genetics , DNA Methylation/genetics , Enhancer Elements, Genetic/genetics , Methyl-CpG-Binding Protein 2/genetics , Repressor Proteins/genetics , Animals , Brain/physiology , Female , Gene Expression Regulation/genetics , Genome/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats
12.
Development ; 151(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38655654

ABSTRACT

Foxg1 masters telencephalic development via a pleiotropic control over its progression. Expressed within the central nervous system (CNS), L1 retrotransposons are implicated in progression of its histogenesis and tuning of its genomic plasticity. Foxg1 represses gene transcription, and L1 elements share putative Foxg1-binding motifs, suggesting the former might limit telencephalic expression (and activity) of the latter. We tested such a prediction, in vivo as well as in engineered primary neural cultures, using loss- and gain-of-function approaches. We found that Foxg1-dependent, transcriptional L1 repression specifically occurs in neopallial neuronogenic progenitors and post-mitotic neurons, where it is supported by specific changes in the L1 epigenetic landscape. Unexpectedly, we discovered that Foxg1 physically interacts with L1-mRNA and positively regulates neonatal neopallium L1-DNA content, antagonizing the retrotranscription-suppressing activity exerted by Mov10 and Ddx39a helicases. To the best of our knowledge, Foxg1 represents the first CNS patterning gene acting as a bimodal retrotransposon modulator, limiting transcription of L1 elements and promoting their amplification, within a specific domain of the developing mouse brain.


Subject(s)
Forkhead Transcription Factors , Gene Expression Regulation, Developmental , Neocortex , Nerve Tissue Proteins , RNA, Messenger , Animals , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Mice , Neocortex/metabolism , Neocortex/embryology , Neocortex/growth & development , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , Retroelements/genetics , DNA/metabolism , DNA/genetics , Neurons/metabolism
13.
EMBO Rep ; 25(7): 3090-3115, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38871984

ABSTRACT

The mammalian neocortex is formed by sequential radial migration of newborn excitatory neurons. Migrating neurons undergo a multipolar-to-bipolar transition at the subplate (SP) layer, where extracellular matrix (ECM) components are abundantly expressed. Here, we investigate the role of the ECM at the SP layer. We show that TGF-ß signaling-related ECM proteins, and their downstream effector, p-smad2/3, are selectively expressed in the SP layer. We also find that migrating neurons express a disintegrin and metalloproteinase with thrombospondin motif 2 (ADAMTS2), an ECM metalloproteinase, just below the SP layer. Knockdown and knockout of Adamts2 suppresses the multipolar-to-bipolar transition of migrating neurons and disturbs radial migration. Time-lapse luminescence imaging of TGF-ß signaling indicates that ADAMTS2 activates this signaling pathway in migrating neurons during the multipolar-to-bipolar transition at the SP layer. Overexpression of TGF-ß2 in migrating neurons partially rescues migration defects in ADAMTS2 knockout mice. Our data suggest that ADAMTS2 secreted by the migrating multipolar neurons activates TGF-ß signaling by ECM remodeling of the SP layer, which might drive the multipolar to bipolar transition.


Subject(s)
ADAMTS Proteins , Cell Movement , Mice, Knockout , Neocortex , Neurons , Signal Transduction , Transforming Growth Factor beta , Animals , Neocortex/metabolism , Neocortex/cytology , ADAMTS Proteins/metabolism , ADAMTS Proteins/genetics , Mice , Transforming Growth Factor beta/metabolism , Neurons/metabolism , Extracellular Matrix/metabolism
14.
Mol Biol Evol ; 41(7)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38913688

ABSTRACT

The outstanding human cognitive capacities are computed in the cerebral cortex, a mammalian-specific brain region and the place of massive biological innovation. Long noncoding RNAs have emerged as gene regulatory elements with higher evolutionary turnover than mRNAs. The many long noncoding RNAs identified in neural tissues make them candidates for molecular sources of cerebral cortex evolution and disease. Here, we characterized the genomic and cellular shifts that occurred during the evolution of the long noncoding RNA repertoire expressed in the developing cerebral cortex and explored putative roles for these long noncoding RNAs in the evolution of the human brain. Using transcriptomics and comparative genomics, we comprehensively annotated the cortical transcriptomes of humans, rhesus macaques, mice, and chickens and classified human cortical long noncoding RNAs into evolutionary groups as a function of their predicted minimal ages. Long noncoding RNA evolutionary groups showed differences in expression levels, splicing efficiencies, transposable element contents, genomic distributions, and transcription factor binding to their promoters. Furthermore, older long noncoding RNAs showed preferential expression in germinative zones, outer radial glial cells, and cortical inhibitory (GABAergic) neurons. In comparison, younger long noncoding RNAs showed preferential expression in cortical excitatory (glutamatergic) neurons, were enriched in primate and human-specific gene co-expression modules, and were dysregulated in neurodevelopmental disorders. These results suggest different evolutionary routes for older and younger cortical long noncoding RNAs, highlighting old long noncoding RNAs as a possible source of molecular evolution of conserved developmental programs; conversely, we propose that the de novo expression of primate- and human-specific young long noncoding RNAs is a putative source of molecular evolution and dysfunction of cortical excitatory neurons, warranting further investigation.


Subject(s)
Cerebral Cortex , Macaca mulatta , Neurons , RNA, Long Noncoding , RNA, Long Noncoding/genetics , Humans , Cerebral Cortex/metabolism , Animals , Mice , Neurons/metabolism , Chickens/genetics , Evolution, Molecular , Transcriptome
15.
EMBO J ; 40(21): e107532, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34549820

ABSTRACT

Astrocytes regulate brain-wide functions and also show region-specific differences, but little is known about how general and region-specific functions are aligned at the single-cell level. To explore this, we isolated adult mouse diencephalic astrocytes by ACSA-2-mediated magnetic-activated cell sorting (MACS). Single-cell RNA-seq revealed 7 gene expression clusters of astrocytes, with 4 forming a supercluster. Within the supercluster, cells differed by gene expression related to ion homeostasis or metabolism, with the former sharing gene expression with other regions and the latter being restricted to specific regions. All clusters showed expression of proliferation-related genes, and proliferation of diencephalic astrocytes was confirmed by immunostaining. Clonal analysis demonstrated low level of astrogenesis in the adult diencephalon, but not in cerebral cortex grey matter. This led to the identification of Smad4 as a key regulator of diencephalic astrocyte in vivo proliferation and in vitro neurosphere formation. Thus, astrocytes show diverse gene expression states related to distinct functions with some subsets being more widespread while others are more regionally restricted. However, all share low-level proliferation revealing the novel concept of adult astrogenesis in the diencephalon.


Subject(s)
Astrocytes/metabolism , Cell Lineage/genetics , Diencephalon/metabolism , Gene Expression Regulation, Developmental , Neurogenesis/genetics , Smad4 Protein/genetics , Animals , Astrocytes/classification , Astrocytes/cytology , Cell Cycle/genetics , Cell Differentiation , Cell Proliferation , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Diencephalon/cytology , Diencephalon/growth & development , Gene Ontology , Gene Regulatory Networks , Gray Matter/cytology , Gray Matter/growth & development , Gray Matter/metabolism , Metabolic Networks and Pathways , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Annotation , Multigene Family , Signal Transduction , Smad4 Protein/metabolism
16.
Annu Rev Neurosci ; 40: 629-652, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28661727

ABSTRACT

The cerebral cortex is the source of our most complex cognitive capabilities and a vulnerable target of many neurological and neuropsychiatric disorders. Transcriptomics offers a new approach to understanding the cortex at the level of its underlying genetic code, and rapid technological advances have propelled this field to the high-throughput study of the complete set of transcribed genes at increasingly fine resolution to the level of individual cells. These tools have revealed features of the genetic architecture of adult cortical areas, layers, and cell types, as well as spatiotemporal patterning during development. This has allowed a fresh look at comparative anatomy as well, illustrating surprisingly large differences between mammals while at the same time revealing conservation of some features from avians to mammals. Finally, transcriptomics is fueling progress in understanding the causes of neurodevelopmental diseases such as autism, linking genetic association studies to specific molecular pathways and affected brain regions.


Subject(s)
Autism Spectrum Disorder/genetics , Cerebral Cortex/pathology , Transcriptome , Animals , Autism Spectrum Disorder/pathology , Biological Evolution , Cerebral Cortex/physiopathology , Genetic Association Studies , Humans , Species Specificity
17.
Cereb Cortex ; 34(1)2024 01 14.
Article in English | MEDLINE | ID: mdl-38037843

ABSTRACT

Human brain structure shows heterogeneous patterns of change across adults aging and is associated with cognition. However, the relationship between cortical structural changes during aging and gene transcription signatures remains unclear. Here, using structural magnetic resonance imaging data of two separate cohorts of healthy participants from the Cambridge Centre for Aging and Neuroscience (n = 454, 18-87 years) and Dallas Lifespan Brain Study (n = 304, 20-89 years) and a transcriptome dataset, we investigated the link between cortical morphometric similarity network and brain-wide gene transcription. In two cohorts, we found reproducible morphometric similarity network change patterns of decreased morphological similarity with age in cognitive related areas (mainly located in superior frontal and temporal cortices), and increased morphological similarity in sensorimotor related areas (postcentral and lateral occipital cortices). Changes in morphometric similarity network showed significant spatial correlation with the expression of age-related genes that enriched to synaptic-related biological processes, synaptic abnormalities likely accounting for cognitive decline. Transcription changes in astrocytes, microglia, and neuronal cells interpreted most of the age-related morphometric similarity network changes, which suggest potential intervention and therapeutic targets for cognitive decline. Taken together, by linking gene transcription signatures to cortical morphometric similarity network, our findings might provide molecular and cellular substrates for cortical structural changes related to cognitive decline across adults aging.


