Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Dev Cell ; 2024 May 14.
Article in English | MEDLINE | ID: mdl-38772376

ABSTRACT

Neural stem cells (NSCs) differentiate into neuron-fated intermediate progenitor cells (IPCs) via cell division. Although differentiation from NSCs to IPCs is a discrete process, recent transcriptome analyses identified a continuous transcriptional trajectory during this process, raising the question of how to reconcile these contradictory observations. In mouse NSCs, Hes1 expression oscillates, regulating the oscillatory expression of the proneural gene Neurog2, while Hes1 expression disappears in IPCs. Thus, the transition from Hes1 oscillation to suppression is involved in the differentiation of NSCs to IPCs. Here, we found that Neurog2 oscillations induce the accumulation of Tbr2, which suppresses Hes1 expression, generating an IPC-like gene expression state in NSCs. In the absence of Tbr2, Hes1 expression is up-regulated, decreasing the formation of IPCs. These results indicate that the Neurog2-Tbr2 axis forms a continuous transcriptional trajectory to an IPC-like neurogenic state in NSCs, which then differentiate into IPCs via cell division.

2.
Dev Growth Differ ; 65(6): 360-369, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37309238

ABSTRACT

The first stage of cell differentiation during mouse development is the differentiation into the trophectoderm and inner cell mass, which occurs during the 8-32-cell stages of preimplantation embryos. This differentiation is regulated by the Hippo signaling pathway. At the 32-cell stage, embryos establish a position-dependent distribution of the Hippo pathway coactivator, Yes-associated protein 1 (YAP, encoded by Yap1). The outer and inner cells showed nuclear and cytoplasmic localization of YAP, respectively. However, the process by which embryos establish position-dependent YAP localization remains elusive. Here, we established a YAP-reporter mouse line, Yap1mScarlet , and examined YAP-mScarlet protein dynamics during the 8-32-cell stages using live imaging. During mitosis, YAP-mScarlet diffused throughout the cells. YAP-mScarlet dynamics in daughter cells varied depending on the cell division patterns. YAP-mScarlet localization in daughter cells at the completion of cell division coincided with that in mother cells. Experimental manipulation of YAP-mScarlet localization in mother cells also altered its localization in daughter cells upon completion of cell division. In daughter cells, YAP-mScarlet localization gradually changed to the final pattern. In some divisions during the 8-16-cell stages, the cytoplasmic YAP-mScarlet localization preceded cell internalization. These results suggest that cell position is not a primary determinant of YAP localization and that the Hippo signaling status of the mother cell is inherited by the daughter cells, which likely contributes to the stabilization of the cell fate specification process beyond cell division.


Subject(s)
Blastocyst , Protein Serine-Threonine Kinases , YAP-Signaling Proteins , Animals , Mice , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Blastocyst/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Stem Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , YAP-Signaling Proteins/metabolism
3.
Physiology (Bethesda) ; 38(2): 0, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36256636

ABSTRACT

The significance of the coupling delay, which is the time required for interactions between coupled oscillators, in various oscillatory dynamics has been investigated mathematically for more than three decades, but its biological significance has been revealed only recently. In the segmentation clock, which regulates the periodic formation of somites in embryos, Hes7 expression oscillates synchronously between neighboring presomitic mesoderm (PSM) cells, and this synchronized oscillation is controlled by Notch signaling-mediated coupling between PSM cells. Recent studies have shown that inappropriate coupling delays dampen and desynchronize Hes7 oscillations, as simulated mathematically, leading to the severe fusion of somites and somite-derived tissues such as the vertebrae and ribs. These results indicate the biological significance of the coupling delay in synchronized Hes7 oscillations in the segmentation clock. The recent development of an in vitro PSM-like system will facilitate the detailed analysis of the coupling delay in synchronized oscillations.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Somites , Humans , Basic Helix-Loop-Helix Transcription Factors/metabolism , Somites/metabolism , Signal Transduction/physiology
5.
Nat Commun ; 12(1): 1318, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33637744

ABSTRACT

Cell-cell interactions mediated by Notch are critical for the maintenance of skeletal muscle stem cells. However, dynamics, cellular source and identity of functional Notch ligands during expansion of the stem cell pool in muscle growth and regeneration remain poorly characterized. Here we demonstrate that oscillating Delta-like 1 (Dll1) produced by myogenic cells is an indispensable Notch ligand for self-renewal of muscle stem cells in mice. Dll1 expression is controlled by the Notch target Hes1 and the muscle regulatory factor MyoD. Consistent with our mathematical model, our experimental analyses show that Hes1 acts as the oscillatory pacemaker, whereas MyoD regulates robust Dll1 expression. Interfering with Dll1 oscillations without changing its overall expression level impairs self-renewal, resulting in premature differentiation of muscle stem cells during muscle growth and regeneration. We conclude that the oscillatory Dll1 input into Notch signaling ensures the equilibrium between self-renewal and differentiation in myogenic cell communities.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Differentiation/physiology , Muscle Development/physiology , Muscles/metabolism , Stem Cells/physiology , Animals , Calcium-Binding Proteins/genetics , Female , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Mice , Mice, Knockout , Muscle Development/genetics , Mutation , MyoD Protein/genetics , MyoD Protein/metabolism , Signal Transduction/physiology , Transcription Factor HES-1/metabolism , Transcriptome
6.
Mol Psychiatry ; 26(6): 2633-2650, 2021 06.
Article in English | MEDLINE | ID: mdl-32350390

ABSTRACT

Calcium, the most versatile second messenger, regulates essential biology including crucial cellular events in embryogenesis. We investigated impacts of calcium channels and purinoceptors on neuronal differentiation of normal mouse embryonic stem cells (ESCs), with outcomes being compared to those of in vitro models of Huntington's disease (HD). Intracellular calcium oscillations tracked via real-time fluorescence and luminescence microscopy revealed a significant correlation between calcium transient activity and rhythmic proneuronal transcription factor expression in ESCs stably expressing ASCL-1 or neurogenin-2 promoters fused to luciferase reporter genes. We uncovered that pharmacological manipulation of L-type voltage-gated calcium channels (VGCCs) and purinoceptors induced a two-step process of neuronal differentiation. Specifically, L-type calcium channel-mediated augmentation of spike-like calcium oscillations first promoted stable expression of ASCL-1 in differentiating ESCs, which following P2Y2 purinoceptor activation matured into GABAergic neurons. By contrast, there was neither spike-like calcium oscillations nor responsive P2Y2 receptors in HD-modeling stem cells in vitro. The data shed new light on mechanisms underlying neurogenesis of inhibitory neurons. Moreover, our approach may be tailored to identify pathogenic triggers of other developmental neurological disorders for devising targeted therapies.


Subject(s)
Huntington Disease , Neural Stem Cells , Adenosine Triphosphate , Animals , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Calcium Signaling , Cell Differentiation , Embryonic Stem Cells/metabolism , GABAergic Neurons/metabolism , Huntington Disease/genetics , Mice , Neural Stem Cells/metabolism , Neurogenesis
7.
Proc Jpn Acad Ser B Phys Biol Sci ; 96(8): 351-363, 2020.
Article in English | MEDLINE | ID: mdl-33041269

ABSTRACT

Neural stem cells (NSCs) actively proliferate and generate neurons and glial cells (active state) in the embryonic brain, whereas they are mostly dormant (quiescent state) in the adult brain. The expression dynamics of Hes1 are different between active and quiescent NSCs. In active NSCs, Hes1 expression oscillates and periodically represses the expression of proneural genes such as Ascl1, thereby driving their oscillations. By contrast, in quiescent NSCs, Hes1 oscillations maintain expression at higher levels even at trough phases (thus continuous), thereby continuously suppressing proneural gene expression. High levels of Hes1 expression and the resultant suppression of Ascl1 promote the quiescent state of NSCs, whereas oscillatory Hes1 expression and the resultant oscillatory Ascl1 expression regulate their active state. Furthermore, in other developmental contexts, high, continuous Hes1 expression induces astrocyte differentiation or the formation of boundaries, which function as signaling centers. Thus, the expression dynamics of Hes1 are a key regulatory mechanism generating and maintaining various cell types in the nervous system.


Subject(s)
Neural Stem Cells/metabolism , Transcriptome , Animals , Humans , Neural Stem Cells/cytology , Signal Transduction/genetics , Transcription Factor HES-1/genetics
8.
Development ; 147(4)2020 02 26.
Article in English | MEDLINE | ID: mdl-32094111

ABSTRACT

The expression of the transcriptional repressor Hes1 oscillates in many cell types, including neural progenitor cells (NPCs), but the significance of Hes1 oscillations in development is not fully understood. To examine the effect of altered oscillatory dynamics of Hes1, we generated two types of Hes1 knock-in mice, a shortened (type-1) and an elongated (type-2) Hes1 gene, and examined their phenotypes focusing on neural development. Although both mutations affected Hes1 oscillations, the type-1 mutation dampened Hes1 oscillations more severely, resulting in much lower amplitudes. The average levels of Hes1 expression in type-1 mutant NPCs were also lower than in wild-type NPCs but similar to or slightly higher than those in Hes1 heterozygous mutant mice, which exhibit no apparent defects. Whereas type-2 mutant mice were apparently normal, type-1 mutant mice displayed smaller brains than wild-type mice and upregulated proneural gene expression. Furthermore, proliferation of NPCs decreased and cell death increased in type-1 mutant embryos. When Hes3 and Hes5 were additionally deleted, neuronal differentiation was also accelerated, leading to microcephaly. Thus, robust Hes1 oscillations are required for maintenance and proliferation of NPCs and the normal timing of neurogenesis, thereby regulating brain morphogenesis.


Subject(s)
Brain/embryology , Neurons/physiology , Oscillometry , Transcription Factor HES-1/physiology , Animals , Cell Death , Cell Differentiation , Cell Proliferation , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Homozygote , Image Processing, Computer-Assisted , Introns , Male , Mice , Models, Theoretical , Mutation , Neural Stem Cells/cytology , Neurogenesis
9.
Dev Cell ; 52(6): 731-747.e8, 2020 03 23.
Article in English | MEDLINE | ID: mdl-32059775

ABSTRACT

Notch signaling controls proliferation of multipotent pancreatic progenitor cells (MPCs) and their segregation into bipotent progenitors (BPs) and unipotent pro-acinar cells (PACs). Here, we showed that fast ultradian oscillations of the ligand Dll1 and the transcriptional effector Hes1 were crucial for MPC expansion, and changes in Hes1 oscillation parameters were associated with selective adoption of BP or PAC fate. Conversely, Jag1, a uniformly expressed ligand, restrained MPC growth. However, when its expression later segregated to PACs, Jag1 became critical for the specification of all but the most proximal BPs, and BPs were entirely lost in Jag1; Dll1 double mutants. Anatomically, ductal morphogenesis and organ architecture are minimally perturbed in Jag1 mutants until later stages, when ductal remodeling fails, and signs of acinar-to-ductal metaplasia appear. Our study thus uncovers that oscillating Notch activity in the developing pancreas, modulated by Jag1, is required to coordinate MPC growth and fate.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/metabolism , Jagged-1 Protein/metabolism , Pancreas/cytology , Signal Transduction , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Lineage , Embryonic Stem Cells/cytology , Female , Gene Expression Regulation, Developmental , Jagged-1 Protein/genetics , Male , Mice , Mice, Inbred C57BL , Mutation , Pancreas/embryology , Pancreas/metabolism , Periodicity , Receptors, Notch/genetics , Receptors, Notch/metabolism , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism
10.
J Vis Exp ; (154)2019 12 12.
Article in English | MEDLINE | ID: mdl-31885384

ABSTRACT

Notch signaling regulates the maintenance of neural stem/progenitor cells by cell-cell interactions. The components of Notch signaling exhibit dynamic expression. Notch signaling effector Hes1 and the Notch ligand Delta-like1 (Dll1) are expressed in an oscillatory manner in neural stem/progenitor cells. Because the period of the oscillatory expression of these genes is very short (2 h), it is difficult to monitor their cyclic expression. To examine such rapid changes in the gene expression or protein dynamics, fast response reporters are required. Because of its fast maturation kinetics and high sensitivity, the bioluminescence reporter luciferase is suitable to monitor rapid gene expression changes in living cells. We used a destabilized luciferase reporter for monitoring the promoter activity and a luciferase-fused reporter for visualization of protein dynamics at single cell resolution. These bioluminescence reporters show rapid turnover and generate very weak signals; therefore, we have developed a highly sensitive bioluminescence imaging system to detect such faint signals. These methods enable us to monitor various gene expression dynamics in living cells and tissues, which are important information to help understand the actual cellular states.


Subject(s)
Luminescent Measurements/methods , Neurogenesis/physiology , Receptors, Notch/physiology , Animals , Female , Gene Expression Regulation, Developmental , Mice , Mice, Inbred ICR , Neural Stem Cells/physiology , Signal Transduction/physiology
11.
Neurosci Res ; 138: 12-18, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30227160

ABSTRACT

During brain development, neural stem cells change their competency to give sequential rise to neurons and glial cells. Expression of the basic helix-loop-helix (bHLH)-type cell-fate determination factors Ascl1, Olig2, and Hes1 is oscillatory in neural stem cells. Conversely, sustained expression of these factors mediates cell-fate determination. Optogenetic analyses suggest that oscillatory expression regulates maintenance and proliferation of neural stem cells, and that sustained expression induces cell-fate determination. Expression of the Notch ligand Delta-like1 (Dll1), which is controlled by Hes1 and Ascl1, is also oscillatory in neural stem cells. Mathematical modeling showed that if the timing of Dll1 expression is changed, Hes1 oscillations are severely dampened, resulting in impaired maintenance and proliferation of neural stem cells and causing microcephaly. Another bHLH factor, Hes5, also shows oscillatory expression in neural stem cells. Hes5 overexpression and knock-out result in abnormal expression of Hmga1 and Hmga2, which are essential for timing the switching of neural stem-cell competency. These data indicate that oscillatory expression of bHLH factors is important for normal neural stem-cell function in the developing nervous system.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Gene Expression Regulation, Developmental/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Animals , Models, Biological
12.
Adv Exp Med Biol ; 1066: 265-277, 2018.
Article in English | MEDLINE | ID: mdl-30030831

ABSTRACT

The Notch effectors Hes1 and Hes7 and the Notch ligand Delta-like1 (Dll1) are expressed in an oscillatory manner during neurogenesis and somitogenesis. These two biological events exhibit different types of oscillations: anti-/out-of-phase oscillation in neural stem cells during neurogenesis and in-phase oscillation in presomitic mesoderm (PSM) cells during somitogenesis. Accelerated or delayed Dll1 expression by shortening or elongating the size of the Dll1 gene, respectively, dampens or quenches Dll1 oscillation at intermediate levels, a phenomenon known as "amplitude/oscillation death" of coupled oscillators. Under this condition, both Hes1 oscillation in neural stem cells and Hes7 oscillation in PSM cells are also dampened. As a result, maintenance of neural stem cells is impaired, leading to microcephaly, while somite segmentation is impaired, leading to severe fusion of somites and their derivatives, such as vertebrae and ribs. Thus, the appropriate timing of Dll1 expression is critical for the oscillatory expression in Notch signaling and normal processes of neurogenesis and somitogenesis. Optogenetic analysis indicated that Dll1 oscillations transfer the oscillatory information between neighboring cells, which may induce anti-/out-of-phase and in-phase oscillations depending on the delay in signaling transmission. These oscillatory dynamics can be described in a unified manner by mathematical modeling.


Subject(s)
Biological Clocks/physiology , Embryo, Mammalian/embryology , Gene Expression Regulation, Developmental/physiology , Models, Biological , Receptors, Notch/metabolism , Signal Transduction/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Calcium-Binding Proteins , Embryo, Mammalian/cytology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mesoderm/cytology , Mesoderm/embryology , Receptors, Notch/genetics , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism
13.
J Biol Chem ; 293(21): 8285-8294, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29523683

ABSTRACT

The transcription factor Hes family basic helix-loop-helix transcription factor 1 (Hes1) is a downstream effector of Notch signaling and plays a crucial role in orchestrating developmental processes during the embryonic stage. However, its aberrant signaling in adulthood is linked to the pathogenesis of cancer. In the present study, we report the discovery of small organic molecules (JI051 and JI130) that impair the ability of Hes1 to repress transcription. Hes1 interacts with the transcriptional corepressor transducing-like enhancer of split 1 (TLE1) via an interaction domain comprising two tryptophan residues, prompting us to search a chemical library of 1,800 small molecules enriched for indole-like π-electron-rich pharmacophores for a compound that blocks Hes1-mediated transcriptional repression. This screening identified a lead compound whose extensive chemical modification to improve potency yielded JI051, which inhibited HEK293 cell proliferation with an EC50 of 0.3 µm Unexpectedly, using immunomagnetic isolation and nanoscale LC-MS/MS, we found that JI051 does not bind TLE1 but instead interacts with prohibitin 2 (PHB2), a cancer-associated protein chaperone. We also found that JI051 stabilizes PHB2's interaction with Hes1 outside the nucleus, inducing G2/M cell-cycle arrest. Of note, JI051 dose-dependently reduced cell growth of the human pancreatic cancer cell line MIA PaCa-2, and JI130 treatment significantly reduced tumor volume in a murine pancreatic tumor xenograft model. These results suggest a previously unrecognized role for PHB2 in the regulation of Hes1 and may inform potential strategies for managing pancreatic cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , High-Throughput Screening Assays , Pancreatic Neoplasms/drug therapy , Repressor Proteins/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Transcription Factor HES-1/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Cell Cycle , Cell Differentiation , Cell Proliferation , Female , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prohibitins , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism , Transcription, Genetic , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Dev Cell ; 36(4): 358-9, 2016 Feb 22.
Article in English | MEDLINE | ID: mdl-26906731

ABSTRACT

A study by Tsiairis and Aulehla (2016), published in Cell, reveals that randomized mixtures of dissociated presomitic mesoderm (PSM) cells reestablish wave-like propagating patterns of oscillatory gene expression by tuning the oscillation dynamics in response to surrounding cells. Thus, PSM cells self-organize the phase-coupled oscillators.


Subject(s)
Biological Clocks , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Mesoderm/metabolism , Organizers, Embryonic/metabolism , Animals
15.
Semin Cell Dev Biol ; 49: 76-82, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26818178

ABSTRACT

During somite segmentation, mRNA expression of the mouse Notch ligand Delta-like1 (Dll1) oscillates synchronously in the presomitic mesoderm (PSM). However, the dynamics of Dll1 protein expression were rather controversial, and their functional significance was not known. Recent live-imaging analysis showed that Dll1 protein expression also oscillates synchronously in the PSM. Interestingly, accelerated or delayed Dll1 expression by shortening or elongating the Dll1 gene, respectively, dampens or quenches Dll1 oscillation at intermediate levels, a phenomenon known as "amplitude/oscillation death" of coupled oscillators in mathematical modeling. Under this condition, oscillation of the Notch effector Hes7 is also dampened, leading to severe fusion of somites and their derivatives, such as vertebrae and ribs. Thus, the appropriate timing of Dll1 expression is critical for its oscillatory expression, pointing to the functional significance of Dll1-mediated oscillatory cell-cell interactions in the segmentation clock. In neural stem cells, Dll1 expression is also oscillatory, but non-synchronous, and when Dll1 oscillation is dampened, oscillation of another Notch effector, Hes1, is also dampened, leading to defects of neural development. In this review, we discuss the underlying mechanism for the different oscillatory dynamics (synchronous versus non-synchronous) in the PSM and neural stem cells in a unified manner.


Subject(s)
Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Neurogenesis , Receptors, Notch/physiology , Somites/metabolism , Animals , Body Patterning , Calcium-Binding Proteins , Embryonic Development , Gene Expression , Gene Expression Regulation, Developmental , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neural Stem Cells/physiology , Signal Transduction
16.
Genes Dev ; 30(1): 102-16, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26728556

ABSTRACT

Notch signaling regulates tissue morphogenesis through cell-cell interactions. The Notch effectors Hes1 and Hes7 are expressed in an oscillatory manner and regulate developmental processes such as neurogenesis and somitogenesis, respectively. Expression of the mRNA for the mouse Notch ligand Delta-like1 (Dll1) is also oscillatory. However, the dynamics of Dll1 protein expression are controversial, and their functional significance is unknown. Here, we developed a live-imaging system and found that Dll1 protein expression oscillated in neural progenitors and presomitic mesoderm cells. Notably, when Dll1 expression was accelerated or delayed by shortening or elongating the Dll1 gene, Dll1 oscillations became severely dampened or quenched at intermediate levels, as modeled mathematically. Under this condition, Hes1 and Hes7 oscillations were also dampened. In the presomitic mesoderm, steady Dll1 expression led to severe fusion of somites and their derivatives, such as vertebrae and ribs. In the developing brain, steady Dll1 expression inhibited proliferation of neural progenitors and accelerated neurogenesis, whereas optogenetic induction of Dll1 oscillation efficiently maintained neural progenitors. These results indicate that the appropriate timing of Dll1 expression is critical for the oscillatory networks and suggest the functional significance of oscillatory cell-cell interactions in tissue morphogenesis.


Subject(s)
Gene Expression Regulation, Developmental , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Morphogenesis/physiology , Neurons/metabolism , Stem Cells/metabolism , Animals , Calcium-Binding Proteins , Cell Communication , Cell Proliferation , Cells, Cultured , Gene Knock-In Techniques , Mesoderm/cytology , Mesoderm/embryology , Mesoderm/metabolism , Mice , Models, Biological , Mutation , Neurogenesis/genetics , Neurons/cytology , Receptors, Notch/genetics , Signal Transduction/genetics , Somites/embryology , Stem Cells/cytology , Time-Lapse Imaging
17.
Cell Tissue Res ; 359(1): 125-33, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24850276

ABSTRACT

The basic helix-loop-helix factors Hes1 and Hes5 repress the expression of proneural factors such as Ascl1, thereby inhibiting neuronal differentiation and maintaining neural progenitor cells (NPCs). Hes1 expression oscillates by negative feedback with a period of about 2-3 h in proliferating NPCs. Induction of sustained expression of Hes1 in NPCs inhibits their cell-cycle progression, suggesting that the oscillatory expression of Hes1 is important for the proliferation of NPCs. Hes1 oscillation drives the oscillatory expression of proneural factors such as Ascl1 by periodic repression. By contrast, in differentiating neurons, Hes1 expression disappears and the expression of proneural factors is up-regulated and sustained. A new optogenetics approach that induces Ascl1 expression by blue light illumination demonstrated that sustained expression of Ascl1 induces neuronal differentiation, whereas oscillatory expression of Ascl1 activates the proliferation of NPCs. These results together indicate that Hes1 regulates the oscillatory versus sustained expression of the proneural factor Ascl1, which in turn regulates the proliferation of NPCs and the subsequent processes of cell-cycle exit and neuronal fate determination, depending on the expression dynamics.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Nervous System/growth & development , Neurogenesis , Animals , Humans , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Optogenetics , Receptors, Notch/metabolism
18.
Methods Mol Biol ; 1187: 169-79, 2014.
Article in English | MEDLINE | ID: mdl-25053489

ABSTRACT

The Notch signaling effectors Hes1 and Hes7 exhibit oscillatory expression with a period of about 2-3 h during embryogenesis. Hes1 oscillation is important for proliferation and differentiation of neural stem cells, whereas Hes7 oscillation regulates periodic formation of somites. Continuous expression of Hes1 and Hes7 inhibits these developmental processes. Thus, expression dynamics are very important for gene functions, but it is difficult to distinguish between oscillatory and persistent expression by conventional methods such as in situ hybridization and immunostaining. Here, we describe time-lapse imaging methods using destabilized luciferase reporters and a highly sensitive cooled charge-coupled device camera, which can monitor dynamic gene expression. Furthermore, the expression of two genes can be examined simultaneously by a dual reporter system using two-color luciferase reporters. Time-lapse imaging analyses reveal how dynamically gene expression changes in many biological events.


Subject(s)
Gene Expression Regulation, Developmental , Optical Imaging/methods , Receptors, Notch/metabolism , Signal Transduction , Animals , Basic Helix-Loop-Helix Transcription Factors/analysis , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Culture Techniques/methods , Cell Separation/methods , Cells, Cultured , Embryo Culture Techniques/methods , Genes, Reporter , Homeodomain Proteins/analysis , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Luciferases/analysis , Luciferases/genetics , Luciferases/metabolism , Luminescent Agents/analysis , Luminescent Agents/metabolism , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis , Receptors, Notch/analysis , Receptors, Notch/genetics , Tissue Culture Techniques/methods , Transcription Factor HES-1
19.
Semin Cell Dev Biol ; 34: 85-90, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24865153

ABSTRACT

Somites, metameric structures, give rise to the vertebral column, ribs, skeletal muscles and subcutaneous tissues. In mouse embryos, a pair of somites is formed every 2h by segmentation of the anterior parts of the presomitic mesoderm. This periodic event is regulated by a biological clock called the segmentation clock, which involves cyclic expression of the basic helix-loop-helix gene Hes7. Hes7 oscillation is regulated by negative feedback with a delayed timing. This process has been mathematically simulated by differential-delay equations, which predict that negative feedback with shorter delays would abolish oscillations or produce dampened but more rapid oscillations. We found that reducing the number of introns within the Hes7 gene shortens the delay and abolishes Hes7 oscillation or results in a more rapid tempo of Hes7 oscillation, increasing the number of somites and vertebrae in the cervical and upper thoracic region. We also found that Hes1, a Hes7-related gene, is expressed in an oscillatory manner by many cell types, including fibroblasts and neural stem cells. In these cells, Hes1 expression oscillates with a period of about 2-3h, and this oscillation is important for cell cycle progression. Furthermore, in neural stem cells, Hes1 oscillation drives cyclic expression of the proneural genes Ascl1 and Neurogenin2 and regulates multipotency. Hes1 expression oscillates more slowly in embryonic stem cells, and Hes1 oscillation regulates their fate preferences. Taken together, these results suggest that oscillatory expression with short periods (ultradian oscillation) is important for many biological events.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Homeodomain Proteins/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Circadian Rhythm , Feedback, Physiological , Gene Expression Regulation , Homeodomain Proteins/metabolism , Humans , Mice , Neural Stem Cells/physiology , Protein Stability , Signal Transduction , Transcription Factor HES-1
20.
Cell Mol Life Sci ; 70(12): 2045-57, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22971775

ABSTRACT

Notch signaling plays crucial roles in fate determination and the differentiation of neural stem cells in embryonic and adult brains. It is now clear that the notch pathway is under more complex and dynamic regulation than previously thought. To understand the functional details of notch signaling more precisely, it is important to reveal when, where, and how notch signaling is dynamically communicated between cells, for which the visualization of notch signaling is essential. In this review, we introduce recent technical advances in the visualization of notch signaling during neural development and in the adult brain, and we discuss the physiological significance of dynamic regulation of notch signaling.


Subject(s)
Brain/embryology , Brain/growth & development , Gene Expression Regulation/physiology , Neurogenesis/physiology , Receptors, Notch/metabolism , Signal Transduction/physiology , Animals , Gene Expression Regulation/genetics , Green Fluorescent Proteins , Humans , Luciferases , Mice , Mice, Transgenic , Models, Biological , Neurogenesis/genetics , Receptors, Notch/genetics , Signal Transduction/genetics , beta-Galactosidase
SELECTION OF CITATIONS
SEARCH DETAIL
...