Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.860
Filter
1.
BMC Genomics ; 25(1): 757, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095712

ABSTRACT

BACKGROUND: It is known that the neurodevelopmental disorder associated gene, Satb2, plays important roles in determining the upper layer neuron specification. However, it is not well known how this gene regulates other neocortical regions during the development. It is also lack of comprehensive delineation of its spatially regulatory pathways in neocortical development. RESULTS: In this work, we utilized spatial transcriptomics and immuno-staining to systematically investigate the region-specific gene regulation of Satb2 by comparing the Satb2+/+ and Satb2-/- mice at embryonic stages, including the ventricle zone (VZ) or subventricle zone (SVZ), intermediate zone (IZ) and cortical plate (CP) respectively. The staining result reveals that these three regions become moderately or significantly thinner in the Satb2-/- mice. In the cellular level, the cell number increases in the VZ/SVZ, whereas the cell number decreases in the CP. The spatial transcriptomics data show that many important genes and relevant pathways are dysregulated in Satb2-/- mice in a region-specific manner. In the VZ/SVZ, the key genes involved in neural precursor cell proliferation, including the intermediate progenitor marker Tbr2 and the lactate production related gene Ldha, are up-regulated in Satb2-/- mice. In the IZ, the key genes in regulating neuronal differentiation and migration, such as Rnd2, exhibit ectopic expressions in the Satb2-/- mice. In the CP, the lineage-specific genes, Tbr1 and Bcl11b, are abnormally expressed. The neuropeptide related gene Npy is down-regulated in Satb2-/- mice. Finally, we validated the abnormal expressions of key regulators by using immunofluorescence or qPCR. CONCLUSIONS: In summary, our work provides insights on the region-specific genes and pathways which are regulated by Satb2 in neocortical development.


Subject(s)
Gene Expression Regulation, Developmental , Matrix Attachment Region Binding Proteins , Neocortex , Transcription Factors , Transcriptome , Animals , Neocortex/metabolism , Neocortex/growth & development , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism , Mice , Transcription Factors/genetics , Transcription Factors/metabolism , Gene Expression Profiling , Mice, Knockout , Repressor Proteins , Tumor Suppressor Proteins
2.
Commun Biol ; 7(1): 885, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39033173

ABSTRACT

Rhythmic brain activity is critical to many brain functions and is sensitive to neuromodulation, but so far very few studies have investigated this activity on the cellular level in vitro in human brain tissue samples. This study reveals and characterizes a novel rhythmic network activity in the human neocortex. Using intracellular patch-clamp recordings of human cortical neurons, we identify large rhythmic depolarizations (LRDs) driven by glutamate release but not by GABA. These LRDs are intricate events made up of multiple depolarizing phases, occurring at ~0.3 Hz, have large amplitudes and long decay times. Unlike human tissue, rat neocortex layers 2/3 exhibit no such activity under identical conditions. LRDs are mainly observed in a subset of L2/3 interneurons that receive substantial excitatory inputs and are likely large basket cells based on their morphology. LRDs are highly sensitive to norepinephrine (NE) and acetylcholine (ACh), two neuromodulators that affect network dynamics. NE increases LRD frequency through ß-adrenergic receptor activity while ACh decreases it via M4 muscarinic receptor activation. Multi-electrode array recordings show that NE enhances and synchronizes oscillatory network activity, whereas ACh causes desynchronization. Thus, NE and ACh distinctly modulate LRDs, exerting specific control over human neocortical activity.


Subject(s)
Acetylcholine , Neocortex , Norepinephrine , Humans , Acetylcholine/pharmacology , Norepinephrine/pharmacology , Neocortex/physiology , Neocortex/metabolism , Neocortex/cytology , Neocortex/drug effects , Male , Female , Animals , Middle Aged , Rats , Aged , Periodicity , Neurons/physiology , Neurons/drug effects , Neurons/metabolism , Interneurons/physiology , Interneurons/drug effects , Interneurons/metabolism , Adult
3.
Elife ; 122024 Jul 08.
Article in English | MEDLINE | ID: mdl-38976495

