Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 15(700): eabq7721, 2023 06 14.
Article in English | MEDLINE | ID: mdl-37315111

ABSTRACT

Intracranial aneurysms (IAs) are a high-risk factor for life-threatening subarachnoid hemorrhage. Their etiology, however, remains mostly unknown at present. We conducted screening for sporadic somatic mutations in 65 IA tissues (54 saccular and 11 fusiform aneurysms) and paired blood samples by whole-exome and targeted deep sequencing. We identified sporadic mutations in multiple signaling genes and examined their impact on downstream signaling pathways and gene expression in vitro and an arterial dilatation model in mice in vivo. We identified 16 genes that were mutated in at least one IA case and found that these mutations were highly prevalent (92%: 60 of 65 IAs) among all IA cases examined. In particular, mutations in six genes (PDGFRB, AHNAK, OBSCN, RBM10, CACNA1E, and OR5P3), many of which are linked to NF-κB signaling, were found in both fusiform and saccular IAs at a high prevalence (43% of all IA cases examined). We found that mutant PDGFRBs constitutively activated ERK and NF-κB signaling, enhanced cell motility, and induced inflammation-related gene expression in vitro. Spatial transcriptomics also detected similar changes in vessels from patients with IA. Furthermore, virus-mediated overexpression of a mutant PDGFRB induced a fusiform-like dilatation of the basilar artery in mice, which was blocked by systemic administration of the tyrosine kinase inhibitor sunitinib. Collectively, this study reveals a high prevalence of somatic mutations in NF-κB signaling pathway-related genes in both fusiform and saccular IAs and opens a new avenue of research for developing pharmacological interventions.


Subject(s)
Intracranial Aneurysm , NF-kappa B , Animals , Mice , Intracranial Aneurysm/genetics , Mutation/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Signal Transduction/genetics , Humans
2.
J Neurosci ; 42(44): 8373-8392, 2022 11 02.
Article in English | MEDLINE | ID: mdl-36127134

ABSTRACT

The chromatin remodeler CHD8 represents a high-confidence risk factor in autism, a multistage progressive neurologic disorder, however the underlying stage-specific functions remain elusive. In this study, by analyzing Chd8 conditional knock-out mice (male and female), we find that CHD8 controls cortical neural stem/progenitor cell (NSC) proliferation and survival in a stage-dependent manner. Strikingly, inducible genetic deletion reveals that CHD8 is required for the production and fitness of transit-amplifying intermediate progenitors (IPCs) essential for upper-layer neuron expansion in the embryonic cortex. p53 loss of function partially rescues apoptosis and neurogenesis defects in the Chd8-deficient brain. Further, transcriptomic and epigenomic profiling indicates that CHD8 regulates the chromatin accessibility landscape to activate neurogenesis-promoting factors including TBR2, a key regulator of IPC neurogenesis, while repressing DNA damage- and p53-induced apoptotic programs. In the adult brain, CHD8 depletion impairs forebrain neurogenesis by impeding IPC differentiation from NSCs in both subventricular and subgranular zones; however, unlike in embryos, it does not affect NSC proliferation and survival. Treatment with an antidepressant approved by the Federal Drug Administration (FDA), fluoxetine, partially restores adult hippocampal neurogenesis in Chd8-ablated mice. Together, our multistage functional studies identify temporally specific roles for CHD8 in developmental and adult neurogenesis, pointing to a potential strategy to enhance neurogenesis in the CHD8-deficient brain.SIGNIFICANCE STATEMENT The role of the high-confidence autism gene CHD8 in neurogenesis remains incompletely understood. Here, we identify a stage-specific function of CHD8 in development of NSCs in developing and adult brains by conserved, yet spatiotemporally distinct, mechanisms. In embryonic cortex, CHD8 is critical for the proliferation, survival, and differentiation of both NSC and IPCs during cortical neurogenesis. In adult brain, CHD8 is required for IPC generation but not the proliferation and survival of adult NSCs. Treatment with FDA-approved antidepressant fluoxetine partially rescues the adult neurogenesis defects in CHD8 mutants. Thus, our findings help resolve CHD8 functions throughout life during embryonic and adult neurogenesis and point to a potential avenue to promote neurogenesis in CHD8 deficiency.


Subject(s)
Autistic Disorder , Chromatin , DNA-Binding Proteins , Neurogenesis , Animals , Female , Male , Mice , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fluoxetine , Hippocampus/metabolism , Mice, Knockout , Neurogenesis/physiology , Tumor Suppressor Protein p53 , Prosencephalon
3.
Development ; 149(5)2022 03 01.
Article in English | MEDLINE | ID: mdl-35132995

ABSTRACT

Distinct neural stem cells (NSCs) reside in different regions of the subventricular zone (SVZ) and generate multiple olfactory bulb (OB) interneuron subtypes in the adult brain. However, the molecular mechanisms underlying such NSC heterogeneity remain largely unknown. Here, we show that the basic helix-loop-helix transcription factor Olig2 defines a subset of NSCs in the early postnatal and adult SVZ. Olig2-expressing NSCs exist broadly but are most enriched in the ventral SVZ along the dorsoventral axis complementary to dorsally enriched Gsx2-expressing NSCs. Comparisons of Olig2-expressing NSCs from early embryonic to adult stages using single cell transcriptomics reveal stepwise developmental changes in their cell cycle and metabolic properties. Genetic studies further show that cross-repression contributes to the mutually exclusive expression of Olig2 and Gsx2 in NSCs/progenitors during embryogenesis, but that their expression is regulated independently from each other in adult NSCs. Finally, lineage-tracing and conditional inactivation studies demonstrate that Olig2 plays an important role in the specification of OB interneuron subtypes. Altogether, our study demonstrates that Olig2 defines a unique subset of adult NSCs enriched in the ventral aspect of the adult SVZ.


Subject(s)
Interneurons/metabolism , Lateral Ventricles/growth & development , Lateral Ventricles/metabolism , Neural Stem Cells/metabolism , Olfactory Bulb/growth & development , Olfactory Bulb/metabolism , Oligodendrocyte Transcription Factor 2/metabolism , Animals , Cell Cycle/genetics , Cell Lineage/genetics , Cells, Cultured , Female , Gene Knockout Techniques , Lateral Ventricles/embryology , Male , Mice , Mice, Knockout , Neurogenesis/genetics , Olfactory Bulb/embryology , Oligodendrocyte Transcription Factor 2/genetics , Signal Transduction/genetics , Transcriptome/genetics
4.
Genes Dev ; 35(1-2): 157-174, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33334823

ABSTRACT

How homeodomain proteins gain sufficient specificity to control different cell fates has been a long-standing problem in developmental biology. The conserved Gsx homeodomain proteins regulate specific aspects of neural development in animals from flies to mammals, and yet they belong to a large transcription factor family that bind nearly identical DNA sequences in vitro. Here, we show that the mouse and fly Gsx factors unexpectedly gain DNA binding specificity by forming cooperative homodimers on precisely spaced and oriented DNA sites. High-resolution genomic binding assays revealed that Gsx2 binds both monomer and homodimer sites in the developing mouse ventral telencephalon. Importantly, reporter assays showed that Gsx2 mediates opposing outcomes in a DNA binding site-dependent manner: Monomer Gsx2 binding represses transcription, whereas homodimer binding stimulates gene expression. In Drosophila, the Gsx homolog, Ind, similarly represses or stimulates transcription in a site-dependent manner via an autoregulatory enhancer containing a combination of monomer and homodimer sites. Integrating these findings, we test a model showing how the homodimer to monomer site ratio and the Gsx protein levels defines gene up-regulation versus down-regulation. Altogether, these data serve as a new paradigm for how cooperative homeodomain transcription factor binding can increase target specificity and alter regulatory outcomes.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/embryology , Drosophila/genetics , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/metabolism , Animals , Drosophila Proteins/genetics , Genome/genetics , Genome-Wide Association Study , Homeodomain Proteins/genetics , Mice , Protein Binding , Telencephalon/embryology
5.
Development ; 147(7)2020 04 10.
Article in English | MEDLINE | ID: mdl-32122989

ABSTRACT

The Gsx2 homeodomain transcription factor promotes neural progenitor identity in the lateral ganglionic eminence (LGE), despite upregulating the neurogenic factor Ascl1. How this balance in maturation is maintained is unclear. Here, we show that Gsx2 and Ascl1 are co-expressed in subapical progenitors that have unique transcriptional signatures in LGE ventricular zone (VZ) cells. Moreover, whereas Ascl1 misexpression promotes neurogenesis in dorsal telencephalic progenitors, the co-expression of Gsx2 with Ascl1 inhibits neurogenesis. Using luciferase assays, we found that Gsx2 reduces the ability of Ascl1 to activate gene expression in a dose-dependent and DNA binding-independent manner. Furthermore, Gsx2 physically interacts with the basic helix-loop-helix (bHLH) domain of Ascl1, and DNA-binding assays demonstrated that this interaction interferes with the ability of Ascl1 to bind DNA. Finally, we modified a proximity ligation assay for tissue sections and found that Ascl1-Gsx2 interactions are enriched within LGE VZ progenitors, whereas Ascl1-Tcf3 (E-protein) interactions predominate in the subventricular zone. Thus, Gsx2 contributes to the balance between progenitor maintenance and neurogenesis by physically interacting with Ascl1, interfering with its DNA binding and limiting neurogenesis within LGE progenitors.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/embryology , Cell Proliferation , Homeodomain Proteins/metabolism , Neural Stem Cells/physiology , Neurogenesis/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain/metabolism , Cell Proliferation/genetics , Cells, Cultured , Drosophila , Embryo, Mammalian , Female , Ganglia/cytology , Ganglia/embryology , Homeodomain Proteins/genetics , Homeostasis/genetics , Male , Mice , Mice, Transgenic , Protein Binding , Telencephalon/cytology , Telencephalon/embryology
6.
Wiley Interdiscip Rev Dev Biol ; 9(3): e369, 2020 05.
Article in English | MEDLINE | ID: mdl-31825170

ABSTRACT

The mature mammalian brain has long been thought to be a structurally rigid, static organ since the era of Ramón y Cajal in the early 20th century. Evidence accumulated over the past three decades, however, has completely overturned this long-held view. We now know that new neurons and glia are continuously added to the brain at postnatal stages, even in mature adults of various mammalian species, including humans. Moreover, these newly added cells contribute to structural plasticity and play important roles in higher order brain function, as well as repair after damage. A major source of these new neurons and glia is neural stem cells (NSCs) that persist in specialized niches in the brain throughout life. With this new view, our understanding of normal brain physiology and interventional approaches to various brain disorders has changed markedly in recent years. This article provides a brief overview on the historical changes in our understanding of the developmental dynamics of neurogenesis and gliogenesis in the postnatal and adult mammalian brain and discusses the roles of NSCs and other progenitor populations in such cellular dynamics in health and disease of the postnatal mammalian brain. This article is categorized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cell Differentiation and Reversion Adult Stem Cells, Tissue Renewal, and Regeneration > Tissue Stem Cells and Niches Adult Stem Cells, Tissue Renewal, and Regeneration > Regeneration Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cells and Disease.


Subject(s)
Brain/growth & development , Neurogenesis , Oligodendroglia/cytology , Animals , Brain/cytology , Brain/pathology , Humans , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Oligodendrocyte Precursor Cells/cytology , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/metabolism
7.
Dev Biol ; 442(1): 115-126, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29990475

ABSTRACT

The homeobox gene Gsx2 has previously been shown to inhibit oligodendroglial specification in dorsal lateral ganglionic eminence (dLGE) progenitors of the ventral telencephalon. The precocious specification of oligodendrocyte progenitor cells (OPCs) observed in Gsx2 mutants, however, is transient and begins to normalize by late stages of embryogenesis. Interestingly, this normalization correlates with the expansion of Gsx1, a close homolog of Gsx2, in a subset of progenitors in the Gsx2 mutant LGE. Here, we interrogated the mechanisms underlying oligodendroglial specification in Gsx2 mutants in relation to Gsx1. We found that Gsx1/2 double mutant embryos exhibit a more robust expansion of Olig2+ cells (i.e. OPCs) in the subventricular zone (SVZ) of the dLGE than Gsx2 mutants. Moreover, misexpression of Gsx1 throughout telencephalic VZ progenitors from E15 and onward resulted in a significant reduction of cortical OPCs. These results demonstrate redundant roles of Gsx1 and Gsx2 in suppressing early OPC specification in LGE VZ progenitors. However, Gsx1/2 mutants did not show a significant increase in adjacent cortical OPCs at later stages compared to Gsx2 mutants. This is likely due to reduced proliferation of OPCs within the SVZ of the Gsx1/2 double mutant LGE, suggesting a novel role for Gsx1 in expansion of migrating OPCs in the ventral telencephalon. We further investigated the glial specification mechanisms downstream of Gsx2 by generating Olig2/Gsx2 double mutants. Consistent with the known essential role for Olig2 in OPC specification, ectopic production of cortical OPCs observed in Gsx2 mutants disappeared in Olig2/Gsx2 double mutants. These mutants, however, maintained the expanded expression of gliogenic markers Zbtb20 and Bcan in the VZ of the LGE similarly to Gsx2 single mutants, suggesting that Gsx2 suppresses gliogenesis via Olig2-dependent and -independent mechanisms.


Subject(s)
Homeodomain Proteins/genetics , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Lineage , Embryo, Mammalian/metabolism , Ganglia/metabolism , Ganglia/physiology , Homeodomain Proteins/metabolism , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Neurogenesis/physiology , Neuroglia/metabolism , Neuroglia/physiology , Neurons/metabolism , Neurons/physiology , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/cytology , Oligodendroglia/physiology , Stem Cells/metabolism , Stem Cells/physiology , Telencephalon/metabolism , Transcription Factors
8.
Genesis ; 54(10): 542-549, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27618396

ABSTRACT

In this study, we generated a transgenic mouse line driving Cre and EGFP expression with two putative cis-regulatory modules (CRMs) (i.e., hs687 and hs678) upstream of the homeobox gene Gsx2 (formerly Gsh2), a critical gene for establishing lateral ganglionic eminence (LGE) identity. The combination of these two CRMs drives transgene expression within the endogenous Gsx2 expression domains along the anterior-posterior neuraxis. By crossing this transgenic line with the RosatdTomato (Ai14) reporter mouse line, we observed a unique recombination pattern in the lateral ventral telencephalon, namely the LGE and the dorsal half of the medial GE (MGE), but not in the septum. We found robust recombination in many cell types derived from these embryonic regions, including olfactory bulb and amygdala interneurons and striatal projection neurons from the LGE, as well as cortical interneurons from the MGE and caudal GE (CGE). In summary, this transgenic mouse line represents a new tool for genetic manipulation in the LGE/CGE and the dorsal half of MGE.


Subject(s)
Ganglion Cysts/genetics , Green Fluorescent Proteins/genetics , Homeodomain Proteins/genetics , Telencephalon/growth & development , Amygdala/growth & development , Animals , Ganglion Cysts/pathology , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/biosynthesis , Integrases/genetics , Mice , Mice, Transgenic , Neurons/metabolism , Neurons/pathology , Olfactory Bulb/growth & development , Telencephalon/metabolism
9.
Article in English | MEDLINE | ID: mdl-27235475

ABSTRACT

Adult neurogenesis in the mammalian brain is often viewed as a continuation of neurogenesis at earlier, developmental stages. Here, we will critically review the extent to which this is the case highlighting similarities as well as key differences. Although many transcriptional regulators are shared in neurogenesis at embryonic and adult stages, recent findings on the molecular mechanisms by which these neuronal fate determinants control fate acquisition and maintenance have revealed profound differences between development and adulthood. Importantly, adult neurogenesis occurs in a gliogenic environment, hence requiring adult-specific additional and unique mechanisms of neuronal fate specification and maintenance. Thus, a better understanding of the molecular logic for continuous adult neurogenesis provides important clues to develop strategies to manipulate endogenous stem cells for the purpose of repair.


Subject(s)
Neurogenesis , Adult , Animals , Cell Cycle , Cell Lineage , Humans
10.
Proc Natl Acad Sci U S A ; 112(44): 13717-22, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26483457

ABSTRACT

Müller glial cells are the source of retinal regeneration in fish and birds; although this process is efficient in fish, it is less so in birds and very limited in mammals. It has been proposed that factors necessary for providing neurogenic competence to Müller glia in fish and birds after retinal injury are not expressed in mammals. One such factor, the proneural transcription factor Ascl1, is necessary for retinal regeneration in fish but is not expressed after retinal damage in mice. We previously reported that forced expression of Ascl1 in vitro reprograms Müller glia to a neurogenic state. We now test whether forced expression of Ascl1 in mouse Müller glia in vivo stimulates their capacity for retinal regeneration. We find that transgenic expression of Ascl1 in adult Müller glia in undamaged retina does not overtly affect their phenotype; however, when the retina is damaged, the Ascl1-expressing glia initiate a response that resembles the early stages of retinal regeneration in zebrafish. The reaction to injury is even more pronounced in Müller glia in young mice, where the Ascl1-expressing Müller glia give rise to amacrine and bipolar cells and photoreceptors. DNaseI-seq analysis of the retina and Müller glia shows progressive reduction in accessibility of progenitor gene cis-regulatory regions consistent with the reduction in their reprogramming. These results show that at least one of the differences between mammal and fish Müller glia that bears on their difference in regenerative potential is the proneural transcription factor Ascl1.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Ependymoglial Cells/metabolism , Regeneration , Retina/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Mice , Mice, Transgenic
11.
eNeuro ; 2(5)2015.
Article in English | MEDLINE | ID: mdl-26730402

ABSTRACT

Cell reprogramming technologies have enabled the generation of various specific cell types including neurons from readily accessible patient cells, such as skin fibroblasts, providing an intriguing novel cell source for autologous cell transplantation. However, cell transplantation faces several difficult hurdles such as cell production and purification, long-term survival, and functional integration after transplantation. Recently, in vivo reprogramming, which makes use of endogenous cells for regeneration purpose, emerged as a new approach to circumvent cell transplantation. There has been evidence for in vivo reprogramming in the mouse pancreas, heart, and brain and spinal cord with various degrees of success. This mini review summarizes the latest developments presented in the first symposium on in vivo reprogramming glial cells into functional neurons in the brain and spinal cord, held at the 2014 annual meeting of the Society for Neuroscience in Washington, DC.


Subject(s)
Brain/physiopathology , Cellular Reprogramming Techniques/methods , Nerve Regeneration/physiology , Spinal Cord/physiopathology , Animals , Cellular Reprogramming/physiology , Congresses as Topic , District of Columbia , Humans , Neurogenesis/physiology , Neurons/physiology
12.
Neuron ; 83(5): 1085-97, 2014 Sep 03.
Article in English | MEDLINE | ID: mdl-25189209

ABSTRACT

The activity of adult stem cells is regulated by signals emanating from the surrounding tissue. Many niche signals have been identified, but it is unclear how they influence the choice of stem cells to remain quiescent or divide. Here we show that when stem cells of the adult hippocampus receive activating signals, they first induce the expression of the transcription factor Ascl1 and only subsequently exit quiescence. Moreover, lowering Ascl1 expression reduces the proliferation rate of hippocampal stem cells, and inactivating Ascl1 blocks quiescence exit completely, rendering them unresponsive to activating stimuli. Ascl1 promotes the proliferation of hippocampal stem cells by directly regulating the expression of cell-cycle regulatory genes. Ascl1 is similarly required for stem cell activation in the adult subventricular zone. Our results support a model whereby Ascl1 integrates inputs from both stimulatory and inhibitory signals and converts them into a transcriptional program activating adult neural stem cells.


Subject(s)
Adult Stem Cells/cytology , Gene Expression Regulation, Developmental/genetics , Hippocampus/cytology , Neurogenesis/genetics , Adult Stem Cells/metabolism , Age Factors , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Proliferation , Cerebral Ventricles/cytology , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 1/metabolism , Gene Expression Regulation, Developmental/drug effects , Glial Fibrillary Acidic Protein/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Kainic Acid/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Box Domain Proteins/metabolism , p300-CBP Transcription Factors/metabolism
13.
J Neurosci ; 34(10): 3767-78, 2014 Mar 05.
Article in English | MEDLINE | ID: mdl-24599474

ABSTRACT

The protein tyrosine phosphatase Shp2 (PTPN11) is crucial for normal brain development and has been implicated in dorsal telencephalic neuronal and astroglia cell fate decisions. However, its roles in the ventral telencephalon and during oligodendrogenesis in the telencephalon remain largely unknown. Shp2 gain-of-function (GOF) mutations are observed in Noonan syndrome, a type of RASopathy associated with multiple phenotypes, including cardiovascular, craniofacial, and neurocognitive abnormalities. To gain insight into requirements for Shp2 (LOF) and the impact of abnormal Shp2 GOF mutations, we used a Shp2 conditional mutant allele (LOF) and a cre inducible Shp2-Q79R GOF transgenic mouse in combination with Olig2(cre/+) mice to target embryonic ventral telencephalic progenitors and the oligodendrocyte lineage. In the absence of Shp2 (LOF), neuronal cell types originating from progenitors in the ventral telencephalon were generated, but oligodendrocyte progenitor cell (OPC) generation was severely impaired. Late embryonic and postnatal Shp2 cKOs showed defects in the generation of OPCs throughout the telencephalon and subsequent reductions in white matter myelination. Conversely, transgenic expression of the Shp2 GOF Noonan syndrome mutation resulted in elevated OPC numbers in the embryo and postnatal brain. Interestingly, expression of this mutation negatively influenced myelination as mice displayed abnormal myelination and fewer myelinated axons in the white matter despite elevated OPC numbers. Increased proliferating OPCs and elevated MAPK activity were also observed during oligodendrogenesis after expression of Shp2 GOF mutation. These results support the notion that appropriate Shp2 activity levels control the number as well as the differentiation of oligodendrocytes during development.


Subject(s)
Nerve Fibers, Myelinated/enzymology , Oligodendroglia/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/biosynthesis , Stem Cells/enzymology , Telencephalon/embryology , Telencephalon/enzymology , Animals , Cell Differentiation/physiology , Mice , Mice, Transgenic , Telencephalon/cytology
14.
Nat Commun ; 4: 2373, 2013.
Article in English | MEDLINE | ID: mdl-23974433

ABSTRACT

Direct reprogramming of non-neuronal cells to generate new neurons is a promising approach to repair damaged brains. Impact of the in vivo environment on neuronal reprogramming, however, is poorly understood. Here we show that regional differences and injury conditions have significant influence on the efficacy of reprogramming and subsequent survival of the newly generated neurons in the adult rodent brain. A combination of local exposure to growth factors and retrovirus-mediated overexpression of the neurogenic transcription factor Neurogenin2 can induce new neurons from non-neuronal cells in the adult neocortex and striatum where neuronal turnover is otherwise very limited. These two regions respond to growth factors and Neurogenin2 differently and instruct new neurons to exhibit distinct molecular phenotypes. Moreover, ischaemic insult differentially affects differentiation of new neurons in these regions. These results demonstrate strong environmental impact on direct neuronal reprogramming in vivo.


Subject(s)
Aging/pathology , Brain/pathology , Cellular Reprogramming , Neurons/metabolism , Animals , Brain/drug effects , Cellular Reprogramming/drug effects , Gene Expression Profiling , Green Fluorescent Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Ischemia/pathology , Male , Neocortex/drug effects , Neocortex/metabolism , Neocortex/pathology , Neostriatum/drug effects , Neostriatum/metabolism , Neostriatum/pathology , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , Neurons/drug effects , Organ Specificity/drug effects , Rats , Rats, Sprague-Dawley , Retroviridae/drug effects , Retroviridae/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology
15.
Genes Dev ; 27(11): 1272-87, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23723414

ABSTRACT

Neural stem cells (NSCs) reside in widespread regions along the lateral ventricle and generate diverse olfactory bulb (OB) interneuron subtypes in the adult mouse brain. Molecular mechanisms underlying their regional diversity, however, are not well understood. Here we show that the homeodomain transcription factor Gsx2 plays a crucial role in the region-specific control of adult NSCs in both persistent and injury-induced neurogenesis. In the intact brain, Gsx2 is expressed in a regionally restricted subset of NSCs and promotes the activation and lineage progression of stem cells, thereby controlling the production of selective OB neuron subtypes. Moreover, Gsx2 is ectopically induced in damaged brains outside its normal expression domains and is required for injury-induced neurogenesis in the subventricular zone (SVZ). These results demonstrate that mobilization of adult NSCs is controlled in a region-specific manner and that distinct mechanisms operate in continuous and injury-induced neurogenesis in the adult brain.


Subject(s)
Homeodomain Proteins/metabolism , Lateral Ventricles/cytology , Lateral Ventricles/injuries , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis , Adult Stem Cells/metabolism , Animals , Cell Lineage , Lateral Ventricles/metabolism , Mice , Neural Stem Cells/classification , Olfactory Bulb/cytology , Organ Specificity , Stem Cell Niche , Transcription Factors/metabolism
16.
J Neurosci ; 33(23): 9752-9768, 2013 Jun 05.
Article in English | MEDLINE | ID: mdl-23739972

ABSTRACT

Oligodendrocytes are the myelin-forming cells of the CNS. They differentiate from oligodendrocyte precursor cells (OPCs) that are produced from progenitors throughout life but more actively during the neonatal period and in response to demyelinating insults. An accurate regulation of oligodendrogenesis is required to generate oligodendrocytes during these developmental or repair processes. We hypothesized that this regulation implicates transcription factors, which are expressed by OPCs and/or their progenitors. Ascl1/Mash1 is a proneural transcription factor previously implicated in embryonic oligodendrogenesis and operating in genetic interaction with Olig2, an essential transcriptional regulator in oligodendrocyte development. Herein, we have investigated the contribution of Ascl1 to oligodendrocyte development and remyelination in the postnatal cortex. During the neonatal period, Ascl1 expression was detected in progenitors of the cortical subventricular zone and in cortical OPCs. Different genetic approaches to delete Ascl1 in cortical progenitors or OPCs reduced neonatal oligodendrogenesis, showing that Ascl1 positively regulated both OPC specification from subventricular zone progenitors as well as the balance between OPC differentiation and proliferation. Examination of remyelination processes, both in the mouse model for focal demyelination of the corpus callosum and in multiple sclerosis lesions in humans, indicated that Ascl1 activity was upregulated along with increased oligodendrogenesis observed in remyelinating lesions. Additional genetic evidence indicated that remyelinating oligodendrocytes derived from Ascl1(+) progenitors/OPCs and that Ascl1 was required for proper remyelination. Together, our results show that Ascl1 function modulates multiple steps of OPC development in the postnatal brain and in response to demyelinating insults.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Brain/physiology , Myelin Sheath/physiology , Oligodendroglia/metabolism , Animals , Brain/cytology , Female , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Nerve Fibers, Myelinated/metabolism , Neural Stem Cells/metabolism , Oligodendroglia/cytology
17.
Development ; 140(11): 2289-98, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23637331

ABSTRACT

The homeobox gene Gsx2 has previously been shown to be required for the specification of distinct neuronal subtypes derived from lateral ganglionic eminence (LGE) progenitors at specific embryonic time points. However, its role in the subsequent generation of oligodendrocytes from these progenitors remains unclear. We have utilized conditional gain-of-function and loss-of-function approaches in order to elucidate the role of Gsx2 in the switch between neurogenesis and oligodendrogenesis within the embryonic ventral telencephalon. In the absence of Gsx2 expression, an increase in oligodendrocyte precursor cells (OPCs) with a concomitant decrease in neurogenesis is observed in the subventricular zone of the LGE at mid-stages of embryogenesis (i.e. E12.5-15.5), which subsequently leads to an increased number of Gsx2-derived OPCs within the adjacent mantle regions of the cortex before birth at E18.5. Moreover, using Olig2(cre) to conditionally inactivate Gsx2 throughout the ventral telencephalon with the exception of the dorsal (d)LGE, we found that the increase in cortical OPCs in Gsx2 germline mutants are derived from dLGE progenitors. We also show that Ascl1 is required for the expansion of these dLGE-derived OPCs in the cortex of Gsx2 mutants. Complementing these results, gain-of-function experiments in which Gsx2 was expressed throughout most of the late-stage embryonic telencephalon (i.e. E15.5-18.5) result in a significant decrease in the number of cortical OPCs. These results support the notion that high levels of Gsx2 suppress OPC specification in dLGE progenitors and that its downregulation is required for the transition from neurogenesis to oligodendrogenesis.


Subject(s)
Ganglia/growth & development , Gene Expression Regulation, Developmental , Homeodomain Proteins/physiology , Oligodendroglia/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Lineage , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Mice , Microscopy, Fluorescence , Mutation , Nerve Tissue Proteins/metabolism , Neurons/physiology , Oligodendrocyte Transcription Factor 2 , Receptor, Platelet-Derived Growth Factor alpha/metabolism , SOXE Transcription Factors/metabolism , Stem Cells/physiology , Telencephalon/physiology , Time Factors
18.
J Neurosci ; 33(3): 1241-58, 2013 Jan 16.
Article in English | MEDLINE | ID: mdl-23325260

ABSTRACT

Appropriately targeted manipulation of endogenous neural stem progenitor (NSP) cells may contribute to therapies for trauma, stroke, and neurodegenerative disease. A prerequisite to such therapies is a better understanding of the mechanisms regulating adult NSP cells in vivo. Indirect data suggest that endogenous ciliary neurotrophic factor (CNTF) receptor signaling may inhibit neuronal differentiation of NSP cells. We challenged subventricular zone (SVZ) cells in vivo with low concentrations of CNTF to anatomically characterize cells containing functional CNTF receptors. We found that type B "stem" cells are highly responsive, whereas type C "transit-amplifying" cells and type A neuroblasts are remarkably unresponsive, as are GFAP(+) astrocytes found outside the SVZ. CNTF was identified in a subset of type B cells that label with acute BrdU administration. Disruption of in vivo CNTF receptor signaling in SVZ NSP cells, with a "floxed" CNTF receptor α (CNTFRα) mouse line and a gene construct driving Cre recombinase (Cre) expression in NSP cells, led to increases in SVZ-associated neuroblasts and new olfactory bulb neurons, as well as a neuron subtype-specific, adult-onset increase in olfactory bulb neuron populations. Adult-onset receptor disruption in SVZ NSP cells with a recombinant adeno-associated virus (AAV-Cre) also led to increased neurogenesis. However, the maintenance of type B cell populations was apparently unaffected by the receptor disruption. Together, the data suggest that endogenous CNTF receptor signaling in type B stem cells inhibits adult neurogenesis, and further suggest that the regulation may occur in a neuron subtype-specific manner.


Subject(s)
Lateral Ventricles/physiology , Neurogenesis/physiology , Neurons/physiology , Prosencephalon/physiology , Receptor, Ciliary Neurotrophic Factor/metabolism , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Ciliary Neurotrophic Factor/metabolism , Lateral Ventricles/cytology , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Olfactory Bulb/cytology , Olfactory Bulb/physiology , Receptor, Ciliary Neurotrophic Factor/genetics , Signal Transduction/physiology
19.
Development ; 138(3): 409-19, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21205786

ABSTRACT

GABAergic neurons and oligodendrocytes originate from progenitors within the ventral telencephalon. However, the molecular mechanisms that control neuron-glial cell-fate segregation, especially how extrinsic factors regulate cell-fate changes, are poorly understood. We have discovered that the Wnt receptor Ryk promotes GABAergic neuron production while repressing oligodendrocyte formation in the ventral telencephalon. We demonstrate that Ryk controls the cell-fate switch by negatively regulating expression of the intrinsic oligodendrogenic factor Olig2 while inducing expression of the interneuron fate determinant Dlx2. In addition, we demonstrate that Ryk is required for GABAergic neuron induction and oligodendrogenesis inhibition caused by Wnt3a stimulation. Furthermore, we showed that the cleaved intracellular domain of Ryk is sufficient to regulate the cell-fate switch by regulating the expression of intrinsic cell-fate determinants. These results identify Ryk as a multi-functional receptor that is able to transduce extrinsic cues into progenitor cells, promote GABAergic neuron formation, and inhibit oligodendrogenesis during ventral embryonic brain development.


Subject(s)
Neurons/cytology , Neurons/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Telencephalon/cytology , gamma-Aminobutyric Acid/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/cytology , Brain/embryology , Brain/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/drug effects , Receptor Protein-Tyrosine Kinases/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Wnt Proteins/pharmacology , Wnt3 Protein , Wnt3A Protein
20.
Proc Natl Acad Sci U S A ; 108(4): 1675-80, 2011 Jan 25.
Article in English | MEDLINE | ID: mdl-21205889

ABSTRACT

Homeobox genes Gsx1 and Gsx2 (formerly Gsh1 and Gsh2) are among the earliest transcription factors expressed in neuronal progenitors of the lateral ganglionic eminence (LGE) in the ventral telencephalon. Gsx2 is required for the early specification of LGE progenitor cells and recently has been shown to specify different LGE neuronal subtypes at distinct time points. In Gsx2 mutants, Gsx1 compensates, at least in part, for the loss of Gsx2 in the specification of LGE neuronal subtypes. Because no specific phenotype has been described in Gsx1 mutants, it is unclear what role this factor plays in the development of the ventral telencephalon. Here, we used a gain-of-function approach to express either Gsx1 or Gsx2 throughout the telencephalon and found that Gsx1 functions similarly to Gsx2 in the specification of LGE identity. However, our results show that Gsx1 and Gsx2 differentially regulate the maturation of LGE progenitors. Specifically, Gsx2 maintains LGE progenitors in an undifferentiated state, whereas Gsx1 promotes progenitor maturation and the acquisition of neuronal phenotypes, at least in part, through the down-regulation of Gsx2. These unique results indicate that the two closely related Gsx genes similarly regulate LGE patterning but oppositely control the balance between proliferation and differentiation in the neuronal progenitor pool.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Neural Stem Cells/metabolism , Telencephalon/metabolism , Animals , Cell Cycle , Cell Differentiation , Cell Proliferation , Cells, Cultured , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Male , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Mutation , Neural Stem Cells/cytology , Reverse Transcriptase Polymerase Chain Reaction , Telencephalon/cytology , Telencephalon/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...