Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Hum Genet ; 66(3): 333-338, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32920601

ABSTRACT

The PAX6 is essential for ocular morphogenesis and is known to be highly sensitive to changes in gene expression, where neither over- nor under-expression ensures normal ocular development. Two unrelated probands with classical aniridia who were previously considered "PAX6-negative", were studied by whole-genome sequencing. Through the use of multiple in silico deep learning-based algorithms, we identified two novel putative causal mutations, c.-133_-132del in the 5' untranslated region (5'-UTR) and c.-52 + 5G>A in an intron upstream of the PAX6 gene. The luciferase activity was significantly increased and VAX2 binding was disrupted with the former 5'-UTR variant compared with wild-type sequence, which resulted in a striking overexpression of PAX6. The minigene assay showed that the c.-52 + 5G>A mutation caused defective splicing, which resulted in the formation of truncated transcripts.


Subject(s)
Aniridia/genetics , Mutation , PAX6 Transcription Factor/genetics , 5' Untranslated Regions/genetics , Algorithms , Causality , Deep Learning , Electrophoretic Mobility Shift Assay , Eye/embryology , Gene Expression Regulation , Genes, Reporter , Homeodomain Proteins/metabolism , Humans , Introns/genetics , Molecular Sequence Annotation , PAX6 Transcription Factor/biosynthesis , PAX6 Transcription Factor/physiology , Recombinant Proteins/genetics , Sequence Deletion , Whole Genome Sequencing
2.
Sci Rep ; 10(1): 16075, 2020 09 30.
Article in English | MEDLINE | ID: mdl-32999322

ABSTRACT

Intra-retinal axon guidance involves a coordinated expression of transcription factors, axon guidance genes, and secretory molecules within the retina. Pax6, the master regulator gene, has a spatio-temporal expression typically restricted till neurogenesis and fate-specification. However, our observation of persistent expression of Pax6 in mature RGCs led us to hypothesize that Pax6 could play a major role in axon guidance after fate specification. Here, we found significant alteration in intra-retinal axon guidance and fasciculation upon knocking out of Pax6 in E15.5 retina. Through unbiased transcriptome profiling between Pax6fl/fl and Pax6-/- retinas, we revealed the mechanistic insight of its role in axon guidance. Our results showed a significant increase in the expression of extracellular matrix molecules and decreased expression of retinal fate specification and neuron projection guidance molecules. Additionally, we found that EphB1 and Sema5B are directly regulated by Pax6 owing to the guidance defects and improper fasciculation of axons. We conclude that Pax6 expression post fate specification of RGCs is necessary for regulating the expression of axon guidance genes and most importantly for maintaining a conducive ECM through which the nascent axons get guided and fasciculate to reach the optic disc.


Subject(s)
Axon Fasciculation/physiology , Axon Guidance/physiology , PAX6 Transcription Factor/physiology , Retinal Ganglion Cells/physiology , Animals , Axon Fasciculation/genetics , Axon Guidance/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Extracellular Matrix/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogenesis/genetics , Neurogenesis/physiology , PAX6 Transcription Factor/deficiency , PAX6 Transcription Factor/genetics , Pregnancy , RNA-Seq , Receptor, EphB1/genetics , Receptor, EphB1/physiology , Retina/embryology , Retina/growth & development , Retina/physiology , Retinal Ganglion Cells/cytology , Semaphorins/genetics , Semaphorins/physiology
3.
Biochem Biophys Res Commun ; 528(3): 561-566, 2020 07 30.
Article in English | MEDLINE | ID: mdl-32505347

ABSTRACT

Cardiac fibroblast (CF) differentiation plays a crucial role in cardiac fibrosis, which is a specific manifestation distinguishing pathological cardiac hypertrophy from physiological hypertrophy. The DNA-binding activity of paired box 6 (Pax6) has been shown to be oppositely regulated in physiological and pathological hypertrophy; however, it remains unclear whether Pax6 is involved in CF differentiation during cardiac fibrosis. We found that Pax6 is expressed in the heart of and CFs isolated from adult mice. Moreover, angiotensin II (Ang II) induced the downregulation of Pax6 mRNA and protein expression in fibrotic heart tissue and cardiac myofibroblasts. Pax6 knockdown in CFs promoted the expression of the myofibroblast marker α-smooth muscle actin (α-SMA) and the synthesis of the extracellular matrix (ECM) proteins collagen I and fibronectin. Furthermore, we validated the ability of Pax6 to bind to the promoter regions of Cxcl10 and Il1r2 and the intronic region of Tgfb1. Pax6 knockdown in CFs decreased CXC chemokine 10 (CXCL10) and interleukin-1 receptor 2 (IL-1R2) expression and increased transforming growth factor ß1 (TGFß1) expression, mimicking the effects of Ang II. In conclusion, Pax6 exerts an inhibitory effect on CF differentiation and ECM synthesis by transcriptionally activating the expression of the anti-fibrotic factors CXCL10 and IL-1R2 and repressing the expression of the pro-fibrotic factor TGFß1. Therefore, maintaining Pax6 expression in CFs is essential for preventing CF differentiation, and provides a new therapeutic target for cardiac fibrosis.


Subject(s)
Cell Differentiation/physiology , Myocardium/cytology , Myocardium/metabolism , PAX6 Transcription Factor/physiology , Angiotensin II/pharmacology , Animals , Cardiomegaly/etiology , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cell Differentiation/genetics , Chemokine CXCL10/genetics , Disease Models, Animal , Extracellular Matrix Proteins/biosynthesis , Fibroblasts/cytology , Fibroblasts/metabolism , Fibrosis , Gene Expression Regulation , Gene Knockdown Techniques , Introns , Male , Mice , Mice, Inbred C57BL , PAX6 Transcription Factor/antagonists & inhibitors , PAX6 Transcription Factor/genetics , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Receptors, Interleukin-1 Type II/genetics , Transforming Growth Factor beta1/genetics
4.
Cereb Cortex ; 30(2): 465-475, 2020 03 21.
Article in English | MEDLINE | ID: mdl-31206158

ABSTRACT

The embryonic mouse cortex displays a striking low caudo-medial and high rostro-lateral graded expression of the homeoprotein transcription factor Pax6, which presents both cell autonomous and direct noncell autonomous activities. Through the genetic induction of anti-Pax6 single-chain antibody secretion, we have analyzed Pax6 noncell autonomous activity on the migration of cortical hem- and septum-derived Cajal-Retzius (CR) neurons by live imaging of flat mount developing cerebral cortices. Blocking extracellular Pax6 disrupts tangential CR cell migration patterns by decreasing the distance traveled and changing both directionality and depth at which CR cells migrate. Tracking of single CR cells in mutant cortices revealed that extracellular Pax6 neutralization enhances contact repulsion in medial regions yet reduces it in lateral regions. This study demonstrates that secreted Pax6 controls neuronal migration and distribution and suggests that it acts as a bona fide morphogen at an early stage of cerebral cortex development.


Subject(s)
Cell Movement , Neocortex/growth & development , Neurons/physiology , PAX6 Transcription Factor/physiology , Animals , Mice, Inbred C57BL , Mice, Transgenic
5.
Dev Biol ; 456(1): 86-103, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31445008

ABSTRACT

The transcription factor Pax6 is an important regulator of early animal development. Loss of function mutations of pax6 in a range of animals result in a reduction or complete loss of the eye, a reduction of a subset of neurons, and defects in axon growth. There are no studies focusing on the role of pax6 during development of any lophotrochozoan representative, however, expression of pax6 in the developing eye and nervous system in a number of species suggest that pax6 plays a highly conserved role in eye and nervous system formation. We investigated the functional role of pax6 during development of the marine annelid Capitella teleta. Expression of pax6 transcripts in C. teleta larvae is similar to patterns found in other animals, with distinct subdomains in the brain and ventral nerve cord as well as in the larval and juvenile eye. To perturb pax6 function, two different splice-blocking morpholinos and a translation-blocking morpholino were used. Larvae resulting from microinjections with either splice-blocking morpholino show a reduction of the pax6 transcript. Development of both the larval eyes and the central nervous system architecture are highly disrupted following microinjection of each of the three morpholinos. The less severe phenotype observed when only the homeodomain is disrupted suggests that presence of the paired domain is sufficient for partial function of the Pax6 protein. Preliminary downstream target analysis confirms disruption in expression of some components of the retinal gene regulatory network, as well as disruption of genes involved in nervous system development. Results from this study, taken together with studies from other species, reveal an evolutionarily conserved role for pax6 in eye and neural specification and development.


Subject(s)
Eye/embryology , Nervous System/embryology , PAX6 Transcription Factor/metabolism , Animals , Annelida/genetics , Annelida/metabolism , Eye/metabolism , Eye Proteins/genetics , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Morpholinos/genetics , Mutation , Nervous System/metabolism , Organogenesis/genetics , PAX6 Transcription Factor/genetics , PAX6 Transcription Factor/physiology , Promoter Regions, Genetic/genetics , Repressor Proteins/metabolism , Retina/metabolism
6.
Brain Res ; 1705: 95-103, 2019 02 15.
Article in English | MEDLINE | ID: mdl-29499177

ABSTRACT

Pax6 transcription factor is a key player in several aspects of brain development and function. Autism spectrum disorder (ASD) is a neurodevelopmental disorder in which several loci and/or genes have been suggested as causative candidate factors. Based on data obtained from meta-analyses of the transcriptome and ChIP analyses, we hypothesized that the neurodevelopmental gene PAX6 regulates and/or binds to a large number of genes (including many ASD-related ones) that modulate the fate of neural stem/progenitor cells and functions of neuronal cells, subsequently affecting animal behavior. Network analyses of PAX6/ASD-related molecules revealed significant clusters of molecular interactions involving regulation of cell-cell adhesion, ion transport, and transcriptional regulation. We discuss a novel function of Pax6 as a chromatin modulator that alters the chromatin status of ASD genes, thereby inducing diverse phenotypes of ASD and related neurodevelopmental diseases.


Subject(s)
Autism Spectrum Disorder/metabolism , Brain/embryology , PAX6 Transcription Factor/metabolism , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/physiopathology , Brain/metabolism , Chromatin/metabolism , Gene Expression Regulation/genetics , Gene Regulatory Networks/genetics , Humans , Neural Stem Cells/metabolism , Neurodevelopmental Disorders , Neurons/metabolism , Neurons/physiology , PAX6 Transcription Factor/physiology , Stem Cells/metabolism , Transcription Factors/metabolism , Transcriptome
7.
Exp Eye Res ; 179: 8-17, 2019 02.
Article in English | MEDLINE | ID: mdl-30292490

ABSTRACT

PAX6-related Aniridia is a sight-threatening disease involving progression of secondary glaucoma and aniridia related keratopathy (ARK). Change or loss of limbal epithelial progenitors causes epithelial surface defects. We analyzed the effect of PAX6 on mRNA expression changes with a two-step approach, as follows. First, we sequenced mRNA from limbal epithelial cells isolated from controls and aniridia patients. Second, we confirmed the bioinformatics and literature-based result list for a small interfering RNA (siRNA)-based primary aniridia cell model (PAX6 knockdown). With this approach, we expected that the genes directly influenced by PAX6 would be distinguishable from those affected secondarily by the ARK disease state. Therefore, epithelial cells were isolated from the limbus region of two patients with aniridia and cultured in keratinocyte serum-free medium. Normal control cells were obtained from the limbus region of corneal donors. For the siRNA-based aniridia cell model, cells were transfected with Lipofectamine and 5 nM siRNA against PAX6 or control treatment. All cells were lysed to yield DNA, RNA, and protein. Reduction of PAX6 protein was assessed by western blot. Aniridia and control Poly-A-enriched RNA libraries were subjected to next-generation sequencing. The differential analysis was a combination of quantification with RSEM and differential tests with edgeR. Gene lists were filtered by comparison to NCBI GEO datasets, annotated with DAVID, and manually annotated using a literature search. Based on the resulting filtered gene list, qPCR primers were purchased, and candidate genes (TP63, ABCG2, ADH7, ALDH1A1, PITX1, DKK1, DSG1, KRT12, KRT3, KRT13, SPINK6, SPINK7, CTSV, SERPINB1) were verified by qPCR on the siRNA-based aniridia cell model. We identified genes that might be regulated by PAX6 and showed that SPINK7 mRNA, which codes for a protease inhibitor, is downregulated in patients as well as in our primary aniridia cell model. ALDH1A1 and AHD7 mRNA levels were reduced in limbal epithelial cells of aniridia patients, and both transcripts were downregulated by PAX6 knockdown in our cell model. This siRNA-based aniridia cell model is a valuable tool for confirming identified PAX6-affected genes that might promote ARK pathogenesis. The model recapitulated expression changes for SPINK7, ADH7, and ALDH1A1 that were also observed in patient samples. These results provide evidence that PAX6 might drive corneal epithelial differentiation by direct or indirect control of retinoic acid signaling processes through ADH7 and ALDH1A1.


Subject(s)
Alcohol Dehydrogenase/genetics , Aldehyde Dehydrogenase/genetics , Aniridia/genetics , Epithelium, Corneal/metabolism , Limbus Corneae/metabolism , Signal Transduction/physiology , Tretinoin/metabolism , Aldehyde Dehydrogenase 1 Family , Blotting, Western , Cell Differentiation , Cells, Cultured , Corneal Diseases/genetics , Corneal Diseases/metabolism , Gene Expression Regulation/physiology , High-Throughput Nucleotide Sequencing , Humans , Models, Biological , PAX6 Transcription Factor/physiology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Retinal Dehydrogenase , Serine Peptidase Inhibitors, Kazal Type/genetics , Transfection
8.
J Neurosci ; 39(1): 44-62, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30425119

ABSTRACT

Control of neuronal precursor cell proliferation is essential for normal brain development, and deregulation of this fundamental developmental event contributes to brain diseases. Typically, neuronal precursor cell proliferation extends over long periods of time during brain development. However, how neuronal precursor proliferation is regulated in a temporally specific manner remains to be elucidated. Here, we report that conditional KO of the transcriptional regulator SnoN in cerebellar granule neuron precursors robustly inhibits the proliferation of these cells and promotes their cell cycle exit at later stages of cerebellar development in the postnatal male and female mouse brain. In laser capture microdissection followed by RNA-Seq, designed to profile gene expression specifically in the external granule layer of the cerebellum, we find that SnoN promotes the expression of cell proliferation genes and concomitantly represses differentiation genes in granule neuron precursors in vivo Remarkably, bioinformatics analyses reveal that SnoN-regulated genes contain binding sites for the transcription factors N-myc and Pax6, which promote the proliferation and differentiation of granule neuron precursors, respectively. Accordingly, we uncover novel physical interactions of SnoN with N-myc and Pax6 in cells. In behavior analyses, conditional KO of SnoN impairs cerebellar-dependent learning in a delayed eye-blink conditioning paradigm, suggesting that SnoN-regulation of granule neuron precursor proliferation bears functional consequences at the organismal level. Our findings define a novel function and mechanism for the major transcriptional regulator SnoN in the control of granule neuron precursor proliferation in the mammalian brain.SIGNIFICANCE STATEMENT This study reports the discovery that the transcriptional regulator SnoN plays a crucial role in the proliferation of cerebellar granule neuron precursors in the postnatal mouse brain. Conditional KO of SnoN in granule neuron precursors robustly inhibits the proliferation of these cells and promotes their cycle exit specifically at later stages of cerebellar development, with biological consequences of impaired cerebellar-dependent learning. Genomics and bioinformatics analyses reveal that SnoN promotes the expression of cell proliferation genes and concomitantly represses cell differentiation genes in vivo Although SnoN has been implicated in distinct aspects of the development of postmitotic neurons, this study identifies a novel function for SnoN in neuronal precursors in the mammalian brain.


Subject(s)
Brain/cytology , Cell Proliferation , Cerebellum/physiology , Neural Stem Cells/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Animals , Behavior, Animal , Blinking/physiology , Brain/growth & development , Cell Differentiation/genetics , Cerebellum/cytology , Computational Biology , Cytoplasmic Granules/physiology , Female , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Genes, myc/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PAX6 Transcription Factor/genetics , PAX6 Transcription Factor/physiology
9.
Development ; 145(15)2018 08 02.
Article in English | MEDLINE | ID: mdl-29980566

ABSTRACT

The transcription factor Pax6 is considered the master control gene for eye formation because (1) it is present within the genomes and retina/lens of all animals with a visual system; (2) severe retinal defects accompany its loss; (3) Pax6 genes have the ability to substitute for one another across the animal kingdom; and (4) Pax6 genes are capable of inducing ectopic eye/lens in flies and mammals. Many roles of Pax6 were first elucidated in Drosophila through studies of the gene eyeless (ey), which controls both growth of the entire eye-antennal imaginal disc and fate specification of the eye. We show that Ey also plays a surprising role within cells of the peripodial epithelium to control pattern formation. It regulates the expression of decapentaplegic (dpp), which is required for initiation of the morphogenetic furrow in the eye itself. Loss of Ey within the peripodial epithelium leads to the loss of dpp expression within the eye, failure of the furrow to initiate, and abrogation of retinal development. These findings reveal an unexpected mechanism for how Pax6 controls eye development in Drosophila.


Subject(s)
DNA-Binding Proteins/physiology , Drosophila Proteins/physiology , Epithelium/embryology , Eye/embryology , Morphogenesis/genetics , PAX6 Transcription Factor/physiology , Animals , Animals, Genetically Modified , Body Patterning/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Embryo, Nonmammalian , Epithelium/metabolism , Eye/metabolism , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Imaginal Discs/embryology , Imaginal Discs/metabolism , PAX6 Transcription Factor/genetics
10.
Mol Biol Rep ; 45(4): 533-540, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29770908

ABSTRACT

The Pax6 is a multifunctional pairedbox and homeobox containing transcription factor which is involved in several functions of brain, eyes, and pancreas. It regulates expression of genes involved in cell proliferation, differentiation, inflammation, oxidative stress management, and neuropathy. Dynamic changes in the sub-cellular localization of Pax6 are proposed to regulate its activity, however, the underlying mechanism remains poorly understood. The oxidative stress mediated changes were studied in sub-cellular localization of Pax6 in cultured cells derived from the eye (cornea) and pancreas. The impact of induced oxidative stress was investigated on reactive oxygen species scavenger molecules, Superoxide dismutase1 (SOD1) and Catalase, and a critical cell signalling molecule Transforming growth factor-beta (TGF-ß1). The cells were treated with three different concentrations of H2O2, viz., 0.3, 1.5, and 3.0 mM. The cell viability was analysed through Trypan blue dye exclusion assay. The localization of Pax6 was observed by immunofluorescence labeling, and alterations in levels of Pax6, SOD1, Catalase, and TGF-ß1 were investigated by semi-quantitative RT-PCR. Nucleo-cytoplasmic shuttling of Pax6 was observed in cells of corneal epithelial (SIRC) and pancreatic origins (MIA-PaCa2). The percentage distribution of Pax6 in nuclear and cytoplasmic compartments of SIRC and MIA-PaCa2 cells was analyzed through ImageJ software. Level of hydrogen peroxide affects expression and sub-cellular localization of Pax6. Expression of Pax6 and TGF-ß1 are directly associated with changes in sub-cellular localization of Pax6 and modulation in expression of Catalase. This may be the result of a cellular protective mechanism against peroxide-dependent cellular stress.


Subject(s)
Hydrogen Peroxide/metabolism , PAX6 Transcription Factor/genetics , PAX6 Transcription Factor/physiology , Active Transport, Cell Nucleus/genetics , Active Transport, Cell Nucleus/physiology , Animals , Brain/metabolism , Catalase/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Cell Survival/drug effects , Eye/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Humans , Oxidation-Reduction , Oxidative Stress/genetics , Rabbits , Reactive Oxygen Species , Signal Transduction , Superoxide Dismutase-1/metabolism , Transforming Growth Factor beta/metabolism
11.
Development ; 145(8)2018 04 16.
Article in English | MEDLINE | ID: mdl-29661783

ABSTRACT

The evolution of unique organ structures is associated with changes in conserved developmental programs. However, characterizing the functional conservation and variation of homologous transcription factors (TFs) that dictate species-specific cellular dynamics has remained elusive. Here, we dissect shared and divergent functions of Pax6 during amniote brain development. Comparative functional analyses revealed that the neurogenic function of Pax6 is highly conserved in the developing mouse and chick pallium, whereas stage-specific binary functions of Pax6 in neurogenesis are unique to mouse neuronal progenitors, consistent with Pax6-dependent temporal regulation of Notch signaling. Furthermore, we identified that Pax6-dependent enhancer activity of Dbx1 is extensively conserved between mammals and chick, although Dbx1 expression in the developing pallium is highly divergent in these species. Our results suggest that spatiotemporal changes in Pax6-dependent regulatory programs contributed to species-specific neurogenic patterns in mammalian and avian lineages, which underlie the morphological divergence of the amniote pallial architectures.


Subject(s)
Avian Proteins/physiology , Brain/embryology , Brain/physiology , PAX6 Transcription Factor/physiology , Animals , Animals, Genetically Modified , Avian Proteins/genetics , Chick Embryo , Enhancer Elements, Genetic , Evolution, Molecular , Female , Gene Deletion , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Mice, Transgenic , Neurogenesis/genetics , Neurogenesis/physiology , PAX6 Transcription Factor/deficiency , PAX6 Transcription Factor/genetics , Pregnancy , Receptors, Notch/genetics , Receptors, Notch/physiology , Signal Transduction , Species Specificity
12.
Dev Biol ; 432(1): 140-150, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28993200

ABSTRACT

In the developing retina, as in other regions of the CNS, neural progenitors give rise to individual cell types during discrete temporal windows. Pax6 is expressed in retinal progenitor cells (RPCs) throughout the course of retinogenesis, and has been shown to be required during early retinogenesis for generation of most early-born cell types. In this study, we examined the function of Pax6 in postnatal mouse retinal development. We found that Pax6 is essential for the generation of late-born interneurons, while inhibiting photoreceptor differentiation. Generation of bipolar interneurons requires Pax6 expression in RPCs, while Pax6 is required for the generation of glycinergic, but not for GABAergic or non-GABAergic-non-glycinergic (nGnG) amacrine cell subtypes. In contrast, overexpression of either full-length Pax6 or its 5a isoform in RPCs induces formation of cells with nGnG amacrine features, and suppresses generation of other inner retinal cell types. Moreover, overexpression of both Pax6 variants prevents photoreceptor differentiation, most likely by inhibiting Crx expression. Taken together, these data show that Pax6 acts in RPCs to control differentiation of multiple late-born neuronal cell types.


Subject(s)
Neurons/physiology , PAX6 Transcription Factor/physiology , Photoreceptor Cells, Vertebrate/physiology , Retina/physiology , Amacrine Cells/cytology , Amacrine Cells/metabolism , Amacrine Cells/physiology , Animals , Cell Differentiation/physiology , Female , Interneurons/cytology , Interneurons/metabolism , Interneurons/physiology , Male , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Neurogenesis/physiology , Neurons/cytology , Neurons/metabolism , PAX6 Transcription Factor/metabolism , Photoreceptor Cells, Vertebrate/cytology , Photoreceptor Cells, Vertebrate/metabolism , Retina/cytology , Retina/metabolism , Retinal Neurons/cytology , Retinal Neurons/metabolism , Retinal Neurons/physiology
13.
Neuron ; 95(6): 1319-1333.e5, 2017 Sep 13.
Article in English | MEDLINE | ID: mdl-28867550

ABSTRACT

Changes in social preference of amphibian larvae result from sustained exposure to kinship odorants. To understand the molecular and cellular mechanisms of this neuroplasticity, we investigated the effects of olfactory system activation on neurotransmitter (NT) expression in accessory olfactory bulb (AOB) interneurons during development. We show that protracted exposure to kin or non-kin odorants changes the number of dopamine (DA)- or gamma aminobutyric acid (GABA)-expressing neurons, with corresponding changes in attraction/aversion behavior. Changing the relative number of dopaminergic and GABAergic AOB interneurons or locally introducing DA or GABA receptor antagonists alters kinship preference. We then isolate AOB microRNAs (miRs) differentially regulated across these conditions. Inhibition of miR-375 and miR-200b reveals that they target Pax6 and Bcl11b to regulate the dopaminergic and GABAergic phenotypes. The results illuminate the role of NT switching governing experience-dependent social preference. VIDEO ABSTRACT.


Subject(s)
Choice Behavior/physiology , Dopamine/biosynthesis , MicroRNAs/physiology , Neurotransmitter Agents/biosynthesis , Olfactory Bulb/metabolism , Social Behavior , gamma-Aminobutyric Acid/biosynthesis , Animals , Dopamine/physiology , Dopamine Antagonists/pharmacology , GABA Antagonists/pharmacology , Interneurons/physiology , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Neurons/metabolism , Neurons/physiology , Neurotransmitter Agents/physiology , PAX6 Transcription Factor/physiology , Pheromones/physiology , Siblings , Transcription Factors/physiology , Xenopus Proteins/physiology , Xenopus laevis , gamma-Aminobutyric Acid/physiology
14.
Proc Natl Acad Sci U S A ; 114(23): 5846-5853, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28584125

ABSTRACT

Paired box 6 (Pax6) is considered to be the master control gene for eye development in all seeing animals studied so far. In vertebrates, it is required not only for lens/retina formation but also for the development of the CNS, olfactory system, and pancreas. Although Pax6 plays important roles in cell differentiation, proliferation, and patterning during the development of these systems, the underlying mechanism remains poorly understood. In the fruit fly, Drosophila melanogaster, Pax6 also functions in a range of tissues, including the eye and brain. In this report, we describe the function of Pax6 in Drosophila eye-antennal disc development. Previous studies have suggested that the two fly Pax6 genes, eyeless (ey) and twin of eyeless (toy), initiate eye specification, whereas eyegone (eyg) and the Notch (N) pathway independently regulate cell proliferation. Here, we show that Pax6 controls eye progenitor cell survival and proliferation through the activation of teashirt (tsh) and eyg, thereby indicating that Pax6 initiates both eye specification and proliferation. Although simultaneous loss of ey and toy during early eye-antennal disc development disrupts the development of all head structures derived from the eye-antennal disc, overexpression of N or tsh in the absence of Pax6 rescues only antennal and head epidermis development. Furthermore, overexpression of tsh induces a homeotic transformation of the fly head into thoracic structures. Taking these data together, we demonstrate that Pax6 promotes development of the entire eye-antennal disc and that the retinal determination network works to repress alternative tissue fates, which ensures proper development of adult head structures.


Subject(s)
Arthropod Antennae/embryology , Drosophila Proteins/physiology , Drosophila melanogaster/embryology , Eye/embryology , Head/embryology , Models, Biological , PAX6 Transcription Factor/physiology , Animals , Cell Differentiation , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Imaginal Discs/cytology , Imaginal Discs/metabolism , PAX6 Transcription Factor/genetics , PAX6 Transcription Factor/metabolism
16.
Mol Vis ; 22: 990-1004, 2016.
Article in English | MEDLINE | ID: mdl-27563231

ABSTRACT

PURPOSE: Patients with a heterozygous mutation in the gene encoding the transcription factor, PAX6, have a degenerative corneal opacity associated with failure of normal radial epithelial cell migration across the corneal surface and a reported wound healing defect. This study investigated the guidance mechanisms that drive the directed migration of corneal epithelial cells. METHODS: In vivo corneal epithelial wounding was performed in adult wild-type and Pax6(+/-) mice, and the healing migration rates were compared. To investigate the control of the cell migration direction, primary corneal epithelial cells from wild-type and Pax6(+/-) mice were plated on grooved quartz substrates, and alignment relative to the grooves was assayed. A reconstructed corneal culture system was developed in which dissociated wild-type and genetically mutant corneal epithelial cells could be cultured on a de-epithelialized corneal stroma or basement membrane and their migration assayed with time-lapse microscopy. RESULTS: The Pax6(+/-) cells efficiently re-epithelialized corneal wounds in vivo but had mild slowing of healing migration compared to the wild-type. Cells aligned parallel to quartz grooves in vitro, but the Pax6(+/-) cells were less robustly oriented than the wild-type. In the reconstructed corneal culture system, corneal epithelial cells continued to migrate radially, showing that the cells are guided by contact-mediated cues from the basement membrane. Recombining wild-type and Pax6 mutant corneal epithelial cells with wild-type and Pax6 mutant corneal stroma showed that normal Pax6 dosage was required autonomously in the epithelial cells for directed migration. Integrin-mediated attachment to the substrate, and intracellular PI3Kγ activity, were required for migration. Pharmacological inhibition of cAMP signaling randomized migration tracks in reconstructed corneas. CONCLUSIONS: Striking patterns of centripetal migration of corneal epithelial cells observed in vivo are driven by contact-mediated cues operating through an intracellular cAMP pathway, and failure to read these cues underlies the migration defects that accompany corneal degeneration in patients with mutations in PAX6.


Subject(s)
Cell Movement/physiology , Corneal Injuries/physiopathology , Epithelial Cells/physiology , Focal Adhesions/physiology , PAX6 Transcription Factor/physiology , Wound Healing/physiology , Animals , Class Ib Phosphatidylinositol 3-Kinase/physiology , Corneal Stroma/cytology , Cyclic AMP/physiology , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Re-Epithelialization/physiology , Signal Transduction/physiology
17.
PLoS One ; 11(7): e0158210, 2016.
Article in English | MEDLINE | ID: mdl-27387288

ABSTRACT

The habenulae are highly conserved nuclei in the dorsal diencephalon that connect the forebrain to the midbrain and hindbrain. These nuclei have been implicated in a broad variety of behaviours in humans, primates, rodents and zebrafish. Despite this, the molecular mechanisms that control the genesis and differentiation of neural progenitors in the habenulae remain relatively unknown. We have previously shown that, in zebrafish, the timing of habenular neurogenesis is left-right asymmetric and that in the absence of Nodal signalling this asymmetry is lost. Here, we show that habenular neurogenesis requires the homeobox transcription factor Pax6a and the redundant action of two proneural bHLH factors, Neurog1 and Neurod4. We present evidence that Hedgehog signalling is required for the expression of pax6a, which is in turn necessary for the expression of neurog1 and neurod4. Finally, we demonstrate by pharmacological inhibition that Hedgehog signalling is required continuously during habenular neurogenesis and by cell transplantation experiments that pathway activation is required cell autonomously. Our data sheds light on the mechanism underlying habenular development that may provide insights into how Nodal signalling imposes asymmetry on the timing of habenular neurogenesis.


Subject(s)
Habenula/embryology , Hedgehog Proteins/physiology , Neurogenesis , PAX6 Transcription Factor/physiology , Signal Transduction , Zebrafish Proteins/physiology , Zebrafish/embryology , Animals , Animals, Genetically Modified , Basic Helix-Loop-Helix Transcription Factors/physiology , Body Patterning , Gene Expression Regulation, Developmental , Genotype , Heterozygote , Mutation , Nerve Tissue Proteins/physiology , Neurons/metabolism , Polymerase Chain Reaction , Transcription Factors/metabolism
18.
PLoS One ; 11(6): e0158153, 2016.
Article in English | MEDLINE | ID: mdl-27355350

ABSTRACT

Pax6 is a transcription factor that pleiotropically regulates various developmental processes in the central nervous system. In a previous study, we revealed that Pax6 heterozygous mutant (rSey2/+) adult rats exhibit abnormalities in social interaction. However, the brain malformations underlying the behavioral abnormality are unknown. To elucidate the brain malformations in rSey2/+ rats, we morphometrically analyzed brains of rSey2/+ and wild type rats using small-animal magnetic resonance imaging (MRI). Sixty 10-week-old rats underwent brain MRI (29 rSey2/+ rats and 31 wild type rats). SPM8 software was used for image preprocessing and statistical image analysis. Normalized maps of the Jacobian determinant, a parameter for the expansion and/or contraction of brain regions, were obtained for each rat. rSey2/+ rats showed significant volume decreases in various brain regions including the neocortex, corpus callosum, olfactory structures, hippocampal formation, diencephalon, and midbrain compared to wild type rats. Among brain regions, the anterior commissure showed significant interaction between genotype and sex, indicating the effect of genotype difference on the anterior commissure volume was more robust in females than in males. The rSey2/+ rats exhibited decreased volume in various gray and white matter regions of the brain, which may contribute to manifestation of abnormal social behaviors.


Subject(s)
Brain/embryology , Brain/growth & development , Magnetic Resonance Imaging , Mutation , PAX6 Transcription Factor/genetics , Animals , Behavior, Animal , Female , Genotype , Gray Matter/pathology , Heterozygote , Male , PAX6 Transcription Factor/physiology , Rats , Rats, Sprague-Dawley , Sex Factors , Software , White Matter/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...