Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Int J Mol Sci ; 22(16)2021 Aug 19.
Article in English | MEDLINE | ID: mdl-34445655

ABSTRACT

The choroid plexus is an important blood barrier that secretes cerebrospinal fluid, which essential for embryonic brain development and adult brain homeostasis. The OTX2 homeoprotein is a transcription factor that is critical for choroid plexus development and remains highly expressed in adult choroid plexus. Through RNA sequencing analyses of constitutive and conditional knockdown adult mouse models, we reveal putative functional roles for OTX2 in adult choroid plexus function, including cell signaling and adhesion, and show that OTX2 regulates the expression of factors that are secreted into the cerebrospinal fluid, notably transthyretin. We also show that Otx2 expression impacts choroid plexus immune and stress responses, and affects splicing, leading to changes in the mRNA isoforms of proteins that are implicated in the oxidative stress response and DNA repair. Through mass spectrometry analysis of OTX2 protein partners in the choroid plexus, and in known non-cell-autonomous target regions, such as the visual cortex and subventricular zone, we identify putative targets that are involved in cell adhesion, chromatin structure, and RNA processing. Thus, OTX2 retains important roles for regulating choroid plexus function and brain homeostasis throughout life.


Subject(s)
Brain/physiology , Choroid Plexus/metabolism , Gene Expression Regulation , Homeostasis , Lateral Ventricles/metabolism , Otx Transcription Factors/physiology , Visual Cortex/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Transcriptome
2.
Elife ; 92020 04 29.
Article in English | MEDLINE | ID: mdl-32347797

ABSTRACT

During vertebrate retinal development, subsets of progenitor cells generate progeny in a non-stochastic manner, suggesting that these decisions are tightly regulated. However, the gene-regulatory network components that are functionally important in these progenitor cells are largely unknown. Here we identify a functional role for the OTX2 transcription factor in this process. CRISPR/Cas9 gene editing was used to produce somatic mutations of OTX2 in the chick retina and identified similar phenotypes to those observed in human patients. Single cell RNA sequencing was used to determine the functional consequences OTX2 gene editing on the population of cells derived from OTX2-expressing retinal progenitor cells. This confirmed that OTX2 is required for the generation of photoreceptors, but also for repression of specific retinal fates and alternative gene regulatory networks. These include specific subtypes of retinal ganglion and horizontal cells, suggesting that in this context, OTX2 functions to repress sister cell fate choices.


Subject(s)
Otx Transcription Factors/physiology , Photoreceptor Cells, Vertebrate/physiology , Retina/embryology , Animals , CRISPR-Cas Systems/genetics , Chickens , Female , Gene Editing , Gene Regulatory Networks , Male , Mutation , Otx Transcription Factors/genetics , PAX6 Transcription Factor/analysis , Sequence Analysis, RNA , Single-Cell Analysis
3.
eNeuro ; 6(2)2019.
Article in English | MEDLINE | ID: mdl-31064838

ABSTRACT

Proliferation and migration during adult neurogenesis are regulated by a microenvironment of signaling molecules originating from local vasculature, from CSF produced by the choroid plexus, and from local supporting cells including astrocytes. Here, we focus on the function of OTX2 homeoprotein transcription factor in the mouse adult ventricular-subventricular zone (V-SVZ), which generates olfactory bulb neurons. We find that OTX2 secreted by choroid plexus is transferred to the supporting cells of the V-SVZ and rostral migratory stream. Deletion of Otx2 in choroid plexus affects neuroblast migration and reduces the number of olfactory bulb newborn neurons. Adult neurogenesis was also decreased by expressing secreted single-chain antibodies to sequester OTX2 in the CSF, demonstrating the importance of non-cell-autonomous OTX2. We show that OTX2 activity modifies extracellular matrix components and signaling molecules produced by supporting astrocytes. Thus, we reveal a multilevel and non-cell-autonomous role of a homeoprotein and reinforce the choroid plexus and astrocytes as key niche compartments affecting adult neurogenesis.