ABSTRACT

Mammals have evolved sex-specific adaptations to reduce energy usage in times of food scarcity. These adaptations are well described for peripheral tissue, though much less is known about how the energy-expensive brain adapts to food restriction, and how such adaptations differ across the sexes. Here, we examined how food restriction impacts energy usage and function in the primary visual cortex (V1) of adult male and female mice. Molecular analysis and RNA sequencing in V1 revealed that in males, but not in females, food restriction significantly modulated canonical, energy-regulating pathways, including pathways associated waith AMP-activated protein kinase, peroxisome proliferator-activated receptor alpha, mammalian target of rapamycin, and oxidative phosphorylation. Moreover, we found that in contrast to males, food restriction in females did not significantly affect V1 ATP usage or visual coding precision (assessed by orientation selectivity). Decreased serum leptin is known to be necessary for triggering energy-saving changes in V1 during food restriction. Consistent with this, we found significantly decreased serum leptin in food-restricted males but no significant change in food-restricted females. Collectively, our findings demonstrate that cortical function and energy usage in female mice are more resilient to food restriction than in males. The neocortex, therefore, contributes to sex-specific, energy-saving adaptations in response to food restriction.


Subject(s)
Energy Metabolism , Neocortex , Animals , Female , Male , Neocortex/physiology , Neocortex/metabolism , Mice , Visual Cortex/physiology , Visual Cortex/metabolism , Sex Factors , Food Deprivation/physiology , Mice, Inbred C57BL , Sex Characteristics , Leptin/metabolism , Leptin/blood , Adaptation, Physiological , Caloric Restriction
4.
Nat Commun ; 15(1): 6337, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39068160

ABSTRACT

Neuronal anatomy is central to the organization and function of brain cell types. However, anatomical variability within apparently homogeneous populations of cells can obscure such insights. Here, we report large-scale automation of neuronal morphology reconstruction and analysis on a dataset of 813 inhibitory neurons characterized using the Patch-seq method, which enables measurement of multiple properties from individual neurons, including local morphology and transcriptional signature. We demonstrate that these automated reconstructions can be used in the same manner as manual reconstructions to understand the relationship between some, but not all, cellular properties used to define cell types. We uncover gene expression correlates of laminar innervation on multiple transcriptomically defined neuronal subclasses and types. In particular, our results reveal correlates of the variability in Layer 1 (L1) axonal innervation in a transcriptomically defined subpopulation of Martinotti cells in the adult mouse neocortex.


Subject(s)
Axons , Dendrites , Neocortex , Transcriptome , Animals , Axons/metabolism , Mice , Dendrites/metabolism , Neocortex/cytology , Neocortex/metabolism , Neuroanatomy/methods , Neurons/metabolism , Neurons/cytology , Male , Gene Expression Profiling/methods , Mice, Inbred C57BL
5.
Med Gas Res ; 14(3): 115-120, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39073339

ABSTRACT

Sevoflurane has been shown to increase the incidence of emergence delirium in children; however, the mechanism remains unclear. Sevoflurane increases cytoplasmic calcium concentration which in turn may play a role in emergence delirium. This study aimed to investigate the level of intracellular calcium in rats experiencing hyperexcitatory behavior after exposure to sevoflurane, as well as the role of magnesium in preventing this phenomenon. After ethical approval, 2-5-week-old Sprague-Dawley rats (n = 34) were insufflated with sevoflurane in a modified anesthesia chamber. One group received magnesium sulphate intraperitoneally. After termination of sevoflurane exposure, the occurrence of hyperexcitation was observed. Brain tissue samples from the rats were studied for intracellular calcium levels under a two-channel laser scanning confocal microscope and were quantitatively calculated using ratiometric calculation. The presence of inflammation or oxidative stress reaction was assessed using nuclear factor κB and malondialdehyde. The incidence of hyperexcitatory behavior post sevoflurane exposure was 9 in 16 rats in the observation group and none in the magnesium group. Tests for inflammation and oxidative stress were within normal limits in both groups. The rats showing hyperexcitation had a higher level of cytosol calcium concentration compared to the other groups. To conclude, the calcium concentration of neocortical neurons in Sprague-Dawley rats with hyperexcitatory behavior is increased after exposure to sevoflurane. Administration of magnesium sulphate can prevent the occurrence of hyperexcitation in experimental animals.


