Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Invest Ophthalmol Vis Sci ; 57(3): 1488-99, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27031842

ABSTRACT

PURPOSE: Müller glia (MG), the principal glial cells of the vertebrate retina, display quiescent progenitor cell characteristics. They express key progenitor markers, including the high mobility group box transcription factor SOX2 and maintain a progenitor-like morphology. In the embryonic and mature central nervous system, SOX2 maintains neural stem cell identity. However, its function in committed Müller glia has yet to be determined. METHODS: We use inducible, MG-specific genetic ablation of Sox2 in vivo at the peak of MG genesis to analyze its function in the maturation of murine MG and effects on other cells in the retina. Histologic and functional analysis of the Sox2-deficient retinas is conducted at key points in postnatal development. RESULTS: Ablation of Sox2 in the postnatal retina results in disorganization of MG processes in the inner plexiform layer and mislocalized cell bodies in the nuclear layers. This disorganization is concurrent with a thinning of the neural retina and disruption of neuronal processes in the inner and outer plexiform layers. Functional analysis by electroretinography reveals a decrease in the b-wave amplitude. Disruption of MG maturation due to Sox2 ablation therefore negatively affected the function of the retina. CONCLUSIONS: These results demonstrate a novel role for SOX2 in glial process outgrowth and adhesion, and provide new insights into the essential role Müller glia play in the development of retinal cytoarchitecture. Prior to this work, SOX2 was known to have a primary role in determining cell fate. Our experiments bypass cell fate conversion to establish a new role for SOX2 in a committed cell lineage.


Subject(s)
Aging/genetics , Ependymoglial Cells/metabolism , Gene Expression Regulation, Developmental , Neuroglia/metabolism , RNA/genetics , Retina/physiology , SOXB1 Transcription Factors/genetics , Animals , Cell Differentiation , Cell Proliferation , Electroretinography , Ependymoglial Cells/ultrastructure , Immunohistochemistry , Mice , Mice, Transgenic , Microscopy, Electron , Neuroglia/ultrastructure , Retina/ultrastructure , SOXB1 Transcription Factors/biosynthesis
2.
Development ; 142(11): 2037-47, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25953344

ABSTRACT

Organ growth occurs through the integration of external growth signals during the G1 phase of the cell cycle to initiate DNA replication. Although numerous growth factor signals have been shown to be required for the proliferation of cardiomyocytes, genetic studies have only identified a very limited number of transcription factors that act to regulate the entry of cardiomyocytes into S phase. Here, we report that the cardiac para-zinc-finger protein CASZ1 is expressed in murine cardiomyocytes. Genetic fate mapping with an inducible Casz1 allele demonstrates that CASZ1-expressing cells give rise to cardiomyocytes in the first and second heart fields. We show through the generation of a cardiac conditional null mutation that Casz1 is essential for the proliferation of cardiomyocytes in both heart fields and that loss of Casz1 leads to a decrease in cardiomyocyte cell number. We further report that the loss of Casz1 leads to a prolonged or arrested S phase, a decrease in DNA synthesis, an increase in phospho-RB and a concomitant decrease in the cardiac mitotic index. Taken together, these studies establish a role for CASZ1 in mammalian cardiomyocyte cell cycle progression in both the first and second heart fields.


Subject(s)
DNA-Binding Proteins/metabolism , G1 Phase , Heart/embryology , Mammals/embryology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , S Phase , Transcription Factors/metabolism , Animals , Cell Lineage , Cell Proliferation , Embryo, Mammalian/metabolism , Female , Integrases/metabolism , Male , Mice , Myocardium/cytology , Myocardium/metabolism , Myocardium/ultrastructure
3.
Neural Dev ; 9: 27, 2014 Dec 09.
Article in English | MEDLINE | ID: mdl-25488119

ABSTRACT

BACKGROUND: Eye development in vertebrates relies on the critical regulation of SOX2 expression. Humans with mutations in SOX2 often suffer from eye defects including anophthalmia (no eye) and microphthalmia (small eye). In mice, deletion of Sox2 in optic cup progenitor cells results in loss of neural competence and cell fate conversion of the neural retina to a non-neurogenic fate, specifically the acquisition of fate associated with progenitors of the ciliary epithelium. This fate is also promoted with constitutive expression of stabilized ß-Catenin in the optic cup, where the WNT pathway is up-regulated. We addressed whether SOX2 co-ordinates the neurogenic boundary of the retina through modulating the WNT/ß-Catenin pathway by using a genetic approach in the mouse. RESULTS: Upon deletion of Sox2 in the optic cup, response to WNT signaling was expanded, correlating with loss of neural competence, cell fate conversion of the neural retina to ciliary epithelium primordium and, in addition, increased cell cycle time of optic cup progenitors. Removal of Ctnnb1 rescued the cell fate conversion; however, the loss of neural competence and the proliferation defect resulting from lack of SOX2 were not overcome. Lastly, central Sox2-deficient optic cup progenitor cells exhibited WNT-independent up-regulation of D-type Cyclins. CONCLUSION: We propose two distinct roles for SOX2 in the developing retina. Our findings suggest that SOX2 antagonizes the WNT pathway to maintain a neurogenic fate and, in contrast, regulates cycling of optic cup progenitors in a WNT-independent manner. Given that WNT signaling acting upstream of SOX2 has been implicated in the tumorigenicity of embryonic stem cell-derived retinal progenitor cells, our results distinguish the endogenous role of WNT signaling in early optic cup patterning and support a WNT-independent role for SOX2 in maintaining retinal progenitor cell proliferation.


Subject(s)
Retina/cytology , Retina/embryology , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , SOXB1 Transcription Factors/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis , SOXB1 Transcription Factors/genetics , beta Catenin/genetics
4.
Am J Physiol Lung Cell Mol Physiol ; 306(7): L645-60, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24487391

ABSTRACT

Tracheobronchial submucosal glands (SMGs) are derived from one or more multipotent glandular stem cells that coalesce to form a placode in surface airway epithelium (SAE). Wnt/ß-catenin-dependent induction of lymphoid enhancer factor (Lef-1) gene expression during placode formation is an early event required for SMG morphogenesis. We discovered that Sox2 expression is repressed as Lef-1 is induced within airway SMG placodes. Deletion of Lef-1 did not activate Sox2 expression in SMG placodes, demonstrating that Lef-1 activation does not directly inhibit Sox2 expression. Repression of Sox2 protein in SMG placodes occurred posttranscriptionally, since the activity of its endogenous promoter remained unchanged in SMG placodes. Thus we hypothesized that Sox2 transcriptionally represses Lef-1 expression in the SAE and that suppression of Sox2 in SMG placodes activates Wnt/ß-catenin-dependent induction of Lef-1 during SMG morphogenesis. Consistent with this hypothesis, transcriptional reporter assays, ChIP analyses, and DNA-protein binding studies revealed a functional Sox2 DNA binding site in the Lef-1 promoter that is required for suppressing ß-catenin-dependent transcription. In polarized primary airway epithelium, Wnt induction enhanced Lef-1 expression while also inhibiting Sox2 expression. Conditional deletion of Sox2 also enhanced Lef-1 expression in polarized primary airway epithelium, but this induction was significantly augmented by Wnt stimulation. Our findings provide the first evidence that Sox2 acts as a repressor to directly modulate Wnt-responsive transcription of the Lef-1 gene promoter. These studies support a model whereby Wnt signals and Sox2 dynamically regulate the expression of Lef-1 in airway epithelia and potentially also during SMG development.


Subject(s)
Lymphoid Enhancer-Binding Factor 1/biosynthesis , Respiratory System/growth & development , SOXB1 Transcription Factors/physiology , Acute Lung Injury/physiopathology , Animals , Animals, Newborn , Humans , Mice , Mice, Transgenic , Promoter Regions, Genetic/physiology , Wnt Proteins/physiology , beta Catenin/physiology
5.
Stem Cells Dev ; 22(12): 1779-88, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23316968

ABSTRACT

Skin-derived precursors (SKPs) are an attractive stem cell model for cell-based therapies. SKPs can be readily generated from embryonic and adult mice and adult humans, exhibit a high degree of multipotency, and have the potential to serve as a patient autologous stem cell. The advancement of these cells toward therapeutic use depends on the ability to control precisely the self-renewal and differentiation of SKPs. Here we show that two well-known stem cell factors, Foxd3 and Sox2, are critical regulators of the stem cell properties of SKPs. Deletion of Foxd3 completely abolishes the sphere-forming potential of these cells. In the absence of Sox2, SKP spheres can be formed, but with reduced size and frequency. Our results provide entry points into the gene regulatory networks dictating SKP behavior, and pave the way for future studies on a therapeutically relevant stem cell.


Subject(s)
Embryonic Stem Cells/metabolism , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Multipotent Stem Cells/metabolism , Repressor Proteins/genetics , SOXB1 Transcription Factors/genetics , Skin/metabolism , Transcription, Genetic , Animals , Cell Differentiation , Embryo, Mammalian , Embryonic Stem Cells/cytology , Forkhead Transcription Factors/deficiency , Mice , Mice, Transgenic , Multipotent Stem Cells/cytology , Primary Cell Culture , Repressor Proteins/deficiency , SOXB1 Transcription Factors/deficiency , Signal Transduction , Skin/cytology , Skin/embryology
6.
J Clin Invest ; 122(10): 3635-46, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22945632

ABSTRACT

Sex-determining region Y (SRY) box 2 (SOX2) haploinsufficiency causes a form of hypopituitarism in humans that is characterized by gonadotrophin deficiency known as hypogonadotrophic hypogonadism. Here, we conditionally deleted Sox2 in mice to investigate the pathogenesis of hypogonadotrophic hypogonadism. First, we found that absence of SOX2 in the developing Rathke pouch of conditional embryos led to severe anterior lobe hypoplasia with drastically reduced expression of the pituitary-specific transcription factor POU class 1 homeobox 1 (POU1F1) as well as severe disruption of somatotroph and thyrotroph differentiation. In contrast, corticotrophs, rostral-tip POU1F1-independent thyrotrophs, and, interestingly, lactotrophs and gonadotrophs were less affected. Second, we identified a requirement for SOX2 in normal proliferation of periluminal progenitors; in its absence, insufficient precursors were available to produce all cell lineages of the anterior pituitary. Differentiated cells derived from precursors exiting cell cycle at early stages, including corticotrophs, rostral-tip thyrotrophs, and gonadotrophs, were generated, while hormone-producing cells originating from late-born precursors, such as somatotrophs and POU1F1-dependent thyrotrophs, were severely reduced. Finally, we found that 2 previously characterized patients with SOX2 haploinsufficiency and associated hypogonadotrophic hypogonadism had a measurable response to gonadotropin-releasing hormone (GnRH) stimulation, suggesting that it is not the absence of gonadotroph differentiation, but rather the deficient hypothalamic stimulation of gonadotrophs, that underlies typical hypogonadotrophic hypogonadism.


Subject(s)
Hypogonadism/genetics , Hypothalamo-Hypophyseal System/physiology , SOXB1 Transcription Factors/physiology , Animals , Cell Differentiation , Cell Lineage , Female , Gene Expression Regulation, Developmental , Gonadotropin-Releasing Hormone/therapeutic use , Heterozygote , Homeodomain Proteins/genetics , Humans , Hypogonadism/drug therapy , Hypogonadism/physiopathology , Mice , Mice, Knockout , Organogenesis/genetics , Organogenesis/physiology , Pituitary Gland, Anterior/abnormalities , Pituitary Gland, Anterior/embryology , Pituitary Gland, Anterior/metabolism , Pituitary Gland, Anterior/pathology , Repressor Proteins/genetics , SOXB1 Transcription Factors/deficiency , SOXB1 Transcription Factors/genetics , Somatotrophs/pathology , Thyrotrophs/pathology , Transcription Factor Pit-1/deficiency
7.
Mol Cell ; 46(5): 573-83, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22560721

ABSTRACT

Human embryonic stem (hES) cells activate a rapid apoptotic response after DNA damage but the underlying mechanisms are unknown. A critical mediator of apoptosis is Bax, which is reported to become active and translocate to the mitochondria only after apoptotic stimuli. Here we show that undifferentiated hES cells constitutively maintain Bax in its active conformation. Surprisingly, active Bax was maintained at the Golgi rather than at the mitochondria, thus allowing hES cells to effectively minimize the risks associated with having preactivated Bax. After DNA damage, active Bax rapidly translocated to the mitochondria by a p53-dependent mechanism. Interestingly, upon differentiation, Bax was no longer active, and cells were not acutely sensitive to DNA damage. Thus, maintenance of Bax in its active form is a unique mechanism that can prime hES cells for rapid death, likely to prevent the propagation of mutations during the early critical stages of embryonic development.


Subject(s)
Apoptosis , Embryonic Stem Cells/metabolism , Golgi Apparatus/metabolism , bcl-2-Associated X Protein/metabolism , Acetylation , Antigens, Nuclear/metabolism , Biological Transport , DNA Damage , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Gene Silencing , Genes, bcl-2 , Humans , Ku Autoantigen , Mitochondria/metabolism , Tumor Suppressor Protein p53/physiology , bcl-2-Associated X Protein/analysis
8.
Acta Neuropathol ; 124(2): 259-71, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22349813

ABSTRACT

Activating mutations in the gene encoding ß-catenin have been identified in the paediatric form of human craniopharyngioma (adamantinomatous craniopharyngioma, ACP), a histologically benign but aggressive pituitary tumour accounting for up to 10% of paediatric intracranial tumours. Recently, we generated an ACP mouse model and revealed that, as in human ACP, nucleocytoplasmic accumulation of ß-catenin (ß-cat(nc)) and over-activation of the Wnt/ß-catenin pathway occurs only in a very small proportion of cells, which form clusters. Here, combining mouse genetics, fluorescence labelling and flow-sorting techniques, we have isolated these cells from tumorigenic mouse pituitaries and shown that the ß-cat(nc) cells are enriched for colony-forming cells when cultured in stem cell-promoting media, and have longer telomeres, indicating shared properties with normal pituitary progenitors/stem cells (PSCs). Global gene profiling analysis has revealed that these ß-cat(nc) cells express high levels of secreted mitogenic signals, such as members of the SHH, BMP and FGF family, in addition to several chemokines and their receptors, suggesting an important autocrine/paracrine role of these cells in the pathogenesis of ACP and a reciprocal communication with their environment. Finally, we highlight the clinical relevance of these findings by showing that these pathways are also up-regulated in the ß-cat(nc) cell clusters identified in human ACP. As well as providing further support to the concept that pituitary stem cells may play an important role in the oncogenesis of human ACP, our data reveal novel disease biomarkers and potential pharmacological targets for the treatment of these devastating childhood tumours.


Subject(s)
Craniopharyngioma/genetics , Neoplastic Stem Cells , Pituitary Neoplasms/genetics , beta Catenin/genetics , Animals , Cells, Cultured , Craniopharyngioma/metabolism , Craniopharyngioma/pathology , Disease Models, Animal , Humans , Mice , Mutation , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Signal Transduction , Telomere/genetics , Telomere/metabolism , beta Catenin/metabolism
9.
PLoS One ; 6(11): e27520, 2011.
Article in English | MEDLINE | ID: mdl-22114676

ABSTRACT

Human embryonic stem cells (hESCs) are primed for rapid apoptosis following mild forms of genotoxic stress. A natural form of such cellular stress occurs in response to recombinant adeno-associated virus (rAAV) single-strand DNA genomes, which exploit the host DNA damage response for replication and genome persistence. Herein, we discovered a unique DNA damage response induced by rAAV transduction specific to pluripotent hESCs. Within hours following rAAV transduction, host DNA damage signaling was elicited as measured by increased gamma-H2AX, ser15-p53 phosphorylation, and subsequent p53-dependent transcriptional activation. Nucleotide incorporation assays demonstrated that rAAV transduced cells accumulated in early S-phase followed by the induction of apoptosis. This lethal signaling sequalae required p53 in a manner independent of transcriptional induction of Puma, Bax and Bcl-2 and was not evident in cells differentiated towards a neural lineage. Consistent with a lethal DNA damage response induced upon rAAV transduction of hESCs, empty AAV protein capsids demonstrated no toxicity. In contrast, DNA microinjections demonstrated that the minimal AAV origin of replication and, in particular, a 40 nucleotide G-rich tetrad repeat sequence, was sufficient for hESC apoptosis. Our data support a model in which rAAV transduction of hESCs induces a p53-dependent lethal response that is elicited by a telomeric sequence within the AAV origin of replication.


Subject(s)
Apoptosis , DNA Replication , DNA, Single-Stranded/genetics , Dependovirus/genetics , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Tumor Suppressor Protein p53/metabolism , Blotting, Western , Cells, Cultured , DNA Damage/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Histones/genetics , Histones/metabolism , Humans , Phosphorylation , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction , Tumor Suppressor Protein p53/genetics
10.
Nat Genet ; 43(6): 607-11, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21532573

ABSTRACT

The HMG-box transcription factor Sox2 plays a role throughout neurogenesis and also acts at other stages of development, as illustrated by the multiple organs affected in the anophthalmia syndrome caused by SOX2 mutations. Here we combined proteomic and genomic approaches to characterize gene regulation by Sox2 in neural stem cells. Chd7, a chromatin remodeling ATPase associated with CHARGE syndrome, was identified as a Sox2 transcriptional cofactor. Sox2 and Chd7 physically interact, have overlapping genome-wide binding sites and regulate a set of common target genes including Jag1, Gli3 and Mycn, genes mutated in Alagille, Pallister-Hall and Feingold syndromes, which show malformations also associated with SOX2 anophthalmia syndrome or CHARGE syndrome. Regulation of disease-associated genes by a Sox2-Chd7 complex provides a plausible explanation for several malformations associated with SOX2 anophthalmia syndrome or CHARGE syndrome. Indeed, we found that Chd7-haploinsufficient embryos showed severely reduced expression of Jag1 in the developing inner ear.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Neural Stem Cells/metabolism , SOXB1 Transcription Factors/metabolism , Animals , Anophthalmos/genetics , CHARGE Syndrome/genetics , Calcium-Binding Proteins/metabolism , Ear, Inner/metabolism , Hedgehog Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Membrane Proteins/metabolism , Mice , Mutation , Receptors, Notch/metabolism , Serrate-Jagged Proteins
11.
Dev Biol ; 352(1): 40-7, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21256837

ABSTRACT

The HMG-Box transcription factor SOX2 is expressed in neural progenitor populations throughout the developing and adult central nervous system and is necessary to maintain their progenitor identity. However, it is unclear whether SOX2 levels are uniformly expressed across all neural progenitor populations. In the developing dorsal telencephalon, two distinct populations of neural progenitors, radial glia and intermediate progenitor cells, are responsible for generating a majority of excitatory neurons found in the adult neocortex. Here we demonstrate, using both cellular and molecular analyses, that SOX2 is differentially expressed between radial glial and intermediate progenitor populations. Moreover, utilizing a SOX2(EGFP) mouse line, we show that this differential expression can be used to prospectively isolate distinct, viable populations of radial glia and intermediate cells for in vitro analysis. Given the limited repertoire of cell-surface markers currently available for neural progenitor cells, this provides an invaluable tool for prospectively identifying and isolating distinct classes of neural progenitor cells from the central nervous system.


Subject(s)
Neural Stem Cells/cytology , Neural Stem Cells/metabolism , SOXB1 Transcription Factors/metabolism , Telencephalon/cytology , Telencephalon/embryology , Animals , Biomarkers/metabolism , Cell Aggregation , Cell Proliferation , Cell Separation , Cell Size , Female , Flow Cytometry , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism , SOXB1 Transcription Factors/genetics , Telencephalon/metabolism
12.
Development ; 138(3): 443-54, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21205789

ABSTRACT

In humans, haploinsufficiency of either SOX2 or PAX6 is associated with microphthalmia, anophthalmia or aniridia. In this study, through the genetic spatiotemporal specific ablation of SOX2 on both wild-type and Pax6-haploinsufficent backgrounds in the mouse, we have uncovered a transcriptionally distinct and developmentally transient stage of eye development. We show that genetic ablation of SOX2 in the optic cup results in complete loss of neural competence and eventual cell fate conversion to non-neurogenic ciliary epithelium. This cell fate conversion is associated with a striking increase in PAX6, and genetically ablating SOX2 on a Pax6-haploinsufficient background partially rescues the Sox2-mutant phenotype. Collectively, these results demonstrate that precise regulation of the ratio of SOX2 to PAX6 is necessary to ensure accurate progenitor cell specification, and place SOX2 as a decisive factor of neural competence in the retina.


Subject(s)
Ciliary Body/cytology , Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Eye Proteins/genetics , Homeodomain Proteins/genetics , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Mutant Strains , Neurons/cytology , Neurons/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Repressor Proteins/genetics , Retina/cytology , Retina/metabolism , SOXB1 Transcription Factors/genetics
13.
Int J Biochem Cell Biol ; 42(3): 421-4, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19733254

ABSTRACT

Throughout vertebrate evolution, Sox2 marks the developing nervous system from its earliest developmental stages and, therein, the most undifferentiated precursor cells, including stem cells. Recent gene targeting studies investigated the function of Sox2 in two neuronal systems: the developing eye and brain. These studies uncovered a requirement for Sox2 in the maintenance of neural stem cells, as well as a downstream role in the differentiation of specific neuron sub-types. In both systems, Sox2 action is markedly dose-dependent, and downstream-target gene studies are beginning to reveal the mechanisms of Sox2 function.


Subject(s)
Neurons/cytology , SOXB1 Transcription Factors/metabolism , Stem Cells/metabolism , Animals , Cell Differentiation , Humans , Neoplastic Stem Cells/metabolism , Stem Cells/cytology
14.
Cell Stem Cell ; 5(1): 64-75, 2009 Jul 02.
Article in English | MEDLINE | ID: mdl-19570515

ABSTRACT

The cellular mechanisms that regulate the maintenance of adult tissue stem cells are still largely unknown. We show here that the p53 family member, TAp63, is essential for maintenance of epidermal and dermal precursors and that, in its absence, these precursors senesce and skin ages prematurely. Specifically, we have developed a TAp63 conditional knockout mouse and used it to ablate TAp63 in the germline (TAp63(-/-)) or in K14-expressing cells in the basal layer of the epidermis (TAp63(fl/fl);K14cre+). TAp63(-/-) mice age prematurely and develop blisters, skin ulcerations, senescence of hair follicle-associated dermal and epidermal cells, and decreased hair morphogenesis. These phenotypes are likely due to loss of TAp63 in dermal and epidermal precursors since both cell types show defective proliferation, early senescence, and genomic instability. These data indicate that TAp63 serves to maintain adult skin stem cells by regulating cellular senescence and genomic stability, thereby preventing premature tissue aging.


Subject(s)
Adult Stem Cells/physiology , Aging, Premature/etiology , Dermis/cytology , Epidermal Cells , Phosphoproteins/genetics , Phosphoproteins/physiology , Trans-Activators/genetics , Trans-Activators/physiology , Adult Stem Cells/cytology , Aging, Premature/pathology , Animals , Cellular Senescence , DNA Damage , Genes, p53 , Genomic Instability , Hair Follicle/cytology , Keratinocytes/cytology , Mice , Mice, Knockout , Skin Aging/genetics , Skin Aging/pathology , Wound Healing/genetics
15.
Proc Natl Acad Sci U S A ; 105(47): 18396-401, 2008 Nov 25.
Article in English | MEDLINE | ID: mdl-19011097

ABSTRACT

Sox2 is a high-mobility transcription factor that is one of the earliest markers of developing inner ear prosensory domains. In humans, mutations in SOX2 cause sensorineural hearing loss and a loss of function study in mice showed that Sox2 is required for prosensory formation in the cochlea. However, the specific roles of Sox2 have not been determined. Here we illustrate a dynamic role of Sox2 as an early permissive factor in prosensory domain formation followed by a mutually antagonistic relationship with Atoh1, a bHLH protein necessary for hair cell development. We demonstrate that decreased levels of Sox2 result in precocious hair cell differentiation and an over production of inner hair cells and that these effects are likely mediated through an antagonistic interaction between Sox2 and the bHLH molecule Atoh1. Using gain- and loss-of-function experiments we provide evidence for the molecular pathway responsible for the formation of the cochlear prosensory domain. Sox2 expression is promoted by Notch signaling and Prox1, a homeobox transcription factor, is a downstream target of Sox2. These results demonstrate crucial and diverse roles for Sox2 in the development, specification, and maintenance of sensory cells within the cochlea.


Subject(s)
Cell Differentiation , Cochlea/cytology , Hair Cells, Auditory, Inner/cytology , SOXB1 Transcription Factors/metabolism , Signal Transduction , Animals , Cochlea/growth & development , Mice , Receptors, Notch/metabolism
16.
CSH Protoc ; 2008: pdb.prot5077, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-21356718

ABSTRACT

INTRODUCTIONThe ability to prospectively identify and characterize neural progenitor cells in vivo has been difficult due to a lack of cell-surface markers specific for these cell types. A widely used in vitro culture method, known as the Neurosphere Assay (NSA), has provided a means to retrospectively identify neural progenitor cells as well as to determine both their self-renewal capacity and their ability to generate the three primary cell types of the nervous system: neurons, astrocytes, and oligodendrocytes. Today, combined with the establishment of multiple transgenic mouse strains expressing fluorescent markers and advances in cell isolation techniques such as fluorescence-activated cell sorting (FACS), the NSA provides a powerful system to prospectively elucidate neural progenitor characteristics and functions. Here we describe methods for the isolation, culture, and differentiation of neural progenitors from the developing mouse and adult cortex.

17.
Development ; 134(13): 2521-31, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17522155

ABSTRACT

Sox2 is expressed in developing foregut endoderm, with highest levels in the future esophagus and anterior stomach. By contrast, Nkx2.1 (Titf1) is expressed ventrally, in the future trachea. In humans, heterozygosity for SOX2 is associated with anopthalmia-esophageal-genital syndrome (OMIM 600992), a condition including esophageal atresia (EA) and tracheoesophageal fistula (TEF), in which the trachea and esophagus fail to separate. Mouse embryos heterozygous for the null allele, Sox2(EGFP), appear normal. However, further reductions in Sox2, using Sox2(LP) and Sox2(COND) hypomorphic alleles, result in multiple abnormalities. Approximately 60% of Sox2(EGFP/COND) embryos have EA with distal TEF in which Sox2 is undetectable by immunohistochemistry or western blot. The mutant esophagus morphologically resembles the trachea, with ectopic expression of Nkx2.1, a columnar, ciliated epithelium, and very few p63(+) basal cells. By contrast, the abnormal foregut of Nkx2.1-null embryos expresses elevated Sox2 and p63, suggesting reciprocal regulation of Sox2 and Nkx2.1 during early dorsal/ventral foregut patterning. Organ culture experiments further suggest that FGF signaling from the ventral mesenchyme regulates Sox2 expression in the endoderm. In the 40% Sox2(EGFP/COND) embryos in which Sox2 levels are approximately 18% of wild type there is no TEF. However, the esophagus is still abnormal, with luminal mucus-producing cells, fewer p63(+) cells, and ectopic expression of genes normally expressed in glandular stomach and intestine. In all hypomorphic embryos the forestomach has an abnormal phenotype, with reduced keratinization, ectopic mucus cells and columnar epithelium. These findings suggest that Sox2 plays a second role in establishing the boundary between the keratinized, squamous esophagus/forestomach and glandular hindstomach.


Subject(s)
Body Patterning , Cell Differentiation , DNA-Binding Proteins/metabolism , Digestive System/embryology , Digestive System/metabolism , Endoderm/cytology , Endoderm/metabolism , Trans-Activators/metabolism , Animals , DNA-Binding Proteins/genetics , Esophageal Atresia/embryology , Esophageal Atresia/genetics , Esophageal Atresia/metabolism , Esophageal Atresia/pathology , Fibroblast Growth Factor 10/genetics , Fibroblast Growth Factor 10/metabolism , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Mutation/genetics , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phenotype , SOXB1 Transcription Factors , Thyroid Nuclear Factor 1 , Time Factors , Tracheoesophageal Fistula/embryology , Tracheoesophageal Fistula/genetics , Tracheoesophageal Fistula/metabolism , Tracheoesophageal Fistula/pathology , Trans-Activators/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Genes Dev ; 20(19): 2654-9, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-17015430

ABSTRACT

Sox2 is expressed in basal epithelial cells of the tongue, with high levels in taste bud placodes, fungiform papillae, and mature taste cells, and low levels in filiform papillae. High Sox2 expression appears to lie downstream from canonical Wnt signaling. In hypomorphic Sox2(EGFP/LP) embryos, placodes form but no mature taste buds develop. In contrast, transgenic overexpression of Sox2 in the basal cells inhibits differentiation of filiform keratinocytes. Together, our loss-of-function and gain-of-function studies suggest that Sox2 functions in a dose-dependent manner to regulate the differentiation of endodermal progenitor cells of the tongue into taste bud sensory cells versus keratinocytes.


Subject(s)
DNA-Binding Proteins/genetics , Taste Buds/embryology , Trans-Activators/genetics , Animals , DNA-Binding Proteins/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental/genetics , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Mice, Mutant Strains , Mice, Transgenic , Models, Biological , Phenotype , SOXB1 Transcription Factors , Taste Buds/cytology , Taste Buds/metabolism , Tongue/cytology , Tongue/embryology , Tongue/metabolism , Trans-Activators/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism
19.
Genes Dev ; 20(9): 1187-202, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16651659

ABSTRACT

Approximately 10% of humans with anophthalmia (absent eye) or severe microphthalmia (small eye) show haploid insufficiency due to mutations in SOX2, a SOXB1-HMG box transcription factor. However, at present, the molecular or cellular mechanisms responsible for these conditions are poorly understood. Here, we directly assessed the requirement for SOX2 during eye development by generating a gene-dosage allelic series of Sox2 mutations in the mouse. The Sox2 mutant mice display a range of eye phenotypes consistent with human syndromes and the severity of these phenotypes directly relates to the levels of SOX2 expression found in progenitor cells of the neural retina. Retinal progenitor cells with conditionally ablated Sox2 lose competence to both proliferate and terminally differentiate. In contrast, in Sox2 hypomorphic/null mice, a reduction of SOX2 expression to <40% of normal causes variable microphthalmia as a result of aberrant neural progenitor differentiation. Furthermore, we provide genetic and molecular evidence that SOX2 activity, in a concentration-dependent manner, plays a key role in the regulation of the NOTCH1 signaling pathway in retinal progenitor cells. Collectively, these results show that precise regulation of SOX2 dosage is critical for temporal and spatial regulation of retinal progenitor cell differentiation and provide a cellular and molecular model for understanding how hypomorphic levels of SOX2 cause retinal defects in humans.


Subject(s)
DNA-Binding Proteins/physiology , Gene Dosage , Retina/abnormalities , Stem Cells/physiology , Trans-Activators/physiology , Alleles , Animals , Anophthalmos/genetics , Cell Differentiation , Cell Proliferation , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Mice , Mice, Knockout , Microphthalmos/genetics , Mutation , Neurons/metabolism , Neurons/physiology , Receptor, Notch1/metabolism , Retina/embryology , Retina/metabolism , SOXB1 Transcription Factors , Signal Transduction , Stem Cells/metabolism , Trans-Activators/biosynthesis , Trans-Activators/genetics
20.
Genes Dev ; 17(1): 126-40, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12514105

ABSTRACT

Each cell lineage specified in the preimplantation mammalian embryo depends on intrinsic factors for its development, but there is also mutual interdependence between them. OCT4 is required for the ICM/epiblast lineage, and at transient high levels for extraembryonic endoderm, but also indirectly through its role in regulating Fgf4 expression, for the establishment and proliferation of extraembryonic ectoderm from polar trophectoderm. The transcription factor SOX2 has also been implicated in the regulation of Fgf4 expression. We have used gene targeting to inactivate Sox2, examining the phenotypic consequences in mutant embryos and in chimeras in which the epiblast is rescued with wild-type ES cells. We find a cell-autonomous requirement for the gene in both epiblast and extraembryonic ectoderm, the multipotent precursors of all embryonic and trophoblast cell types, respectively. However, an earlier role within the ICM may be masked by the persistence of maternal protein, whereas the lack of SOX2 only becomes critical in the chorion after 7.5 days postcoitum. Our data suggest that maternal components could be involved in establishing early cell fate decisions and that a combinatorial code, requiring SOX2 and OCT4, specifies the first three lineages present at implantation.


Subject(s)
Cell Lineage/genetics , DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , Multipotent Stem Cells/cytology , Nuclear Proteins/physiology , Transcription Factors , Animals , Blastocyst/metabolism , Chimera , Chorion/cytology , Crosses, Genetic , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Ectoderm/cytology , Embryo Transfer , Embryonic and Fetal Development/genetics , Female , Fibroblast Growth Factors/physiology , Gene Targeting , Genes, Lethal , HMGB Proteins , Heterozygote , Male , Maternal-Fetal Exchange , Mice , Microscopy, Confocal , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Octamer Transcription Factor-3 , Phenotype , Pregnancy , SOXB1 Transcription Factors , Trophoblasts/cytology , Trophoblasts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...