Subject(s)
Astrocytes/metabolism , Cerebrospinal Fluid/metabolism , Choroid Plexus/metabolism , Extracellular Matrix/metabolism , Lateral Ventricles , Neurogenesis/physiology , Olfactory Bulb , Otx Transcription Factors/physiology , Signal Transduction/physiology , Animals , Cell Movement/physiology , Female , Lateral Ventricles/cytology , Lateral Ventricles/metabolism , Male , Mice, 129 Strain , Mice, Inbred C57BL , Olfactory Bulb/cytology , Olfactory Bulb/metabolism , Otx Transcription Factors/deficiency , Otx Transcription Factors/metabolism
4.
J Neurosci ; 39(6): 1005-1019, 2019 02 06.
Article in English | MEDLINE | ID: mdl-30593496

ABSTRACT

The habenulo-interpeduncular system (HIPS) is now recognized as a critical circuit modulating aversion, reward, and social behavior. There is evidence that dysfunction of this circuit leads to psychiatric disorders. Because psychiatric diseases may originate in developmental abnormalities, it is crucial to investigate the developmental mechanisms controlling the formation of the HIPS. Thus far, this issue has been the focus of limited studies. Here, we explored the developmental processes underlying the formation of the medial habenula (MHb) and its unique output, the interpeduncular nucleus (IPN), in mice independently of their gender. We report that the Otx2 homeobox gene is essential for the proper development of both structures. We show that MHb and IPN neurons require Otx2 at different developmental stages and, in both cases, Otx2 deletion leads to disruption of HIPS subcircuits. Finally, we show that Otx2+ neurons tend to be preferentially interconnected. This study reveals that synaptically connected components of the HIPS, despite radically different developmental strategies, share high sensitivity to Otx2 expression.SIGNIFICANCE STATEMENT Brain reward circuits are highly complex and still poorly understood. In particular, it is important to understand how these circuits form as many psychiatric diseases may arise from their abnormal development. This work shows that Otx2, a critical evolutionary conserved gene implicated in brain development and a predisposing factor for psychiatric diseases, is required for the formation of the habenulo-interpeduncular system (HIPS), an important component of the reward circuit. Otx2 deletion affects multiple processes such as proliferation and migration of HIPS neurons. Furthermore, neurons expressing Otx2 are preferentially interconnected. Therefore, Otx2 expression may represent a code that specifies the connectivity of functional subunits of the HIPS. Importantly, the Otx2 conditional knock-out animals used in this study might represent a new genetic model of psychiatric diseases.


Subject(s)
Habenula/growth & development , Interpeduncular Nucleus/growth & development , Neural Pathways/growth & development , Otx Transcription Factors/physiology , Animals , Cell Movement/physiology , Female , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Habenula/physiology , Interpeduncular Nucleus/physiology , Male , Mice , Mice, 129 Strain , Mice, Knockout , Neural Pathways/physiology , Neurons/physiology , Synapses/physiology
5.
J Biol Chem ; 293(6): 2137-2148, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29273633

ABSTRACT

The progenitor cells in the cerebral cortex coordinate proliferation and mitotic exit to generate the correct number of neurons and glial cells during development. However, mechanisms for regulating the mitotic cycle of cortical progenitors are not fully understood. Otx1 is one of the homeobox-containing transcription factors frequently implicated in the development of the central nervous system. Mice bearing a targeted deletion of Otx1 exhibit brain hypoplasia and a decrease in the number of cortical neurons. We hypothesized that Otx1 might be crucial to the proliferation and differentiation of cortical progenitors. Otx1 knockdown by in utero electroporation in the mouse brain reduced the proportion of the G1 phase while increasing the S and M phases of progenitor cells. The knockdown diminished Tbr1+ neurons but increased GFAP+ astrocytes in the early postnatal cortex as revealed by lineage tracing study. Tbr2+ basal progenitors lacking Otx1 were held at the transit-amplifying stage. In contrast, overexpression of wildtype Otx1 but not an astrocytoma-related mutant Y320C inhibited proliferation of the progenitor cells in embryonic cortex. This study demonstrates that Otx1 is one of the key elements regulating cortical neurogenesis, and a loss-of-function in Otx1 may contribute to the overproduction of astrocytes in vivo.


Subject(s)
Cell Cycle , Cerebral Cortex/cytology , Neural Stem Cells/cytology , Otx Transcription Factors/physiology , Animals , Astrocytes/cytology , Cell Count , Cell Differentiation , Cell Proliferation , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Mice , Neurogenesis , Neurons/cytology
6.
Neural Plast ; 2016: 7931693, 2016.
Article in English | MEDLINE | ID: mdl-26881132

ABSTRACT

The ability of the environment to shape cortical function is at its highest during critical periods of postnatal development. In the visual cortex, critical period onset is triggered by the maturation of parvalbumin inhibitory interneurons, which gradually become surrounded by a specialized glycosaminoglycan-rich extracellular matrix: the perineuronal nets. Among the identified factors regulating cortical plasticity in the visual cortex, extracortical homeoprotein Otx2 is transferred specifically into parvalbumin interneurons and this transfer regulates both the onset and the closure of the critical period of plasticity for binocular vision. Here, we review the interaction between the complex sugars of the perineuronal nets and homeoprotein Otx2 and how this interaction regulates cortical plasticity during critical period and in adulthood.


Subject(s)
Critical Period, Psychological , Extracellular Matrix/physiology , Neuronal Plasticity , Otx Transcription Factors/physiology , Visual Cortex/physiology , Animals , Extracellular Matrix Proteins/physiology , Humans , Interneurons/metabolism , Interneurons/physiology , Parvalbumins/metabolism , Visual Cortex/growth & development
7.
Aging (Albany NY) ; 7(11): 928-36, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26554466

ABSTRACT

OTX proteins, homologs of the Drosophila orthodenticle (Otd), are important for the morphogenesis of the neuroectoderm, and for the central nervous system formation. OTX1 and OTX2 are important for the cochlea and macula development, indeed when OTX1 is knocked down, these organs undergo developmental failure. Moreover OTX2 transfection revert this effect in OTX1(-/-) mice. The TA isoform of TP63, involved in Notch regulation pathway, has a critical function in the cochlear neuroepithelium differentiation. TAp63 positively regulates Hes5 and Atoh1 transcription. This pathway has been also demonstrated in p63(-/-) mice, and in patients p63 mutated, affected by Ectodermal Dysplasia (ED, OMIM 129810). These patients are affected by mild sensorineural deafness, most likely related to the mutation in p63 gene impairing the Notch pathway. We demonstrated the role of OTX2 on TAp63 regulation necessary for the correct formation of macular neuroepithelium and we confirmed the impairment of vestibular function caused by p63 mutations. Although the abnormalities found in our patient were still at a subclinical extent, aging could exacerbate this impairment and cause a decrease in quality of life.


Subject(s)
Cochlea/embryology , Gene Expression Regulation, Developmental , Macula Lutea/embryology , Otx Transcription Factors/physiology , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Animals , Cell Differentiation , Ectodermal Dysplasia/metabolism , Humans , Mice , Otx Transcription Factors/chemistry , Vestibule, Labyrinth/physiology
8.
Int J Biol Sci ; 11(6): 688-700, 2015.
Article in English | MEDLINE | ID: mdl-25999792

ABSTRACT

The homeobox transcription factor orthodenticle homolog 2 (otx2) is supposed as an organizer that orchestrates a transcription factor network during photoreceptor development. However, its regulation in the process remains unclear. In this study, we have identified a zebrafish limb bud and heart-like gene (lbh-like), which is expressed initially at 30 hours post fertilization (hpf) in the developing brain and eyes. Lbh-like knockdown by morpholinos specifically inhibits expression of multiple photoreceptor-specific genes, such as opsins, gnat1, gnat2 and irbp. Interestingly, otx2 expression in the morphants is not significantly reduced until 32 hpf when lbh-like begins to express, but its expression level in 72 hpf morphants is higher than that in wild type embryos. Co-injection of otx2 and its downstream target neuroD mRNAs can rescue the faults in eyes of Lbh-like morphants. Combined with the results of promoter-reporter assay, we suggest that lbh-like is a new regulator of photoreceptor differentiation directly through affecting otx2 expression in zebrafish. Furthermore, knockdown of lbh-like increases the activity of Notch pathway and perturbs the balance among proliferation, differentiation and survival of photoreceptor precursors.


Subject(s)
Cell Differentiation/genetics , Otx Transcription Factors/physiology , Photoreceptor Cells, Vertebrate/cytology , Trans-Activators/physiology , Zebrafish Proteins/physiology , Zebrafish/growth & development , Amino Acid Sequence , Animals , Apoptosis , Cell Proliferation , Eye/anatomy & histology , Eye/embryology , Gene Knockdown Techniques , Genetic Markers , Molecular Sequence Data , Otx Transcription Factors/genetics , Otx Transcription Factors/metabolism , Sequence Alignment , Trans-Activators/genetics , Trans-Activators/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
9.
Dev Biol ; 396(1): 107-20, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25281935

ABSTRACT

In this study, we investigated the gene regulatory network that governs formation of the Zona limitans intrathalamica (ZLI), a signaling center that secretes Sonic Hedgehog (Shh) to control the growth and regionalization of the caudal forebrain. Using loss- and gain-of-function, explants and grafting experiments in amphibians, we demonstrate that barhl2 acts downstream of otx2 and together with the iroquois (irx)-3 gene in establishment of the ZLI compartment initiated by Shh influence. We find that the presumptive (pre)-ZLI domain expresses barhl2, otx2 and irx3, whereas the thalamus territory caudally bordering the pre-ZLI expresses barhl2, otx2 and irx1/2 and early on irx3. We demonstrate that Barhl2 activity is required for determination of the ZLI and thalamus fates and that within the p2 alar plate the ratio of Irx3 to Irx1/2 contributes to ZLI specification and size determination. We show that when continuously exposed to Shh, neuroepithelial cells coexpressing barhl2, otx2 and irx3 acquire two characteristics of the ZLI compartment-the competence to express shh and the ability to segregate from anterior neural plate cells. In contrast, neuroepithelial cells expressing barhl2, otx2 and irx1/2, are not competent to express shh. Noteworthy in explants, under Shh influence, ZLI-like cells segregate from thalamic-like cells. Our study establishes that Barhl2 activity plays a key role in p2 alar plate patterning, specifically ZLI formation, and provides new insights on establishment of the signaling center of the caudal forebrain.


Subject(s)
Gene Expression Regulation, Developmental , Hedgehog Proteins/physiology , Homeodomain Proteins/physiology , Nerve Tissue Proteins/physiology , Otx Transcription Factors/physiology , Prosencephalon/embryology , Thalamus/embryology , Transcription Factors/physiology , Xenopus Proteins/physiology , Animals , Blastomeres/ultrastructure , Body Patterning , Gene Expression Profiling , Genes, Homeobox , HEK293 Cells , Hedgehog Proteins/metabolism , Humans , Neural Crest/cytology , Neuroepithelial Cells/cytology , Oligonucleotides, Antisense/chemistry , Rats , Signal Transduction , Time Factors , Xenopus laevis
10.
Development ; 141(20): 3859-67, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25231759

ABSTRACT

The Otx2 gene encodes a paired-type homeobox transcription factor that is essential for the induction and the patterning of the anterior structures in the mouse embryo. Otx2 knockout embryos fail to form a head. Whereas previous studies have shown that Otx2 is required in the anterior visceral endoderm and the anterior neuroectoderm for head formation, its role in the anterior mesendoderm (AME) has not been assessed specifically. Here, we show that tissue-specific ablation of Otx2 in the AME phenocopies the truncation of the embryonic head of the Otx2 null mutant. Expression of Dkk1 and Lhx1, two genes that are also essential for head formation, is disrupted in the AME of the conditional Otx2-deficient embryos. Consistent with the fact that Dkk1 is a direct target of OTX2, we showed that OTX2 can interact with the H1 regulatory region of Dkk1 to activate its expression. Cross-species comparative analysis, RT-qPCR, ChIP-qPCR and luciferase assays have revealed two conserved regions in the Lhx1 locus to which OTX2 can bind to activate Lhx1 expression. Abnormal development of the embryonic head in Otx2;Lhx1 and Otx2;Dkk1 compound mutant embryos highlights the functional intersection of Otx2, Dkk1 and Lhx1 in the AME for head formation.


Subject(s)
Gene Expression Regulation, Developmental , Head/embryology , Intercellular Signaling Peptides and Proteins/metabolism , LIM-Homeodomain Proteins/metabolism , Mesoderm/physiology , Otx Transcription Factors/physiology , Transcription Factors/metabolism , 3T3 Cells , Animals , Crosses, Genetic , Luciferases/metabolism , Mice , Mutation , Phenotype
11.
Semin Cell Dev Biol ; 35: 189-202, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25194659

ABSTRACT

Morphogenesis and axonal targeting are key processes during development that depend on complex interactions at molecular, cellular and tissue level. Mathematical modeling is essential to bridge this multi-scale gap in order to understand how the emergence of large structures is controlled at molecular level by interactions between various signaling pathways. We summarize mathematical modeling and computational methods for time evolution and precision of morphogenetic gradient formation. We discuss tissue patterning and the formation of borders between regions labeled by different morphogens. Finally, we review models and algorithms that reveal the interplay between morphogenetic gradients and patterned activity for axonal pathfinding and the generation of the retinotopic map in the visual system.


Subject(s)
Algorithms , Computational Biology/methods , Models, Neurological , Morphogenesis/physiology , Animals , Axons/metabolism , Axons/physiology , Brain/growth & development , Brain/metabolism , Brain/physiology , Homeodomain Proteins/metabolism , Homeodomain Proteins/physiology , Humans , Otx Transcription Factors/metabolism , Otx Transcription Factors/physiology
12.
Cell Rep ; 8(4): 1018-25, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25127144

ABSTRACT

Distinct midbrain dopamine (mDA) neuron subtypes are found in the substantia nigra pars compacta (SNc) and the ventral tegmental area (VTA), but it is mainly SNc neurons that degenerate in Parkinson's disease. Interest in how mDA neurons develop has been stimulated by the potential use of stem cells in therapy or disease modeling. However, very little is known about how specific dopaminergic subtypes are generated. Here, we show that the expression profiles of the transcription factors Sox6, Otx2, and Nolz1 define subpopulations of mDA neurons already at the neural progenitor cell stage. After cell-cycle exit, Sox6 selectively localizes to SNc neurons, while Otx2 and Nolz1 are expressed in a subset of VTA neurons. Importantly, Sox6 ablation leads to decreased expression of SNc markers and a corresponding increase in VTA markers, while Otx2 ablation has the opposite effect. Moreover, deletion of Sox6 affects striatal innervation and dopamine levels. We also find reduced Sox6 levels in Parkinson's disease patients. These findings identify Sox6 as a determinant of SNc neuron development and should facilitate the engineering of relevant mDA neurons for cell therapy and disease modeling.


Subject(s)
Dopaminergic Neurons/physiology , Otx Transcription Factors/physiology , SOXD Transcription Factors/physiology , Substantia Nigra/cytology , Ventral Tegmental Area/cytology , Animals , Body Patterning , Humans , Mice, Transgenic , Organ Specificity , Substantia Nigra/embryology , Substantia Nigra/metabolism , Ventral Tegmental Area/embryology , Ventral Tegmental Area/metabolism
13.
Dev Cell ; 30(5): 513-27, 2014 Sep 08.
Article in English | MEDLINE | ID: mdl-25155555

ABSTRACT

Gene regulatory networks (GRNs) regulate critical events during development. In complex tissues, such as the mammalian central nervous system (CNS), networks likely provide the complex regulatory interactions needed to direct the specification of the many CNS cell types. Here, we dissect a GRN that regulates a binary fate decision between two siblings in the murine retina, the rod photoreceptor and bipolar interneuron. The GRN centers on Blimp1, one of the transcription factors (TFs) that regulates the rod versus bipolar cell fate decision. We identified a cis-regulatory module (CRM), B108, that mimics Blimp1 expression. Deletion of genomic B108 by CRISPR/Cas9 in vivo using electroporation abolished the function of Blimp1. Otx2 and RORß were found to regulate Blimp1 expression via B108, and Blimp1 and Otx2 were shown to form a negative feedback loop that regulates the level of Otx2, which regulates the production of the correct ratio of rods and bipolar cells.


Subject(s)
Gene Expression Regulation, Developmental , Gene Regulatory Networks , Retina/embryology , Retinal Bipolar Cells/cytology , Retinal Rod Photoreceptor Cells/cytology , Animals , Cell Differentiation/physiology , Cell Lineage , Enhancer Elements, Genetic , Homeodomain Proteins/physiology , In Situ Hybridization, Fluorescence , Mice , Nuclear Receptor Subfamily 1, Group F, Member 2/physiology , Otx Transcription Factors/physiology , Positive Regulatory Domain I-Binding Factor 1 , Receptor, Notch1/physiology , Transcription Factors/physiology
14.
PLoS One ; 9(2): e89110, 2014.
Article in English | MEDLINE | ID: mdl-24558479

ABSTRACT

During mouse retinal development and into adulthood, the transcription factor Otx2 is expressed in pigment epithelium, photoreceptors and bipolar cells. In the mature retina, Otx2 ablation causes photoreceptor degeneration through a non-cell-autonomous mechanism involving Otx2 function in the supporting RPE. Surprisingly, photoreceptor survival does not require Otx2 expression in the neural retina, where the related Crx homeobox gene, a major regulator of photoreceptor development, is also expressed. To get a deeper view of mouse Otx2 activities in the neural retina, we performed chromatin-immunoprecipitation followed by massively parallel sequencing (ChIP-seq) on Otx2. Using two independent ChIP-seq assays, we identified consistent sets of Otx2-bound cis-regulatory elements. Comparison with our previous RPE-specific Otx2 ChIP-seq data shows that Otx2 occupies different functional domains of the genome in RPE cells and in neural retina cells and regulates mostly different sets of genes. To assess the potential redundancy of Otx2 and Crx, we compared our data with Crx ChIP-seq data. While Crx genome occupancy markedly differs from Otx2 genome occupancy in the RPE, it largely overlaps that of Otx2 in the neural retina. Thus, in accordance with its essential role in the RPE and its non-essential role in the neural retina, Otx2 regulates different gene sets in the RPE and the neural retina, and shares an important part of its repertoire with Crx in the neural retina. Overall, this study provides a better understanding of gene-regulatory networks controlling photoreceptor homeostasis and disease.


Subject(s)
Gene Expression Regulation/physiology , Gene Regulatory Networks/genetics , Otx Transcription Factors/genetics , Otx Transcription Factors/physiology , Retina/physiology , Animals , Base Sequence , Chromatin Immunoprecipitation , Gene Expression Regulation/genetics , Gene Ontology , High-Throughput Nucleotide Sequencing/methods , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Microarray Analysis , Molecular Sequence Data , Otx Transcription Factors/metabolism , Regulatory Elements, Transcriptional/genetics , Retina/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
15.
J Clin Invest ; 124(2): 631-43, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24382353

ABSTRACT

Leber congenital amaurosis (LCA) encompasses a set of early-onset blinding diseases that are characterized by vision loss, involuntary eye movement, and nonrecordable electroretinogram (ERG). At least 19 genes are associated with LCA, which is typically recessive; however, mutations in homeodomain transcription factor CRX lead to an autosomal dominant form of LCA. The mechanism of CRX-associated LCA is not understood. Here, we identified a spontaneous mouse mutant with a frameshift mutation in Crx (CrxRip). We determined that CrxRip is a dominant mutation that results in congenital blindness with nonrecordable response by ERG and arrested photoreceptor differentiation with no associated degeneration. Expression of LCA-associated dominant CRX frameshift mutations in mouse retina mimicked the CrxRip phenotype, which was rescued by overexpression of WT CRX. Whole-transcriptome profiling using deep RNA sequencing revealed progressive and complete loss of rod differentiation factor NRL in CrxRip retinas. Expression of NRL partially restored rod development in CrxRip/+ mice. We show that the binding of homeobox transcription factor OTX2 at the Nrl promoter was obliterated in CrxRip mice and ectopic expression of OTX2 rescued the rod differentiation defect. Together, our data indicate that OTX2 maintains Nrl expression in developing rods to consolidate rod fate. Our studies provide insights into CRX mutation-associated congenital blindness and should assist in therapeutic design.


Subject(s)
Blindness/genetics , Homeodomain Proteins/genetics , Otx Transcription Factors/genetics , Otx Transcription Factors/physiology , Retinal Rod Photoreceptor Cells/metabolism , Trans-Activators/genetics , Amino Acid Sequence , Animals , Blindness/congenital , Cell Differentiation , Codon , Crosses, Genetic , Disease Models, Animal , Electroretinography , Frameshift Mutation , Gene Deletion , Gene Expression Regulation , Genotype , High-Throughput Nucleotide Sequencing , Homozygote , Leber Congenital Amaurosis/genetics , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Phenotype , Retina/metabolism , Sequence Homology, Amino Acid , Transcriptome
16.
Development ; 140(5): 1055-66, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23364326

ABSTRACT

The choroid plexuses (ChPs) are the main regulators of cerebrospinal fluid (CSF) composition and thereby also control the composition of a principal source of signaling molecules that is in direct contact with neural stem cells in the developing brain. The regulators of ChP development mediating the acquisition of a fate that differs from the neighboring neuroepithelial cells are poorly understood. Here, we demonstrate in mice a crucial role for the transcription factor Otx2 in the development and maintenance of ChP cells. Deletion of Otx2 by the Otx2-CreERT2 driver line at E9 resulted in a lack of all ChPs, whereas deletion by the Gdf7-Cre driver line affected predominately the hindbrain ChP, which was reduced in size, primarily owing to an increase in apoptosis upon Otx2 deletion. Strikingly, Otx2 was still required for the maintenance of hindbrain ChP cells at later stages when Otx2 deletion was induced at E15, demonstrating a central role of Otx2 in ChP development and maintenance. Moreover, the predominant defects in the hindbrain ChP mediated by Gdf7-Cre deletion of Otx2 revealed its key role in regulating early CSF composition, which was altered in protein content, including the levels of Wnt4 and the Wnt modulator Tgm2. Accordingly, proliferation and Wnt signaling levels were increased in the distant cerebral cortex, suggesting a role of the hindbrain ChP in regulating CSF composition, including key signaling molecules. Thus, Otx2 acts as a master regulator of ChP development, thereby influencing one of the principal sources of signaling in the developing brain, the CSF.


Subject(s)
Choroid Plexus/embryology , Choroid Plexus/growth & development , Choroid Plexus/physiology , Otx Transcription Factors/physiology , Animals , Animals, Newborn , Cells, Cultured , Cerebrospinal Fluid/chemistry , Cerebrospinal Fluid/metabolism , Choroid Plexus/metabolism , Embryo, Mammalian , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Mice , Mice, Transgenic , Otx Transcription Factors/genetics , Otx Transcription Factors/metabolism , Pregnancy , Rhombencephalon/embryology , Rhombencephalon/growth & development , Rhombencephalon/metabolism , Rhombencephalon/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/physiology , Transcriptome/genetics
17.
Development ; 140(1): 43-55, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23154415

ABSTRACT

Mouse embryonic stem cells (ESCs) represent the naïve ground state of the preimplantation epiblast and epiblast stem cells (EpiSCs) represent the primed state of the postimplantation epiblast. Studies have revealed that the ESC state is maintained by a dynamic mechanism characterized by cell-to-cell spontaneous and reversible differences in sensitivity to self-renewal and susceptibility to differentiation. This metastable condition ensures indefinite self-renewal and, at the same time, predisposes ESCs for differentiation to EpiSCs. Despite considerable advances, the molecular mechanism controlling the ESC state and pluripotency transition from ESCs to EpiSCs have not been fully elucidated. Here we show that Otx2, a transcription factor essential for brain development, plays a crucial role in ESCs and EpiSCs. Otx2 is required to maintain the ESC metastable state by antagonizing ground state pluripotency and promoting commitment to differentiation. Furthermore, Otx2 is required for ESC transition into EpiSCs and, subsequently, to stabilize the EpiSC state by suppressing, in pluripotent cells, the mesendoderm-to-neural fate switch in cooperation with BMP4 and Fgf2. However, according to its central role in neural development and differentiation, Otx2 is crucially required for the specification of ESC-derived neural precursors fated to generate telencephalic and mesencephalic neurons. We propose that Otx2 is a novel intrinsic determinant controlling the functional integrity of ESCs and EpiSCs.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Otx Transcription Factors/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Genes, Reporter , Mice , Mice, Knockout , Mice, Transgenic , Otx Transcription Factors/biosynthesis , Otx Transcription Factors/deficiency , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology
18.
PLoS One ; 7(11): e48879, 2012.
Article in English | MEDLINE | ID: mdl-23145006

ABSTRACT

The retinal pigment epithelium (RPE) shares its developmental origin with the neural retina (NR). When RPE development is disrupted, cells in the presumptive RPE region abnormally differentiate into NR-like cells. Therefore, the prevention of NR differentiation in the presumptive RPE area seems to be essential for regionalizing the RPE during eye development. However, its molecular mechanisms are not fully understood. In this study, we conducted a functional inhibition of a transcription factor Otx2, which is required for RPE development, using early chick embryos. The functional inhibition of Otx2 in chick eyes, using a recombinant gene encoding a dominant negative form of Otx2, caused the outer layer of the optic cup (the region forming the RPE, when embryos normally develop) to abnormally form an ectopic NR. In that ectopic NR, the characteristics of the RPE did not appear and NR markers were ectopically expressed. Intriguingly, the repression of Otx2 function also caused the ectopic expression of Fgf8 and Sox2 in the outer layer of the optic cup (the presumptive RPE region of normally developing eyes). These two factors are known to be capable of inducing NR cell differentiation in the presumptive RPE region, and are not expressed in the normally developing RPE region. Here, we suggest that Otx2 prevents the presumptive RPE region from forming the NR by repressing the expression of both Fgf8 and Sox2 which induce the NR cell fate.


Subject(s)
Otx Transcription Factors/physiology , Retinal Pigment Epithelium/embryology , Animals , Apoptosis/genetics , Avian Proteins/metabolism , Cell Differentiation , Cell Proliferation , Chick Embryo , Eye/embryology , Eye/metabolism , Eye/pathology , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , Otx Transcription Factors/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , SOXB1 Transcription Factors/metabolism , Telencephalon/embryology , Transfection
19.
J Neurosci ; 32(27): 9429-37, 2012 Jul 04.
Article in English | MEDLINE | ID: mdl-22764251

ABSTRACT

Specific transfer of (orthodenticle homeobox 2) Otx2 homeoprotein into GABAergic interneurons expressing parvalbumin (PV) is necessary and sufficient to open, then close, a critical period (CP) of plasticity in the developing mouse visual cortex. The accumulation of endogenous Otx2 in PV cells suggests the presence of specific Otx2 binding sites. Here, we find that perineuronal nets (PNNs) on the surfaces of PV cells permit the specific, constitutive capture of Otx2. We identify a 15 aa domain containing an arginine-lysine doublet (RK peptide) within Otx2, bearing prototypic traits of a glycosaminoglycan (GAG) binding sequence that mediates Otx2 binding to PNNs, and specifically to chondroitin sulfate D and E, with high affinity. Accordingly, PNN hydrolysis by chondroitinase ABC reduces the amount of endogenous Otx2 in PV cells. Direct infusion of RK peptide similarly disrupts endogenous Otx2 localization to PV cells, reduces PV and PNN expression, and reopens plasticity in adult mice. The closure of one eye during this transient window reduces cortical acuity and is specific to the RK motif, as an Alanine-Alanine variant or a scrambled peptide fails to reactivate plasticity. Conversely, this transient reopening of plasticity in the adult restores binocular vision in amblyopic mice. Thus, one function of PNNs is to facilitate the persistent internalization of Otx2 by PV cells to maintain CP closure. The pharmacological use of the Otx2 GAG binding domain offers a novel, potent therapeutic tool with which to restore cortical plasticity in the mature brain.


Subject(s)
Extracellular Matrix/metabolism , Interneurons/metabolism , Neuronal Plasticity/physiology , Otx Transcription Factors/metabolism , Visual Cortex/metabolism , Age Factors , Animals , GABAergic Neurons/metabolism , Male , Mice , Mice, Inbred C57BL , Otx Transcription Factors/physiology , Protein Binding/physiology , Visual Cortex/cytology
20.
PLoS One ; 7(6): e39770, 2012.
Article in English | MEDLINE | ID: mdl-22761896

ABSTRACT

BACKGROUND: Precise coordination of the hypothalamic-pituitary-gonadal axis orchestrates the normal reproductive function. As a central regulator, the appropriate synthesis and secretion of gonadotropin-releasing hormone I (GnRH-I) from the hypothalamus is essential for the coordination. Recently, emerging evidence indicates that histone deacetylases (HDACs) play an important role in maintaining normal reproductive function. In this study, we identify the potential effects of HDACs on Gnrh1 gene transcription. METHODOLOGY/PRINCIPAL FINDINGS: Inhibition of HDACs activities by trichostatin A (TSA) and valproic acid (VPA) promptly and dramatically repressed transcription of Gnrh1 gene in the mouse immortalized mature GnRH neuronal cells GT1-7. The suppression was connected with a specific region of Gnrh1 gene promoter, which contains two consensus Otx2 binding sites. Otx2 has been known to activate the basal and also enhancer-driven transcription of Gnrh1 gene. The transcriptional activity of Otx2 is negatively modulated by Grg4, a member of the Groucho-related-gene (Grg) family. In the present study, the expression of Otx2 was downregulated by TSA and VPA in GT1-7 cells, accompanied with the opposite changes of Grg4 expression. Chromatin immunoprecipitation and electrophoretic mobility shift assays demonstrated that the DNA-binding activity of Otx2 to Gnrh1 gene was suppressed by TSA and VPA. Overexpression of Otx2 partly abolished the TSA- and VPA-induced downregulation of Gnrh1 gene expression. CONCLUSIONS/SIGNIFICANCE: Our data indicate that HDAC inhibitors downregulate Gnrh1 gene expression via repressing Otx2-driven transcriptional activity. This study should provide an insight for our understanding on the effects of HDACs in the reproductive system and suggests that HDACs could be potential novel targets for the therapy of GnRH-related diseases.


Subject(s)
Gene Expression Regulation/physiology , Gonadotropin-Releasing Hormone/genetics , Histone Deacetylases/metabolism , Otx Transcription Factors/physiology , Animals , Blotting, Western , Cell Line , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Histone Deacetylases/physiology , Hydroxamic Acids/pharmacology , Mice , Promoter Regions, Genetic , Transcription, Genetic/physiology , Valproic Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL