Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Neurosci ; 38(42): 9105-9121, 2018 10 17.
Article in English | MEDLINE | ID: mdl-30143575

ABSTRACT

Specification of dorsoventral regional identity in progenitors of the developing telencephalon is a first pivotal step in the development of the cerebral cortex and basal ganglia. Previously, we demonstrated that the two zinc finger doublesex and mab-3 related (Dmrt) genes, Dmrt5 (Dmrta2) and Dmrt3, which are coexpressed in high caudomedial to low rostrolateral gradients in the cerebral cortical primordium, are separately needed for normal formation of the cortical hem, hippocampus, and caudomedial neocortex. We have now addressed the role of Dmrt3 and Dmrt5 in controlling dorsoventral division of the telencephalon in mice of either sex by comparing the phenotypes of single knock-out (KO) with double KO embryos and by misexpressing Dmrt5 in the ventral telencephalon. We find that DMRT3 and DMRT5 act as critical regulators of progenitor cell dorsoventral identity by repressing ventralizing regulators. Early ventral fate transcriptional regulators expressed in the dorsal lateral ganglionic eminence, such as Gsx2, are upregulated in the dorsal telencephalon of Dmrt3;Dmrt5 double KO embryos and downregulated when ventral telencephalic progenitors express ectopic Dmrt5 Conditional overexpression of Dmrt5 throughout the telencephalon produces gene expression and structural defects that are highly consistent with reduced GSX2 activity. Further, Emx2;Dmrt5 double KO embryos show a phenotype similar to Dmrt3;Dmrt5 double KO embryos, and both DMRT3, DMRT5 and the homeobox transcription factor EMX2 bind to a ventral telencephalon-specific enhancer in the Gsx2 locus. Together, our findings uncover cooperative functions of DMRT3, DMRT5, and EMX2 in dividing dorsal from ventral in the telencephalon.SIGNIFICANCE STATEMENT We identified the DMRT3 and DMRT5 zinc finger transcription factors as novel regulators of dorsoventral patterning in the telencephalon. Our data indicate that they have overlapping functions and compensate for one another. The double, but not the single, knock-out produces a dorsal telencephalon that is ventralized, and olfactory bulb tissue takes over most remaining cortex. Conversely, overexpressing Dmrt5 throughout the telencephalon causes expanded expression of dorsal gene determinants and smaller olfactory bulbs. Furthermore, we show that the homeobox transcription factor EMX2 that is coexpressed with DMRT3 and DMRT5 in cortical progenitors cooperates with them to maintain dorsoventral patterning in the telencephalon. Our study suggests that DMRT3/5 function with EMX2 in positioning the pallial-subpallial boundary by antagonizing the ventral homeobox transcription factor GSX2.


Subject(s)
Homeodomain Proteins/physiology , Neural Stem Cells/physiology , Neurons/physiology , Telencephalon/embryology , Transcription Factors/physiology , Animals , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/metabolism , Neurons/metabolism , Telencephalon/metabolism , Transcription Factors/genetics
2.
Development ; 142(19): 3416-28, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26443638

ABSTRACT

V1 interneurons are inhibitory neurons that play an essential role in vertebrate locomotion. The molecular mechanisms underlying their genesis remain, however, largely undefined. Here, we show that the transcription factor Prdm12 is selectively expressed in p1 progenitors of the hindbrain and spinal cord in the frog embryo, and that a similar restricted expression profile is observed in the nerve cord of other vertebrates as well as of the cephalochordate amphioxus. Using frog, chick and mice, we analyzed the regulation of Prdm12 and found that its expression in the caudal neural tube is dependent on retinoic acid and Pax6, and that it is restricted to p1 progenitors, due to the repressive action of Dbx1 and Nkx6-1/2 expressed in the adjacent p0 and p2 domains. Functional studies in the frog, including genome-wide identification of its targets by RNA-seq and ChIP-Seq, reveal that vertebrate Prdm12 proteins act as a general determinant of V1 cell fate, at least in part, by directly repressing Dbx1 and Nkx6 genes. This probably occurs by recruiting the methyltransferase G9a, an activity that is not displayed by the amphioxus Prdm12 protein. Together, these findings indicate that Prdm12 promotes V1 interneurons through cross-repressive interactions with Dbx1 and Nkx6 genes, and suggest that this function might have only been acquired after the split of the vertebrate and cephalochordate lineages.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation, Developmental/physiology , Morphogenesis/physiology , Nerve Tissue Proteins/metabolism , Renshaw Cells/physiology , Xenopus/embryology , Animals , Base Sequence , Chick Embryo , Chromatin Immunoprecipitation , Computational Biology , DNA Primers/genetics , DNA, Complementary/genetics , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Mice , Molecular Sequence Data , Rhombencephalon/metabolism , Sequence Analysis, RNA , Species Specificity , Spinal Cord/metabolism
3.
Dev Biol ; 386(2): 340-57, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24370451