Subject(s)
Calcium , Neocortex , Neurons , Rats, Sprague-Dawley , Sevoflurane , Animals , Sevoflurane/pharmacology , Sevoflurane/adverse effects , Calcium/metabolism , Neurons/drug effects , Neurons/metabolism , Rats , Neocortex/drug effects , Neocortex/metabolism , Male , Anesthetics, Inhalation/pharmacology , Anesthetics, Inhalation/adverse effects , Methyl Ethers/pharmacology , Methyl Ethers/adverse effects , Behavior, Animal/drug effects , Oxidative Stress/drug effects
6.
Neurobiol Aging ; 141: 160-170, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38964013

ABSTRACT

Women have a higher incidence of Alzheimer's disease (AD), even after adjusting for increased longevity. Thus, there is an urgent need to identify genes that underpin sex-associated risk of AD. PIN1 is a key regulator of the tau phosphorylation signaling pathway; however, potential differences in PIN1 expression, in males and females, are still unknown. We analyzed brain transcriptomic datasets focusing on sex differences in PIN1 mRNA levels in an aging and AD cohort, which revealed reduced PIN1 levels primarily within females. We validated this observation in an independent dataset (ROS/MAP), which also revealed that PIN1 is negatively correlated with multiregional neurofibrillary tangle density and global cognitive function in females only. Additional analysis revealed a decrease in PIN1 in subjects with mild cognitive impairment (MCI) compared with aged individuals, again driven predominantly by female subjects. Histochemical analysis of PIN1 in AD and control male and female neocortex revealed an overall decrease in axonal PIN1 protein levels in females. These findings emphasize the importance of considering sex differences in AD research.


Subject(s)
Alzheimer Disease , Cognition , Cognitive Dysfunction , NIMA-Interacting Peptidylprolyl Isomerase , Neocortex , Neurofibrillary Tangles , Sex Characteristics , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/genetics , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Humans , Female , Neocortex/pathology , Neocortex/metabolism , Male , Cognitive Dysfunction/genetics , Cognitive Dysfunction/pathology , Cognitive Dysfunction/metabolism , Aged , Aged, 80 and over , Neurofibrillary Tangles/pathology , Neurofibrillary Tangles/metabolism , Phenotype , Limbic System/pathology , Limbic System/metabolism , Gene Expression , Aging/pathology , Aging/genetics , Aging/metabolism , RNA, Messenger/metabolism , RNA, Messenger/genetics , tau Proteins/metabolism , tau Proteins/genetics , Phosphorylation
7.
J Comp Neurol ; 532(6): e25630, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38852043

ABSTRACT

Mitochondria play critical roles in neural stem/progenitor cell proliferation and fate decisions. The subcellular localization of mitochondria in neural stem/progenitor cells during mitosis potentially influences the distribution of mitochondria to the daughter cells and thus their fates. Therefore, understanding the spatial dynamics of mitochondria provides important knowledge about brain development. In this study, we analyzed the subcellular localization of mitochondria in the fetal human neocortex with a particular focus on the basal radial glial cells (bRGCs), a neural stem/progenitor cell subtype attributed to the evolutionary expansion of the human neocortex. During interphase, bRGCs exhibit a polarized localization of mitochondria that is localized at the base of the process or the proximal part of the process. Thereafter, mitochondria in bRGCs at metaphase show unpolarized distribution in which the mitochondria are randomly localized in the cytoplasm. During anaphase and telophase, mitochondria are still localized evenly, but mainly in the periphery of the cytoplasm. Mitochondria start to accumulate at the cleavage furrow during cytokinesis. These results suggest that the mitochondrial localization in bRGCs is tightly regulated during the cell cycle, which may ensure the proper distribution of mitochondria to the daughter cells and, thus in turn, influence their fates.


