Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
PLoS Biol ; 20(9): e3001563, 2022 09.
Article in English | MEDLINE | ID: mdl-36067211

ABSTRACT

The development of stable specialized cell types in multicellular organisms relies on mechanisms controlling inductive intercellular signals and the competence of cells to respond to such signals. In developing cerebral cortex, progenitors generate only glutamatergic excitatory neurons despite being exposed to signals with the potential to initiate the production of other neuronal types, suggesting that their competence is limited. Here, we tested the hypothesis that this limitation is due to their expression of transcription factor Pax6. We used bulk and single-cell RNAseq to show that conditional cortex-specific Pax6 deletion from the onset of cortical neurogenesis allowed some progenitors to generate abnormal lineages resembling those normally found outside the cortex. Analysis of selected gene expression showed that the changes occurred in specific spatiotemporal patterns. We then compared the responses of control and Pax6-deleted cortical cells to in vivo and in vitro manipulations of extracellular signals. We found that Pax6 loss increased cortical progenitors' competence to generate inappropriate lineages in response to extracellular factors normally present in developing cortex, including the morphogens Shh and Bmp4. Regional variation in the levels of these factors could explain spatiotemporal patterns of fate change following Pax6 deletion in vivo. We propose that Pax6's main role in developing cortical cells is to minimize the risk of their development being derailed by the potential side effects of morphogens engaged contemporaneously in other essential functions.


Subject(s)
Homeodomain Proteins , Paired Box Transcription Factors , Cerebral Cortex/metabolism , Eye Proteins/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , PAX6 Transcription Factor/genetics , PAX6 Transcription Factor/metabolism , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism
2.
Front Cell Neurosci ; 12: 419, 2018.
Article in English | MEDLINE | ID: mdl-30498434

ABSTRACT

Pax6 is a key regulator of the rates of progenitor cell division in cerebral corticogenesis. Previous work has suggested that this action is mediated at least in part by regulation of the cell cycle gene Cdk6, which acts largely on the transition from G1 to S phase. We began the present study by investigating whether, in addition to Cdk6, other Pax6-regulated cell cycle genes are likely to be primary mediators of Pax6's actions on cortical progenitor cell cycles. Following acute cortex-specific deletion of Pax6, Cdk6 showed changes in expression a day earlier than any other Pax6-regulated cell cycle gene suggesting that it is the primary mediator of Pax6's actions. We then used flow cytometry to show that progenitors lacking Pax6 have a shortened G1 phase and that their Cdk6 levels are increased in all phases. We found that Cdk6 levels oscillate during the cell cycle, increasing from G1 to M phase. We propose a model in which loss of Pax6 shortens G1 phase by raising overall Cdk6 levels, thereby shortening the time taken for Cdk6 levels to cross a threshold triggering transition to S phase.

3.
iScience ; 10: 171-191, 2018 Dec 21.
Article in English | MEDLINE | ID: mdl-30529950

ABSTRACT

Differences in the growth and maturation of diverse forebrain tissues depend on region-specific transcriptional regulation. Individual transcription factors act simultaneously in multiple regions that develop very differently, raising questions about the extent to which their actions vary regionally. We found that the transcription factor Pax6 affects the transcriptomes and the balance between proliferation and differentiation in opposite directions in the diencephalon versus cerebral cortex. We tested several possible mechanisms to explain Pax6's tissue-specific actions and found that the presence of the transcription factor Foxg1 in the cortex but not in the diencephalon was most influential. We found that Foxg1 is responsible for many of the differences in cell cycle gene expression between the diencephalon and cortex and, in cortex lacking Foxg1, Pax6's action on the balance of proliferation versus differentiation becomes diencephalon like. Our findings reveal a mechanism for generating regional forebrain diversity in which one transcription factor completely reverses the actions of another.