ABSTRACT

The basic helix-loop-helix (bHLH) transcriptional activator Ptf1a determines inhibitory GABAergic over excitatory glutamatergic neuronal cell fate in progenitors of the vertebrate dorsal spinal cord, cerebellum and retina. In an in situ hybridization expression survey of PR domain containing genes encoding putative chromatin-remodeling zinc finger transcription factors in Xenopus embryos, we identified Prdm13 as a histone methyltransferase belonging to the Ptf1a synexpression group. Gain and loss of Ptf1a function analyses in both frog and mice indicates that Prdm13 is positively regulated by Ptf1a and likely constitutes a direct transcriptional target. We also showed that this regulation requires the formation of the Ptf1a-Rbp-j complex. Prdm13 knockdown in Xenopus embryos and in Ptf1a overexpressing ectodermal explants lead to an upregulation of Tlx3/Hox11L2, which specifies a glutamatergic lineage and a reduction of the GABAergic neuronal marker Pax2. It also leads to an upregulation of Prdm13 transcription, suggesting an autonegative regulation. Conversely, in animal caps, Prdm13 blocks the ability of the bHLH factor Neurog2 to activate Tlx3. Additional gain of function experiments in the chick neural tube confirm that Prdm13 suppresses Tlx3(+)/glutamatergic and induces Pax2(+)/GABAergic neuronal fate. Thus, Prdm13 is a novel crucial component of the Ptf1a regulatory pathway that, by modulating the transcriptional activity of bHLH factors such as Neurog2, controls the balance between GABAergic and glutamatergic neuronal fate in the dorsal and caudal part of the vertebrate neural tube.


Subject(s)
Cell Differentiation/physiology , GABAergic Neurons/physiology , Gene Expression Regulation, Developmental/physiology , Histone-Lysine N-Methyltransferase/metabolism , Neural Tube/embryology , Xenopus Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chick Embryo , DNA Primers/genetics , Electroporation , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Immunohistochemistry , Immunoprecipitation , In Situ Hybridization , Mice , Neural Tube/cytology , PAX2 Transcription Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Xenopus Proteins/genetics , Xenopus laevis
4.
Dev Biol ; 373(1): 39-52, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23064029

ABSTRACT

The Dmrt (doublesex and mab-3 related transcription factor) genes encode a large family of evolutionarily conserved transcription factors whose function in sex specific differentiation has been well studied in all animal lineages. In vertebrates, their function is not restricted to the developing gonads. For example, Xenopus Dmrt4 is essential for neurogenesis in the olfactory system. Here we have isolated and characterized Xenopus Dmrt5 and found that it is coexpressed with Dmrt4 in the developing olfactory placodes. As Dmrt4, Dmrt5 is positively regulated in the ectoderm by neural inducers and negatively by proneural factors. Both Dmrt5 and Dmrt4 genes are also activated by the combined action of the transcription factor Otx2, broadly transcribed in the head ectoderm and of Notch signaling, activated in the anterior neural ridge. As for Dmrt4, knockdown of Dmrt5 impairs neurogenesis in the embryonic olfactory system and in neuralized animal caps. Conversely, its overexpression promotes neuronal differentiation in animal caps, a property that requires the conserved C-terminal DMA and DMB domains. We also found that the sea anenome Dmrt4/5 related gene NvDmrtb also induces neurogenesis in Xenopus animal caps and that conversely, its knockdown in Nematostella reduces elav-1 positive neurons. Together, our data identify Dmrt5 as a novel important regulator of neurogenesis whose function overlaps with that of Dmrt4 during Xenopus olfactory system development. They also suggest that Dmrt may have had a role in neurogenesis in the last common ancestor of cnidarians and bilaterians.