Subject(s)
Aging , Brain , Adult , Humans , Brain/physiology , Aging/physiology , Cognition/physiology , Temporal Lobe , Magnetic Resonance Imaging/methods
18.
Cereb Cortex ; 34(4)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38610088

ABSTRACT

The axons of neocortical pyramidal neurons are frequently myelinated. Heterogeneity in the topography of axonal myelination in the cerebral cortex has been attributed to a combination of electrophysiological activity, axonal morphology, and neuronal-glial interactions. Previously, we showed that axonal segment length and caliber are critical local determinants of fast-spiking interneuron myelination. However, the factors that determine the myelination of individual axonal segments along neocortical pyramidal neurons remain largely unexplored. Here, we used structured illumination microscopy to examine the extent to which axonal morphology is predictive of the topography of myelination along neocortical pyramidal neurons. We identified critical thresholds for axonal caliber and interbranch distance that are necessary, but not sufficient, for myelination of pyramidal cell axons in mouse primary somatosensory cortex (S1). Specifically, we found that pyramidal neuron axonal segments with a caliber < 0.24 µm or interbranch distance < 18.10 µm are rarely myelinated. Moreover, we further confirmed that these findings in mice are similar for human neocortical pyramidal cell myelination (caliber < 0.25 µm, interbranch distance < 19.00 µm), suggesting that this mechanism is evolutionarily conserved. Taken together, our findings suggest that axonal morphology is a critical correlate of the topography and cell-type specificity of neocortical myelination.


Subject(s)
Neocortex , Pyramidal Cells , Humans , Animals , Mice , Axons , Myelin Sheath , Interneurons
19.
Cereb Cortex ; 34(2)2024 01 31.
Article in English | MEDLINE | ID: mdl-38385890

ABSTRACT

Epidemiologic studies suggest that prenatal exposures to certain viruses may influence early neurodevelopment, predisposing offspring to neuropsychiatric conditions later in life. The long-term effects of maternal COVID-19 infection in pregnancy on early brain development, however, remain largely unknown. We prospectively enrolled infants in an observational cohort study for a single-site study in the Washington, DC Metropolitan Area from June 2020 to November 2021 and compared these infants to pre-pandemic controls (studied March 2014-February 2020). The primary outcomes are measures of cortical morphometry (tissue-specific volumes), along with global and regional measures of local gyrification index, and sulcal depth. We studied 210 infants (55 infants of COVID-19 unexposed mothers, 47 infants of COVID-19-positive mothers, and 108 pre-pandemic healthy controls). We found increased cortical gray matter volume (182.45 ± 4.81 vs. 167.29 ± 2.92) and accelerated sulcal depth of the frontal lobe (5.01 ± 0.19 vs. 4.40 ± 0.13) in infants of COVID-19-positive mothers compared to controls. We found additional differences in infants of COVID-19 unexposed mothers, suggesting both maternal viral exposures, as well as non-viral stressors associated with the pandemic, may influence early development and warrant ongoing follow-up.


Subject(s)
COVID-19 , Infant , Infant, Newborn , Female , Pregnancy , Humans , SARS-CoV-2 , Brain/diagnostic imaging , Gray Matter , Mothers
20.
Cereb Cortex ; 34(2)2024 01 31.
Article in English | MEDLINE | ID: mdl-38216525

ABSTRACT

Observational studies have reported that osteoporosis is associated with cortical changes in the brain. However, the inherent limitations of observational studies pose challenges in eliminating confounding factors and establishing causal relationships. And previous observational studies have not reported changes in specific brain regions. By employing Mendelian randomization, we have been able to infer a causal relationship between osteoporosis and a reduction in the surficial area (SA) of the brain cortical. This effect is partially mediated by vascular calcification. We found that osteoporosis significantly decreased the SA of global brain cortical (ß = -1587.62 mm2, 95%CI: -2645.94 mm2 to -529.32 mm2, P = 0.003) as well as the paracentral gyrus without global weighted (ß = - 19.42 mm2, 95%CI: -28.90 mm2 to -9.95 mm2, P = 5.85 × 10-5). Furthermore, we estimated that 42.25% and 47.21% of the aforementioned effects are mediated through vascular calcification, respectively. Osteoporosis leads to a reduction in the SA of the brain cortical, suggesting the presence of the bone-brain axis. Vascular calcification plays a role in mediating this process to a certain extent. These findings establish a theoretical foundation for further investigations into the intricate interplay between bone, blood vessels, and the brain.


Subject(s)
Osteoporosis , Vascular Calcification , Humans , Mendelian Randomization Analysis , Brain/diagnostic imaging , Osteoporosis/diagnostic imaging , Osteoporosis/genetics , Genome-Wide Association Study , Polymorphism, Single Nucleotide
SELECTION OF CITATIONS
SEARCH DETAIL