4.
BMC Res Notes ; 11(1): 705, 2018 Oct 05.
Article in English | MEDLINE | ID: mdl-30290846

ABSTRACT

OBJECTIVE: The corneas of heterozygous Pax6+/- mice develop abnormally and deteriorate further after birth but it is not known whether the postnatal deterioration is predetermined by abnormal development. Our objective was to identify whether depletion of Pax6 in adult mice caused any corneal abnormalities, similar to those in Pax6+/- mice, where Pax6 levels are low throughout development and adulthood. We used two tamoxifen-inducible, Cre-loxP experimental strategies to deplete Pax6 either ubiquitously or in a restricted range of cell types. RESULTS: In a preliminary study, ubiquitous depletion of Pax6 by tamoxifen treatment of E9.5 CAG-CreERTg/-;Pax6fl/fl embryos affected eye development. Tamoxifen treatment of 12-week old, adult CAG-CreERTg/-;Pax6fl/+ and CAG-CreERTg/-;Pax6fl/fl mice resulted in weak and/or patchy Pax6 immunostaining in the corneal epithelium but caused no corneal abnormalities. GFP staining in tamoxifen-treated CAG-CreERTg/-;RCE:loxP reporter mice was also patchy. We attribute patchy Pax6 staining to mosaic deletion of the Pax6fl allele, probably caused by mosaic CAG-CreERTg expression. In a parallel study, we treated adult Krt19-CreERTg/-;Pax6fl/+ mice with tamoxifen to try to deplete Pax6 in limbal epithelial stem cells (LESCs) which replenish the corneal epithelium. However, Pax6 staining remained strong after a 12-week chase period so the Krt19-CreERTg/- transgene may have failed to target LESCs.


Subject(s)
Corneal Diseases/metabolism , Corneal Diseases/pathology , Epithelium, Corneal/pathology , PAX6 Transcription Factor/deficiency , Age Factors , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic
5.
Neural Dev ; 10: 26, 2015 Oct 31.
Article in English | MEDLINE | ID: mdl-26520399

ABSTRACT

BACKGROUND: In mouse embryos, the Pax6 transcription factor is expressed in the progenitors of thalamic neurons but not in thalamic neurons themselves. Its null-mutation causes early mis-patterning of thalamic progenitors. It is known that thalamic neurons generated by Pax6 (-/-) progenitors do not develop their normal connections with the cortex, but it is not clear why. We investigated the extent to which defects intrinsic to the thalamus are responsible. RESULTS: We first confirmed that, in constitutive Pax6 (-/-) mutants, the axons of thalamic neurons fail to enter the telencephalon and, instead, many of them take an abnormal path to the hypothalamus, whose expression of Slits would normally repel them. We found that thalamic neurons show reduced expression of the Slit receptor Robo2 in Pax6 (-/-) mutants, which might enhance the ability of their axons to enter the hypothalamus. Remarkably, however, in chimeras comprising a mixture of Pax6 (-/-) and Pax6 (+/+) cells, Pax6 (-/-) thalamic neurons are able to generate axons that exit the diencephalon, take normal trajectories through the telencephalon and avoid the hypothalamus. This occurs despite abnormalities in their molecular patterning (they express Nkx2.2, unlike normal thalamic neurons) and their reduced expression of Robo2. In conditional mutants, acute deletion of Pax6 from the forebrain at the time when thalamic axons are starting to grow does not prevent the development of the thalamocortical tract, suggesting that earlier extra-thalamic patterning and /or morphological defects are the main cause of thalamocortical tract failure in Pax6 (-/-) constitutive mutants. CONCLUSIONS: Our results indicate that Pax6 is required by thalamic progenitors for the normal molecular patterning of the thalamic neurons that they generate but thalamic neurons do not need normal Pax6-dependent patterning to become competent to grow axons that can be guided appropriately.