Subject(s)
Cell Cycle , Ependymoglial Cells , Mitochondria , Neocortex , Humans , Neocortex/cytology , Neocortex/metabolism , Mitochondria/metabolism , Cell Cycle/physiology , Ependymoglial Cells/metabolism , Ependymoglial Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/cytology
8.
eNeuro ; 11(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38886064

ABSTRACT

Preparing acute brain slices produces trauma that mimics severe penetrating brain injury. In neonatal acute brain slices, the spatiotemporal characteristics of trauma-induced calcium dynamics in neurons and its effect on network activity are relatively unknown. Using multiphoton laser scanning microscopy of the somatosensory neocortex in acute neonatal mouse brain slices (P8-12), we simultaneously imaged neuronal Ca2+ dynamics (GCaMP6s) and cytotoxicity (propidium iodide or PI) to determine the relationship between cytotoxic Ca2+ loaded neurons (GCaMP-filled) and cell viability at different depths and incubation times. PI+ cells and GCaMP-filled neurons were abundant at the surface of the slices, with an exponential decrease with depth. Regions with high PI+ cells correlated with elevated neuronal and neuropil Ca2+ The number of PI+ cells and GCaMP-filled neurons increased with prolonged incubation. GCaMP-filled neurons did not participate in stimulus-evoked or seizure-evoked network activity. Significantly, the superficial tissue, with a higher degree of trauma-induced injury, showed attenuated seizure-related neuronal Ca2+ responses. Calpain inhibition prevented the increase in PI+ cells and GCaMP-filled neurons in the deep tissue and during prolonged incubation times. Isoform-specific pharmacological inhibition implicated calpain-2 as a significant contributor to trauma-induced injury in acute slices. Our results show a calpain-mediated spatiotemporal relationship between cell death and aberrant neuronal Ca2+ load in acute neonatal brain slices. Also, we demonstrate that neurons in acute brain slices exhibit altered physiology depending on the degree of trauma-induced injury. Blocking calpains may be a therapeutic option to prevent acute neuronal death during traumatic brain injury in the young brain.


Subject(s)
Animals, Newborn , Calcium , Calpain , Cell Death , Neurons , Animals , Calpain/metabolism , Cell Death/physiology , Neurons/metabolism , Calcium/metabolism , Mice , Mice, Inbred C57BL , Female , Male , Neocortex/metabolism
9.
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
10.
Nat Commun ; 15(1): 4879, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849354

ABSTRACT

The mammalian neocortex comprises an enormous diversity regarding cell types, morphology, and connectivity. In this work, we discover a post-transcriptional mechanism of gene expression regulation, protein translation, as a determinant of cortical neuron identity. We find specific upregulation of protein synthesis in the progenitors of later-born neurons and show that translation rates and concomitantly protein half-lives are inherent features of cortical neuron subtypes. In a small molecule screening, we identify Ire1α as a regulator of Satb2 expression and neuronal polarity. In the developing brain, Ire1α regulates global translation rates, coordinates ribosome traffic, and the expression of eIF4A1. Furthermore, we demonstrate that the Satb2 mRNA translation requires eIF4A1 helicase activity towards its 5'-untranslated region. Altogether, we show that cortical neuron diversity is generated by mechanisms operating beyond gene transcription, with Ire1α-safeguarded proteostasis serving as an essential regulator of brain development.


Subject(s)
Matrix Attachment Region Binding Proteins , Neocortex , Neurons , Protein Biosynthesis , Protein Serine-Threonine Kinases , Animals , Neocortex/metabolism , Neocortex/cytology , Neocortex/embryology , Neurons/metabolism , Neurons/cytology , Mice , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Matrix Attachment Region Binding Proteins/metabolism , Matrix Attachment Region Binding Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Gene Expression Regulation, Developmental , Proteostasis , Neurogenesis/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , 5' Untranslated Regions/genetics , Ribosomes/metabolism , Ribosomes/genetics , Humans , Endoribonucleases/metabolism , Endoribonucleases/genetics , Cell Differentiation/genetics
11.
Science ; 384(6698): eadh7688, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38781356