Subject(s)
Neurogenesis/physiology , Olfactory Mucosa/embryology , Transcription Factors/metabolism , Xenopus Proteins/metabolism , Xenopus/embryology , Animals , COS Cells , Chlorocebus aethiops , DNA Primers/genetics , DNA, Complementary/genetics , Electrophoretic Mobility Shift Assay , Gene Knockdown Techniques , In Situ Nick-End Labeling , Otx Transcription Factors/metabolism , Plasmids/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sea Anemones/genetics , Species Specificity , Transcription Factors/genetics , Transcription Factors/physiology , Xenopus/genetics , Xenopus Proteins/genetics , Xenopus Proteins/physiology
5.
Cereb Cortex ; 23(11): 2552-67, 2013 Nov.
Article in English | MEDLINE | ID: mdl-22923088

ABSTRACT

Regional patterning of the cerebral cortex is initiated by morphogens secreted by patterning centers that establish graded expression of transcription factors within cortical progenitors. Here, we show that Dmrt5 is expressed in cortical progenitors in a high-caudomedial to low-rostrolateral gradient. In its absence, the cortex is strongly reduced and exhibits severe abnormalities, including agenesis of the hippocampus and choroid plexus and defects in commissural and thalamocortical tracts. Loss of Dmrt5 results in decreased Wnt and Bmp in one of the major telencephalic patterning centers, the dorsomedial telencephalon, and in a reduction of Cajal-Retzius cells. Expression of the dorsal midline signaling center-dependent transcription factors is downregulated, including Emx2, which promotes caudomedial fates, while the rostral determinant Pax6, which is inhibited by midline signals, is upregulated. Consistently, Dmrt5(-/-) brains exhibit patterning defects with a dramatic reduction of the caudomedial cortex. Dmrt5 is increased upon the activation of Wnt signaling and downregulated in Gli3(xt/xt) mutants. We conclude that Dmrt5 is a novel Wnt-dependent transcription factor required for early cortical development and that it may regulate initial cortical patterning by promoting dorsal midline signaling center formation and thereby helping to establish the graded expression of the other transcription regulators of cortical identity.


Subject(s)
Cerebral Cortex/embryology , Transcription Factors/metabolism , Animals , Bone Morphogenetic Protein Receptors/metabolism , Cerebral Cortex/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Transcription Factors/genetics , Wnt Proteins/metabolism
6.
Neural Dev ; 19(1): 1, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38167468

ABSTRACT

Prdm12 is an epigenetic regulator expressed in developing and mature nociceptive neurons, playing a key role in their specification during neurogenesis and modulating pain sensation at adulthood. In vitro studies suggested that Prdm12 recruits the methyltransferase G9a through its zinc finger domains to regulate target gene expression, but how Prdm12 interacts with G9a and whether G9a plays a role in Prdm12's functional properties in sensory ganglia remain unknown. Here we report that Prdm12-G9a interaction is likely direct and that it involves the SET domain of G9a. We show that both proteins are largely co-expressed in dorsal root ganglia during early murine development, opening the possibility that G9a plays a role in DRG and may act as a mediator of Prdm12's function in the development of nociceptive sensory neurons. To test this hypothesis, we conditionally inactivated G9a in neural crest using a Wnt1-Cre transgenic mouse line. We found that the specific loss of G9a in the neural crest lineage does not lead to dorsal root ganglia hypoplasia due to the loss of somatic nociceptive neurons nor to the ectopic expression of the visceral determinant Phox2b as observed upon Prdm12 ablation. These findings suggest that Prdm12 function in the initiation of the nociceptive lineage does not critically involves its interaction with G9a.


Subject(s)
Neurogenesis , Nociceptors , Mice , Animals , Nociceptors/metabolism , Neurogenesis/physiology , Sensory Receptor Cells , Transcription Factors/genetics , Transcription Factors/metabolism , Ganglia, Spinal , Mice, Transgenic , Carrier Proteins/genetics , Carrier Proteins/metabolism , Nerve Tissue Proteins/metabolism
7.
iScience ; 26(12): 108364, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38025786

ABSTRACT

Prdm12 is a transcriptional regulator essential for the emergence of the somatic nociceptive lineage during sensory neurogenesis. The exact mechanisms by which Prdm12 promotes nociceptor development remain, however, poorly understood. Here, we report that the trigeminal and dorsal root ganglia hypoplasia induced by the loss of Prdm12 involves Bax-dependent apoptosis and that it is accompanied by the ectopic expression of the visceral sensory neuron determinants Phox2a and Phox2b, which is, however, not sufficient to impose a complete fate switch in surviving somatosensory neurons. Mechanistically, our data reveal that Prdm12 is required from somatosensory neural precursors to early post-mitotic differentiating nociceptive neurons to repress Phox2a/b and that its repressive function is context dependent. Together, these findings reveal that besides its essential role in nociceptor survival during development, Prdm12 also promotes nociceptor fate via an additional mechanism, by preventing precursors from engaging into an alternate Phox2 driven visceral neuronal type differentiation program.