Subject(s)
Body Patterning/genetics , Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Neural Stem Cells/metabolism , Neurogenesis/genetics , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , Thalamus/embryology , Animals , Axons/metabolism , Eye Proteins/genetics , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , Neural Stem Cells/cytology , Neurons/cytology , Neurons/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction
6.
Front Cell Neurosci ; 9: 70, 2015.
Article in English | MEDLINE | ID: mdl-25805971

ABSTRACT

Understanding brain development remains a major challenge at the heart of understanding what makes us human. The neocortex, in evolutionary terms the newest part of the cerebral cortex, is the seat of higher cognitive functions. Its normal development requires the production, positioning, and appropriate interconnection of very large numbers of both excitatory and inhibitory neurons. Pax6 is one of a relatively small group of transcription factors that exert high-level control of cortical development, and whose mutation or deletion from developing embryos causes major brain defects and a wide range of neurodevelopmental disorders. Pax6 is very highly conserved between primate and non-primate species, is expressed in a gradient throughout the developing cortex and is essential for normal corticogenesis. Our understanding of Pax6's functions and the cellular processes that it regulates during mammalian cortical development has significantly advanced in the last decade, owing to the combined application of genetic and biochemical analyses. Here, we review the functional importance of Pax6 in regulating cortical progenitor proliferation, neurogenesis, and formation of cortical layers and highlight important differences between rodents and primates. We also review the pathological effects of PAX6 mutations in human neurodevelopmental disorders. We discuss some aspects of Pax6's molecular actions including its own complex transcriptional regulation, the distinct molecular functions of its splice variants and some of Pax6's known direct targets which mediate its actions during cortical development.

7.
Cell Rep ; 8(5): 1405-18, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25176648

ABSTRACT

During development, region-specific patterns of regulatory gene expression are controlled by signaling centers that release morphogens providing positional information to surrounding cells. Regulation of signaling centers themselves is therefore critical. The size and the influence of a Shh-producing forebrain organizer, the zona limitans intrathalamica (ZLI), are limited by Pax6. By studying mouse chimeras, we find that Pax6 acts cell autonomously to block Shh expression in cells around the ZLI. Immunoprecipitation and luciferase assays indicate that Pax6 can bind the Shh promoter and repress its function. An analysis of chimeras suggests that many of the regional gene expression pattern defects that occur in Pax6(-/-) diencephalic cells result from a non-cell-autonomous position-dependent defect of local intercellular signaling. Blocking Shh signaling in Pax6(-/-) mutants reverses major diencephalic patterning defects. We conclude that Pax6's cell-autonomous repression of Shh expression around the ZLI is critical for many aspects of normal diencephalic patterning.


Subject(s)
Diencephalon/metabolism , Eye Proteins/metabolism , Hedgehog Proteins/metabolism , Homeodomain Proteins/metabolism , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , Animals , Diencephalon/cytology , Diencephalon/embryology , Eye Proteins/genetics , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Homeodomain Proteins/genetics , Mice , Neurogenesis , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Promoter Regions, Genetic , Protein Binding , Repressor Proteins/genetics
8.
Neuron ; 78(2): 269-84, 2013 Apr 24.
Article in English | MEDLINE | ID: mdl-23622063

ABSTRACT

The mechanisms by which early spatiotemporal expression patterns of transcription factors such as Pax6 regulate cortical progenitors in a region-specific manner are poorly understood. Pax6 is expressed in a gradient across the developing cortex and is essential for normal corticogenesis. We found that constitutive or conditional loss of Pax6 increases cortical progenitor proliferation by amounts that vary regionally with normal Pax6 levels. We compared the gene expression profiles of equivalent Pax6-expressing progenitors isolated from Pax6⁺/⁺ and Pax6⁻/⁻ cortices and identified many negatively regulated cell-cycle genes, including Cyclins and Cdks. Biochemical assays indicated that Pax6 directly represses Cdk6 expression. Cyclin/Cdk repression inhibits retinoblastoma protein (pRb) phosphorylation, thereby limiting the transcription of genes that directly promote the mechanics of the cell cycle, and we found that Pax6 inhibits pRb phosphorylation and represses genes involved in DNA replication. Our results indicate that Pax6's modulation of cortical progenitor cell cycles is regional and direct.


Subject(s)
Body Patterning/genetics , Cerebral Cortex/cytology , Cyclin-Dependent Kinase 6/metabolism , Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , Retinoblastoma Protein/metabolism , Stem Cells/physiology , Animals , Bromodeoxyuridine , Cell Cycle/genetics , Cell Proliferation , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase 6/genetics , Embryo, Mammalian , Eye Proteins/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , PAX6 Transcription Factor , PAX7 Transcription Factor/genetics , Paired Box Transcription Factors/genetics , Phosphorylation , Protein Binding/genetics , Repressor Proteins/genetics , Retinoblastoma Protein/genetics , Transcription Factors/genetics
9.
Neural Dev ; 6: 9, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21418559

ABSTRACT

BACKGROUND: The transcription factor Foxg1 is an important regulator of telencephalic cell cycles. Its inactivation causes premature lengthening of telencephalic progenitor cell cycles and increased neurogenic divisions, leading to severe hypoplasia of the telencephalon. These proliferation defects could be a secondary consequence of the loss of Foxg1 caused by the abnormal expression of several morphogens (Fibroblast growth factor 8, bone morphogenetic proteins) in the telencephalon of Foxg1 null mutants. Here we investigated whether Foxg1 has a cell autonomous role in the regulation of telencephalic progenitor proliferation. We analysed Foxg1+/+↔Foxg1-/- chimeras, in which mutant telencephalic cells have the potential to interact with, and to have any cell non-autonomous defects rescued by, normal wild-type cells. RESULTS: Our analysis showed that the Foxg1-/- cells are under-represented in the chimeric telencephalon and the proportion of them in S-phase is significantly smaller than that of their wild-type neighbours, indicating that their under-representation is caused by a cell autonomous reduction in their proliferation. We then analysed the expression of the cell-cycle regulator Pax6 and found that it is cell-autonomously downregulated in Foxg1-/- dorsal telencephalic cells. We went on to show that the introduction into Foxg1-/- embryos of a transgene designed to reverse Pax6 expression defects resulted in a partial rescue of the telencephalic progenitor proliferation defects. CONCLUSIONS: We conclude that Foxg1 exerts control over telencephalic progenitor proliferation by cell autonomous mechanisms that include the regulation of Pax6, which itself is known to regulate proliferation cell autonomously in a regional manner.


Subject(s)
Eye Proteins/biosynthesis , Eye Proteins/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/physiology , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Neural Stem Cells/physiology , Paired Box Transcription Factors/biosynthesis , Paired Box Transcription Factors/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Telencephalon/cytology , Animals , Antimetabolites , Bromodeoxyuridine , Cell Count , Cell Proliferation , Chimera , Down-Regulation/genetics , Down-Regulation/physiology , Female , Gene Expression Regulation , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , PAX6 Transcription Factor , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction
10.
Cereb Cortex ; 21(1): 81-94, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20413449

ABSTRACT

The ventricular zone (VZ) of the embryonic dorsal telencephalon is a major site for generating cortical projection neurons. The transcription factor Pax6 is highly expressed in apical progenitors (APs) residing in the VZ from the earliest stages of corticogenesis. Previous studies mainly focused on Pax6(-/-) mice have implicated Pax6 in regulating cortical progenitor proliferation, neurogenesis, and formation of superficial cortical layers. We analyzed the developing cortex of PAX77 transgenic mice that overexpress Pax6 in its normal domains of expression. We show that Pax6 overexpression increases cell cycle length of APs and drives the system toward neurogenesis. These effects are specific to late stages of corticogenesis, when superficial layer neurons are normally generated, in cortical regions that express Pax6 at the highest levels. The number of superficial layer neurons is reduced in postnatal PAX77 mice, whereas radial migration and lamina specification of cortical neurons are not affected by Pax6 overexpression. Conditional deletion of Pax6 in cortical progenitors at midstages of corticogenesis, by using a tamoxifen-inducible Emx1-CreER line, affected both numbers and specification of late-born neurons in superficial layers of the mutant cortex. Our analyses suggest that correct levels of Pax6 are essential for normal production of superficial layers of the cortex.


Subject(s)
Body Patterning , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Eye Proteins/physiology , Homeodomain Proteins/physiology , Neurogenesis , Paired Box Transcription Factors/physiology , Repressor Proteins/physiology , Animals , Body Patterning/genetics , Cell Movement/genetics , Cerebral Cortex/metabolism , Eye Proteins/biosynthesis , Eye Proteins/genetics , Female , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/genetics , PAX6 Transcription Factor , Paired Box Transcription Factors/biosynthesis , Paired Box Transcription Factors/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics
11.
Development ; 137(3): 487-97, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20081193

ABSTRACT

Foxg1 is required for development of the ventral telencephalon in the embryonic mammalian forebrain. Although one existing hypothesis suggests that failed ventral telencephalic development in the absence of Foxg1 is due to reduced production of the morphogens sonic hedgehog (Shh) and fibroblast growth factor 8 (Fgf8), the possibility that telencephalic cells lacking Foxg1 are intrinsically incompetent to generate the ventral telencephalon has remained untested. We examined the ability of Foxg1(-/-) telencephalic cells to respond to Shh and Fgf8 by examining the expression of genes whose activation requires Shh or Fgf8 in vivo and by testing their responses to Shh and Fgf8 in culture. We found that many elements of the Shh and Fgf8 signalling pathways continue to function in the absence of Foxg1 but, nevertheless, we were unable to elicit normal responses of key ventral telencephalic marker genes in Foxg1(-/-) telencephalic tissue following a range of in vivo and in vitro manipulations. We explored the development of Foxg1(-/-) cells in Foxg1(-/-) Foxg1(+/+) chimeric embryos that contained ventral telencephalon created by normally patterned wild-type cells. We found that Foxg1(-/-) cells contributed to the chimeric ventral telencephalon, but that they retained abnormal specification, expressing dorsal rather than ventral telencephalic markers. These findings indicate that, in addition to regulating the production of ventralising signals, Foxg1 acts cell-autonomously in the telencephalon to ensure that cells develop the competence to adopt ventral identities.


Subject(s)
Forkhead Transcription Factors/physiology , Nerve Tissue Proteins/physiology , Telencephalon/cytology , Animals , Cells, Cultured , Fibroblast Growth Factor 8/physiology , Gene Expression Regulation, Developmental , Hedgehog Proteins/physiology , Kruppel-Like Transcription Factors , Mice , Mice, Knockout , Telencephalon/embryology , Telencephalon/growth & development , Transcription Factors , Zinc Finger Protein Gli3
12.
BMC Dev Biol ; 8: 59, 2008 May 28.
Article in English | MEDLINE | ID: mdl-18507827

ABSTRACT

BACKGROUND: The transcription factor Pax6 is expressed by many cell types in the developing eye. Eyes do not form in homozygous loss-of-function mouse mutants (Pax6Sey/Sey) and are abnormally small in Pax6Sey/+ mutants. Eyes are also abnormally small in PAX77 mice expressing multiple copies of human PAX6 in addition to endogenous Pax6; protein sequences are identical in the two species. The developmental events that lead to microphthalmia in PAX77 mice are not well-characterised, so it is not clear whether over- and under-expression of Pax6/PAX6 cause microphthalmia through similar mechanisms. Here, we examined the consequences of over-expression for the eye and its axonal connections. RESULTS: Eyes form in PAX77+/+ embryos but subsequently degenerate. At E12.5, we found no abnormalities in ocular morphology, retinal cell cycle parameters and the incidence of retinal cell death. From E14.5 on, we observed malformations of the optic disc. From E16.5 into postnatal life there is progressively more severe retinal dysplasia and microphthalmia. Analyses of patterns of gene expression indicated that PAX77+/+ retinae produce a normal range of cell types, including retinal ganglion cells (RGCs). At E14.5 and E16.5, quantitative RT-PCR with probes for a range of molecules associated with retinal development showed only one significant change: a slight reduction in levels of mRNA encoding the secreted morphogen Shh at E16.5. At E16.5, tract-tracing with carbocyanine dyes in PAX77+/+ embryos revealed errors in intraretinal navigation by RGC axons, a decrease in the number of RGC axons reaching the thalamus and an increase in the proportion of ipsilateral projections among those RGC axons that do reach the thalamus. A survey of embryos with different Pax6/PAX6 gene dosage (Pax6Sey/+, Pax6+/+, PAX77+ and PAX77+/+) showed that (1) the total number of RGC axons projected by the retina and (2) the proportions that are sorted into the ipsilateral and contralateral optic tracts at the optic chiasm vary differently with gene dosage. Increasing dosage increases the proportion projecting ipsilaterally regardless of the size of the total projection. CONCLUSION: Pax6 overexpression does not obviously impair the initial formation of the eye and its major cell-types but prevents normal development of the retina from about E14.5, leading eventually to severe retinal degeneration in postnatal life. This sequence is different to that underlying microphthalmia in Pax6+/- heterozygotes, which is due primarily to defects in the initial stages of lens formation. Before the onset of severe retinal dysplasia, Pax6 overexpression causes defects of retinal axons, preventing their normal growth and navigation through the optic chiasm.


Subject(s)
Eye Proteins/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Microphthalmos/genetics , Paired Box Transcription Factors/genetics , Repressor Proteins/genetics , Retinal Dysplasia/genetics , Retinal Ganglion Cells/metabolism , Animals , Axons , Cell Count , Embryo, Mammalian , Gene Dosage , Humans , Immunohistochemistry , Mice , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmos/embryology , Morphogenesis , Optic Chiasm/embryology , PAX6 Transcription Factor , Retinal Dysplasia/embryology , Reverse Transcriptase Polymerase Chain Reaction
13.
Development ; 134(3): 545-55, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17202185

ABSTRACT

Levels of expression of the transcription factor Pax6 vary throughout corticogenesis in a rostro-lateral(high) to caudo-medial(low) gradient across the cortical proliferative zone. Previous loss-of-function studies have indicated that Pax6 is required for normal cortical progenitor proliferation, neuronal differentiation, cortical lamination and cortical arealization, but whether and how its level of expression affects its function is unclear. We studied the developing cortex of PAX77 YAC transgenic mice carrying several copies of the human PAX6 locus with its full complement of regulatory regions. We found that PAX77 embryos express Pax6 in a normal spatial pattern, with levels up to three times higher than wild type. By crossing PAX77 mice with a new YAC transgenic line that reports Pax6 expression (DTy54), we showed that increased expression is limited by negative autoregulation. Increased expression reduces proliferation of late cortical progenitors specifically, and analysis of PAX77<---->wild-type chimeras indicates that the defect is cell autonomous. We analyzed cortical arealization in PAX77 mice and found that, whereas the loss of Pax6 shifts caudal cortical areas rostrally, Pax6 overexpression at levels predicted to shift rostral areas caudally has very little effect. These findings indicate that Pax6 levels are stabilized by autoregulation, that the proliferation of cortical progenitors is sensitive to altered Pax6 levels and that cortical arealization is not.


Subject(s)
Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Eye Proteins/genetics , Homeodomain Proteins/genetics , Paired Box Transcription Factors/genetics , Repressor Proteins/genetics , Animals , Base Sequence , Cell Proliferation , Cerebral Cortex/cytology , DNA Primers/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Eye Proteins/metabolism , Gene Dosage , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Homeostasis , Humans , Mice , Mice, Transgenic , Models, Neurological , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/metabolism
14.
Brain Res Bull ; 66(4-6): 387-93, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16144620

ABSTRACT

Pax6 is a highly conserved transcription factor essential for the development of the eyes in vertebrate and invertebrate species. It is also required for normal development of many regions of the central nervous system, including the mammalian forebrain, hindbrain and spinal cord. In the forebrain, it is expressed in a gradient in the dorsal telencephalon, where it is required for the expression of genes that confer dorsal characteristics and where it might play a role in regionalization of the cerebral cortex. It is expressed in the diencephalon, where it is essential for the specification of its derivatives. While the ancestral function of Pax6 may have been to specify a structure sensitive to light, it has been co-opted into the regulation of a broader range of processes in development of the vertebrate nervous system.