ABSTRACT

RNA splicing is highly prevalent in the brain and has strong links to neuropsychiatric disorders; yet, the role of cell type-specific splicing and transcript-isoform diversity during human brain development has not been systematically investigated. In this work, we leveraged single-molecule long-read sequencing to deeply profile the full-length transcriptome of the germinal zone and cortical plate regions of the developing human neocortex at tissue and single-cell resolution. We identified 214,516 distinct isoforms, of which 72.6% were novel (not previously annotated in Gencode version 33), and uncovered a substantial contribution of transcript-isoform diversity-regulated by RNA binding proteins-in defining cellular identity in the developing neocortex. We leveraged this comprehensive isoform-centric gene annotation to reprioritize thousands of rare de novo risk variants and elucidate genetic risk mechanisms for neuropsychiatric disorders.


Subject(s)
Mental Disorders , Neocortex , Neurogenesis , Protein Isoforms , RNA Splicing , Single-Cell Analysis , Transcriptome , Humans , Alternative Splicing , Genetic Predisposition to Disease , Mental Disorders/genetics , Molecular Sequence Annotation , Neocortex/metabolism , Neocortex/embryology , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Neurogenesis/genetics
12.
J Nucl Med ; 65(7): 1122-1128, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38782458

ABSTRACT

The widespread deposition of amyloid-ß (Aß) plaques in late-stage Alzheimer disease is well defined and confirmed by in vivo PET. However, there are discrepancies between which regions contribute to the earliest topographic Aß deposition within the neocortex. Methods: This study investigated Aß signals in the perithreshold SUV ratio range using Pittsburgh compound B (PiB) PET in a population-based study cross-sectionally and longitudinally. PiB PET scans from 1,088 participants determined the early patterns of PiB loading in the neocortex. Results: Early-stage Aß loading is seen first in the temporal, cingulate, and occipital regions. Regional early deposition patterns are similar in both apolipoprotein ε4 carriers and noncarriers. Clustering analysis shows groups with different patterns of early amyloid deposition. Conclusion: These findings of initial Aß deposition patterns may be of significance for diagnostics and understanding the development of Alzheimer disease phenotypes.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Neocortex , Positron-Emission Tomography , Thiazoles , Humans , Neocortex/diagnostic imaging , Neocortex/metabolism , Amyloid beta-Peptides/metabolism , Male , Female , Aged , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Aniline Compounds , Middle Aged , Aged, 80 and over , Cross-Sectional Studies , Longitudinal Studies , Radiopharmaceuticals
13.
Cell Rep ; 43(5): 114212, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38743567

ABSTRACT

Diverse types of inhibitory interneurons (INs) impart computational power and flexibility to neocortical circuits. Whereas markers for different IN types in cortical layers 2-6 (L2-L6) have been instrumental for generating a wealth of functional insights, only the recent identification of a selective marker (neuron-derived neurotrophic factor [NDNF]) has opened comparable opportunities for INs in L1 (L1INs). However, at present we know very little about the connectivity of NDNF L1INs with other IN types, their input-output conversion, and the existence of potential NDNF L1IN subtypes. Here, we report pervasive inhibition of L2/3 INs (including parvalbumin INs and vasoactive intestinal peptide INs) by NDNF L1INs. Intersectional genetics revealed similar physiology and connectivity in the NDNF L1IN subpopulation co-expressing neuropeptide Y. Finally, NDNF L1INs prominently and selectively engage in persistent firing, a physiological hallmark disconnecting their output from the current input. Collectively, our work therefore identifies NDNF L1INs as specialized master regulators of superficial neocortex according to their pervasive top-down afferents.


Subject(s)
Interneurons , Interneurons/metabolism , Animals , Mice , Neuropeptide Y/metabolism , Neocortex/metabolism , Neocortex/cytology , Neocortex/physiology , Vasoactive Intestinal Peptide/metabolism , Male , Parvalbumins/metabolism
14.
Neurology ; 102(12): e209447, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38810211