8.
Pain ; 163(8): e927-e941, 2022 08 01.
Article in English | MEDLINE | ID: mdl-34961757

ABSTRACT

ABSTRACT: Prdm12 is a conserved epigenetic transcriptional regulator that displays restricted expression in nociceptors of the developing peripheral nervous system. In mice, Prdm12 is required for the development of the entire nociceptive lineage. In humans, PRDM12 mutations cause congenital insensitivity to pain, likely because of the loss of nociceptors. Prdm12 expression is maintained in mature nociceptors suggesting a yet-to-be explored functional role in adults. Using Prdm12 inducible conditional knockout mouse models, we report that in adult nociceptors Prdm12 is no longer required for cell survival but continues to play a role in the transcriptional control of a network of genes, many of them encoding ion channels and receptors. We found that disruption of Prdm12 alters the excitability of dorsal root ganglion neurons in culture. Phenotypically, we observed that mice lacking Prdm12 exhibit normal responses to thermal and mechanical nociceptive stimuli but a reduced response to capsaicin and hypersensitivity to formalin-induced inflammatory pain. Together, our data indicate that Prdm12 regulates pain-related behavior in a complex way by modulating gene expression in adult nociceptors and controlling their excitability. The results encourage further studies to assess the potential of Prdm12 as a target for analgesic development.


Subject(s)
Carrier Proteins , Ganglia, Spinal , Nerve Tissue Proteins , Nociceptors , Animals , Carrier Proteins/genetics , Ganglia, Spinal/metabolism , Gene Expression , Humans , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nociceptors/physiology , Pain/genetics , Pain/metabolism
9.
Cell Rep ; 26(13): 3522-3536.e5, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30917309

ABSTRACT

In humans, many cases of congenital insensitivity to pain (CIP) are caused by mutations of components of the NGF/TrkA signaling pathway, which is required for survival and specification of nociceptors and plays a major role in pain processing. Mutations in PRDM12 have been identified in CIP patients that indicate a putative role for this transcriptional regulator in pain sensing. Here, we show that Prdm12 expression is restricted to developing and adult nociceptors and that its genetic ablation compromises their viability and maturation. Mechanistically, we find that Prdm12 is required for the initiation and maintenance of the expression of TrkA by acting as a modulator of Neurogenin1/2 transcription factor activity, in frogs, mice, and humans. Altogether, our results identify Prdm12 as an evolutionarily conserved key regulator of nociceptor specification and as an actionable target for new pain therapeutics.


Subject(s)
Carrier Proteins/physiology , Nerve Tissue Proteins/physiology , Neurogenesis/physiology , Nociceptors/cytology , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carrier Proteins/genetics , Cell Line , Evolution, Molecular , Female , Ganglia, Sensory/cytology , Gene Knockout Techniques , Human Embryonic Stem Cells , Humans , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Crest/cytology , Nociceptors/metabolism , Receptor, trkA/metabolism , Tretinoin/physiology , Xenopus laevis
10.
Mech Dev ; 110(1-2): 113-24, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11744373

ABSTRACT

The Notch signaling pathway plays an important role in many cell-fate decisions during development. Here we investigate the regulation and function of the conserved gene XNAP, which is a member of the Delta-Notch synexpression group in Xenopus. XNAP encodes a small protein with two C-terminal tandem ankyrin repeats which is expressed in the neurectoderm and in the presomitic mesoderm in a pattern that resembles that of other component of the Notch pathway. When a myc-tag form of XNAP is overexpressed in Xenopus or Hela cells, XNAP protein is detected both in the nucleus and the cytoplasm. In embryos and in animal cap assays, XNAP expression is activated, perhaps directly, by the Notch pathway and this activation appears to be Su(H) dependent. Overexpression of XNAP in embryos decreases Notch signaling, which leads to an increase in the number of primary neurons that form within the domains of the neural plate where neurogenesis normally occurs. In culture Hela cells, XNAP overexpression interferes with ICD activation of a Notch regulated reporter gene. Together, these data indicate that XNAP is a novel target of the Notch pathway that may, in a feedback loop, modulate its activity.