Subject(s)
Eye Proteins/physiology , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/physiology , Paired Box Transcription Factors/physiology , Prosencephalon/physiology , Repressor Proteins/physiology , Animals , Humans , PAX6 Transcription Factor , Prosencephalon/anatomy & histology
15.
Eur J Biochem ; 269(10): 2527-37, 2002 May.
Article in English | MEDLINE | ID: mdl-12027891

ABSTRACT

Heat shock transcription factors (HSFs) are characterized by their ability, upon activation, to bind to heat shock response elements (HSE) present in the promoter of their target genes. HSE are composed of inverted repeats of the pentamer nGAAm. In this study, we compare the embryonic HSF2 protein, purified from F9 embryonal carcinoma cells tumor, and the in vitro synthesized HSF2. We show that the context of HSF2 synthesis influences its thermosensitivity and DNA-binding properties. Therefore, we determined the consensus binding sequence for the purified embryonic HSF2 by the technique of systematic evolution of ligands by exponential enrichment (SELEX). We show that embryonic HSF2 prefers sites containing three or four nGAAm inverted pentamers and that its optimal binding sequence contains the 8-mer palindromic core 5'-TTCTAGAA-3'. The consensus binding sequence for the embryonic HSF2 will be very helpful to identify new targets for this factor, during developmental and differentiation processes.


Subject(s)
Heat-Shock Proteins/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , Cloning, Molecular , Consensus Sequence , DNA/metabolism , Gene Library , Heat-Shock Proteins/genetics , Heat-Shock Proteins/isolation & purification , Hot Temperature , Mice , Protein Binding , Transcription Factors/genetics , Transcription Factors/isolation & purification , Tumor Cells, Cultured
16.
EMBO J ; 21(11): 2591-601, 2002 Jun 03.
Article in English | MEDLINE | ID: mdl-12032072

ABSTRACT

Heat shock factor 2, one of the four vertebrate HSFs, transcriptional regulators of heat shock gene expression, is active during embryogenesis and spermatogenesis, with unknown functions and targets. By disrupting the Hsf2 gene, we show that, although the lack of HSF2 is not embryonic lethal, Hsf2(-/-) mice suffer from brain abnormalities, and meiotic and gameto genesis defects in both genders. The disturbances in brain are characterized by the enlargement of lateral and third ventricles and the reduction of hippocampus and striatum, in correlation with HSF2 expression in proliferative cells of the neuroepithelium and in some ependymal cells in adults. Many developing spermatocytes are eliminated via apoptosis in a stage-specific manner in Hsf2(-/-) males, and pachytene spermatocytes also display structural defects in the synaptonemal complexes between homologous chromosomes. Hsf2(-/-) females suffer from multiple fertility defects: the production of abnormal eggs, the reduction in ovarian follicle number and the presence of hemorrhagic cystic follicles are consistent with meiotic defects. Hsf2(-/-) females also display hormone response defects, that can be rescued by superovulation treatment, and exhibit abnormal rates of luteinizing hormone receptor mRNAs.


Subject(s)
Brain/abnormalities , Brain/metabolism , Chromosomes/ultrastructure , Heat-Shock Proteins/genetics , Infertility, Female/genetics , Meiosis , Transcription Factors/genetics , Alleles , Animals , Apoptosis , Blotting, Western , Embryo, Mammalian/metabolism , Female , Fertility/genetics , Genetic Vectors , Genotype , Heterozygote , Immunohistochemistry , Lac Operon , Male , Mice , Mice, Transgenic , Microscopy, Fluorescence , Models, Genetic , Ovary/metabolism , Promoter Regions, Genetic , Recombination, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors , Testis/metabolism , Time Factors , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...