ABSTRACT

BACKGROUND AND OBJECTIVES: Self-reported cognitive decline is an early behavioral manifestation of Alzheimer disease (AD) at the preclinical stage, often believed to precede concerns reported by a study partner. Previous work shows cross-sectional associations with ß-amyloid (Aß) status and self-reported and study partner-reported cognitive decline, but less is known about their associations with tau deposition, particularly among those with preclinical AD. METHODS: This cross-sectional study included participants from the Anti-Amyloid Treatment in Asymptomatic AD/Longitudinal Evaluation of Amyloid Risk and Neurodegeneration studies (N = 444) and the Harvard Aging Brain Study and affiliated studies (N = 231), which resulted in a cognitively unimpaired (CU) sample of individuals with both nonelevated (Aß-) and elevated Aß (Aß+). All participants and study partners completed the Cognitive Function Index (CFI). Two regional tau composites were derived by averaging flortaucipir PET uptake in the medial temporal lobe (MTL) and neocortex (NEO). Global Aß PET was measured in Centiloids (CLs) with Aß+ >26 CL. We conducted multiple linear regression analyses to test associations between tau PET and CFI, covarying for amyloid, age, sex, education, and cohort. We also controlled for objective cognitive performance, measured using the Preclinical Alzheimer Cognitive Composite (PACC). RESULTS: Across 675 CU participants (age = 72.3 ± 6.6 years, female = 59%, Aß+ = 60%), greater tau was associated with greater self-CFI (MTL: ß = 0.28 [0.12, 0.44], p < 0.001, and NEO: ß = 0.26 [0.09, 0.42], p = 0.002) and study partner CFI (MTL: ß = 0.28 [0.14, 0.41], p < 0.001, and NEO: ß = 0.31 [0.17, 0.44], p < 0.001). Significant associations between both CFI measures and MTL/NEO tau PET were driven by Aß+. Continuous Aß showed an independent effect on CFI in addition to MTL and NEO tau for both self-CFI and study partner CFI. Self-CFI (ß = 0.01 [0.001, 0.02], p = 0.03), study partner CFI (ß = 0.01 [0.003, 0.02], p = 0.01), and the PACC (ß = -0.02 [-0.03, -0.01], p < 0.001) were independently associated with MTL tau, but for NEO tau, PACC (ß = -0.02 [-0.03, -0.01], p < 0.001) and study partner report (ß = 0.01 [0.004, 0.02], p = 0.002) were associated, but not self-CFI (ß = 0.01 [-0.001, 0.02], p = 0.10). DISCUSSION: Both self-report and study partner report showed associations with tau in addition to Aß. Additionally, self-report and study partner report were associated with tau above and beyond performance on a neuropsychological composite. Stratification analyses by Aß status indicate that associations between self-reported and study partner-reported cognitive concerns with regional tau are driven by those at the preclinical stage of AD, suggesting that both are useful to collect on the early AD continuum.


Subject(s)
Amyloid beta-Peptides , Cognitive Dysfunction , Positron-Emission Tomography , tau Proteins , Humans , Female , Male , Aged , tau Proteins/metabolism , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/etiology , Cross-Sectional Studies , Aged, 80 and over , Amyloid beta-Peptides/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/diagnostic imaging , Self Report , Cohort Studies , Temporal Lobe/metabolism , Temporal Lobe/diagnostic imaging , Middle Aged , Neocortex/metabolism , Neocortex/diagnostic imaging
15.
Biomolecules ; 14(5)2024 May 17.
Article in English | MEDLINE | ID: mdl-38786001

ABSTRACT

During the first and second stages of postnatal development, neocortical neurons exhibit a wide range of spontaneous synchronous activity (SSA). Towards the end of the second postnatal week, the SSA is replaced by a more sparse and desynchronized firing pattern. The developmental desynchronization of neocortical spontaneous neuronal activity is thought to be intrinsically generated, since sensory deprivation from the periphery does not affect the time course of this transition. The extracellular protein reelin controls various aspects of neuronal development through multimodular signaling. However, so far it is unclear whether reelin contributes to the developmental desynchronization transition of neocortical neurons. The present study aims to investigate the role of reelin in postnatal cortical developmental desynchronization using a conditional reelin knockout (RelncKO) mouse model. Conditional reelin deficiency was induced during early postnatal development, and Ca2+ recordings were conducted from organotypic cultures (OTCs) of the somatosensory cortex. Our results show that both wild type (wt) and RelncKO exhibited an SSA pattern during the early postnatal week. However, at the end of the second postnatal week, wt OTCs underwent a transition to a desynchronized network activity pattern, while RelncKO activity remained synchronous. This changing activity pattern suggests that reelin is involved in regulating the developmental desynchronization of cortical neuronal network activity. Moreover, the developmental desynchronization impairment observed in RelncKO was rescued when RelncKO OTCs were co-cultured with wt OTCs. Finally, we show that the developmental transition to a desynchronized state at the end of the second postnatal week is not dependent on glutamatergic signaling. Instead, the transition is dependent on GABAAR and GABABR signaling. The results suggest that reelin controls developmental desynchronization through GABAAR and GABABR signaling.


Subject(s)
Extracellular Matrix Proteins , Mice, Knockout , Neocortex , Nerve Tissue Proteins , Reelin Protein , Serine Endopeptidases , Animals , Mice , Neocortex/metabolism , Neocortex/growth & development , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Serine Endopeptidases/metabolism , Serine Endopeptidases/genetics , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Neurons/metabolism , Nerve Net/metabolism , Nerve Net/growth & development , Somatosensory Cortex/metabolism , Somatosensory Cortex/growth & development
16.
Nature ; 629(8011): 402-409, 2024 May.
Article in English | MEDLINE | ID: mdl-38632412

ABSTRACT

Throughout life, neuronal networks in the mammalian neocortex maintain a balance of excitation and inhibition, which is essential for neuronal computation1,2. Deviations from a balanced state have been linked to neurodevelopmental disorders, and severe disruptions result in epilepsy3-5. To maintain balance, neuronal microcircuits composed of excitatory and inhibitory neurons sense alterations in neural activity and adjust neuronal connectivity and function. Here we identify a signalling pathway in the adult mouse neocortex that is activated in response to increased neuronal network activity. Overactivation of excitatory neurons is signalled to the network through an increase in the levels of BMP2, a growth factor that is well known for its role as a morphogen in embryonic development. BMP2 acts on parvalbumin-expressing (PV) interneurons through the transcription factor SMAD1, which controls an array of glutamatergic synapse proteins and components of perineuronal nets. PV-interneuron-specific disruption of BMP2-SMAD1 signalling is accompanied by a loss of glutamatergic innervation in PV cells, underdeveloped perineuronal nets and decreased excitability. Ultimately, this impairment of the functional recruitment of PV interneurons disrupts the cortical excitation-inhibition balance, with mice exhibiting spontaneous epileptic seizures. Our findings suggest that developmental morphogen signalling is repurposed to stabilize cortical networks in the adult mammalian brain.


Subject(s)
Bone Morphogenetic Protein 2 , Interneurons , Neocortex , Nerve Net , Neural Inhibition , Neurons , Signal Transduction , Smad1 Protein , Animals , Female , Humans , Male , Mice , Bone Morphogenetic Protein 2/metabolism , Epilepsy/metabolism , Epilepsy/physiopathology , Interneurons/metabolism , Neocortex/metabolism , Neocortex/cytology , Nerve Net/metabolism , Neurons/metabolism , Parvalbumins/metabolism , Smad1 Protein/metabolism , Synapses/metabolism , Glutamic Acid/metabolism
17.
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
18.
eNeuro ; 11(5)2024 May.
Article in English | MEDLINE | ID: mdl-38664009

ABSTRACT

Seizures are generally associated with epilepsy but may also be a symptom of many other neurological conditions. A hallmark of a seizure is the intensity of the local neuronal activation, which can drive large-scale gene transcription changes. Such changes in the transcriptional profile likely alter neuronal function, thereby contributing to the pathological process. Therefore, there is a strong clinical imperative to characterize how gene expression is changed by seizure activity. To this end, we developed a simplified ex vivo technique for studying seizure-induced transcriptional changes. We compared the RNA sequencing profile in mouse neocortical tissue with up to 3 h of epileptiform activity induced by 4-aminopyridine (4AP) relative to control brain slices not exposed to the drug. We identified over 100 genes with significantly altered expression after 4AP treatment, including multiple genes involved in MAPK, TNF, and neuroinflammatory signaling pathways, all of which have been linked to epilepsy previously. Notably, the patterns in male and female brain slices were almost identical. Various immediate early genes were among those showing the largest upregulation. The set of down-regulated genes included ones that might be expected either to increase or to decrease neuronal excitability. In summary, we found the seizure-induced transcriptional profile complex, but the changes aligned well with an analysis of published epilepsy-associated genes. We discuss how simple models may provide new angles for investigating seizure-induced transcriptional changes.


Subject(s)
4-Aminopyridine , Neocortex , Transcriptome , Animals , Neocortex/metabolism , Neocortex/drug effects , Female , Male , Mice , 4-Aminopyridine/pharmacology , Seizures/genetics , Seizures/metabolism , Seizures/physiopathology , Sequence Analysis, RNA/methods , Epilepsy/genetics , Epilepsy/metabolism , Epilepsy/physiopathology , Mice, Inbred C57BL
19.
Genes (Basel) ; 15(4)2024 03 26.
Article in English | MEDLINE | ID: mdl-38674342

ABSTRACT

Hypophosphatasia is a rare inherited metabolic disorder caused by the deficiency of tissue-nonspecific alkaline phosphatase. More severe and early onset cases present symptoms of muscle weakness, diminished motor coordination, and epileptic seizures. These neurological manifestations are poorly characterized. Thus, it is urgent to discover novel differentially expressed genes for investigating the genetic mechanisms underlying the neurological manifestations of hypophosphatasia. RNA-sequencing data offer a high-resolution and highly accurate transcript profile. In this study, we apply an empirical Bayes model to RNA-sequencing data acquired from the spinal cord and neocortex tissues of a mouse model, individually, to more accurately estimate the genetic effects without bias. More importantly, we further develop two integration methods, weighted gene approach and weighted Z method, to incorporate two RNA-sequencing data into a model for enhancing the effects of genetic markers in the diagnostics of hypophosphatasia disease. The simulation and real data analysis have demonstrated the effectiveness of our proposed integration methods, which can maximize genetic signals identified from the spinal cord and neocortex tissues, minimize the prediction error, and largely improve the prediction accuracy in risk prediction.


Subject(s)
Alkaline Phosphatase , Bayes Theorem , Hypophosphatasia , Hypophosphatasia/genetics , Animals , Mice , Alkaline Phosphatase/genetics , Sequence Analysis, RNA/methods , Spinal Cord/metabolism , Spinal Cord/pathology , Humans , Disease Models, Animal , Neocortex/metabolism , Neocortex/pathology
20.
Cereb Cortex ; 34(4)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38572735

ABSTRACT

Many studies indicate a broad role of various classes of GABAergic interneurons in the processes related to learning. However, little is known about how the learning process affects intrinsic excitability of specific classes of interneurons in the neocortex. To determine this, we employed a simple model of conditional learning in mice where vibrissae stimulation was used as a conditioned stimulus and a tail shock as an unconditioned one. In vitro whole-cell patch-clamp recordings showed an increase in intrinsic excitability of low-threshold spiking somatostatin-expressing interneurons (SST-INs) in layer 4 (L4) of the somatosensory (barrel) cortex after the conditioning paradigm. In contrast, pseudoconditioning reduced intrinsic excitability of SST-LTS, parvalbumin-expressing interneurons (PV-INs), and vasoactive intestinal polypeptide-expressing interneurons (VIP-INs) with accommodating pattern in L4 of the barrel cortex. In general, increased intrinsic excitability was accompanied by narrowing of action potentials (APs), whereas decreased intrinsic excitability coincided with AP broadening. Altogether, these results show that both conditioning and pseudoconditioning lead to plastic changes in intrinsic excitability of GABAergic interneurons in a cell-specific manner. In this way, changes in intrinsic excitability can be perceived as a common mechanism of learning-induced plasticity in the GABAergic system.


Subject(s)
Neocortex , Mice , Animals , Neocortex/metabolism , Interneurons/physiology , Learning/physiology , Conditioning, Classical/physiology , Parvalbumins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL