49 results on '"NEUROG2"'
Search Results
2. Requirements for Neurogenin2 during mouse postnatal retinal neurogenesis
- Author
-
Kowalchuk, Angelica M, Maurer, Kate A, Shoja-Taheri, Farnaz, and Brown, Nadean L
- Subjects
Biomedical and Clinical Sciences ,Ophthalmology and Optometry ,Neurosciences ,Stem Cell Research - Nonembryonic - Non-Human ,Eye Disease and Disorders of Vision ,Stem Cell Research ,Genetics ,Pediatric ,1.1 Normal biological development and functioning ,Eye ,Animals ,Basic Helix-Loop-Helix Transcription Factors ,Cell Differentiation ,Female ,Mice ,Mice ,Inbred C57BL ,Mice ,Inbred ICR ,Nerve Tissue Proteins ,Neurogenesis ,Pregnancy ,Repressor Proteins ,Retina ,Retinal Cone Photoreceptor Cells ,Retinal Neurons ,Retinal Rod Photoreceptor Cells ,Transcription Factors ,Neurog2 ,Blimp1/Prdm1 ,Rod photoreceptor ,Bipolar cell ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
During embryonic retinal development, the bHLH factor Neurog2 regulates the temporal progression of neurogenesis, but no role has been assigned for this gene in the postnatal retina. Using Neurog2 conditional mutants, we found that Neurog2 is necessary for the development of an early, embryonic cohort of rod photoreceptors, but also required by both a subset of cone bipolar subtypes, and rod bipolars. Using transcriptomics, we identified a subset of downregulated genes in P2 Neurog2 mutants, which act during rod differentiation, outer segment morphogenesis or visual processing. We also uncovered defects in neuronal cell culling, which suggests that the rod and bipolar cell phenotypes may arise via more complex mechanisms rather than a simple cell fate shift. However, given an overall phenotypic resemblance between Neurog2 and Blimp1 mutants, we explored the relationship between these two factors. We found that Blimp1 is downregulated between E12-birth in Neurog2 mutants, which probably reflects a dependence on Neurog2 in embryonic progenitor cells. Overall, we conclude that the Neurog2 gene is expressed and active prior to birth, but also exerts an influence on postnatal retinal neuron differentiation.
- Published
- 2018
3. Integral bHLH factor regulation of cell cycle exit and RGC differentiation
- Author
-
Maurer, Kate A, Kowalchuk, Angelica, Shoja‐Taheri, Farnaz, and Brown, Nadean L
- Subjects
Biochemistry and Cell Biology ,Bioinformatics and Computational Biology ,Evolutionary Biology ,Biological Sciences ,Stem Cell Research ,Neurosciences ,Genetics ,Eye Disease and Disorders of Vision ,Stem Cell Research - Nonembryonic - Non-Human ,Animals ,Basic Helix-Loop-Helix Transcription Factors ,Cell Cycle ,Cell Differentiation ,Embryo ,Mammalian ,Mice ,Nerve Tissue Proteins ,Neurogenesis ,Retinal Ganglion Cells ,Transcriptome ,Neurog2 ,Atoh7 ,Ascl ,retinal ganglion cell ,neurogenesis ,bHLH factor ,Medical and Health Sciences ,Developmental Biology ,Biochemistry and cell biology ,Bioinformatics and computational biology ,Evolutionary biology - Abstract
BACKGROUND:In the developing mouse embryo, the bHLH transcription factor Neurog2 is transiently expressed by retinal progenitor cells and required for the initial wave of neurogenesis. Remarkably, another bHLH factor, Ascl1, normally not present in the embryonic Neurog2 retinal lineage, can rescue the temporal phenotypes of Neurog2 mutants. RESULTS:Here we show that Neurog2 simultaneously promotes terminal cell cycle exit and retinal ganglion cell differentiation, using mitotic window labeling and integrating these results with retinal marker quantifications. We also analyzed the transcriptomes of E12.5 GFP-expressing cells from Neurog2GFP/+ , Neurog2GFP/GFP , and Neurog2Ascl1KI/GFP eyes, and validated the most significantly affected genes using qPCR assays. CONCLUSIONS:Our data support the hypothesis that Neurog2 acts at the top of a retinal bHLH transcription factor hierarchy. The combined expression levels of these downstream factors are sufficiently induced by ectopic Ascl1 to restore RGC genesis, highlighting the robustness of this gene network during retinal ganglion cell neurogenesis. Developmental Dynamics 247:965-975, 2018. © 2018 Wiley Periodicals, Inc.
- Published
- 2018
4. Examining the NEUROG2 lineage and associated gene expression in human cortical organoids.
- Author
-
Vasan L, Chinchalongporn V, Saleh F, Zinyk D, Ke C, Suresh H, Ghazale H, Belfiore L, Touahri Y, Oproescu AM, Patel S, Rozak M, Amemiya Y, Han S, Moffat A, Black SE, McLaurin J, Near J, Seth A, Goubran M, Reiner O, Gillis J, Wang C, Okawa S, and Schuurmans C
- Subjects
- Humans, Neurogenesis genetics, Cell Lineage genetics, Gene Expression Regulation, Developmental, Neural Stem Cells metabolism, Neural Stem Cells cytology, Transcriptome genetics, T-Box Domain Proteins genetics, T-Box Domain Proteins metabolism, Human Embryonic Stem Cells metabolism, Human Embryonic Stem Cells cytology, Animals, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Organoids metabolism, Organoids cytology, Basic Helix-Loop-Helix Transcription Factors genetics, Basic Helix-Loop-Helix Transcription Factors metabolism, Cerebral Cortex metabolism, Cerebral Cortex cytology
- Abstract
Proneural genes are conserved drivers of neurogenesis across the animal kingdom. How their functions have adapted to guide human-specific neurodevelopmental features is poorly understood. Here, we mined transcriptomic data from human fetal cortices and generated from human embryonic stem cell-derived cortical organoids (COs) to show that NEUROG1 and NEUROG2 are most highly expressed in basal neural progenitor cells, with pseudotime trajectory analyses indicating that NEUROG1-derived lineages predominate early and NEUROG2 lineages later. Using ChIP-qPCR, gene silencing and overexpression studies in COs, we show that NEUROG2 is necessary and sufficient to directly transactivate known target genes (NEUROD1, EOMES, RND2). To identify new targets, we engineered NEUROG2-mCherry knock-in human embryonic stem cells for CO generation. The mCherry-high CO cell transcriptome is enriched in extracellular matrix-associated genes, and two genes associated with human-accelerated regions: PPP1R17 and FZD8. We show that NEUROG2 binds COL1A1, COL3A1 and PPP1R17 regulatory elements, and induces their ectopic expression in COs, although NEUROG2 is not required for this expression. Neurog2 similarly induces Col3a1 and Ppp1r17 in murine P19 cells. These data are consistent with a conservation of NEUROG2 function across mammalian species., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2025. Published by The Company of Biologists.)
- Published
- 2025
- Full Text
- View/download PDF
5. The Neurog2-Tbr2 axis forms a continuous transition to the neurogenic gene expression state in neural stem cells.
- Author
-
Shimojo, Hiromi, Masaki, Taimu, and Kageyama, Ryoichiro
- Subjects
- *
NEURAL stem cells , *GENE expression , *PROGENITOR cells , *CELL imaging , *CELL division - 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. [Display omitted] • Hes1-regulated Neurog2 oscillations lead to Tbr2 accumulation in NSCs • Tbr2 represses Hes1 expression, making an IPC-like state in NSCs • The transition from NSC to IPC-like states proceeds continuously in NSCs • Continuous transcriptional trajectory regulates discrete NSC-to-IPC differentiation Shimojo et al. show that Hes1-regulated Neurog2 oscillations lead to an accumulation of Tbr2, which then represses Hes1 in neural stem cells (NSCs) before differentiation into intermediate progenitor cells (IPCs). Thus, the transition from NSC (Hes1+;Tbr2−) to IPC-like gene expression (Hes1−;Tbr2+) states proceeds continuously during discrete NSC-to-IPC differentiation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Initiation of Otx2 expression in the developing mouse retina requires a unique enhancer and either Ascl1 or Neurog2 activity.
- Author
-
Kaufman, Michael L., Goodson, Noah B., Ko Uoon Park, Schwanke, Michael, Office, Emma, Schneider, Sophia R., Abraham, Joy, Hensley, Austin, Jones, Kenneth L., and Brzezinski, Joseph A.
- Subjects
- *
RETINA , *RNA sequencing , *BIPOLAR cells , *PROGENITOR cells , *CRISPRS , *PHOTORECEPTORS , *CIS-regulatory elements (Genetics) - Abstract
During retinal development, a large subset of progenitors upregulates the transcription factor Otx2, which is required for photoreceptor and bipolar cell formation. How these retinal progenitor cells initially activate Otx2 expression is unclear. To address this, we investigated the cis-regulatory network that controls Otx2 expression in mice. We identified a minimal enhancer element, DHS-4D, that drove expression in newly formed OTX2+ cells. CRISPR/Cas9-mediated deletion of DHS-4D reduced OTX2 expression, but this effect was diminished in postnatal development. Systematic mutagenesis of the enhancer revealed that three basic helix-loop-helix (bHLH) transcription factor-binding sites were required for its activity. Single cell RNA-sequencing of nascent Otx2+ cells identified the bHLH factors Ascl1 and Neurog2 as candidate regulators. CRISPR/Cas9 targeting of these factors showed that only the simultaneous loss of Ascl1 and Neurog2 prevented OTX2 expression. Our findings suggest that Ascl1 and Neurog2 act either redundantly or in a compensatory fashion to activate the DHS-4D enhancer and Otx2 expression. We observed redundancy or compensation at both the transcriptional and enhancer utilization levels, suggesting that the mechanisms governing Otx2 regulation in the retina are flexible and robust. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
7. Striped Distribution Pattern of Purkinje Cells of Different Birthdates in the Mouse Cerebellar Cortex Studied with the Neurog2-CreER Transgenic Line.
- Author
-
Zhang, Jingyun, Tran-Anh, Khoa, Hirata, Tatsumi, and Sugihara, Izumi
- Subjects
- *
CEREBELLAR cortex , *PURKINJE cells , *CELL aggregation , *MICE , *STRIPES - Abstract
[Display omitted] • Birthdates of Purkinje cells (PCs) were mapped in Neurog2 -CreER (G2A)::AldocV mice. • The birthdate-specific distribution was arranged into unique longitudinal stripes. • Each stripe contained PCs born in a particular period between E10.0 and E13.5. • A birthdate stripe either matched, included or subdivided a zebrin stripe. • The PC birthdate partially determines the zebrin stripe to which a PC belongs. Heterogeneity of Purkinje cells (PCs) that are arranged into discrete longitudinally-striped compartments in the cerebellar cortex is related to the timing of PC generation. To understand the cerebellar compartmental organization, we mapped the PC birthdate (or differentiation timing) in the entire cerebellar cortex. We used the birthdate-tagging system of Neurog2 -CreER (G2A) mice hybridized with the AldocV strain which visualizes the zebrin (aldolase C) longitudinal striped pattern. The birthdate-specific distribution pattern of PCs was arranged into longitudinally-oriented stripes consistently throughout almost all lobules except for the nodulus, paraflocculus, and flocculus, in which distinct stripes were observed. Boundaries of the birthdate stripes coincided with the boundary of zebrin stripes or located in the middle of a zebrin stripe. Each birthdate stripe contained PCs born in a particular period between embryonic day (E) 10.0 and E 13.5. In the vermis, PCs were chronologically distributed from lateral to medial stripes. In the paravermis, PCs of early birthdates were distributed in the long lateral zebrin-positive stripe (stripe 4+//5+) and the medially neighboring narrow zebrin-negative substripe (3d−//e2−), while PCs of late birthdates were distributed in the rest of all paravermal areas. In the hemisphere, PCs of early and late birthdates were intermingled in the majority of areas. The results indicate that the birthdate of a PC is a partial determinant for the zebrin compartment in which it is located. However, the correlation between the PC birthdate and the zebrin compartmentalization is complex and distinct among the vermis, paravermis, hemisphere, nodulus, and flocculus. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
8. New Insights Into the Intricacies of Proneural Gene Regulation in the Embryonic and Adult Cerebral Cortex
- Author
-
Ana-Maria Oproescu, Sisu Han, and Carol Schuurmans
- Subjects
Neurog1 ,Neurog2 ,Ascl1 ,phosphorylation ,protein–protein interactions ,protein stability ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Historically, the mammalian brain was thought to lack stem cells as no new neurons were found to be made in adulthood. That dogma changed ∼25 years ago with the identification of neural stem cells (NSCs) in the adult rodent forebrain. However, unlike rapidly self-renewing mature tissues (e.g., blood, intestinal crypts, skin), the majority of adult NSCs are quiescent, and those that become ‘activated’ are restricted to a few neurogenic zones that repopulate specific brain regions. Conversely, embryonic NSCs are actively proliferating and neurogenic. Investigations into the molecular control of the quiescence-to-proliferation-to-differentiation continuum in the embryonic and adult brain have identified proneural genes encoding basic-helix-loop-helix (bHLH) transcription factors (TFs) as critical regulators. These bHLH TFs initiate genetic programs that remove NSCs from quiescence and drive daughter neural progenitor cells (NPCs) to differentiate into specific neural cell subtypes, thereby contributing to the enormous cellular diversity of the adult brain. However, new insights have revealed that proneural gene activities are context-dependent and tightly regulated. Here we review how proneural bHLH TFs are regulated, with a focus on the murine cerebral cortex, drawing parallels where appropriate to other organisms and neural tissues. We discuss upstream regulatory events, post-translational modifications (phosphorylation, ubiquitinylation), protein–protein interactions, epigenetic and metabolic mechanisms that govern bHLH TF expression, stability, localization, and consequent transactivation of downstream target genes. These tight regulatory controls help to explain paradoxical findings of changes to bHLH activity in different cellular contexts.
- Published
- 2021
- Full Text
- View/download PDF
9. New Insights Into the Intricacies of Proneural Gene Regulation in the Embryonic and Adult Cerebral Cortex.
- Author
-
Oproescu, Ana-Maria, Han, Sisu, and Schuurmans, Carol
- Subjects
POST-translational modification ,CEREBRAL cortex ,GENETIC regulation ,NEURAL stem cells ,PROGENITOR cells ,PROTEIN-protein interactions - Abstract
Historically, the mammalian brain was thought to lack stem cells as no new neurons were found to be made in adulthood. That dogma changed ∼25 years ago with the identification of neural stem cells (NSCs) in the adult rodent forebrain. However, unlike rapidly self-renewing mature tissues (e.g., blood, intestinal crypts, skin), the majority of adult NSCs are quiescent, and those that become 'activated' are restricted to a few neurogenic zones that repopulate specific brain regions. Conversely, embryonic NSCs are actively proliferating and neurogenic. Investigations into the molecular control of the quiescence-to-proliferation-to-differentiation continuum in the embryonic and adult brain have identified proneural genes encoding basic-helix-loop-helix (bHLH) transcription factors (TFs) as critical regulators. These bHLH TFs initiate genetic programs that remove NSCs from quiescence and drive daughter neural progenitor cells (NPCs) to differentiate into specific neural cell subtypes, thereby contributing to the enormous cellular diversity of the adult brain. However, new insights have revealed that proneural gene activities are context-dependent and tightly regulated. Here we review how proneural bHLH TFs are regulated, with a focus on the murine cerebral cortex, drawing parallels where appropriate to other organisms and neural tissues. We discuss upstream regulatory events, post-translational modifications (phosphorylation, ubiquitinylation), protein–protein interactions, epigenetic and metabolic mechanisms that govern bHLH TF expression, stability, localization, and consequent transactivation of downstream target genes. These tight regulatory controls help to explain paradoxical findings of changes to bHLH activity in different cellular contexts. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
10. Neurog2 Acts as a Classical Proneural Gene in the Ventromedial Hypothalamus and Is Required for the Early Phase of Neurogenesis.
- Author
-
Aslanpour, Shaghayegh, Sisu Han, Schuurmans, Carol, and Kurrasch, Deborah M.
- Subjects
- *
HYPOTHALAMUS , *DEVELOPMENTAL neurobiology , *GENES , *PREOPTIC area , *PHYSIOLOGY , *NEURONS - Abstract
The tuberal hypothalamus is comprised of the dorsomedial, ventromedial, and arcuate nuclei, as well as parts of the lateral hypothalamic area, and it governs a wide range of physiologies. During neurogenesis, tuberal hypothalamic neurons are thought to be born in a dorsal-to-ventral and outside-in pattern, although the accuracy of this description has been questioned over the years. Moreover, the intrinsic factors that control the timing of neurogenesis in this region are poorly characterized. Proneural genes, including Achate-scute-like 1 (Ascii) and Neurogenin 3 (Neurog3) are widely expressed in hypothalamic progenitors and contribute to lineage commitment and subtype-specific neuronal identifies, but the potential role of Neurogenin 2 (Neurog2) remains unexplored. Birthdating in male and female mice showed that tuberal hypothalamic neurogenesis begins as early as E9.5 in the lateral hypothalamic and arcuate and rapidly expands to dorsomedial and ventromedial neurons by E10.5, peaking throughout the region by El 1.5. We confirmed an outside-in trend, except for neurons born at E9.5, and uncovered a rostrocaudal progression but did not confirm a dorsal-ventral patterning to tuberal hypothalamic neuronal birth. In the absence of Neurog2, neurogenesis stalls, with a significant reduction in early-born BrdU+ cells but no change at later time points. Further, the loss of Ascii yielded a similar delay in neuronal birth, suggesting that Ascii cannot rescue the loss of Neurog2 and that these proneural genes act independently in the tuberal hypothalamus. Together, our findings show that Neurog2 functions as a classical proneural gene to regulate the temporal progression of tuberal hypothalamic neurogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
11. Fbxo9 functions downstream of Sox10 to determine neuron-glial fate choice in the dorsal root ganglia through Neurog2 destabilization.
- Author
-
Aijia Liu, Jessica, Tai, Andrew, Jialin Hong, May Pui Lai Cheung, Mai Har Sham, Cheah, Kathryn S. E., Chi Wai Cheung, and Cheung, Martin
- Subjects
- *
DORSAL root ganglia , *NEURAL crest , *NEUROGLIA , *SENSORY neurons , *CELL determination - Abstract
The transcription factor Sox10 is a key regulator in the fate determination of a subpopulation of multipotent trunk neural crest (NC) progenitors toward glial cells instead of sensory neurons in the dorsal root ganglia (DRG). However, the mechanism by which Sox10 regulates glial cell fate commitment during lineage segregation remains poorly understood. In our study, we showed that the neurogenic determinant Neurogenin 2 (Neurog2) exhibited transient overlapping expression with Sox10 in avian trunk NC progenitors, which progressively underwent lineage segregation during migration toward the forming DRG. Gain- and loss-of-function studies revealed that the temporary expression of Neurog2 was due to Sox10 regulation of its protein stability. Transcriptional profiling identified Sox10-regulated F-box only protein (Fbxo9), which is an SCF (Skp1-Cul-F-box)-type ubiquitin ligase for Neurog2. Consistently, overexpression of Fbxo9 in NC progenitors down-regulated Neurog2 protein expression through ubiquitination and promoted the glial lineage at the expense of neuronal differentiation, whereas Fbxo9 knockdown resulted in the opposite phenomenon. Mechanistically, we found that Fbxo9 interacted with Neurog2 to promote its destabilization through the F-box motif. Finally, epistasis analysis further demonstrated that Fbxo9 and probably other F-box members mediated the role of Sox10 in destabilizing Neurog2 protein and directing the lineage of NC progenitors toward glial cells rather than sensory neurons. Altogether, these findings unravel a Sox10-Fbxo9 regulatory axis in promoting the glial fate of NC progenitors through Neurog2 destabilization. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
12. ETV5 is Essential for Neuronal Differentiation of Human Neural Progenitor Cells by Repressing NEUROG2 Expression.
- Author
-
Liu, Yang and Zhang, Yuanyuan
- Subjects
- *
PROGENITOR cells , *NEURONAL differentiation , *GABAERGIC neurons , *TRANSCRIPTION factors , *EMBRYONIC stem cells , *BINDING sites , *NEUROGLIA - Abstract
Neural progenitor cells (NPCs) are multipotent cells that have the potential to produce neurons and glial cells in the neural system. NPCs undergo identity maintenance or differentiation regulated by different kinds of transcription factors. Here we present evidence that ETV5, which is an ETS transcription factor, promotes the generation of glial cells and drives the neuronal subtype-specific genes in newly differentiated neurons from the human embryonic stem cells-derived NPCs. Next, we find a new role for ETV5 in the repression of NEUROG2 expression in NPCs. ETV5 represses NEUROG2 transcription via NEUROG2 promoter and requires the ETS domain. We identify ETV5 has the binding sites and is implicated in silent chromatin in NEUROG2 promoter by chromatin immunoprecipitation (ChIP) assays. Further, NEUROG2 transcription repression by ETV5 was shown to be dependent on a transcriptional corepressor (CoREST). During NPC differentiation toward neurons, ETV5 represses NEUROG2 expression and blocks the appearance of glutamatergic neurons. This finding suggests that ETV5 negatively regulates NEUROG2 expression and increases the number of GABAergic subtype neurons derived from NPCs. Thus, ETV5 represents a potent new candidate protein with benefits for the generation of GABAergic neurons. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
13. Small Molecules Modulate Chromatin Accessibility to Promote NEUROG2-Mediated Fibroblast-to-Neuron Reprogramming
- Author
-
Derek K. Smith, Jianjing Yang, Meng-Lu Liu, and Chun-Li Zhang
- Subjects
NEUROG2 ,SOX4 ,SWI/SNF ,CREB1 ,small molecules ,reprogramming ,transdifferentiation ,ATAC-seq ,ChIP-seq ,RNA-seq ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Pro-neural transcription factors and small molecules can induce the reprogramming of fibroblasts into functional neurons; however, the immediate-early molecular events that catalyze this conversion have not been well defined. We previously demonstrated that neurogenin 2 (NEUROG2), forskolin (F), and dorsomorphin (D) can reprogram fibroblasts into functional neurons with high efficiency. Here, we used this model to define the genetic and epigenetic events that initiate an acquisition of neuronal identity. We demonstrate that NEUROG2 is a pioneer factor, FD enhances chromatin accessibility and H3K27 acetylation, and synergistic transcription activated by these factors is essential to successful reprogramming. CREB1 promotes neuron survival and acts with NEUROG2 to upregulate SOX4, which co-activates NEUROD1 and NEUROD4. In addition, SOX4 targets SWI/SNF subunits and SOX4 knockdown results in extensive loss of open chromatin and abolishes reprogramming. Applying these insights, adult human glioblastoma cell and skin fibroblast reprogramming can be improved using SOX4 or chromatin-modifying chemicals.
- Published
- 2016
- Full Text
- View/download PDF
14. TAZ Represses the Neuronal Commitment of Neural Stem Cells
- Author
-
Natalia Robledinos-Antón, Maribel Escoll, Kun-Liang Guan, and Antonio Cuadrado
- Subjects
neural stem cells ,neurogenesis ,ASCL ,NEUROG2 ,NEUROD1 ,SOX2 ,Cytology ,QH573-671 - Abstract
The mechanisms involved in regulation of quiescence, proliferation, and reprogramming of Neural Stem Progenitor Cells (NSPCs) of the mammalian brain are still poorly defined. Here, we studied the role of the transcriptional co-factor TAZ, regulated by the WNT and Hippo pathways, in the homeostasis of NSPCs. We found that, in the murine neurogenic niches of the striatal subventricular zone and the dentate gyrus granular zone, TAZ is highly expressed in NSPCs and declines with ageing. Moreover, TAZ expression is lost in immature neurons of both neurogenic regions. To characterize mechanistically the role of TAZ in neuronal differentiation, we used the midbrain-derived NSPC line ReNcell VM to replicate in a non-animal model the factors influencing NSPC differentiation to the neuronal lineage. TAZ knock-down and forced expression in NSPCs led to increased and reduced neuronal differentiation, respectively. TEADs-knockdown indicated that these TAZ co-partners are required for the suppression of NSPCs commitment to neuronal differentiation. Genetic manipulation of the TAZ/TEAD system showed its participation in transcriptional repression of SOX2 and the proneuronal genes ASCL1, NEUROG2, and NEUROD1, leading to impediment of neurogenesis. TAZ is usually considered a transcriptional co-activator promoting stem cell proliferation, but our study indicates an additional function as a repressor of neuronal differentiation.
- Published
- 2020
- Full Text
- View/download PDF
15. Direct Lineage Reprogramming Reveals Disease-Specific Phenotypes of Motor Neurons from Human ALS Patients
- Author
-
Meng-Lu Liu, Tong Zang, and Chun-Li Zhang
- Subjects
ALS disease ,direct reprogramming ,motor neuron ,NEUROG2 ,small molecules ,kenpaullone ,Biology (General) ,QH301-705.5 - Abstract
Subtype-specific neurons obtained from adult humans will be critical to modeling neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS). Here, we show that adult human skin fibroblasts can be directly and efficiently converted into highly pure motor neurons without passing through an induced pluripotent stem cell stage. These adult human induced motor neurons (hiMNs) exhibit the cytological and electrophysiological features of spinal motor neurons and form functional neuromuscular junctions (NMJs) with skeletal muscles. Importantly, hiMNs converted from ALS patient fibroblasts show disease-specific degeneration manifested through poor survival, soma shrinkage, hypoactivity, and an inability to form NMJs. A chemical screen revealed that the degenerative features of ALS hiMNs can be remarkably rescued by the small molecule kenpaullone. Taken together, our results define a direct and efficient strategy to obtain disease-relevant neuronal subtypes from adult human patients and reveal their promising value in disease modeling and drug identification.
- Published
- 2016
- Full Text
- View/download PDF
16. Instructing neuronal identity during CNS development and astroglial-lineage reprogramming: Roles of NEUROG2 and ASCL1.
- Author
-
Chouchane, Malek and Costa, Marcos R.
- Subjects
- *
MOLECULAR probes , *ASTROCYTES - Abstract
Highlights • NEUROG2/ASCL1 instruct different neuronal fates in NPCs and reprogrammed astroglia. • Origin of astroglia affects the acquisition of final neuronal phenotype of iNs. • Reprogramming of astroglia is a powerful tool to probe neuronal fate specification. Abstract The adult mammalian brain contains an enormous variety of neuronal types, which are generally categorized in large groups, based on their neurochemical identity, hodological properties and molecular markers. This broad classification has allowed the correlation between individual neural progenitor populations and their neuronal progeny, thus contributing to probe the cellular and molecular mechanisms involved in neuronal identity determination during central nervous system (CNS) development. In this review, we discuss the contribution of the proneural genes Neurogenin2 (Neurog2) and Achaete-scute homolog 1 (Ascl1) for the specification of neuronal phenotypes in the developing neocortex, cerebellum and retina. Then, we revise recent data on astroglia cell lineage reprogramming into induced neurons using the same proneural proteins to compare the neuronal phenotypes obtained from astroglial cells originated in those CNS regions. We conclude that Ascl1 and Neurog2 have different contributions to determine neuronal fates, depending on the neural progenitor or astroglial population expressing those proneural factors. Finally, we discuss some possible explanations for these seemingly conflicting effects of Ascl1 and Neurog2 and propose future approaches to further dissect the molecular mechanisms of neuronal identity specification. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
17. A long range distal enhancer controls temporal fine-tuning of PAX6 expression in neuronal precursors.
- Author
-
Lacomme, Marine, Medevielle, François, Bourbon, Henri-Marc, Thierion, Elodie, Kleinjan, Dirk-Jan, Roussat, Mélanie, Pituello, Fabienne, and Bel-Vialar, Sophie
- Subjects
- *
GENE expression , *PROTEIN precursors , *EMBRYOLOGY , *CELLULAR signal transduction , *NEURONS - Abstract
Proper embryonic development relies on a tight control of spatial and temporal gene expression profiles in a highly regulated manner. One good example is the ON/OFF switching of the transcription factor PAX6 that governs important steps of neurogenesis. In the neural tube PAX6 expression is initiated in neural progenitors through the positive action of retinoic acid signaling and downregulated in neuronal precursors by the bHLH transcription factor NEUROG2. How these two regulatory inputs are integrated at the molecular level to properly fine tune temporal PAX6 expression is not known. In this study we identified and characterized a 940-bp long distal cis -regulatory module (CRM), located far away from the PAX6 transcription unit and which conveys positive input from RA signaling pathway and indirect repressive signal(s) from NEUROG2. These opposing regulatory signals are integrated through HOMZ, a 94 bp core region within E940 which is evolutionarily conserved in distant organisms such as the zebrafish. We show that within HOMZ, NEUROG2 and RA exert their opposite temporal activities through a short 60 bp region containing a functional RA-responsive element (RARE). We propose a model in which retinoic acid receptors (RARs) and NEUROG2 repressive target(s) compete on the same DNA motif to fine tune temporal PAX6 expression during the course of spinal neurogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
18. KDM3A-mediated demethylation of histone H3 lysine 9 facilitates the chromatin binding of Neurog2 during neurogenesis.
- Author
-
Hao Lin, Xuechen Zhu, Geng Chen, Lei Song, Li Gao, Khand, Aftab A., Ying Chen, Lin, Gufa, and Qinghua Tao
- Subjects
- *
DEMETHYLATION , *HISTONES , *DEVELOPMENTAL neurobiology - Abstract
Neurog2 is a crucial regulator of neuronal fate specification and differentiation in vivo and in vitro. However, it remains unclear how Neurog2 transactivates neuronal genes that are silenced by repressive chromatin. Here, we provide evidence that the histone H3 lysine 9 demethylase KDM3A facilitates the Xenopus Neurog2 (formerly known as Xngnr1) chromatin accessibility during neuronal transcription. Loss-of-function analyses reveal that KDM3A is not required for the transition of naive ectoderm to neural progenitor cells but is essential for primary neuron formation. ChIP series followed by qPCR analyses reveal that Neurog2 promotes the removal of the repressive H3K9me2 marks and addition of active histone marks, including H3K27ac and H3K4me3, at the NeuroD1 and Tubb2b promoters; this activity depends on the presence of KDM3A because Neurog2, via its C-terminal domain, interacts with KDM3A. Interestingly, KDM3A is dispensable for the neuronal transcription initiated by Ascl1, a proneural factor related to neurogenin in the bHLH family. In summary, our findings uncover a crucial role for histone H3K9 demethylation during Neurog2-mediated neuronal transcription and help in the understanding of the different activities of Neurog2 and Ascl1 in initiating neuronal development. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
19. DISC1 Regulates the Proliferation and Migration of Mouse Neural Stem/Progenitor Cells through Pax5, Sox2, Dll1 and Neurog2.
- Author
-
Qian Wu, Weiting Tang, Zhaohui Luo, Yi Li, Yi Shu, Zongwei Yue, Bo Xiao, and Li Feng
- Subjects
NEURAL stem cells ,DEVELOPMENTAL neurobiology ,CELL proliferation ,CELL migration ,PROGENITOR cells ,LABORATORY mice - Abstract
Disrupted-in-schizophrenia 1 (DISC1) regulates neurogenesis and is a genetic risk factor for major psychiatric disorders. However, how DISC1 dysfunction affects neurogenesis and cell cycle progression at the molecular level is still unknown. Here, we investigated the role of DISC1 in regulating proliferation, migration, cell cycle progression and apoptosis in mouse neural stem/progenitor cells (MNSPCs) in vitro. Methods: MNSPCs were isolated and cultured from mouse fetal hippocampi. Retroviral vectors or siRNAs were used to manipulate DISC1 expression in MNSPCs. Proliferation, migration, cell cycle progression and apoptosis of altered MNSPCs were analyzed in cell proliferation assays (MTS), transwell system and flow cytometry. A neurogenesis specific polymerase chain reaction (PCR) array was used to identify genes downstream of DISC1, and functional analysis was performed through transfection of expression plasmids and siRNAs. Results: Loss of DISC1 reduced proliferation and migration of MNSPCs, while an increase in DISC1 led to increased proliferation and migration. Meanwhile, an increase in the proportion of cells in G0/G1 phase was concomitant with reduced levels of DISC1, but significant changes were not observed in the number MNSPCs undergoing apoptosis. Paired box gene 5 (Pax5), sex determining region Y-box 2 (Sox2), deltalike1 (Dll1) and Neurogenin2 (Neurog2) emerged as candidate molecules downstream of DISC1, and rescue experiments demonstrated that increased or decreased expression of either molecule regulated proliferation and migration in DISC1-altered MNSPCs. Conclusion: These results suggest that Pax5, Sox2, Dll1 and Neurog2 mediate DISC1 activity in MNSPC proliferation and migration. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
20. Oligodendrocyte development in the embryonic tuberal hypothalamus and the influence of Ascl1.
- Author
-
Marsters, Candace M., Rosin, Jessica M., Thornton, Hayley F., Aslanpour, Shaghayegh, Klenin, Natasha, Wilkinson, Grey, Schuurmans, Carol, Pittman, Quentin J., and Kurrasch, Deborah M.
- Subjects
- *
HYPOTHALAMUS , *OLIGODENDROGLIA , *DEVELOPMENTAL cytology , *CENTRAL nervous system , *NEUROGLIA , *NEUROGENETICS - Abstract
Background: Although the vast majority of cells in our brains are glia, we are only beginning to understand programs governing their development, especially within the embryonic hypothalamus. In mice, gliogenesis is a protracted process that begins during embryonic stages and continues into the early postnatal period, with glial progenitors first producing oligodendrocyte precursor cells, which then differentiate into pro-oligodendrocytes, promyelinating oligodendrocytes, and finally, mature myelinating oligodendrocytes. The exact timing of the transition from neurogenesis to gliogenesis and the subsequent differentiation of glial lineages remains unknown for most of the Central Nervous System (CNS), and is especially true for the hypothalamus. Methods: Here we used mouse embryonic brain samples to determine the onset of gliogenesis and expansion of glial populations in the tuberal hypothalamus using glial markers Sox9, Sox10, Olig2, PdgfRα, Aldh1L1, and MBP. We further employed Ascl1 and Neurog2 mutant mice to probe the influence of these proneural genes on developing embryonic gliogenic populations. Results: Using marker analyses for glial precursors, we found that gliogenesis commences just prior to E13.5 in the tuberal hypothalamus, beginning with the detection of glioblast and oligodendrocyte precursor cell markers in a restricted domain adjacent to the third ventricle. Sox9+ and Olig2+ glioblasts are also observed in the mantle region from E13.5 onwards, many of which are Ki67+ proliferating cells, and peaks at E17.5. Using Ascl1 and Neurog2 mutant mice to investigate the influence of these bHLH transcription factors on the progression of gliogenesis in the tuberal hypothalamus, we found that the elimination of Ascl1 resulted in an increase in oligodendrocyte cells throughout the expansive period of oligodendrogenesis. Conclusion: Our results are the first to define the timing of gliogenesis in the tuberal hypothalamus and indicate that Ascl1 is required to repress oligodendrocyte differentiation within this brain region. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
21. Fbxo9 functions downstream of Sox10 to determine neuron-glial fate choice in the dorsal root ganglia through Neurog2 destabilization
- Author
-
Mai Har Sham, Martin Cheung, Jialin Hong, Jessica Aijia Liu, Andrew Tai, Kathryn S.E. Cheah, May Pui Lai Cheung, and Chi Wai Cheung
- Subjects
Male ,Neurogenesis ,SOX10 ,Sox10 ,Amino Acid Motifs ,Nerve Tissue Proteins ,Neurog2 ,Chick Embryo ,Mice ,Ubiquitin ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Neurogenin-2 ,Transcription factor ,Mice, Knockout ,Neurons ,Multidisciplinary ,biology ,Protein Stability ,SOXE Transcription Factors ,F-Box Proteins ,Neural crest ,Gene Expression Regulation, Developmental ,neural crest progenitors ,Biological Sciences ,Fbxo9 ,Ubiquitin ligase ,Cell biology ,medicine.anatomical_structure ,nervous system ,Neural Crest ,embryonic structures ,biology.protein ,Female ,Neuron ,Spinal Nerve Roots ,Glial cell fate commitment ,Neuroglia ,Developmental Biology ,Protein Binding - Abstract
Significance The neuron–glial lineage segregation of NC progenitors forming the DRG is essential for peripheral nervous system development. However, hierarchical transcriptional control does not explain how this lineage bifurcation occurs. Here, we provide in vivo evidence that a glial lineage specifier, Sox10, inhibits neurogenesis via a degradation mechanism. A ubiquitin-ligase, Fbxo9, acting downstream of Sox10 destabilizes the proneural Neurog2 protein, leading to the loss of neurogenic capacity in NC progenitors while leaving their glial potential intact. Functional inhibition of Fbxo9 and possibly other members of the F-box family lead to prolonged expression of Neurog2 protein that causes both the expansion of NC progenitors and precocious neuronal differentiation. Our results unravel a previously unrecognized mechanism in neuron–glial cell fate decision., The transcription factor Sox10 is a key regulator in the fate determination of a subpopulation of multipotent trunk neural crest (NC) progenitors toward glial cells instead of sensory neurons in the dorsal root ganglia (DRG). However, the mechanism by which Sox10 regulates glial cell fate commitment during lineage segregation remains poorly understood. In our study, we showed that the neurogenic determinant Neurogenin 2 (Neurog2) exhibited transient overlapping expression with Sox10 in avian trunk NC progenitors, which progressively underwent lineage segregation during migration toward the forming DRG. Gain- and loss-of-function studies revealed that the temporary expression of Neurog2 was due to Sox10 regulation of its protein stability. Transcriptional profiling identified Sox10-regulated F-box only protein (Fbxo9), which is an SCF (Skp1-Cul-F-box)-type ubiquitin ligase for Neurog2. Consistently, overexpression of Fbxo9 in NC progenitors down-regulated Neurog2 protein expression through ubiquitination and promoted the glial lineage at the expense of neuronal differentiation, whereas Fbxo9 knockdown resulted in the opposite phenomenon. Mechanistically, we found that Fbxo9 interacted with Neurog2 to promote its destabilization through the F-box motif. Finally, epistasis analysis further demonstrated that Fbxo9 and probably other F-box members mediated the role of Sox10 in destabilizing Neurog2 protein and directing the lineage of NC progenitors toward glial cells rather than sensory neurons. Altogether, these findings unravel a Sox10–Fbxo9 regulatory axis in promoting the glial fate of NC progenitors through Neurog2 destabilization.
- Published
- 2020
22. Direct Lineage Reprogramming Reveals Disease-Specific Phenotypes of Motor Neurons from Human ALS Patients.
- Author
-
Liu, Meng-Lu, Zang, Tong, and Zhang, Chun-Li
- Abstract
Summary Subtype-specific neurons obtained from adult humans will be critical to modeling neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS). Here, we show that adult human skin fibroblasts can be directly and efficiently converted into highly pure motor neurons without passing through an induced pluripotent stem cell stage. These adult human induced motor neurons (hiMNs) exhibit the cytological and electrophysiological features of spinal motor neurons and form functional neuromuscular junctions (NMJs) with skeletal muscles. Importantly, hiMNs converted from ALS patient fibroblasts show disease-specific degeneration manifested through poor survival, soma shrinkage, hypoactivity, and an inability to form NMJs. A chemical screen revealed that the degenerative features of ALS hiMNs can be remarkably rescued by the small molecule kenpaullone. Taken together, our results define a direct and efficient strategy to obtain disease-relevant neuronal subtypes from adult human patients and reveal their promising value in disease modeling and drug identification. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
23. Transcriptional Regulatory Events Initiated by Ascl1 and Neurog2 During Neuronal Differentiation of P19 Embryonic Carcinoma Cells.
- Author
-
Huang, Holly, Redmond, Tanya, Kubish, Ginger, Gupta, Shweta, Thompson, Robert, Turner, David, and Uhler, Michael
- Abstract
As members of the proneural basic-helix-loop-helix (bHLH) family of transcription factors, Ascl1 and Neurog2 direct the differentiation of specific populations of neurons at various times and locations within the developing nervous system. In order to characterize the mechanisms employed by these two bHLH factors, we generated stable, doxycycline-inducible lines of P19 embryonic carcinoma cells that express comparable levels of Ascl1 and Neurog2. Upon induction, both Ascl1 and Neurog2 directed morphological and immunocytochemical changes consistent with initiation of neuronal differentiation. Comparison of Ascl1- and Neurog2-regulated genes by microarray analyses showed both shared and distinct transcriptional changes for each bHLH protein. In both Ascl1- and Neurog2-differentiating cells, repression of Oct4 mRNA levels was accompanied by increased Oct4 promoter methylation. However, DNA demethylation was not detected for genes induced by either bHLH protein. Neurog2-induced genes included glutamatergic marker genes while Ascl1-induced genes included GABAergic marker genes. The Neurog2-specific induction of a gene encoding a protein phosphatase inhibitor, Ppp1r14a, was dependent on distinct, canonical E-box sequences within the Ppp1r14a promoter and the nucleotide sequences within these E-boxes were partially responsible for Neurog2-specific regulation. Our results illustrate multiple novel mechanisms by which Ascl1 and Neurog2 regulate gene repression during neuronal differentiation in P19 cells. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
24. Heterogeneity of neurons reprogrammed from spinal cord astrocytes by the proneural factors Ascl1 and Neurogenin2
- Author
-
Magdalena Götz, V. Bednarova, J. Kempf, Therese Riedemann, David Petrik, B.A. Hersbach, Tatiana Simon-Ebert, Giacomo Masserdotti, Pawel Smialowski, Wolfgang Enard, Aleksandar Janjic, and K. Knelles
- Subjects
Transcription, Genetic ,Nerve Tissue Proteins ,Neurog2 ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Ascl1 ,Gene ,030304 developmental biology ,Neurons ,0303 health sciences ,single-cell RNA-seq ,Correction ,patterning genes ,Spinal cord ,Cellular Reprogramming ,direct reprogramming ,Electrophysiological Phenomena ,Mice, Inbred C57BL ,ASCL1 ,Electrophysiology ,medicine.anatomical_structure ,Spinal Cord ,Organ Specificity ,Astrocytes ,patch-seq ,Direct Reprogramming ,Patch-seq ,Patterning Genes ,Single-cell Rna-seq ,Neuroscience ,Biomarkers ,030217 neurology & neurosurgery ,Astrocyte - Abstract
Summary Astrocytes are a viable source for generating new neurons via direct conversion. However, little is known about the neurogenic cascades triggered in astrocytes from different regions of the CNS. Here, we examine the transcriptome induced by the proneural factors Ascl1 and Neurog2 in spinal cord-derived astrocytes in vitro. Each factor initially elicits different neurogenic programs that later converge to a V2 interneuron-like state. Intriguingly, patch sequencing (patch-seq) shows no overall correlation between functional properties and the transcriptome of the heterogenous induced neurons, except for K-channels. For example, some neurons with fully mature electrophysiological properties still express astrocyte genes, thus calling for careful molecular and functional analysis. Comparing the transcriptomes of spinal cord- and cerebral-cortex-derived astrocytes reveals profound differences, including developmental patterning cues maintained in vitro. These relate to the distinct neuronal identity elicited by Ascl1 and Neurog2 reflecting their developmental functions in subtype specification of the respective CNS region., Graphical abstract, Highlights • Ascl1 and Neurog2 induce initially distinct transcriptomes in spinal cord astrocytes • Neurons induced by Ascl1 or Neurog2 converge to a V2 interneuron-like state • Patch-seq shows functional and transcriptional heterogeneity with low correlation • Developmentally established patterning genes are maintained in astrocytes in vitro, Kempf et al. show that Ascl1 and Neurog2 elicit initially divergent transcriptional programs in spinal cord astrocytes converging later to a V2 interneuron-like state, according to their developmental role and patterning cues in astrocytes. Patch-seq analysis reveals the heterogeneity of fate conversion with little correlation between transcriptome and electrophysiological properties.
- Published
- 2021
- Full Text
- View/download PDF
25. Notch signaling differentially regulates Atoh7 and Neurog2 in the distal mouse retina.
- Author
-
Maurer, Kate A., Riesenberg, Amy N., and Brown, Nadean L.
- Subjects
- *
RETINA , *MICE , *DEVELOPMENTAL neurobiology , *CELLULAR signal transduction , *GENETIC mutation - Abstract
Notch signaling regulates basic helix-loop-helix (bHLH) factors as an evolutionarily conserved module, but the tissue-specific mechanisms are incompletely elucidated. In the mouse retina, bHLH genes Atoh7 and Neurog2 have distinct functions, with Atoh7 regulating retinal competence and Neurog2 required for progression of neurogenesis. These transcription factors are extensively co-expressed, suggesting similar regulation.We directly compared Atoh7 and Neurog2 regulation at the earliest stages of retinal neurogenesis in a broad spectrum of Notch pathway mutants. Notch1 and Rbpj normally block Atoh7 and Neurog2 expression. However, the combined activities of Notch1, Notch3 and Rbpj regulate Neurog2 patterning in the distal retina. Downstream of the Notch complex, we found the Hes1 repressor mediates Atoh7 suppression, but Hes1,Hes3 and Hes5 do not regulate Neurog2 expression.We alsotestedNotch-mediated regulationof Jag1 and Pax6 in the distal retina, to establish the appropriate context for Neurog2 patterning. We found that Notch1;Notch3 and Rbpj block co-expression of Jag1 and Neurog2, while specifically stimulatingPax6 within an adjacent domain. Our data suggest that Notch signaling controls the overall tempo of retinogenesis, by integrating cell fate specification, the wave of neurogenesis and the developmental status of cells ahead of this wave. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
26. TAZ Represses the Neuronal Commitment of Neural Stem Cells
- Author
-
Antonio Cuadrado, Natalia Robledinos-Antón, Maribel Escoll, Kun-Liang Guan, Ministerio de Economía y Competitividad (España), and Comunidad de Madrid
- Subjects
Neurogenesis ,SOX2 ,Subventricular zone ,Biology ,Article ,Mice ,Hippo ,ASCL ,medicine ,NEUROG2 ,Animals ,Humans ,Progenitor cell ,lcsh:QH301-705.5 ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,neural stem cells ,Neural stem cells ,Wnt signaling pathway ,Cell Differentiation ,General Medicine ,Neural stem cell ,Cell biology ,neurogenesis ,medicine.anatomical_structure ,Neuronal differentiation ,nervous system ,lcsh:Biology (General) ,NEUROD1 ,Stem cell - Abstract
© 2020 by the authors., The mechanisms involved in regulation of quiescence, proliferation, and reprogramming of Neural Stem Progenitor Cells (NSPCs) of the mammalian brain are still poorly defined. Here, we studied the role of the transcriptional co-factor TAZ, regulated by the WNT and Hippo pathways, in the homeostasis of NSPCs. We found that, in the murine neurogenic niches of the striatal subventricular zone and the dentate gyrus granular zone, TAZ is highly expressed in NSPCs and declines with ageing. Moreover, TAZ expression is lost in immature neurons of both neurogenic regions. To characterize mechanistically the role of TAZ in neuronal differentiation, we used the midbrain-derived NSPC line ReNcell VM to replicate in a non-animal model the factors influencing NSPC differentiation to the neuronal lineage. TAZ knock-down and forced expression in NSPCs led to increased and reduced neuronal differentiation, respectively. TEADs-knockdown indicated that these TAZ co-partners are required for the suppression of NSPCs commitment to neuronal differentiation. Genetic manipulation of the TAZ/TEAD system showed its participation in transcriptional repression of SOX2 and the proneuronal genes ASCL1, NEUROG2, and NEUROD1, leading to impediment of neurogenesis. TAZ is usually considered a transcriptional co-activator promoting stem cell proliferation, but our study indicates an additional function as a repressor of neuronal differentiation., This study was funded by the Spanish Ministry of Economy and Competitiveness (MINECO) (Grant SAF2016-76520-R) and The Autonomous Community of Madrid (grant B2017/BMD-3827). N.R.A was recipient of an FPU contract of MINECO; M.E was the recipient of a postdoctoral contract Juan de la Cierva
- Published
- 2020
27. TAZ Represses the Neuronal Commitment of Neural Stem Cells
- Author
-
Ministerio de Economía y Competitividad (España), Comunidad de Madrid, Robledinos-Antón, Natalia, Escoll, Maribel, Guan, Kun-Liang, Cuadrado, Antonio, Ministerio de Economía y Competitividad (España), Comunidad de Madrid, Robledinos-Antón, Natalia, Escoll, Maribel, Guan, Kun-Liang, and Cuadrado, Antonio
- Abstract
The mechanisms involved in regulation of quiescence, proliferation, and reprogramming of Neural Stem Progenitor Cells (NSPCs) of the mammalian brain are still poorly defined. Here, we studied the role of the transcriptional co-factor TAZ, regulated by the WNT and Hippo pathways, in the homeostasis of NSPCs. We found that, in the murine neurogenic niches of the striatal subventricular zone and the dentate gyrus granular zone, TAZ is highly expressed in NSPCs and declines with ageing. Moreover, TAZ expression is lost in immature neurons of both neurogenic regions. To characterize mechanistically the role of TAZ in neuronal differentiation, we used the midbrain-derived NSPC line ReNcell VM to replicate in a non-animal model the factors influencing NSPC differentiation to the neuronal lineage. TAZ knock-down and forced expression in NSPCs led to increased and reduced neuronal differentiation, respectively. TEADs-knockdown indicated that these TAZ co-partners are required for the suppression of NSPCs commitment to neuronal differentiation. Genetic manipulation of the TAZ/TEAD system showed its participation in transcriptional repression of SOX2 and the proneuronal genes ASCL1, NEUROG2, and NEUROD1, leading to impediment of neurogenesis. TAZ is usually considered a transcriptional co-activator promoting stem cell proliferation, but our study indicates an additional function as a repressor of neuronal differentiation.
- Published
- 2020
28. The Prdm13 histone methyltransferase encoding gene is a Ptf1a–Rbpj downstream target that suppresses glutamatergic and promotes GABAergic neuronal fate in the dorsal neural tube.
- Author
-
Hanotel, Julie, Bessodes, Nathalie, Thélie, Aurore, Hedderich, Marie, Parain, Karine, Driessche, Benoit Van, Brandão, Karina De Oliveira, Kricha, Sadia, Jorgensen, Mette C., Grapin-Botton, Anne, Serup, Palle, Lint, Carine Van, Perron, Muriel, Pieler, Tomas, Henningfeld, Kristine A., and Bellefroid, Eric J.
- Subjects
- *
HISTONE methyltransferases , *GENE targeting , *EXCITATORY amino acid agents , *GABA agents , *NEURAL tube , *SPINAL cord physiology , *PROGENITOR cells - Abstract
Abstract: The basic helix–loop–helix (bHLH) transcriptional activator Ptf1a determines inhibitory GABAergic over excitatory glutamatergic neuronal cell fate in progenitors of the vertebrate dorsal spinal cord, cerebellum and retina. In an in situ hybridization expression survey of PR domain containing genes encoding putative chromatin-remodeling zinc finger transcription factors in Xenopus embryos, we identified Prdm13 as a histone methyltransferase belonging to the Ptf1a synexpression group. Gain and loss of Ptf1a function analyses in both frog and mice indicates that Prdm13 is positively regulated by Ptf1a and likely constitutes a direct transcriptional target. We also showed that this regulation requires the formation of the Ptf1a–Rbp-j complex. Prdm13 knockdown in Xenopus embryos and in Ptf1a overexpressing ectodermal explants lead to an upregulation of Tlx3/Hox11L2, which specifies a glutamatergic lineage and a reduction of the GABAergic neuronal marker Pax2. It also leads to an upregulation of Prdm13 transcription, suggesting an autonegative regulation. Conversely, in animal caps, Prdm13 blocks the ability of the bHLH factor Neurog2 to activate Tlx3. Additional gain of function experiments in the chick neural tube confirm that Prdm13 suppresses Tlx3+/glutamatergic and induces Pax2+/GABAergic neuronal fate. Thus, Prdm13 is a novel crucial component of the Ptf1a regulatory pathway that, by modulating the transcriptional activity of bHLH factors such as Neurog2, controls the balance between GABAergic and glutamatergic neuronal fate in the dorsal and caudal part of the vertebrate neural tube. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
29. Neuropilin2 regulates the guidance of post-crossing spinal commissural axons in a subtype-specific manner.
- Author
-
Tran, Tracy S., Carlin, Edward, Lin, Ruihe, Martinez, Edward, Johnson, Jane E., and Kaprielian, Zaven
- Subjects
- *
NEUROPILINS , *CENTRAL nervous system , *AXONS , *INTERNEURONS , *SEMAPHORINS , *SPINAL cord - Abstract
Background: Spinal commissural axons represent a model system for deciphering the molecular logic that regulates the guidance of midline-crossing axons in the developing central nervous system (CNS). Whether the same or specific sets of guidance signals control the navigation of molecularly distinct subtypes of these axons remains an open and largely unexplored question. Although it is well established that post-crossing commissural axons alter their responsiveness to midline-associated guidance cues, our understanding of the repulsive mechanisms that drive the post-crossing segments of these axons away from the midline and whether the underlying guidance systems operate in a commissural axon subtype-specific manner, remains fragmentary at best. Results: Here, we utilize axonally targeted transgenic reporter mice to visualize genetically distinct dorsal interneuron (dI)1 and dI4 commissural axons and show that the repulsive class 3 semaphorin (Sema3) guidance receptor Neuropilin 2 (Npn2), is selectively expressed on the dI1 population and is required for the guidance of post-crossing dI1, but not dI4, axons. Consistent with these observations, the midline-associated Npn2 ligands, Sema3F and Sema3B, promote the collapse of dI1, but not dI4, axon-associated growth cones in vitro. We also identify, for the first time, a discrete GABAergic population of ventral commissural neurons/axons in the embryonic mouse spinal cord that expresses Npn2, and show that Npn2 is required for the proper guidance of their post-crossing axons. Conclusions: Together, our findings indicate that Npn2 is selectively expressed in distinct populations of commissural neurons in both the dorsal and ventral spinal cord, and suggest that Sema3-Npn2 signaling regulates the guidance of post-crossing commissural axons in a population-specific manner. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
30. Direct visualization of the transition status during neural differentiation by dual-fluorescent reporter human pluripotent stem cells.
- Author
-
Park G, Shin M, Lee W, Hotta A, Kobayashi T, and Kosodo Y
- Subjects
- Animals, Basic Helix-Loop-Helix Transcription Factors, Cell Differentiation genetics, Humans, Mice, Nerve Tissue Proteins genetics, Neurons, Organoids, Induced Pluripotent Stem Cells, Neural Stem Cells
- Abstract
Human induced pluripotent stem cells (hiPSCs) can differentiate into neurons and glia via neural progenitor cells and are widely used for neurogenic studies. However, directly visualizing the transition status during the neural differentiation of live cells is difficult. Here, targeting NEUROG2 (NGN2) and TUBB3 as markers of neurogenic cells and neurons, respectively, we established TUBB3
EGFP /NGN2TagRFP dual-reporter hiPSCs using CRISPR-Cas9 technology. We induced the differentiation of cortical neurons from dual-reporter hiPSCs, successfully visualizing cell-fate conversion in two-dimensional (2D) culture and 3D organoids. The reporter cells were used to monitor drug effects to enhance neural induction, responses to gene knockdown, transplantation to the embryonic mouse brain, and live imaging at single-cell resolution. Notably, the earliest REELIN-positive neurons showed a distinctive migration pattern, and their production was accelerated by HES1-function loss. Together, these results demonstrate the potential for dual-reporter hiPSCs in therapeutic neural regeneration strategies and studies on human cortical development., Competing Interests: Conflicts of interests The authors have no conflicts of interest to declare., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
31. Neurogenin 2 regulates progenitor cell-cycle progression and Purkinje cell dendritogenesis in cerebellar development.
- Author
-
Florio, Marta, Leto, Ketty, Muzio, Luca, Tinterri, Andrea, Badaloni, Aurora, Croci, Laura, Zordan, Paola, Barili, Valeria, Albieri, Ilaria, Guillemot, François, Rossi, Ferdinando, and Consalez, G. Giacomo
- Subjects
- *
NEUROGENINS , *PROGENITOR cells , *CELL cycle , *PURKINJE cells , *CEREBELLAR cortex , *NEURONS , *NEURODEGENERATION , *RECOMBINASES - Abstract
By serving as the sole output of the cerebellar cortex, integrating a myriad of afferent stimuli, Purkinje cells (PCs) constitute the principal neuron in cerebellar circuits. Several neurodegenerative cerebellar ataxias feature a selective cell-autonomous loss of PCs, warranting the development of regenerative strategies. To date, very little is known as to the regulatory cascades controlling PC development. During central nervous system development, the proneural gene neurogenin 2 (Neurog2) contributes to many distinct neuronal types by specifying their fate and/or dictating development of their morphological features. By analyzing a mouse knock-in line expressing Cre recombinase under the control of Neurog2 cis-acting sequences we show that, in the cerebellar primordium, Neurog2 is expressed by cycling progenitors cell-autonomously fated to become PCs, even when transplanted heterochronically. During cerebellar development, Neurog2 is expressed in G1 phase by progenitors poised to exit the cell cycle. We demonstrate that, in the absence of Neurog2, both cell-cycle progression and neuronal output are significantly affected, leading to an overall reduction of the mature cerebellar volume. Although PC fate identity is correctly specified, the maturation of their dendritic arbor is severely affected in the absence of Neurog2, as null PCs develop stunted and poorly branched dendrites, a defect evident from the early stages of dendritogenesis. Thus, Neurog2 represents a key regulator of PC development and maturation [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
32. Epigenetic regulation of sensory neurogenesis in the dorsal root ganglion cell line ND7 by folic acid.
- Author
-
Boshnjaku, Vanda, Ichi, Shunsuke, Yueh-Wei Shen, Puranmalka, Rahul, Mania-Farnell, Barbara, McLone, David G., Tomita, Tadanori, and Mayanil, Chandra S. K.
- Published
- 2011
- Full Text
- View/download PDF
33. Interaction of MTG family proteins with NEUROG2 and ASCL1 in the developing nervous system
- Author
-
Aaker, Joshua D., Patineau, Andrea L., Yang, Hyun-jin, Ewart, David T., Nakagawa, Yasushi, McLoon, Steven C., and Koyano-Nakagawa, Naoko
- Subjects
- *
TRANSCRIPTION factors , *HELIX-loop-helix motifs , *PROTEINS , *DEVELOPMENTAL neurobiology , *GENETIC repressors , *NEURON development , *GENE expression - Abstract
Abstract: During neural development, members of MTG family of transcriptional repressors are induced by proneural basic helix-loop-helix (bHLH) transcription factors and in turn inhibit the activity of the bHLH proteins, forming a negative feedback loop that regulates the normal progression of neurogenesis. Three MTG genes, MTG8, MTG16 and MTGR1, are expressed in distinct patterns in the developing nervous system. Various bHLH proteins are also expressed in distinct patterns. We asked whether there is a functional relationship between specific MTG and bHLH proteins in developing chick spinal cord. First, we examined if each MTG gene is induced by specific bHLH proteins. Although expression of NEUROG2, ASCL1 and MTG genes overlapped, the boundaries of gene expression did not match. Ectopic expression analysis showed that MTGR1 and NEUROD4, which show similar expression patterns, are regulated differently by NEUROG2 and ASCL1. Thus, our results show that expression of MTG genes is not regulated by a single upstream bHLH protein, but represents an integration of the activity of multiple regulators. Next, we asked if each MTG protein inhibits specific bHLH proteins. Transcription assay showed that NEUROG2 and ASCL1 are inhibited by MTGR1 and MTG16, and less efficiently by MTG8. Deletion mapping of MTGR1 showed that MTGR1 binds NEUROG2 and ASCL1 using multiple interaction surfaces, and all conserved domains are required for its repressor activity. These results support the model that MTG proteins form a higher-order repressor complex and modulate transcriptional activity of bHLH proteins during neurogenesis. [Copyright &y& Elsevier]
- Published
- 2010
- Full Text
- View/download PDF
34. Neurog2 controls the leading edge of neurogenesis in the mammalian retina
- Author
-
Hufnagel, Robert B., Le, Tien T., Riesenberg, Ashley L., and Brown, Nadean L.
- Subjects
- *
DEVELOPMENTAL neurobiology , *RETINAL ganglion cells , *TRANSCRIPTION factors , *CELL differentiation , *CELL populations , *PROTEIN structure , *NEURON development - Abstract
Abstract: In the mammalian retina, neuronal differentiation begins in the dorso-central optic cup and sweeps peripherally and ventrally. While certain extrinsic factors have been implicated, little is known about the intrinsic factors that direct this process. In this study, we evaluate the expression and function of proneural bHLH transcription factors during the onset of mouse retinal neurogenesis. Dorso-central retinal progenitor cells that give rise to the first postmitotic neurons express Neurog2/Ngn2 and Atoh7/Math5. In the absence of Neurog2, the spread of neurogenesis stalls, along with Atoh7 expression and RGC differentiation. However, neurogenesis is eventually restored, and at birth Neurog2 mutant retinas are reduced in size, with only a slight increase in the retinal ganglion cell population. We find that the re-establishment of neurogenesis coincides with the onset of Ascl1 expression, and that Ascl1 can rescue the early arrest of neural development in the absence of Neurog2. Together, this study supports the hypothesis that the intrinsic factors Neurog2 and Ascl1 regulate the temporal progression of retinal neurogenesis by directing overlapping waves of neuron formation. [Copyright &y& Elsevier]
- Published
- 2010
- Full Text
- View/download PDF
35. Feedback regulation of NEUROG2 activity by MTGR1 is required for progression of neurogenesis
- Author
-
Aaker, Joshua D., Patineau, Andrea L., Yang, Hyun-jin, Ewart, David T., Gong, Wuming, Li, Tongbin, Nakagawa, Yasushi, McLoon, Steven C., and Koyano-Nakagawa, Naoko
- Subjects
- *
CELLULAR mechanics , *DEVELOPMENTAL neurobiology , *TRANSCRIPTION factors , *GENETIC regulation , *GENETIC repressors , *NERVOUS system , *PROTEIN binding - Abstract
Abstract: The sequential steps of neurogenesis are characterized by highly choreographed changes in transcription factor activity. In contrast to the well-studied mechanisms of transcription factor activation during neurogenesis, much less is understood regarding how such activity is terminated. We previously showed that MTGR1, a member of the MTG family of transcriptional repressors, is strongly induced by a proneural basic helix–loop–helix transcription factor, NEUROG2 in developing nervous system. In this study, we describe a novel feedback regulation of NEUROG2 activity by MTGR1. We show that MTGR1 physically interacts with NEUROG2 and represses transcriptional activity of NEUROG2. MTGR1 also prevents DNA binding of the NEUROG2/E47 complex. In addition, we provide evidence that proper termination of NEUROG2 activity by MTGR1 is necessary for normal progression of neurogenesis in the developing spinal cord. These results highlight the importance of feedback regulation of proneural gene activity in neurodevelopment. [Copyright &y& Elsevier]
- Published
- 2009
- Full Text
- View/download PDF
36. Sequential roles for Mash1 and Ngn2 in the generation of dorsal spinal cord interneurons.
- Author
-
Helms, Amy W., Battiste, James, Henke, R. Michael, Nakada, Yuji, Simplicio, Nicolas, Guillemot, Francois, and Johnson, Jane E.
- Subjects
- *
SPINAL cord , *NEURONS , *MOTOR neurons , *CENTRAL nervous system , *CELLS - Abstract
The dorsal spinal cord contains a diverse array of neurons that connect sensory input from the periphery to spinal cord motoneurons and brain. During development, six dorsal neuronal populations (dI1-dI6) have been defined by expression of homeodomain factors and position in the dorsoventral axis. The bHLH transcription factors Mash1 and Ngn2 have distinct roles in specification of these neurons. Mash1 is necessary and sufficient for generation of most dI3 and all dI5 neurons. Unexpectedly, dI4 neurons are derived from cells expressing low levels or no Mash1, and this population increases in the Mash1 mutant. Ngn2 is not required for any specific neuronal cell type but appears to modulate the composition of neurons that form. In the absence of Ngn2, there is an increase in the number of dI3 and dI5 neurons, in contrast to the effects produced by activity of Mash1. Mash1 is epistatic to Ngn2, and, unlike the relationship between other neural bHLH factors, cross-repression of expression is not detected. Thus, bHLH factors, particularly Mash1 and related family members Math1 and Ngn1, provide a code for generating neuronal diversity in the dorsal spinal cord with Ngn2 serving to modulate the number of neurons in each population formed. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
37. Neurog2 Deficiency Uncovers a Critical Period of Cell Fate Plasticity and Vulnerability among Neural-Crest-Derived Somatosensory Progenitors.
- Author
-
Ventéo, Stéphanie, Desiderio, Simon, Cabochette, Pauline, Deslys, Alexandre, Carroll, Patrick, Pattyn, Alexandre PA, Ventéo, Stéphanie, Desiderio, Simon, Cabochette, Pauline, Deslys, Alexandre, Carroll, Patrick, and Pattyn, Alexandre PA
- Abstract
Functionally distinct classes of dorsal root ganglia (DRG) somatosensory neurons arise from neural crest cells (NCCs) in two successive phases of differentiation assumed to be respectively and independently controlled by the proneural genes Neurog2 and Neurog1. However, the precise role of Neurog2 during this process remains unclear, notably because no neuronal loss has been reported hitherto in Neurog2-/- mutants. Here, we show that at trunk levels, Neurog2 deficiency impairs the production of subsets of all DRG neuron subtypes. We establish that this phenotype is highly dynamic and reflects multiple defects in NCC-derived progenitors, including somatosensory-to-melanocyte fate switch, apoptosis, and delayed differentiation which alters neuronal identity, all occurring during a narrow time window when Neurog2 temporarily controls onset of Neurog1 expression and neurogenesis. Collectively, these findings uncover a critical period of cell fate plasticity and vulnerability among somatosensory progenitors and establish that Neurog2 function in the developing DRG is broader than initially envisaged., info:eu-repo/semantics/published
- Published
- 2019
38. Instructing neuronal identity during CNS development and astroglial-lineage reprogramming: Roles of NEUROG2 and ASCL1
- Author
-
Chouchane, Malek and Costa, Marcos Romualdo
- Subjects
Neuronal identity specification ,Astroglia lineage-reprogramming ,Neurog2 ,Ascl1 ,Neural progenitors - Abstract
The adult mammalian brain contains an enormous variety of neuronal types, which are generally categorized in large groups, based on their neurochemical identity, hodological properties and molecular markers. This broad classification has allowed the correlation between individual neural progenitor populations and their neuronal progeny, thus contributing to probe the cellular and molecular mechanisms involved in neuronal identity determination during central nervous system (CNS) development. In this review, we discuss the contribution of the proneural genes Neurogenin2 (Neurog2) and Achaete-scute homolog 1 (Ascl1) for the specification of neuronal phenotypes in the developing neocortex, cerebellum and retina. Then, we revise recent data on astroglia cell lineage reprogramming into induced neurons using the same proneural proteins to compare the neuronal phenotypes obtained from astroglial cells originated in those CNS regions. We conclude that Ascl1 and Neurog2 have different contributions to determine neuronal fates, depending on the neural progenitor or astroglial population expressing those proneural factors. Finally, we discuss some possible explanations for these seemingly conflicting effects of Ascl1 and Neurog2 and propose future approaches to further dissect the molecular mechanisms of neuronal identity specification.
- Published
- 2018
39. Heterogeneity of neurons reprogrammed from spinal cord astrocytes by the proneural factors Ascl1 and Neurogenin2.
- Author
-
Kempf, J., Knelles, K., Hersbach, B.A., Petrik, D., Riedemann, T., Bednarova, V., Janjic, A., Simon-Ebert, T., Enard, W., Smialowski, P., Götz, M., and Masserdotti, G.
- Abstract
Astrocytes are a viable source for generating new neurons via direct conversion. However, little is known about the neurogenic cascades triggered in astrocytes from different regions of the CNS. Here, we examine the transcriptome induced by the proneural factors Ascl1 and Neurog2 in spinal cord-derived astrocytes in vitro. Each factor initially elicits different neurogenic programs that later converge to a V2 interneuron-like state. Intriguingly, patch sequencing (patch-seq) shows no overall correlation between functional properties and the transcriptome of the heterogenous induced neurons, except for K-channels. For example, some neurons with fully mature electrophysiological properties still express astrocyte genes, thus calling for careful molecular and functional analysis. Comparing the transcriptomes of spinal cord- and cerebral-cortex-derived astrocytes reveals profound differences, including developmental patterning cues maintained in vitro. These relate to the distinct neuronal identity elicited by Ascl1 and Neurog2 reflecting their developmental functions in subtype specification of the respective CNS region. [Display omitted] • Ascl1 and Neurog2 induce initially distinct transcriptomes in spinal cord astrocytes • Neurons induced by Ascl1 or Neurog2 converge to a V2 interneuron-like state • Patch-seq shows functional and transcriptional heterogeneity with low correlation • Developmentally established patterning genes are maintained in astrocytes in vitro Kempf et al. show that Ascl1 and Neurog2 elicit initially divergent transcriptional programs in spinal cord astrocytes converging later to a V2 interneuron-like state, according to their developmental role and patterning cues in astrocytes. Patch-seq analysis reveals the heterogeneity of fate conversion with little correlation between transcriptome and electrophysiological properties. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
40. DISC1 Regulates the Proliferation and Migration of Mouse Neural Stem/Progenitor Cells through Pax5, Sox2, Dll1 and Neurog2
- Author
-
Zhaohui Luo, Yi Shu, Bo Xiao, Qian Wu, Zongwei Yue, Li Feng, Weiting Tang, and Yi Li
- Subjects
0301 basic medicine ,Small interfering RNA ,Dll1 ,proliferation ,Sox2 ,Neurog2 ,Biology ,migration ,lcsh:RC321-571 ,Flow cytometry ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,DISC1 ,SOX2 ,medicine ,Progenitor cell ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Original Research ,Pax5 ,medicine.diagnostic_test ,Neurogenesis ,Transfection ,Cell biology ,030104 developmental biology ,Apoptosis ,Cancer research ,biology.protein ,Neuroscience - Abstract
Background: Disrupted-in-schizophrenia 1 (DISC1) regulates neurogenesis and is a genetic risk factor for major psychiatric disorders. However, how DISC1 dysfunction affects neurogenesis and cell cycle progression at the molecular level is still unknown. Here, we investigated the role of DISC1 in regulating proliferation, migration, cell cycle progression and apoptosis in mouse neural stem/progenitor cells (MNSPCs) in vitro. Methods: MNSPCs were isolated and cultured from mouse fetal hippocampi. Retroviral vectors or siRNAs were used to manipulate DISC1 expression in MNSPCs. Proliferation, migration, cell cycle progression and apoptosis of altered MNSPCs were analyzed in cell proliferation assays (MTS), transwell system and flow cytometry. A neurogenesis specific polymerase chain reaction (PCR) array was used to identify genes downstream of DISC1, and functional analysis was performed through transfection of expression plasmids and siRNAs. Results: Loss of DISC1 reduced proliferation and migration of MNSPCs, while an increase in DISC1 led to increased proliferation and migration. Meanwhile, an increase in the proportion of cells in G0/G1 phase was concomitant with reduced levels of DISC1, but significant changes were not observed in the number MNSPCs undergoing apoptosis. Paired box gene 5 (Pax5), sex determining region Y-box 2 (Sox2), delta-like1 (Dll1) and Neurogenin2 (Neurog2) emerged as candidate molecules downstream of DISC1, and rescue experiments demonstrated that increased or decreased expression of either molecule regulated proliferation and migration in DISC1-altered MNSPCs. Conclusion: These results suggest that Pax5, Sox2, Dll1 and Neurog2 mediate DISC1 activity in MNSPC proliferation and migration.
- Published
- 2017
41. Genetic analysis of the NEUROG2 gene in patients with Parkinson's disease
- Author
-
Deng, Sheng, Deng, Hao, Le, Weidong, Xu, Hongbo, Yang, Huarong, Deng, Xiong, Lv, Hongwei, Xie, Wenjie, Zhu, Shaihong, and Jankovic, Joseph
- Subjects
- *
PARKINSON'S disease & genetics , *NERVE tissue proteins , *TRANSCRIPTION factors , *DOPAMINERGIC neurons , *GENETIC mutation , *HUMAN genetic variation , *CELL differentiation , *GENETIC code - Abstract
Abstract: The proneural protein Neurogenin 2 (NEUROG2) is a transcription factor of importance for the differentiation and survival of midbrain dopaminergic neurons. To determine whether genetic variation in the coding region of the NEUROG2 gene plays a role in the etiology of Parkinson''s disease (PD), we screened DNA samples from 202 PD patients and 201 normal controls. No mutation in the NEUROG2 gene was identified in our PD cohort, except that novel compound heterozygous variants (Gly56Arg and Asp206Glu) were found in a 91-year normal male, suggesting that mutations in the coding region of the NEUROG2 gene play little or no role in the development of PD. [Copyright &y& Elsevier]
- Published
- 2010
- Full Text
- View/download PDF
42. TAZ Represses the Neuronal Commitment of Neural Stem Cells.
- Author
-
Robledinos-Antón, Natalia, Escoll, Maribel, Guan, Kun-Liang, and Cuadrado, Antonio
- Subjects
NEURAL stem cells ,NEURONAL differentiation ,DENTATE gyrus ,STEM cells - Abstract
The mechanisms involved in regulation of quiescence, proliferation, and reprogramming of Neural Stem Progenitor Cells (NSPCs) of the mammalian brain are still poorly defined. Here, we studied the role of the transcriptional co-factor TAZ, regulated by the WNT and Hippo pathways, in the homeostasis of NSPCs. We found that, in the murine neurogenic niches of the striatal subventricular zone and the dentate gyrus granular zone, TAZ is highly expressed in NSPCs and declines with ageing. Moreover, TAZ expression is lost in immature neurons of both neurogenic regions. To characterize mechanistically the role of TAZ in neuronal differentiation, we used the midbrain-derived NSPC line ReNcell VM to replicate in a non-animal model the factors influencing NSPC differentiation to the neuronal lineage. TAZ knock-down and forced expression in NSPCs led to increased and reduced neuronal differentiation, respectively. TEADs-knockdown indicated that these TAZ co-partners are required for the suppression of NSPCs commitment to neuronal differentiation. Genetic manipulation of the TAZ/TEAD system showed its participation in transcriptional repression of SOX2 and the proneuronal genes ASCL1, NEUROG2, and NEUROD1, leading to impediment of neurogenesis. TAZ is usually considered a transcriptional co-activator promoting stem cell proliferation, but our study indicates an additional function as a repressor of neuronal differentiation. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
43. Bcl-2-Assisted Reprogramming of Mouse Astrocytes and Human Fibroblasts into Induced Neurons.
- Author
-
Falco A, Bartolomé-Cabrero R, and Gascón S
- Subjects
- Animals, Astrocytes drug effects, Cell Culture Techniques, Cell Line, Cells, Cultured, Cellular Reprogramming drug effects, Culture Media, Conditioned pharmacology, Gene Expression, Genetic Vectors genetics, Humans, Mice, Neurons drug effects, Retroviridae genetics, Transduction, Genetic, Transfection, Astrocytes cytology, Astrocytes metabolism, Cellular Reprogramming genetics, Cellular Reprogramming Techniques, Fibroblasts cytology, Neurons cytology, Neurons metabolism, Proto-Oncogene Proteins c-bcl-2 genetics
- Abstract
Direct neuronal reprogramming is a promising strategy to generate various types of neurons that are, otherwise, inaccessible for researchers. However, the efficiency of neuronal conversion is highly dependent on the transcription factor used, the identity of the initial cells to convert, their species' background, and the neuronal subtype to which cells will convert. Regardless of these conditioning factors, the apoptotic regulator Bcl-2 acts as a pan-neuronal reprogramming enhancer. Bcl-2 mediates its effect in reprogramming by preventing an overshot of oxidative stress during the acquisition of a neuronal oxidative metabolism, thus reducing cell death by ferroptosis and facilitating the phenotypic conversion. In this chapter, we outline two methods to obtain either mouse or human neurons derived from postnatal astrocytes and skin fibroblasts, respectively. The overall reprogramming strategy is based on the co-expression of Bcl-2 and the transcription factor Neurog2 that produces mostly excitatory neurons. However, the method can be easily adapted to achieve alternative neuronal subtypes by using additional transcription factors, such as Isl1 for motor neurons. Therefore, our approaches provide solid but flexible platforms to obtain human and mouse induced neurons in vitro that can be applied to basic or translational research., (© 2021. Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2021
- Full Text
- View/download PDF
44. Neurog2 Deficiency Uncovers a Critical Period of Cell Fate Plasticity and Vulnerability among Neural-Crest-Derived Somatosensory Progenitors.
- Author
-
Ventéo S, Desiderio S, Cabochette P, Deslys A, Carroll P, and Pattyn A
- Subjects
- Animals, Cell Differentiation physiology, Ganglia, Spinal metabolism, Ganglia, Spinal physiology, Gene Expression Regulation, Developmental, Mice, Neurogenesis physiology, Neurons metabolism, Neurons physiology, Basic Helix-Loop-Helix Transcription Factors metabolism, Cell Plasticity physiology, Nerve Tissue Proteins metabolism, Neural Crest metabolism
- Abstract
Functionally distinct classes of dorsal root ganglia (DRG) somatosensory neurons arise from neural crest cells (NCCs) in two successive phases of differentiation assumed to be respectively and independently controlled by the proneural genes Neurog2 and Neurog1. However, the precise role of Neurog2 during this process remains unclear, notably because no neuronal loss has been reported hitherto in Neurog2
-/- mutants. Here, we show that at trunk levels, Neurog2 deficiency impairs the production of subsets of all DRG neuron subtypes. We establish that this phenotype is highly dynamic and reflects multiple defects in NCC-derived progenitors, including somatosensory-to-melanocyte fate switch, apoptosis, and delayed differentiation which alters neuronal identity, all occurring during a narrow time window when Neurog2 temporarily controls onset of Neurog1 expression and neurogenesis. Collectively, these findings uncover a critical period of cell fate plasticity and vulnerability among somatosensory progenitors and establish that Neurog2 function in the developing DRG is broader than initially envisaged., (Copyright © 2019 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
45. The Prdm13 histone methyltransferase encoding gene is a Ptf1a-Rbpj downstream target that suppresses glutamatergic and promotes GABAergic neuronal fate in the dorsal neural tube
- Author
-
Mette C. Jørgensen, Carine Van Lint, Sadia Kricha, Julie Hanotel, Tomas Pieler, Muriel Perron, Karine Parain, Eric Bellefroid, Benoît Van Driessche, Aurore Thelie, Nathalie Bessodes, Kristine A. Henningfeld, Palle Serup, Anne Grapin-Botton, Marie Hedderich, Karina de Oliveira Brandão, Laboratory of Developmental Genetics, Université libre de Bruxelles (ULB), Department of Developmental Biochemistry (CNMPB), University of Göttingen - Georg-August-Universität Göttingen, Neurobiologie et Développement (N&eD), Centre National de la Recherche Scientifique (CNRS), Laboratory of Molecular Virology, Université libre de Bruxelles (ULB)-Institut de Biologie et de Médecine Moléculaires, The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), Faculty of Health and Medical Sciences, University of Copenhagen = Københavns Universitet (KU)-University of Copenhagen = Københavns Universitet (KU), and Institut de Neurobiologie Alfred Fessard (INAF)
- Subjects
Prdm genes ,Chick Embryo ,Neurog2 ,Xenopus Proteins ,Mice ,Xenopus laevis ,0302 clinical medicine ,MESH: Reverse Transcriptase Polymerase Chain Reaction ,MESH: Basic Helix-Loop-Helix Transcription Factors ,MESH: Gene Expression Regulation, Developmental ,Basic Helix-Loop-Helix Transcription Factors ,GABAergic neuron ,MESH: Animals ,GABAergic Neurons ,MESH: Xenopus Proteins ,In Situ Hybridization ,0303 health sciences ,Spinal cord ,Ptf1a ,Reverse Transcriptase Polymerase Chain Reaction ,Tlx3 ,Synexpression ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Glutamatergic neuron ,MESH: Chick Embryo ,Immunohistochemistry ,Cell biology ,MESH: PAX2 Transcription Factor ,medicine.anatomical_structure ,Electroporation ,Histone methyltransferase ,Histone Methyltransferases ,MESH: GABAergic Neurons ,GABAergic ,[SDV.NEU]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC] ,MESH: Cell Differentiation ,MESH: DNA Primers ,Neural Tube ,Biology ,Cell fate determination ,Retina ,MESH: Neural Tube ,03 medical and health sciences ,Glutamatergic ,MESH: In Situ Hybridization ,MESH: Xenopus laevis ,medicine ,Animals ,Immunoprecipitation ,MESH: Electroporation ,Transcription factor ,Molecular Biology ,MESH: Mice ,030304 developmental biology ,DNA Primers ,Pax2 ,RBPJ ,MESH: Immunoprecipitation ,PAX2 Transcription Factor ,Neural tube ,MESH: Histone-Lysine N-Methyltransferase ,MESH: Immunohistochemistry ,Histone-Lysine N-Methyltransferase ,Cell Biology ,Molecular biology ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
International audience; The basic helix-loop-helix (bHLH) transcriptional activator Ptf1a determines inhibitory GABAergic over excitatory glutamatergic neuronal cell fate in progenitors of the vertebrate dorsal spinal cord, cerebellum and retina. In an in situ hybridization expression survey of PR domain containing genes encoding putative chromatin-remodeling zinc finger transcription factors in Xenopus embryos, we identified Prdm13 as a histone methyltransferase belonging to the Ptf1a synexpression group. Gain and loss of Ptf1a function analyses in both frog and mice indicates that Prdm13 is positively regulated by Ptf1a and likely constitutes a direct transcriptional target. We also showed that this regulation requires the formation of the Ptf1a-Rbp-j complex. Prdm13 knockdown in Xenopus embryos and in Ptf1a overexpressing ectodermal explants lead to an upregulation of Tlx3/Hox11L2, which specifies a glutamatergic lineage and a reduction of the GABAergic neuronal marker Pax2. It also leads to an upregulation of Prdm13 transcription, suggesting an autonegative regulation. Conversely, in animal caps, Prdm13 blocks the ability of the bHLH factor Neurog2 to activate Tlx3. Additional gain of function experiments in the chick neural tube confirm that Prdm13 suppresses Tlx3(+)/glutamatergic and induces Pax2(+)/GABAergic neuronal fate. Thus, Prdm13 is a novel crucial component of the Ptf1a regulatory pathway that, by modulating the transcriptional activity of bHLH factors such as Neurog2, controls the balance between GABAergic and glutamatergic neuronal fate in the dorsal and caudal part of the vertebrate neural tube.
- Published
- 2014
- Full Text
- View/download PDF
46. DISC1 Regulates the Proliferation and Migration of Mouse Neural Stem/Progenitor Cells through Pax5, Sox2, Dll1 and Neurog2.
- Author
-
Wu Q, Tang W, Luo Z, Li Y, Shu Y, Yue Z, Xiao B, and Feng L
- Abstract
Background : Disrupted-in-schizophrenia 1 (DISC1) regulates neurogenesis and is a genetic risk factor for major psychiatric disorders. However, how DISC1 dysfunction affects neurogenesis and cell cycle progression at the molecular level is still unknown. Here, we investigated the role of DISC1 in regulating proliferation, migration, cell cycle progression and apoptosis in mouse neural stem/progenitor cells (MNSPCs) in vitro . Methods : MNSPCs were isolated and cultured from mouse fetal hippocampi. Retroviral vectors or siRNAs were used to manipulate DISC1 expression in MNSPCs. Proliferation, migration, cell cycle progression and apoptosis of altered MNSPCs were analyzed in cell proliferation assays (MTS), transwell system and flow cytometry. A neurogenesis specific polymerase chain reaction (PCR) array was used to identify genes downstream of DISC1 , and functional analysis was performed through transfection of expression plasmids and siRNAs. Results : Loss of DISC1 reduced proliferation and migration of MNSPCs, while an increase in DISC1 led to increased proliferation and migration. Meanwhile, an increase in the proportion of cells in G0/G1 phase was concomitant with reduced levels of DISC1, but significant changes were not observed in the number MNSPCs undergoing apoptosis. Paired box gene 5 (Pax5), sex determining region Y-box 2 (Sox2), delta-like1 (Dll1) and Neurogenin2 (Neurog2) emerged as candidate molecules downstream of DISC1, and rescue experiments demonstrated that increased or decreased expression of either molecule regulated proliferation and migration in DISC1-altered MNSPCs. Conclusion : These results suggest that Pax5, Sox2, Dll1 and Neurog2 mediate DISC1 activity in MNSPC proliferation and migration.
- Published
- 2017
- Full Text
- View/download PDF
47. The Tbr2 Molecular Network Controls Cortical Neuronal Differentiation Through Complementary Genetic and Epigenetic Pathways.
- Author
-
Sessa A, Ciabatti E, Drechsel D, Massimino L, Colasante G, Giannelli S, Satoh T, Akira S, Guillemot F, and Broccoli V
- Subjects
- Animals, Basic Helix-Loop-Helix Transcription Factors genetics, Basic Helix-Loop-Helix Transcription Factors metabolism, Cell Cycle genetics, Cell Movement genetics, Cell Polarity genetics, Embryo, Mammalian, Gene Expression Regulation, Developmental physiology, Gene Regulatory Networks genetics, Hippocampus cytology, Jumonji Domain-Containing Histone Demethylases metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Microarray Analysis, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, T-Box Domain Proteins metabolism, Transcription Factors metabolism, Cell Differentiation genetics, Cerebral Cortex cytology, Gene Expression Regulation, Developmental genetics, Neural Stem Cells physiology, Neurons physiology, T-Box Domain Proteins genetics
- Abstract
The T-box containing Tbr2 gene encodes for a transcription factor essential for the specification of the intermediate neural progenitors (INPs) originating the excitatory neurons of the cerebral cortex. However, its overall mechanism of action, direct target genes and cofactors remain unknown. Herein, we carried out global gene expression profiling combined with genome-wide binding site identification to determine the molecular pathways regulated by TBR2 in INPs. This analysis led to the identification of novel protein-protein interactions that control multiple features of INPs including cell-type identity, morphology, proliferation and migration dynamics. In particular, NEUROG2 and JMJD3 were found to associate with TBR2 revealing unexplored TBR2-dependent mechanisms. These interactions can explain, at least in part, the role of this transcription factor in the implementation of the molecular program controlling developmental milestones during corticogenesis. These data identify TBR2 as a major determinant of the INP-specific traits by regulating both genetic and epigenetic pathways., (© The Author 2016. Published by Oxford University Press. All rights reserved. For Permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2017
- Full Text
- View/download PDF
48. Folate receptor alpha is more than just a folate transporter.
- Author
-
Mohanty V, Siddiqui MR, Tomita T, and Mayanil CS
- Abstract
Until recently folate receptor alpha (FRα) has only been considered as a folate transporter. However, a novel role of FRα as a transcription factor was reported by our lab. More recently our lab showed a novel pleiotropic role of FRα: (a) direct transcriptional activation of Oct4, Sox2 , and Klf4 genes; and (b) repression of biogenesis of miRNAs that target these genes or their effector molecules. These observations beg a question: "Can a simple molecule such as folate be used to manipulate the production and/or differentiation of endogenous neural stem cells (NSCs), which may hold promise for future therapies?" Conditions such as spinal cord injury, motor neuron diseases, Alzheimer's disease and multiple sclerosis may benefit from increasing stem cell pool and promoting specific pathways of differentiation. On the flip-side, these NSCs may also contribute to some CNS tumors therefore promoting differentiation could prove more beneficial. FRα may hold promises for both since it has the potential to remodel chromatin in a context dependent manner. In this commentary we discuss our previous data and new questions arising in the context of the new role for FRα.
- Published
- 2017
- Full Text
- View/download PDF
49. The Role of Basic Helix-Loop-Helix Transcription Factors in Early Retinal Neurogenesis
- Author
-
Hufnagel, Robert B.
- Subjects
- Neurology, retina, n eurogenesis, retinal ganglion cell, Neurog2, Atoh7, Ascl1
- Abstract
The retina converts visual information into neural signals that are processed and transmitted to the brain. During retinal development, seven major cell types, six neuronal and one glial, are generated from a common neuroepithelium during discrete but overlapping time periods. Here, we study the retinal expression and function of three basic helix-loop-helix (bHLH) transcription factors: Atoh7/Math5 (atonal homologue 7), Neurog2/Neurogenin2, and Ascl1/Mash1 (achaete-scute complex like 1). Proneural bHLH transcription factors are critical for neuronal differentiation and cell type specification in the retina. Atoh7 and Neurog2 are expressed at the initiation of retinal development, and Atoh7 is critical for the generation of the first-born cell type, retinal ganglion cells (RGCs), which transmit visual information to the brain via the optic nerve. Ascl1 is expressed later in retinogenesis, and is required for normal bipolar interneuron and Müller glial genesis, two later-born cell types. First, I explored the regulation of Atoh7 expression, using GFP-expressing transgenes under control of Atoh7 regulatory DNA, which expressed GFP in Atoh7-expressing progenitor cells and nascent RGC axons as they sent projections into the optic nerve and established connections with the brain. In addition to the visual system, Math5-GFP transgenic expression was observed ectopically in developing auditory and proprioceptive systems in the developing brain, spinal cord, and inner ear that normally express Atoh1/Math1, the other atonal semi-orthologue. I found similarities in the genetic regulation of the proximal 2.1 Kb of 5’ Atoh7 DNA and the Atoh1 3’ enhancers, and concluded that these highly-related bHLHs share common regulatory features that, during evolution from a common precursor, were restricted to nonoverlapping expression domains by as of yet unknown DNA repressor elements. Second, I examined the function of Neurog2 at the initiation of retinal neurogenesis. Neurog2 and Atoh7 expression was observed sequentially in progenitor cells that give rise to the first neurons in the central retina. I determined that Neurog2, but not Atoh7, is essential for the peripheral expansion of neurogenesis and RGC genesis. In Neurog2 mutant mice, neurogenesis was delayed until the onset of retinal Ascl1/Mash1 expression, but by birth the proportions of early-born cell types are returned to normal. Ascl1 replacement of Neurog2 rescued the delay in both neural differentiation and RGC genesis, signifying that retinal development proceeds as overlapping waves of neurogenesis regulated by these bHLH factors. Finally, I further explored the interchangeability of bHLH transcription factors. To test the hypothesis that Ascl1 and Atoh7 have distinct functions in cell cycle exit and fate specification in retinal progenitor cells, I used a previously constructed mouse model, the Atoh7Ascl1KI allele, which misexpresses Ascl1 in Atoh7-lineage cells. Ascl1 replacement of Atoh7 did not rescue RGC development but increased bipolar interneuron and decreased Müller glia number in adult eyes. During the initiation of neurogenesis, ectopic Ascl1 prolonged proliferation of Atoh7-expressing cells that normally exit the cell cycle, dominant to endogenous Atoh7 function. In sum, this thesis provokes new mechanisms for the divergence of bHLH regulation and function in mouse retinal development. Neurog2 and Atoh7 have separate roles in early retinal progenitor cells during the initiation of neurogenesis. While Ascl1 can compensate for neural differentiation defects in Neurog2 mutant mice, it does not promote cell cycle exit or rescue RGC specification in terminally mitotic Atoh7-lineage cells. Together, bHLH factors have overlapping and distinct functions in the mammalian retina, defined by a combination of evolutionary homology, phase of cell cycle expression, and developmental timing.
- Published
- 2010
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.