Subject(s)
Membrane Proteins/genetics , Nervous System/embryology , Xenopus Proteins/genetics , Xenopus laevis/embryology , Xenopus laevis/genetics , Amino Acid Sequence , Animals , Ankyrin Repeat , Base Sequence , DNA/genetics , HeLa Cells , Humans , In Situ Hybridization , Mice , Molecular Sequence Data , Nervous System/growth & development , Phenotype , Rats , Receptors, Notch , Sequence Homology, Amino Acid , Signal Transduction , Transfection , Xenopus laevis/growth & development
11.
Dev Dyn ; 238(6): 1379-88, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19347954

ABSTRACT

The precise localization of gene expression within the developing embryo, and how it changes over time, is one of the most important sources of information for elucidating gene function. As a searchable resource, this information has up until now been largely inaccessible to the Xenopus community. Here, we present a new database of Xenopus gene expression patterns, queryable by specific location or region in the embryo. Pattern matching can be driven either from an existing in situ image, or from a user-defined pattern based on development stage schematic diagrams. The data are derived from the work of a group of 21 Xenopus researchers over a period of 4 days. We used a novel, rapid manual annotation tool, XenMARK, which exploits the ability of the human brain to make the necessary distortions in transferring data from the in situ images to the standard schematic geometry. Developmental Dynamics 238:1379-1388, 2009. (c) 2009 Wiley-Liss, Inc.


Subject(s)
Databases, Genetic , Gene Expression Regulation, Developmental , Gene Expression , Xenopus laevis/embryology , Xenopus laevis/genetics , Animals , Humans , Software , Xenopus laevis/anatomy & histology
12.
Dev Dyn ; 237(8): 2147-57, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18627098

ABSTRACT

The evolutionarily conserved IA1 (Insm1) gene is strongly expressed in the developing nervous system. Here, we show that IA1 is expressed during Xenopus laevis embryogenesis in neural plate primary neurons as well as in a population of uncharacterized anteroventral cells that form in front of the cement gland and that we identified as noradrenergic neurons. We also show that the formation of those anteroventral cells is dependent on BMPs and inhibited by Notch and that it is regulated by the transcription factors Xash1, Phox2, and Hand2. Finally, we provide functional evidence suggesting that IA1 may also play a role in their formation. Together, our results reveal that IA1 constitutes a novel player downstream of Xash1 in the formation of a previously unidentified population of Xenopus noradrenergic primary neurons.


Subject(s)
Gene Expression Regulation, Developmental , Nervous System/embryology , Norepinephrine/physiology , Transcription Factors/genetics , Xenopus Proteins/genetics , Xenopus laevis/embryology , Zinc Fingers/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Morphogenetic Proteins/metabolism , Cell Differentiation/physiology , Cloning, Molecular , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Homeodomain Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/physiology , Receptors, Notch/metabolism , Stem Cells/physiology , Transcription Factors/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/genetics
13.
Dev Dyn ; 237(11): 3352-60, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18855900

ABSTRACT

BBP proteins constitute a subclass of CUL3 interacting BTB proteins whose in vivo function remains unknown. Here, we show that the Xenopus BBP gene BTBD6 and the single Drosophila homologue of mammalian BBP genes lute are strongly expressed in the developing nervous system. In Xenopus, BTBD6 expression responds positively to proneural and negatively to neurogenic gene overexpression. Knockdown of BTBD6 in Xenopus or loss of Drosophila lute result in embryos with strong defects in late neuronal markers and strongly reduced and disorganized axons while early neural development is unaffected. XBTBD6 knockdown in Xenopus also affects muscle development. Together, these data indicate that BTBD6/lute is required for proper embryogenesis and plays an essential evolutionary conserved role during neuronal development.


Subject(s)
Carrier Proteins/metabolism , Drosophila Proteins/immunology , Muscle Development/physiology , Muscle Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Xenopus Proteins/metabolism , Animals , Carrier Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Gene Knockdown Techniques , Humans , Muscle Proteins/genetics , Nerve Tissue Proteins/genetics , Nervous System/cytology , Nervous System/embryology , Sequence Homology, Amino Acid , Xenopus Proteins/genetics , Xenopus laevis
14.
Dev Biol ; 306(1): 34-49, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17442301

ABSTRACT

The DNA-binding transcription factor Smad-interacting protein-1 (Sip1) (also named Zfhx1b/ZEB2) plays essential roles in vertebrate embryogenesis. In Xenopus, XSip1 is essential at the gastrula stage for neural tissue formation, but the precise molecular mechanisms that underlie this process have not been fully identified yet. Here we show that XSip1 functions as a transcriptional repressor during neural induction. We observed that constitutive activation of BMP signaling prevents neural induction by XSip1 but not the inhibition of several epidermal genes. We provide evidence that XSip1 binds directly to the BMP4 proximal promoter and modulates its activity. Finally, by deletion and mutational analysis, we show that XSip1 possesses multiple repression domains and that CtBPs contribute to its repression activity. Consistent with this, interference with XCtBP function reduced XSip1 neuralizing activity. These results suggest that Sip1 acts in neural tissue formation through direct repression of BMP4 but that BMP-independent mechanisms are involved as well. Our data also provide the first demonstration of the importance of CtBP binding in Sip1 transcriptional activity in vivo.


Subject(s)
Alcohol Oxidoreductases/metabolism , Bone Morphogenetic Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Nervous System/embryology , Repressor Proteins/metabolism , Xenopus Proteins/metabolism , Xenopus/embryology , Alcohol Oxidoreductases/genetics , Animals , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Ectoderm/metabolism , Epidermis/metabolism , Homeodomain Proteins/genetics , Nervous System/metabolism , Promoter Regions, Genetic , Protein Structure, Tertiary , Repressor Proteins/genetics , Xenopus/anatomy & histology , Xenopus/genetics , Xenopus Proteins/genetics
15.
Dev Genes Evol ; 212(10): 491-5, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12424520

ABSTRACT

We have isolated a Xenopus homologue of the mammalian hairy and Enhancer of split related gene HRT1. XHRT1 expression in late gastrula and early neurula embryos is restricted to two stripes of cells in the medial neural plate and in dorsal endodermal cells. At later stages, XHRT1 is expressed in the floor plate, in hypochord cells and in the somitogenic and anterior presomitic mesoderm. By tailbud stage, XHRT1 is also highly expressed in the dorsal hindbrain, telencephalon and eye vesicles, olfactory placodes, pronephros, branchial arches and tail fin. We also show that XHRT1 expression in medial neural cells is induced by Notch signaling and that there are differences in the way XHRT1 and other H/E(spl) genes are regulated.


Subject(s)
Gene Expression Regulation, Developmental , Transcription Factors/genetics , Xenopus Proteins/genetics , Xenopus laevis/genetics , Amino Acid Sequence , Animals , Gastrula/physiology , Molecular Sequence Data , Sequence Homology, Amino Acid , Tissue Distribution , Transcription Factors/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/embryology
16.
Dev Dyn ; 228(4): 716-25, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14648848

ABSTRACT

Hairy-related transcription factor (HRT/Hey) genes encode a novel subfamily of basic helix-loop-helix (bHLH) transcription factors related to the Drosophila hairy and Enhancer-of-split (E(spl)) and the mammalian HES proteins that function as downstream mediators of Notch signaling. Using the yeast two-hybrid approach, a previously uncharacterized protein was identified in Xenopus that interacts with XHRT1 (originally referred to as bc8), one member of the HRT/Hey subclass. This protein is evolutionarily conserved in chordates. It binds to sequences adjacent to the bHLH domain of XHRT1 known as the Orange domain and has been named bc8 Orange interacting protein (BOIP). BOIP shows a rather uniform subcellular localization and is recruited to the nucleus upon binding to XHRT1. In Xenopus, XBOIP mRNA is detected by RNase protection analysis throughout embryogenesis. In the adult, the strongest expression is detected in testis. In the mouse, high levels of BOIP mRNA are also found in adult testis. No expression is detected in the embryo and in any of the other adult organs tested. In situ hybridization revealed that BOIP transcripts were detected almost exclusively in round spermatids and that this expression overlaps with that of Hey1 (HRT1), which is expressed throughout spermatogenesis. In view of the importance of the Orange domain for HRT/Hey function, the newly identified BOIP proteins may serve as regulators specifically of HRT1/Hey1 activity.


Subject(s)
Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Cycle Proteins/chemistry , DNA, Complementary/metabolism , Gene Expression Regulation, Developmental , Spermatogenesis , Spermatozoa/metabolism , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Cycle Proteins/metabolism , Cloning, Molecular , Conserved Sequence , Embryo, Mammalian/metabolism , Embryo, Nonmammalian , Gene Library , HeLa Cells , Humans , Immunohistochemistry , In Situ Hybridization , Luciferases/metabolism , Male , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , RNA/metabolism , RNA, Messenger/metabolism , Receptors, Notch , Ribonucleases/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Testis/metabolism , Transfection , Two-Hybrid System Techniques , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL