Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.023
Filter
1.
Brief Bioinform ; 25(3)2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38739758

ABSTRACT

The complicated process of neuronal development is initiated early in life, with the genetic mechanisms governing this process yet to be fully elucidated. Single-cell RNA sequencing (scRNA-seq) is a potent instrument for pinpointing biomarkers that exhibit differential expression across various cell types and developmental stages. By employing scRNA-seq on human embryonic stem cells, we aim to identify differentially expressed genes (DEGs) crucial for early-stage neuronal development. Our focus extends beyond simply identifying DEGs. We strive to investigate the functional roles of these genes through enrichment analysis and construct gene regulatory networks to understand their interactions. Ultimately, this comprehensive approach aspires to illuminate the molecular mechanisms and transcriptional dynamics governing early human brain development. By uncovering potential links between these DEGs and intelligence, mental disorders, and neurodevelopmental disorders, we hope to shed light on human neurological health and disease. In this study, we have used scRNA-seq to identify DEGs involved in early-stage neuronal development in hESCs. The scRNA-seq data, collected on days 26 (D26) and 54 (D54), of the in vitro differentiation of hESCs to neurons were analyzed. Our analysis identified 539 DEGs between D26 and D54. Functional enrichment of those DEG biomarkers indicated that the up-regulated DEGs participated in neurogenesis, while the down-regulated DEGs were linked to synapse regulation. The Reactome pathway analysis revealed that down-regulated DEGs were involved in the interactions between proteins located in synapse pathways. We also discovered interactions between DEGs and miRNA, transcriptional factors (TFs) and DEGs, and between TF and miRNA. Our study identified 20 significant transcription factors, shedding light on early brain development genetics. The identified DEGs and gene regulatory networks are valuable resources for future research into human brain development and neurodevelopmental disorders.


Subject(s)
Biomarkers , Brain , Gene Regulatory Networks , Human Embryonic Stem Cells , Single-Cell Analysis , Humans , Single-Cell Analysis/methods , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Brain/metabolism , Brain/embryology , Brain/cytology , Biomarkers/metabolism , Neurons/metabolism , Neurons/cytology , Cell Differentiation/genetics , RNA-Seq , Neurogenesis/genetics , Gene Expression Regulation, Developmental , Gene Expression Profiling , Sequence Analysis, RNA/methods , Single-Cell Gene Expression Analysis
2.
Nat Cell Biol ; 26(5): 710-718, 2024 May.
Article in English | MEDLINE | ID: mdl-38714853

ABSTRACT

During brain development, neural progenitors expand through symmetric divisions before giving rise to differentiating cell types via asymmetric divisions. Transition between those modes varies among individual neural stem cells, resulting in clones of different sizes. Imaging-based lineage tracing allows for lineage analysis at high cellular resolution but systematic approaches to analyse clonal behaviour of entire tissues are currently lacking. Here we implement whole-tissue lineage tracing by genomic DNA barcoding in 3D human cerebral organoids, to show that individual stem cell clones produce progeny on a vastly variable scale. By using stochastic modelling we find that variable lineage sizes arise because a subpopulation of lineages retains symmetrically dividing cells. We show that lineage sizes can adjust to tissue demands after growth perturbation via chemical ablation or genetic restriction of a subset of cells in chimeric organoids. Our data suggest that adaptive plasticity of stem cell populations ensures robustness of development in human brain organoids.


Subject(s)
Cell Lineage , Neural Stem Cells , Organoids , Organoids/cytology , Organoids/metabolism , Humans , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Brain/cytology , Brain/growth & development , Brain/metabolism , Cell Differentiation , Cell Proliferation , Clone Cells , Neurogenesis/genetics , DNA Barcoding, Taxonomic , Animals
3.
Development ; 151(9)2024 May 01.
Article in English | MEDLINE | ID: mdl-38727565

ABSTRACT

Proper embryonic development depends on the timely progression of a genetic program. One of the key mechanisms for achieving precise control of developmental timing is to use gene expression oscillations. In this Review, we examine how gene expression oscillations encode temporal information during vertebrate embryonic development by discussing the gene expression oscillations occurring during somitogenesis, neurogenesis, myogenesis and pancreas development. These oscillations play important but varied physiological functions in different contexts. Oscillations control the period of somite formation during somitogenesis, whereas they regulate the proliferation-to-differentiation switch of stem cells and progenitor cells during neurogenesis, myogenesis and pancreas development. We describe the similarities and differences of the expression pattern in space (i.e. whether oscillations are synchronous or asynchronous across neighboring cells) and in time (i.e. different time scales) of mammalian Hes/zebrafish Her genes and their targets in different tissues. We further summarize experimental evidence for the functional role of their oscillations. Finally, we discuss the outstanding questions for future research.


Subject(s)
Embryonic Development , Gene Expression Regulation, Developmental , Somites , Animals , Embryonic Development/genetics , Humans , Somites/metabolism , Somites/embryology , Muscle Development/genetics , Neurogenesis/genetics , Neurogenesis/physiology , Pancreas/embryology , Pancreas/metabolism , Cell Differentiation/genetics
4.
Cells ; 13(7)2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38607059

ABSTRACT

Embryonic neurogenesis can be defined as a period of prenatal development during which divisions of neural stem and progenitor cells give rise to neurons. In the central nervous system of most mammals, including humans, the majority of neocortical neurogenesis occurs before birth. It is a highly spatiotemporally organized process whose perturbations lead to cortical malformations and dysfunctions underlying neurological and psychiatric pathologies, and in which oxygen availability plays a critical role. In case of deprived oxygen conditions, known as hypoxia, the hypoxia-inducible factor (HIF) signaling pathway is activated, resulting in the selective expression of a group of genes that regulate homeostatic adaptations, including cell differentiation and survival, metabolism and angiogenesis. While a physiological degree of hypoxia is essential for proper brain development, imbalanced oxygen levels can adversely affect this process, as observed in common obstetrical pathologies such as prematurity. This review comprehensively explores and discusses the current body of knowledge regarding the role of hypoxia and the HIF pathway in embryonic neurogenesis of the mammalian cortex. Additionally, it highlights existing gaps in our understanding, presents unanswered questions, and provides avenues for future research.


Subject(s)
Neural Stem Cells , Humans , Animals , Pregnancy , Female , Neural Stem Cells/metabolism , Neurons/metabolism , Neurogenesis/genetics , Hypoxia/metabolism , Oxygen/metabolism , Mammals
5.
Nat Commun ; 15(1): 2837, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38565566

ABSTRACT

The adult mammalian brain retains some capacity to replenish neurons and glia, holding promise for brain regeneration. Thus, understanding the mechanisms controlling adult neural stem cell (NSC) differentiation is crucial. Paradoxically, adult NSCs in the subependymal zone transcribe genes associated with both multipotency maintenance and neural differentiation, but the mechanism that prevents conflicts in fate decisions due to these opposing transcriptional programmes is unknown. Here we describe intron detention as such control mechanism. In NSCs, while multiple mRNAs from stemness genes are spliced and exported to the cytoplasm, transcripts from differentiation genes remain unspliced and detained in the nucleus, and the opposite is true under neural differentiation conditions. We also show that m6A methylation is the mechanism that releases intron detention and triggers nuclear export, enabling rapid and synchronized responses. m6A RNA methylation operates as an on/off switch for transcripts with antagonistic functions, tightly controlling the timing of NSCs commitment to differentiation.


Subject(s)
Neural Stem Cells , Animals , Introns/genetics , Cell Differentiation/genetics , Neurons , Neurogenesis/genetics , Mammals
6.
J Cell Biol ; 223(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38652118

ABSTRACT

Chromatin-remodeling protein BRG1/SMARCA4 is pivotal for establishing oligodendrocyte (OL) lineage identity. However, its functions for oligodendrocyte-precursor cell (OPC) differentiation within the postnatal brain and during remyelination remain elusive. Here, we demonstrate that Brg1 loss profoundly impairs OPC differentiation in the brain with a comparatively lesser effect in the spinal cord. Moreover, BRG1 is critical for OPC remyelination after injury. Integrative transcriptomic/genomic profiling reveals that BRG1 exhibits a dual role by promoting OPC differentiation networks while repressing OL-inhibitory cues and proneuronal programs. Furthermore, we find that BRG1 interacts with EED/PRC2 polycomb-repressive-complexes to enhance H3K27me3-mediated repression at gene loci associated with OL-differentiation inhibition and neurogenesis. Notably, BRG1 depletion decreases H3K27me3 deposition, leading to the upregulation of BMP/WNT signaling and proneurogenic genes, which suppresses OL programs. Thus, our findings reveal a hitherto unexplored spatiotemporal-specific role of BRG1 for OPC differentiation in the developing CNS and underscore a new insight into BRG1/PRC2-mediated epigenetic regulation that promotes and safeguards OL lineage commitment and differentiation.


Subject(s)
Cell Differentiation , DNA Helicases , Oligodendroglia , Polycomb Repressive Complex 2 , Animals , Mice , DNA Helicases/metabolism , DNA Helicases/genetics , Epigenesis, Genetic , Histones/metabolism , Histones/genetics , Mice, Inbred C57BL , Neurogenesis/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/metabolism , Polycomb Repressive Complex 2/metabolism , Polycomb Repressive Complex 2/genetics , Remyelination , Transcription Factors/metabolism , Transcription Factors/genetics
7.
EMBO Rep ; 25(5): 2202-2219, 2024 May.
Article in English | MEDLINE | ID: mdl-38600346

ABSTRACT

Neural progenitor cells within the cerebral cortex undergo a characteristic switch between symmetric self-renewing cell divisions early in development and asymmetric neurogenic divisions later. Yet, the mechanisms controlling this transition remain unclear. Previous work has shown that early but not late neural progenitor cells (NPCs) endogenously express the autism-linked transcription factor Foxp1, and both loss and gain of Foxp1 function can alter NPC activity and fate choices. Here, we show that premature loss of Foxp1 upregulates transcriptional programs regulating angiogenesis, glycolysis, and cellular responses to hypoxia. These changes coincide with a premature destabilization of HIF-1α, an elevation in HIF-1α target genes, including Vegfa in NPCs, and precocious vascular network development. In vitro experiments demonstrate that stabilization of HIF-1α in Foxp1-deficient NPCs rescues the premature differentiation phenotype and restores NPC maintenance. Our data indicate that the endogenous decline in Foxp1 expression activates the HIF-1α transcriptional program leading to changes in the tissue environment adjacent to NPCs, which, in turn, might alter their self-renewal and neurogenic capacities.


Subject(s)
Cerebral Cortex , Forkhead Transcription Factors , Hypoxia-Inducible Factor 1, alpha Subunit , Neural Stem Cells , Repressor Proteins , Signal Transduction , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Animals , Mice , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Repressor Proteins/metabolism , Repressor Proteins/genetics , Neovascularization, Physiologic/genetics , Cell Differentiation/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Neurogenesis/genetics , Glycolysis , Angiogenesis
8.
Sci Rep ; 14(1): 9355, 2024 04 23.
Article in English | MEDLINE | ID: mdl-38654093

ABSTRACT

Thyroid hormones (TH) play critical roles during nervous system development and patients carrying coding variants of MCT8 (monocarboxylate transporter 8) or THRA (thyroid hormone receptor alpha) present a spectrum of neurological phenotypes resulting from perturbed local TH action during early brain development. Recently, human cerebral organoids (hCOs) emerged as powerful in vitro tools for disease modelling recapitulating key aspects of early human cortex development. To begin exploring prospects of this model for thyroid research, we performed a detailed characterization of the spatiotemporal expression of MCT8 and THRA in developing hCOs. Immunostaining showed MCT8 membrane expression in neuronal progenitor cell types including early neuroepithelial cells, radial glia cells (RGCs), intermediate progenitors and outer RGCs. In addition, we detected robust MCT8 protein expression in deep layer and upper layer neurons. Spatiotemporal SLC16A2 mRNA expression, detected by fluorescent in situ hybridization (FISH), was highly concordant with MCT8 protein expression across cortical cell layers. FISH detected THRA mRNA expression already in neuroepithelium before the onset of neurogenesis. THRA mRNA expression remained low in the ventricular zone, increased in the subventricular zone whereas strong THRA expression was observed in excitatory neurons. In combination with a robust up-regulation of known T3 response genes following T3 treatment, these observations show that hCOs provide a promising and experimentally tractable model to probe local TH action during human cortical neurogenesis and eventually to model the consequences of impaired TH function for early cortex development.


Subject(s)
Cerebral Cortex , Monocarboxylic Acid Transporters , Neurogenesis , Organoids , RNA, Messenger , Symporters , Thyroid Hormone Receptors alpha , Female , Humans , Pregnancy , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Gene Expression Regulation, Developmental , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Neurogenesis/genetics , Neurons/metabolism , Organoids/metabolism , Pregnancy Trimester, First/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Symporters/genetics , Symporters/metabolism , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormones/metabolism , Thyroid Hormones/genetics
9.
Proc Natl Acad Sci U S A ; 121(12): e2315707121, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38489388

ABSTRACT

KCTD10 belongs to the KCTD (potassiumchannel tetramerization domain) family, many members of which are associated with neuropsychiatric disorders. However, the biological function underlying the association with brain disorders remains to be explored. Here, we reveal that Kctd10 is highly expressed in neuronal progenitors and layer V neurons throughout brain development. Kctd10 deficiency triggers abnormal proliferation and differentiation of neuronal progenitors, reduced deep-layer (especially layer V) neurons, increased upper-layer neurons, and lowered brain size. Mechanistically, we screened and identified a unique KCTD10-interacting protein, KCTD13, associated with neurodevelopmental disorders. KCTD10 mediated the ubiquitination-dependent degradation of KCTD13 and KCTD10 ablation resulted in a considerable increase of KCTD13 expression in the developing cortex. KCTD13 overexpression in neuronal progenitors led to reduced proliferation and abnormal cell distribution, mirroring KCTD10 deficiency. Notably, mice with brain-specific Kctd10 knockout exhibited obvious motor deficits. This study uncovers the physiological function of KCTD10 and provides unique insights into the pathogenesis of neurodevelopmental disorders.


Subject(s)
Brain Diseases , Neurodevelopmental Disorders , Potassium Channels, Voltage-Gated , Animals , Mice , Proteins/metabolism , Brain/metabolism , Neurons/metabolism , Neurodevelopmental Disorders/genetics , Brain Diseases/genetics , Neurogenesis/genetics , Potassium Channels, Voltage-Gated/metabolism
10.
J Physiol ; 602(7): 1427-1442, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38468384

ABSTRACT

Fibroblast growth factor-2 (FGF2) is involved in the regulation of affective behaviour and shows antidepressant effects through the Akt and extracellular signal regulated kinase (ERK) 1/2 pathways. Nudix hydrolase 6 (NUDT6) protein is encoded from FGF2 gene's antisense strand and its role in the regulation of affective behaviour is unknown. Here, we overexpressed NUDT6 in the hippocampus and investigated its behavioural effects and the underlying molecular mechanisms affecting the behaviour. We showed that increasing hippocampal NUDT6 results in depression-like behaviour in rats without changing FGF2 levels or activating its downstream effectors, Akt and ERK1/2. Instead, NUDT6 acted by inducing inflammatory signalling, specifically by increasing S100 calcium binding protein A9 (S100A9) levels, activating nuclear factor-kappa B-p65 (NF-κB-p65), and elevating microglia numbers along with a reduction in neurogenesis. Our results suggest that NUDT6 could play a role in major depression by inducing a proinflammatory state. This is the first report of an antisense protein acting through a different mechanism of action than regulation of its sense protein. The opposite effects of NUDT6 and FGF2 on depression-like behaviour may serve as a mechanism to fine-tune affective behaviour. Our findings open up new venues for studying the differential regulation and functional interactions of sense and antisense proteins in neural function and behaviour, as well as in neuropsychiatric disorders. KEY POINTS: Hippocampal overexpression of nudix hydrolase 6 (NUDT6), the antisense protein of fibroblast growth factor-2 (FGF2), increases depression-like behaviour in rats. Hippocampal NUDT6 overexpression triggers a neuroinflammatory cascade by increasing S100 calcium binding proteinA9 (S100A9) expression and nuclear NF-κB-p65 translocation in neurons, in addition to microglial recruitment and activation. Hippocampal NUDT6 overexpression suppresses neurogenesis. NUDT6 exerts its actions without altering the levels or downstream signalling pathways of FGF2.


Subject(s)
Depression , Fibroblast Growth Factor 2 , NF-kappa B , Animals , Rats , Fibroblast Growth Factor 2/genetics , Inflammation/genetics , Neurogenesis/genetics , NF-kappa B/metabolism , NF-kappa B/pharmacology , Proto-Oncogene Proteins c-akt , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Depression/genetics , Depression/metabolism
11.
Nat Aging ; 4(4): 546-567, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38553564

ABSTRACT

Partial reprogramming (pulsed expression of reprogramming transcription factors) improves the function of several tissues in old mice. However, it remains largely unknown how partial reprogramming impacts the old brain. Here we use single-cell transcriptomics to systematically examine how partial reprogramming influences the subventricular zone neurogenic niche in aged mouse brains. Whole-body partial reprogramming mainly improves neuroblasts (cells committed to give rise to new neurons) in the old neurogenic niche, restoring neuroblast proportion to more youthful levels. Interestingly, targeting partial reprogramming specifically to the neurogenic niche also boosts the proportion of neuroblasts and their precursors (neural stem cells) in old mice and improves several molecular signatures of aging, suggesting that the beneficial effects of reprogramming are niche intrinsic. In old neural stem cell cultures, partial reprogramming cell autonomously restores the proportion of neuroblasts during differentiation and blunts some age-related transcriptomic changes. Importantly, partial reprogramming improves the production of new neurons in vitro and in old brains. Our work suggests that partial reprogramming could be used to rejuvenate the neurogenic niche and counter brain decline in old individuals.


Subject(s)
Neural Stem Cells , Neurons , Mice , Animals , Neurogenesis/genetics , Cell Differentiation/genetics , Cellular Reprogramming/genetics
12.
J Chem Neuroanat ; 137: 102414, 2024 04.
Article in English | MEDLINE | ID: mdl-38490283

ABSTRACT

Rat offspring who are exposed to an amorphous formula of curcumin (CUR) from the embryonic stage have anti-anxiety-like behaviors, enhanced fear extinction learning, and increased synaptic plasticity in the hippocampal dentate gyrus (DG). In the present study, we investigated the links between genes with altered methylation status in the neurogenic niche and enhanced neural functions after CUR exposure. We conducted methylation and RNA sequencing analyses of the DG of CUR-exposed rat offspring on day 77 after delivery. Methylation status and transcript levels of candidate genes were validated using methylation-sensitive high-resolution melting and real-time reverse-transcription PCR, respectively. In the CUR group, we confirmed the hypermethylation and downregulation of Gpr150, Mmp23, Rprml, and Pcdh8 as well as the hypomethylation and upregulation of Ppm1j, Fam222a, and Opn3. Immunohistochemically, reprimo-like+ hilar cells and protocadherin-8+ granule cells were decreased and opsin-3+ hilar cells were increased by CUR exposure. Both reprimo-like and opsin-3 were partially expressed on subpopulations of glutamic acid decarboxylase 67+ γ-aminobutyric acid-ergic interneurons. Furthermore, the transcript levels of genes involved in protocadherin-8-mediated N-cadherin endocytosis were altered with CUR exposure; this was accompanied by Ctnnb1 and Syp upregulation and Mapk14, Map2k3, and Grip1 downregulation, suggesting that CUR-induced enhanced synaptic plasticity is associated with cell adhesion. Together, our results indicate that functionally different genes have altered methylation and expression in different neuronal populations of the hippocampal neurogenic niche, thus enhancing synaptic plasticity after CUR exposure.


Subject(s)
Curcumin , DNA Methylation , Hippocampus , Animals , Curcumin/pharmacology , Rats , DNA Methylation/drug effects , Hippocampus/metabolism , Hippocampus/drug effects , Female , Neurogenesis/drug effects , Neurogenesis/genetics , Male , Pregnancy , Rats, Sprague-Dawley , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/chemically induced
13.
STAR Protoc ; 5(1): 102904, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38427568

ABSTRACT

Neurodevelopmental disorders are characterized by complex phenotypes that often result from concomitant dysregulation of cell proliferation, differentiation, or other crucial developmental processes. Here, we present a protocol to quantify differentiation of progenitor populations during early stages of neurogenesis in induced pluripotent stem cell (iPSC)-derived cerebral organoids. We describe steps for organoid differentiation and maturation, sample preparation, immunofluorescence, and imaging and analysis using epifluorescence microscopy. This protocol can be used to compare cerebral organoids from control and patient-derived iPSCs. For complete details on the use and execution of this protocol, please refer to Rakotomamonjy et al. (2023).1.


Subject(s)
Induced Pluripotent Stem Cells , Neurodevelopmental Disorders , Humans , Cell Differentiation/genetics , Neurogenesis/genetics , Organoids
14.
Development ; 151(4)2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38369736

ABSTRACT

The generation of neurons in the developing neocortex is a major determinant of neocortex size. Crucially, the increase in cortical neuron numbers in the primate lineage, notably in the upper-layer neurons, contributes to increased cognitive abilities. Here, we review major evolutionary changes affecting the apical progenitors in the ventricular zone and focus on the key germinal zone constituting the foundation of neocortical neurogenesis in primates, the outer subventricular zone (OSVZ). We summarize characteristic features of the OSVZ and its key stem cell type, the basal (or outer) radial glia. Next, we concentrate on primate-specific and human-specific genes, expressed in OSVZ-progenitors, the ability of which to amplify these progenitors by targeting the regulation of the cell cycle ultimately underlies the evolutionary increase in upper-layer neurons. Finally, we address likely differences in neocortical development between present-day humans and Neanderthals that are based on human-specific amino acid substitutions in proteins operating in cortical progenitors.


Subject(s)
Neocortex , Neuroglia , Animals , Humans , Neuroglia/metabolism , Neocortex/metabolism , Neurons/metabolism , Stem Cells , Primates/genetics , Neurogenesis/genetics
15.
Int J Mol Sci ; 25(3)2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38338930

ABSTRACT

CXCL14 is not only involved in the immune process but is also closely related to neurodevelopment according to its molecular evolution. However, what role it plays in neurodevelopment remains unclear. In the present research, we found that, by crossbreeding CXCL14+/- and CXCL14-/- mice, the number of CXCL14-/- mice in their offspring was lower than the Mendelian frequency; CXCL14-/- mice had significantly fewer neurons in the external pyramidal layer of cortex than CXCL14+/- mice; and CXCL14 may be involved in synaptic plasticity, neuron projection, and chemical synaptic transmission based on analysis of human clinical transcriptome data. The expression of CXCL14 was highest at day 14.5 in the embryonic phase and after birth in the mRNA and protein levels. Therefore, we hypothesized that CXCL14 promotes the development of neurons in the somatic layer of the pyramidal cells of mice cortex on embryonic day 14.5. In order to further explore its mechanism, CXCR4 and CXCR7 were suggested as receptors by Membrane-Anchored Ligand and Receptor Yeast Two-Hybrid technology. Through metabolomic techniques, we inferred that CXCL14 promotes the development of neurons by regulating fatty acid anabolism and glycerophospholipid anabolism.


Subject(s)
Chemokines, CXC , Multiomics , Neurogenesis , Animals , Humans , Mice , Chemokines, CXC/genetics , Neurons/metabolism , Signal Transduction , Synaptic Transmission , Neurogenesis/genetics
16.
J Cell Sci ; 137(4)2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38305737

ABSTRACT

Tight control over transcription factor activity is necessary for a sensible balance between cellular proliferation and differentiation in the embryo and during tissue homeostasis by adult stem cells, but mechanistic details have remained incomplete. The homeodomain transcription factor MEIS2 is an important regulator of neurogenesis in the ventricular-subventricular zone (V-SVZ) adult stem cell niche in mice. We here identify MEIS2 as direct target of the intracellular protease calpain-2 (composed of the catalytic subunit CAPN2 and the regulatory subunit CAPNS1). Phosphorylation at conserved serine and/or threonine residues, or dimerization with PBX1, reduced the sensitivity of MEIS2 towards cleavage by calpain-2. In the adult V-SVZ, calpain-2 activity is high in stem and progenitor cells, but rapidly declines during neuronal differentiation, which is accompanied by increased stability of MEIS2 full-length protein. In accordance with this, blocking calpain-2 activity in stem and progenitor cells, or overexpression of a cleavage-insensitive form of MEIS2, increased the production of neurons, whereas overexpression of a catalytically active CAPN2 reduced it. Collectively, our results support a key role for calpain-2 in controlling the output of adult V-SVZ neural stem and progenitor cells through cleavage of the neuronal fate determinant MEIS2.


Subject(s)
Neural Stem Cells , Transcription Factors , Animals , Mice , Calpain/genetics , Calpain/metabolism , Cell Differentiation , Cell Proliferation , Endopeptidases/metabolism , Lateral Ventricles/metabolism , Neural Stem Cells/metabolism , Neurogenesis/genetics , Neurons/metabolism , Peptide Hydrolases/metabolism , Transcription Factors/metabolism
17.
Elife ; 122024 Feb 23.
Article in English | MEDLINE | ID: mdl-38391176

ABSTRACT

Neuroblasts in Drosophila divide asymmetrically, sequentially expressing a series of intrinsic factors to generate a diversity of neuron types. These intrinsic factors known as temporal factors dictate timing of neuroblast transitions in response to steroid hormone signaling and specify early versus late temporal fates in neuroblast neuron progeny. After completing their temporal programs, neuroblasts differentiate or die, finalizing both neuron number and type within each neuroblast lineage. From a screen aimed at identifying genes required to terminate neuroblast divisions, we identified Notch and Notch pathway components. When Notch is knocked down, neuroblasts maintain early temporal factor expression longer, delay late temporal factor expression, and continue dividing into adulthood. We find that Delta, expressed in cortex glia, neuroblasts, and after division, their GMC progeny, regulates neuroblast Notch activity. We also find that Delta in neuroblasts is expressed high early, low late, and is controlled by the intrinsic temporal program: early factor Imp promotes Delta, late factors Syp/E93 reduce Delta. Thus, in addition to systemic steroid hormone cues, forward lineage progression is controlled by local cell-cell signaling between neuroblasts and their cortex glia/GMC neighbors: Delta transactivates Notch in neuroblasts bringing the early temporal program and early temporal factor expression to a close.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Drosophila/genetics , Drosophila Proteins/metabolism , Neurogenesis/genetics , Hormones/metabolism , Steroids/metabolism , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental
18.
Curr Opin Genet Dev ; 85: 102156, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38354530

ABSTRACT

Emerging evidence supports the existence of dedicated molecular mechanisms under evolutionary selection to control time during neurogenesis. Here, we briefly review these mechanisms and discuss a potentially useful conceptual framework inspired by computer science to think about how these biological mechanisms operate during brain development and evolution.


Subject(s)
CLOCK Proteins , Neurogenesis , Neurogenesis/genetics , Algorithms , Biological Evolution
19.
Sci Rep ; 14(1): 3395, 2024 02 09.
Article in English | MEDLINE | ID: mdl-38336823

ABSTRACT

Developmental neurogenesis is a tightly regulated spatiotemporal process with its dysregulation implicated in neurodevelopmental disorders. NMDA receptors are glutamate-gated ion channels that are widely expressed in the early nervous system, yet their contribution to neurogenesis is poorly understood. Notably, a variety of mutations in genes encoding NMDA receptor subunits are associated with neurodevelopmental disorders. To rigorously define the role of NMDA receptors in developmental neurogenesis, we used a mutant zebrafish line (grin1-/-) that lacks all NMDA receptors yet survives to 10 days post-fertilization, offering the opportunity to study post-embryonic neurodevelopment in the absence of NMDA receptors. Focusing on the forebrain, we find that these fish have a progressive supernumerary neuron phenotype confined to the telencephalon at the end of embryonic neurogenesis, but which extends to all forebrain regions during postembryonic neurogenesis. This enhanced neuron population does not arise directly from increased numbers or mitotic activity of radial glia cells, the principal neural stem cells. Rather, it stems from a lack of timely maturation of transit-amplifying neuroblasts into post-mitotic neurons, as indicated by a decrease in expression of the ontogenetically-expressed chloride transporter, KCC2. Pharmacological blockade with MK-801 recapitulates the grin1-/- supernumerary neuron phenotype, indicating a requirement for ionotropic signaling. Thus, NMDA receptors are required for suppression of indirect, transit amplifying cell-driven neurogenesis by promoting maturational termination of mitosis. Loss of suppression results in neuronal overpopulation that can fundamentally change brain circuitry and may be a key factor in pathogenesis of neurodevelopmental disorders caused by NMDA receptor dysfunction.


Subject(s)
Neural Stem Cells , Receptors, N-Methyl-D-Aspartate , Animals , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Neurons/metabolism , Neural Stem Cells/metabolism , Neurogenesis/genetics , Telencephalon/metabolism
20.
Brain Struct Funct ; 229(3): 759-773, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38411929

ABSTRACT

Malformation during cortical development can disrupt the balance of excitatory and inhibitory neural circuits, contributing to various psychiatric and developmental disorders. One of the critical factors of cortical neural networks is the fine regulation of neurogenesis through mechanical cues, such as shear stress and substrate stiffness. Piezo1, a mechanically-activated channel, serves as a transducer for these mechanical cues, regulating embryogenesis. However, specific cell-type expression patterns of this channel during cortical development have not yet been characterized. In the present study, we conducted an RNAscope experiment to visualize the location of Piezo1 transcripts with embryonic neuronal/glial lineage cell markers. Our analysis covered coronal sections of the mouse forebrain on embryonic day 12.5 (E12.5), E14.5, E16.5, and E18.5. In addition, applying Yoda1, a specific Piezo1 agonist, evoked distinct calcium elevation in piriform cortices of E16.5 and E18.5 embryonic slices. Furthermore, pharmacological activation or inhibition of this channel significantly modulated the migration of neurosphere-derived cells in vitro. These findings contribute valuable insights to the field of mechanobiology and provide an understanding of the intricate processes underlying embryonic brain development.


Subject(s)
Ion Channels , Neurogenesis , Animals , Mice , Ion Channels/genetics , Ion Channels/metabolism , Mechanotransduction, Cellular/physiology , Neurogenesis/genetics , Prosencephalon/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...