Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Publication year range
1.
bioRxiv ; 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-36993710

ABSTRACT

Attention is required for most higher-order cognitive functions. Prior studies have revealed functional roles for the prefrontal cortex and its extended circuits to enabling attention, but the underlying molecular processes and their impacts on cellular and circuit function remain poorly understood. To develop insights, we here took an unbiased forward genetics approach to identify single genes of large effect on attention. We studied 200 genetically diverse mice on measures of pre-attentive processing and through genetic mapping identified a small locus on chromosome 13 (95%CI: 92.22-94.09 Mb) driving substantial variation (19%) in this trait. Further characterization of the locus revealed a causative gene, Homer1, encoding a synaptic protein, where down-regulation of its short isoforms in prefrontal cortex (PFC) during early postnatal development led to improvements in multiple measures of attention in the adult. Subsequent mechanistic studies revealed that prefrontal Homer1 down-regulation is associated with GABAergic receptor up-regulation in those same cells. This enhanced inhibitory influence, together with dynamic neuromodulatory coupling, led to strikingly low PFC activity at baseline periods of the task but targeted elevations at cue onset, predicting short-latency correct choices. Notably high-Homer1, low-attentional performers, exhibited uniformly elevated PFC activity throughout the task. We thus identify a single gene of large effect on attention - Homer1 - and find that it improves prefrontal inhibitory tone and signal-to-noise (SNR) to enhance attentional performance. A therapeutic strategy focused on reducing prefrontal activity and increasing SNR, rather than uniformly elevating PFC activity, may complement the use of stimulants to improve attention.

2.
Front Neurosci ; 17: 1149603, 2023.
Article in English | MEDLINE | ID: mdl-37456993

ABSTRACT

Adult neural stem cells (NSCs) in the mouse subventricular zone (SVZ) serve as a lifelong reservoir for newborn olfactory bulb neurons. Recent studies have identified a slowly dividing subpopulation of embryonic neural stem-progenitor cells (NPCs) as the embryonic origin of adult NSCs. Yet, little is known about how these slowly dividing embryonic NPCs are maintained until adulthood while other NPCs are extinguished by the completion of brain development. The extracellular matrix (ECM) is an essential component of stem cell niches and thus a key determinant of stem cell fate. Here we investigated tissue inhibitors of metalloproteinases (TIMPs)-regulators of ECM remodeling-for their potential roles in the establishment of adult NSCs. We found that Timp2, Timp3, and Timp4 were expressed at high levels in slowly dividing NPCs compared to rapidly dividing NPCs. Deletion of TIMP3 reduced the number of adult NSCs and neuroblasts in the lateral SVZ. In addition, overexpression of TIMP3 in the embryonic NPCs suppressed neuronal differentiation and upregulated the expression levels of Notch signaling relating genes. These results thus suggest that TIMP3 keeps the undifferentiated state of embryonic NPCs, leading to the establishment and maintenance of adult NSCs.

3.
Nat Commun ; 12(1): 6562, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34772946

ABSTRACT

Quiescent neural stem cells (NSCs) in the adult mouse brain are the source of neurogenesis that regulates innate and adaptive behaviors. Adult NSCs in the subventricular zone are derived from a subpopulation of embryonic neural stem-progenitor cells (NPCs) that is characterized by a slower cell cycle relative to the more abundant rapid cycling NPCs that build the brain. Yet, how slow cell cycle can cause the establishment of adult NSCs remains largely unknown. Here, we demonstrate that Notch and an effector Hey1 form a module that is upregulated by cell cycle arrest in slowly dividing NPCs. In contrast to the oscillatory expression of the Notch effectors Hes1 and Hes5 in fast cycling progenitors, Hey1 displays a non-oscillatory stationary expression pattern and contributes to the long-term maintenance of NSCs. These findings reveal a novel division of labor in Notch effectors where cell cycle rate biases effector selection and cell fate.


Subject(s)
Adult Stem Cells/metabolism , Cell Cycle Checkpoints/physiology , Cell Cycle Proteins/metabolism , Neurogenesis/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/cytology , Cell Cycle/genetics , Cell Cycle/physiology , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/genetics , Embryonic Stem Cells , Gene Expression , Lateral Ventricles/metabolism , Mice , Nervous System , Neurogenesis/genetics , Receptor, Notch1 , Repressor Proteins/metabolism
4.
Stem Cells ; 39(7): 929-944, 2021 07.
Article in English | MEDLINE | ID: mdl-33609411

ABSTRACT

Lysosomes have recently been implicated in regulation of quiescence in adult neural stem cells (NSCs). Whether lysosomes regulate the differentiation of neural stem-progenitor cells (NPCs) in the embryonic brain has remained unknown, however. We here show that lysosomes are more abundant in rapidly dividing NPCs than in differentiating neurons in the embryonic mouse neocortex and ganglionic eminence. The genes for TFEB and TFE3, master regulators of lysosomal biosynthesis, as well as other lysosome-related genes were also expressed at higher levels in NPCs than in differentiating neurons. Anatomic analysis revealed accumulation of lysosomes at the apical and basal endfeet of NPCs. Knockdown of TFEB and TFE3, or that of the lysosomal transporter Slc15a4, resulted in premature differentiation of neocortical NPCs. Conversely, forced expression of an active form of TFEB (TFEB-AA) suppressed neuronal differentiation of NPCs in association with upregulation of NPC-related genes. These results together point to a previously unappreciated role for TFEB and TFE3, and possibly for lysosomes, in maintenance of the undifferentiated state of embryonic NPCs. We further found that lysosomes are even more abundant in an NPC subpopulation that rarely divides and includes the embryonic origin of adult NSCs than in the majority of NPCs that divide frequently for construction of the embryonic brain, and that overexpression of TFEB-AA also suppressed the cell cycle of neocortical NPCs. Our results thus also implicate lysosomes in establishment of the slowly dividing, embryonic origin of adult NSCs.


Subject(s)
Neocortex , Neural Stem Cells , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Differentiation/physiology , Lysosomes/metabolism , Membrane Transport Proteins/metabolism , Mice , Neural Stem Cells/metabolism
5.
Sci Rep ; 11(1): 613, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436697

ABSTRACT

The lateral ventricles of the adult mammalian brain are lined by a single layer of multiciliated ependymal cells, which generate a flow of cerebrospinal fluid through directional beating of their cilia as well as regulate neurogenesis through interaction with adult neural stem cells. Ependymal cells are derived from a subset of embryonic neural stem-progenitor cells (NPCs, also known as radial glial cells) that becomes postmitotic during the late embryonic stage of development. Members of the Geminin family of transcriptional regulators including GemC1 and Mcidas play key roles in the differentiation of ependymal cells, but it remains largely unclear what extracellular signals regulate these factors and ependymal differentiation during embryonic and early-postnatal development. We now show that the levels of Smad1/5/8 phosphorylation and Id1/4 protein expression-both of which are downstream events of bone morphogenetic protein (BMP) signaling-decline in cells of the ventricular-subventricular zone in the mouse lateral ganglionic eminence in association with ependymal differentiation. Exposure of postnatal NPC cultures to BMP ligands or to a BMP receptor inhibitor suppressed and promoted the emergence of multiciliated ependymal cells, respectively. Moreover, treatment of embryonic NPC cultures with BMP ligands reduced the expression level of the ependymal marker Foxj1 and suppressed the emergence of ependymal-like cells. Finally, BMP ligands reduced the expression levels of Gemc1 and Mcidas in postnatal NPC cultures, whereas the BMP receptor inhibitor increased them. Our results thus implicate BMP signaling in suppression of ependymal differentiation from NPCs through regulation of Gemc1 and Mcidas expression during embryonic and early-postnatal stages of mouse telencephalic development.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Differentiation , Embryonic Stem Cells/cytology , Ependyma/cytology , Neural Stem Cells/cytology , Telencephalon/cytology , Animals , Cell Cycle Proteins/metabolism , Embryonic Stem Cells/metabolism , Ependyma/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Neural Stem Cells/metabolism , Neurogenesis , Telencephalon/metabolism
6.
Brain Nerve ; 69(9): 1027-1034, 2017 Sep.
Article in Japanese | MEDLINE | ID: mdl-28900065

ABSTRACT

Neural stem cells (NSCs) in the adult mammalian brain produce new neurons throughout life. Defects in adult neurogenesis can influence neurodegenerative and psychiatric disorders. Hence, understanding long-term maintenance of adult NSCs is crucial. Cell-intrinsic and -extrinsic factors contribute to long-term maintenance of adult NSCs, and we have previously reported that NSCs produce their own niches that send a feedback signal for their own maintenance. In addition, we have identified a slowly dividing subpopulation of embryonic neural progenitor cells that is set aside during development, and later becomes a substantial fraction of NSCs in the adult subventricular zone. Here, we review the mechanisms of long-term maintenance and embryonic origin of adult NSCs. We also discuss current topics on adult NSCs and future perspectives in this field of study.


Subject(s)
Adult Stem Cells/cytology , Neural Stem Cells/cytology , Neurogenesis , Animals , Cell Division , Humans , Stem Cell Niche
7.
Nat Neurosci ; 18(5): 657-65, 2015 May.
Article in English | MEDLINE | ID: mdl-25821910

ABSTRACT

The mechanism by which adult neural stem cells (NSCs) are established during development is unclear. In this study, analysis of cell cycle progression by examining retention of a histone 2B (H2B)-GFP fusion protein revealed that, in a subset of mouse embryonic neural progenitor cells (NPCs), the cell cycle slows between embryonic day (E) 13.5 and E15.5 while other embryonic NPCs continue to divide rapidly. By allowing H2B-GFP expressed at E9.5 to become diluted in dividing cells until the young adult stage, we determined that a majority of NSCs in the young adult subependymal zone (SEZ) originated from these slowly dividing embryonic NPCs. The cyclin-dependent kinase inhibitor p57 is highly expressed in this embryonic subpopulation, and the deletion of p57 impairs the emergence of adult NSCs. Our results suggest that a substantial fraction of adult SEZ NSCs is derived from a slowly dividing subpopulation of embryonic NPCs and identify p57 as a key factor in generating this embryonic origin of adult SEZ NSCs.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p57/physiology , Nerve Tissue Proteins/physiology , Neural Stem Cells/cytology , Neurogenesis , Telencephalon/cytology , Animals , Cell Cycle , Cell Division , Cell Lineage , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Cyclin-Dependent Kinase Inhibitor p57/genetics , Genes, Reporter , Histones/biosynthesis , Histones/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Recombinant Fusion Proteins/metabolism , Telencephalon/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...