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










Publication year range
1.
Neuroscience ; 277: 250-66, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25020121

ABSTRACT

In the mammalian cerebellum, deep cerebellar nuclear (DCN) cells convey all information from cortical Purkinje cells (PCs) to premotor nuclei and other brain regions. However, how DCN cells integrate inhibitory input from PCs with excitatory inputs from other sources has been difficult to assess, in part due to the large spatial separation between cortical PCs and their target cells in the nuclei. To circumvent this problem we have used a Cre-mediated genetic approach to generate mice in which channelrhodopsin-2 (ChR2), fused with a fluorescent reporter, is selectively expressed by GABAergic neurons, including PCs. In recordings from brain slice preparations from this model, mammalian PCs can be robustly depolarized and discharged by brief photostimulation. In recordings of postsynaptic DCN cells, photostimulation of PC axons induces a strong inhibition that resembles these cells' responses to focal electrical stimulation, but without a requirement for the glutamate receptor blockers typically applied in such experiments. In this optogenetic model, laser pulses as brief as 1 ms can reliably induce an inhibition that shuts down the spontaneous spiking of a DCN cell for ∼50 ms. If bursts of such brief light pulses are delivered, a fixed pattern of bistable bursting emerges. If these pulses are delivered continuously to a spontaneously bistable cell, the immediate response to such photostimulation is inhibitory in the cell's depolarized state and excitatory when the membrane has repolarized; a less regular burst pattern then persists after stimulation has been terminated. These results indicate that the spiking activity of DCN cells can be bidirectionally modulated by the optically activated synaptic inhibition of cortical PCs.


Subject(s)
Cerebellar Nuclei/physiology , Neural Inhibition/physiology , Purkinje Cells/physiology , Synaptic Transmission/physiology , Animals , Cerebellar Nuclei/cytology , Cerebellar Nuclei/drug effects , Channelrhodopsins , Mice, Transgenic , Microscopy, Confocal , Neuronal Plasticity/physiology , Optogenetics , Patch-Clamp Techniques , Tissue Culture Techniques
2.
Mech Dev ; 105(1-2): 129-44, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11429288

ABSTRACT

The vertebrate midbrain consists of dorsal and ventral domains, the tectum and tegmentum, which execute remarkably different developmental programs. Tectal development is characterized by radial migration of differentiating neurons to form a laminar structure, while the tegmentum generates functionally diverse nuclei at characteristic positions along the neural axis. Here we show that neurons appearing early in the development of the tectum are characterized either by the expression of the POU-domain transcription factor Brn3.0, or by members of the Pax and LIM families. Early neurons of the rostral tegmentum co-express Brn3.0 and Lim1/2, and caudal tegmental neurons express Islet1/2. Notochord tissue or Shh-transfected epithelial cells, transplanted to the developing tectum, suppress the development of tectal neurons, and induce the differentiation of multiple tegmental cell types. The distance from the midbrain-hindbrain boundary (MHB) determines the specific markers expressed by the tegmental neurons induced in the tectum, and the transplantation of MHB tissue adjacent to the rostral tegmentum also induces caudal markers, demonstrating the role of MHB signals in determining the phenotype of these early midbrain neurons. Co-culture of isolated midbrain neuroepithelium with Shh-expressing cells demonstrates that Shh is sufficient to convert tectal neurons to a tegmental fate. In mice lacking Shh, Brn3.0- and Pax7-expressing neurons typical of the tectum develop throughout the ventral midbrain, and gene expression patterns characteristic of early tegmental development do not appear.


Subject(s)
Brain/embryology , Brain/physiology , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Mesencephalon/embryology , Signal Transduction , Transcription Factors/biosynthesis , Transcription Factors/genetics , Animals , Cell Differentiation , Cell Line , Cell Movement , Cells, Cultured , Chick Embryo , Coculture Techniques , Hedgehog Proteins , Homeodomain Proteins/biosynthesis , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , Neurons/cytology , Neurons/metabolism , Notochord/metabolism , PAX7 Transcription Factor , Protein Structure, Tertiary , Quail , Time Factors , Trans-Activators/metabolism , Transcription Factor Brn-3 , Transcription Factor Brn-3A , Transfection
3.
Arch Biochem Biophys ; 385(2): 397-405, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11368023

ABSTRACT

The regulator of immunoglobulin expression Oct-2 and the related widely expressed transcription factor Oct-1 have been shown to interact with DNA sequences containing an "octamer" motif, ATGC(A/T)AAT. To better understand Oct-2 function we have used random oligonucleotide selection and competition assays to define the optimal recognition site for this protein. The selected site contains an extended sequence that is remarkably similar to octamer-heptamer sequences found in immunoglobulin heavy-chain regulatory sequences, and the affinity of Oct-2 for this site is at least 50-fold greater than for sites containing the octamer motif alone. Fusion to glutathione S-transferase, a widely used model for protein-DNA and protein-protein interaction, does not alter the optimal Oct-2 recognition site, but inhibits Oct-2 POU-domain dimerization, slows the dissociation rate of the GST-Oct-2/DNA complex, and increases the relative importance of the heptamer domain for Oct-2 binding. These data advance our ability to identify in vivo targets of POU-factor regulation and also suggest that GST-fusion proteins should be used with caution in DNA-binding studies.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Recombinant Fusion Proteins/metabolism , Response Elements/genetics , Transcription Factors/metabolism , Amino Acid Sequence , Base Sequence , Binding, Competitive , Consensus Sequence/genetics , DNA/genetics , DNA Probes/genetics , DNA Probes/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Dimerization , Humans , Molecular Sequence Data , Octamer Transcription Factor-2 , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Substrate Specificity , Thermodynamics , Transcription Factors/chemistry , Transcription Factors/genetics , Transcriptional Activation
4.
J Neurosci ; 21(2): 541-9, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11160433

ABSTRACT

Brn3a/Brn-3.0 is a POU-domain transcription factor expressed in primary sensory neurons of the cranial and dorsal root ganglia and in specific neurons in the caudal CNS. Mice lacking Brn3a undergo extensive sensory neural death late in gestation and die at birth. To further examine Brn3a expression and the abnormalities that accompany its absence, we constructed a transgene containing 11 kb of Brn3a upstream regulatory sequence linked to a LacZ reporter. Here we show that these regulatory sequences direct transgene expression specifically to Brn3a peripheral sensory neurons of the cranial and dorsal root ganglia. Furthermore, expression of the 11 kb/LacZ reporter in the sensory neurons of the mesencephalic trigeminal, but not other Brn3a midbrain neurons, demonstrates that cell-specific transgene expression is targeted to a functional class of neurons rather than to an anatomical region. We then interbred the 11 kb/LacZ reporter strain with mice carrying a null mutant allele of Brn3a to generate 11 kb/LacZ, Brn3a knock-out mice. beta-Galactosidase expression in these mice reveals significant axonal growth defects, including excessive and premature branching of the major divisions of the trigeminal nerve and a failure to correctly innervate whisker follicles, all of which precede sensory neural death in these mice. These defects in Brn3a(-/-) mice resemble strongly those seen in mice lacking the mediators of sensory pathfinding semaphorin 3A and neuropilin-1. Here we show, however, that sensory neurons are able to express neuropilin-1 in the absence of Brn3a.


Subject(s)
Axons/pathology , DNA-Binding Proteins/deficiency , Neurons, Afferent/pathology , Peripheral Nervous System Diseases/genetics , Transcription Factors/deficiency , Animals , Brain/metabolism , Brain/pathology , Cell Death , Crosses, Genetic , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Ganglia, Spinal/embryology , Ganglia, Spinal/pathology , Genes, Reporter/genetics , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/biosynthesis , Neurons, Afferent/metabolism , Neuropilin-1 , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/pathology , RNA, Messenger/metabolism , Receptor, trkC/deficiency , Receptor, trkC/genetics , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factor Brn-3 , Transcription Factor Brn-3A , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Transgenes/genetics , Vibrissae/innervation , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics
5.
Development ; 126(16): 3545-59, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10409501

ABSTRACT

Sensory and autonomic neurons of the vertebrate peripheral nervous system are derived from the neural crest. Here we use the expression of lineage-specific transcription factors as a means to identify neuronal subtypes that develop in rat neural crest cultures grown in a defined medium. Sensory neurons, identified by expression of the POU-domain transcription factor Brn-3.0, develop from dividing precursors that differentiate within 2 days following emigration from the neural tube. Most of these precursors generate sensory neurons even when challenged with BMP2, a factor that induces autonomic neurogenesis in many other cells in the explants. Moreover, BMP2 fails to prevent expression of the sensory-specific basic helix-loop-helix (bHLH) transcription factors neurogenin1, neurogenin2 and neuroD, although it induces expression of the autonomic-specific bHLH factor MASH1 and the paired homeodomain factor Phox2a in other cells. These data suggest that there are mitotically active precursors in the mammalian neural crest that can generate sensory neurons even in the presence of a strong autonomic-inducing cue. Further characterization of the neurons generated from such precursors indicates that, under these culture conditions, they exhibit a proprioceptive and/or mechanosensory, but not nociceptive, phenotype. Such precursors may therefore correspond to a lineally (Frank, E. and Sanes, J. (1991) Development 111, 895-908) and genetically (Ma, Q., Fode, C., Guillemot, F. and Anderson, D. J. (1999) Genes Dev. 13, in press) distinct subset of early-differentiating precursors of large-diameter sensory neurons identified in vivo.


Subject(s)
Membrane Glycoproteins , Neural Crest/cytology , Neurons, Afferent/cytology , Stem Cells/cytology , Transcription Factors/analysis , Transforming Growth Factor beta , Animals , Biomarkers , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/pharmacology , Cell Differentiation/drug effects , Cell Movement , Cells, Cultured , Gestational Age , Helix-Loop-Helix Motifs , Homeodomain Proteins/analysis , Intermediate Filament Proteins/analysis , Mitosis , Nerve Tissue Proteins/analysis , Neural Crest/physiology , Neurons, Afferent/physiology , Organ Culture Techniques , Peripherins , Rats , Rats, Sprague-Dawley , Stem Cells/physiology , Transcription Factors/genetics
6.
J Neurosci ; 19(15): 6549-58, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10414983

ABSTRACT

The POU-IV or Brn-3 class of transcription factors exhibit conserved structure, DNA-binding properties, and expression in specific subclasses of neurons across widely diverged species. In the mouse CNS, Brn-3.0 expression characterizes specific neurons from neurogenesis through the life of the cell. This irreversible activation of expression suggests positive autoregulation. To search for cis-acting elements that could mediate autoregulation we used a novel method, complex stability screening, which we applied to rapidly identify functional Brn-3.0 recognition sites within a large genomic region encompassing the mouse brn-3.0 locus. This method is based on the observation that the kinetic stability of Brn-3.0 complexes with specific DNA sequences, as measured by their dissociation half-lives, is highly correlated with the ability of those sequences to mediate transcriptional activation by Brn-3.0. The principal Brn-3.0 autoregulatory region lies approximately 5 kb upstream from the Brn-3.0 transcription start site and contains multiple Brn-3.0-binding sites that strongly resemble the optimal binding site for this protein class. This region also mediates transactivation by the closely related protein Brn-3.2, suggesting a regulatory cascade of POU proteins in specific neurons in which Brn-3.2 expression precedes Brn-3.0.


Subject(s)
Chromosome Mapping/methods , DNA-Binding Proteins/genetics , Genetic Techniques , Homeodomain Proteins , Homeostasis/physiology , Transcription Factors/genetics , Aging/metabolism , Animals , DNA/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Drug Stability , Embryo, Mammalian/metabolism , Embryo, Mammalian/physiology , Embryonic and Fetal Development/physiology , Genome , Immunohistochemistry , Mice , Time Factors , Transcription Factor Brn-3 , Transcription Factor Brn-3A , Transcription Factor Brn-3B , Transcription Factors/metabolism , Transcription Factors/physiology , Transcriptional Activation/physiology
7.
J Biol Chem ; 273(51): 34196-205, 1998 Dec 18.
Article in English | MEDLINE | ID: mdl-9852081

ABSTRACT

POU-domain proteins have been shown to play important roles in the development of the nervous, endocrine, and immune systems. However, the distinctive DNA recognition properties of the six major POU subclasses have not been well defined. Here, we have used random oligonucleotide selection and competitive binding assays to determine the optimal DNA recognition elements for the POU-III and POU-VI protein classes, represented by Brn-2 and Brn-5, respectively. The optimal Brn-5 consensus binding sequence GCATAA(T/A)TTAT strongly resembles that previously determined for the POU-IV (Brn-3) class, whereas Brn-2 exhibits highest affinity for non-octamer sites of the form ATG(A/C)AT(A/T)0-2ATTNAT and for octamer sites that contain a full associated heptamer sequence. Brn-2, Brn-3.0, and their invertebrate homologues all exhibit highly cooperative homodimerization on the Brn-2 consensus sequence, demonstrating that cooperative dimerization is a general property of these neural POU proteins. However, modified sites to which Brn-2 binds only as a monomer mediate the transcriptional effects of Brn-2 better than the consensus sequence, demonstrating that dimerization on these sites diminishes the transactivation ability of the protein. Together with the findings of our prior studies these data greatly facilitate the identification of functional POU recognition elements in the regulatory regions of neural genes.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Oligodeoxyribonucleotides/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Animals , Base Sequence , Binding, Competitive , Consensus Sequence , Dimerization , Homeodomain Proteins , Kinetics , Mice , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Oligodeoxyribonucleotides/chemistry , POU Domain Factors , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Substrate Specificity , Transcription Factor Brn-3 , Transcription Factor Brn-3A
8.
Dev Biol ; 190(1): 18-31, 1997 Oct 01.
Article in English | MEDLINE | ID: mdl-9331328

ABSTRACT

Brn-3.0, a POU-domain transcription factor, is expressed in specific postmitotic neurons in the dorsal part of the neural tube which are among the first spinal cord neurons to appear in development. In the mature spinal cord, the Brn-3.0 cells form a numerous population of scattered neurons in the intermediate spinal gray. Ablation of the notochord in chick embryos extends the domain of Brn-3.0 expression into the ventral neural tube, while ectopic grafts of notochord tissue suppress Brn-3.0 expression. The notochord effects on Brn-3.0 expression are reproduced in vivo by the implantation of a local source of recombinant Shh protein. The down-regulation of Brn-3.0 expression in the dorsal spinal cord by the notochord and Shh contrasts with the known inductive effects of these ventral signals on the approximately simultaneous development of the spinal motor neurons. In cultured explants of neural plate from the region of the presumptive spinal cord, Brn-3.0 neurons develop in the absence of surface ectoderm and ventral midline tissue, suggesting that the Brn-3.0 phenotype may represent a "default" developmental pathway for early spinal cord neurons. Together these results advance the understanding of the mechanism of the generation of neuronal diversity in the developing vertebrate CNS.


Subject(s)
Body Patterning/physiology , DNA-Binding Proteins/analysis , Neurons/cytology , Signal Transduction/physiology , Spinal Cord/embryology , Trans-Activators , Transcription Factors/analysis , Animals , Biomarkers , Cell Differentiation , Central Nervous System/cytology , Central Nervous System/embryology , Chick Embryo , Culture Techniques , Epithelial Cells , Gene Expression Regulation, Developmental , Hedgehog Proteins , Homeodomain Proteins/analysis , LIM-Homeodomain Proteins , MSX1 Transcription Factor , Motor Neurons/chemistry , Motor Neurons/cytology , Nerve Tissue Proteins/analysis , Neurons/chemistry , Notochord/physiology , Notochord/transplantation , Proteins/pharmacology , Recombinant Fusion Proteins/pharmacology , Spinal Cord/chemistry , Spinal Cord/cytology , Transcription Factor Brn-3
9.
Nucleic Acids Res ; 25(13): 2589-94, 1997 Jul 01.
Article in English | MEDLINE | ID: mdl-9185568

ABSTRACT

The restriction of herpes virus latency to mammalian sensory ganglia has led to a search for tissue-specific regulatory molecules in these neurons which alter viral gene expression. We have recently shown that the POU-domain transcriptional regulator Brn-3.0 is abundantly expressed in the adult trigeminal ganglion. To begin to examine the hypothesis that Brn-3.0 might participate in the regulation of the HSV life-cycle, we used Brn-3.0 POU-domain protein as an affinity matrix, and biochemically screened the entire HSV genome for sites of Brn-3.0 binding. This screen identified several sites of the form TA/TA A T N A N TA/T, which significantly do not include the previously identified HSV octamer sequences. All of the selected sites occur in the <25% of the HSV genome which has not been assigned to open reading frames, suggesting that these sites may be transcriptional regulatory elements recognized by Brn-3.0 or another homeobox factor with similar DNA binding properties. However, these sites do not interact with Brn-3.0 with sufficiently high affinity to directly mediate transcriptional activation by Brn-3.0 alone in transfection assays. The experiments described also provide an effective general method for exhaustive screening of large viral genomes or sub-genomic fragments of eukaryotic DNA for sites of interaction with specific transcription factors.


Subject(s)
DNA, Viral/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Simplexvirus/genetics , Transcription Factors/chemistry , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites , Binding, Competitive , DNA, Viral/chemistry , Glutathione Transferase/genetics , Herpesvirus 1, Human/genetics , Humans , Mice , Octamer Transcription Factor-2 , Recombinant Fusion Proteins/metabolism , Transcription Factor Brn-3 , Transcription Factor Brn-3A
10.
Mol Cell Biol ; 17(5): 2391-400, 1997 May.
Article in English | MEDLINE | ID: mdl-9111308

ABSTRACT

To better understand the diversity of function within the POU domain class of transcriptional regulators, we have determined the optimal DNA recognition site of several proteins of the POU-IV (Brn-3) subclass by random oligonucleotide selection. The consensus recognition element derived in this study, ATAATTAAT, is clearly distinct from octamer sites described for the POU factor Oct-1. The optimal POU-IV site determined here also binds Brn-3.0 with significantly higher affinity than consensus recognition sites previously proposed for this POU subclass. The binding affinity of Brn-3.0 on its optimal site, several variants of this site, and several naturally occurring POU recognition elements is highly correlated with the activation of reporter gene expression by Brn-3.0 in transfection assays. The preferred DNA recognition site of Brn-3.0 resembles strongly the optimal sites of another mammalian POU-IV class protein, Brn-3.2, and of the Caenorhabditis elegans Brn-3.0 homolog Unc-86, demonstrating that the site-specific DNA recognition properties of these factors are highly conserved between widely divergent species.


Subject(s)
Caenorhabditis elegans Proteins , DNA-Binding Proteins/metabolism , Evolution, Molecular , Transcription Factors/metabolism , Amino Acid Sequence , Base Sequence , Binding Sites , Conserved Sequence , DNA/metabolism , DNA-Binding Proteins/chemistry , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Host Cell Factor C1 , Molecular Sequence Data , Mutagenesis, Site-Directed , Octamer Transcription Factor-1 , POU Domain Factors , Transcription Factor Brn-3 , Transcription Factors/chemistry , Transcription, Genetic
11.
Schizophr Res ; 27(2-3): 169-80, 1997 Oct 30.
Article in English | MEDLINE | ID: mdl-9416646

ABSTRACT

Schizophrenia is in essence a developmental disorder, but an unusual one in that the onset of symptoms is markedly delayed. Neuropathologic studies of the brain in schizophrenia have revealed subtle abnormalities that may reflect abnormal neuronal development. A more detailed examination of the cellular and molecular pathology of schizophrenia has been limited by the lack of informative markers that might allow a more complete understanding of the brain defects that characterize this disorder. Recent advances in molecular biology have made available a growing number of probes for examining the expression of specific gene products in brain tissue by using in situ hybridization and immunohistochemistry using antibodies to recombinant antigens. Several recently cloned neural genes are expressed in the forebrain regions which have been implicated in schizophrenia, and may have significant roles in brain development or function. Selected neurotransmitter receptors, neurotrophins and their receptors, and transcription factors of the POU and MADS families are promising candidates for future studies of the cellular and molecular neuropathology of schizophrenia.


Subject(s)
Brain/pathology , Schizophrenia/pathology , Biomarkers , Brain/physiopathology , Dopamine/physiology , Glutamic Acid/physiology , Humans , Nerve Growth Factors/physiology , Neurotransmitter Agents/physiology , Neurotrophin 3 , Schizophrenia/physiopathology , Transcription Factors/physiology , gamma-Aminobutyric Acid/physiology
12.
Proc Natl Acad Sci U S A ; 93(26): 15097-101, 1996 Dec 24.
Article in English | MEDLINE | ID: mdl-8986770

ABSTRACT

The POU-IV or "Brn-3" class of POU-domain transcription factors is represented in Drosophila by I-POU and twin-of-I-POU, alternative splice products of the I-POU gene. I-POU has been previously reported to inhibit DNA binding by the POU-III class factor drifter/Cf1a via the formation of heterodimeric complexes. Here we report that expression of the I-POU/tI-POU message is maximal late in the embryonic phase of Drosophila development, and I-POU is the preferred splice variant. Although I-POU lacks two basic amino acid residues in the POU-homeodomain found in tI-POU and Brn-3.0, these three POU-IV class proteins exhibit very similar DNA-binding specificity. In contrast to previously published reports, the results presented here show no effect of I-POU on DNA binding by drifter, and no evidence for I-POU/drifter dimerization. These results suggest that the I-POU/tI-POU gene products function by transcriptional mechanisms similar to those of the homologous POU-IV class factors expressed in other species, not by unique inhibitory mechanism.


Subject(s)
DNA-Binding Proteins , DNA/metabolism , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Nuclear Proteins , Transcription Factors/chemistry , Transcription Factors/metabolism , Alternative Splicing , Amino Acid Sequence , Animals , Antennapedia Homeodomain Protein , Base Sequence , Binding Sites , Conserved Sequence , DNA/chemistry , DNA Primers , Drosophila , Drosophila Proteins , Exons , Homeodomain Proteins/biosynthesis , Host Cell Factor C1 , Insect Hormones/chemistry , Introns , Mammals , Molecular Sequence Data , Octamer Transcription Factor-1 , Oligodeoxyribonucleotides , Protein Structure, Secondary , Sequence Homology, Amino Acid , Transcription Factors/biosynthesis
13.
Neuroreport ; 7(18): 2829-32, 1996 Nov 25.
Article in English | MEDLINE | ID: mdl-9116190

ABSTRACT

The restriction of herpes virus latency to mammalian sensory ganglia has led to a search for tissue-specific regulatory molecules in these neurons which alter viral gene expression. Based primarily on work in sensory-related cell lines, several prior reports have suggested that the POU-domain transcription factor Oct-2 functions in the establishment of viral latency. Here we show that among the POU-domain transcriptional regulators, Brn-3.0 is abundantly expressed in the postnatal trigeminal ganglion. In contrast, Oct-2 message and protein are not detectable by sensitive and specific assays, effectively excluding a direct role for Oct-2 in the viral life cycle in vivo.


Subject(s)
DNA-Binding Proteins/biosynthesis , Gene Expression Regulation, Viral/physiology , Herpesviridae/genetics , Transcription Factors/biosynthesis , Trigeminal Ganglion/metabolism , Animals , DNA-Binding Proteins/genetics , Immunohistochemistry , Mice , Mice, Inbred C57BL , Octamer Transcription Factor-2 , POU Domain Factors , RNA, Messenger/biosynthesis , Ribonucleases/metabolism , Transcription Factor Brn-3 , Transcription Factor Brn-3A , Transcription Factors/genetics
14.
Mech Dev ; 53(3): 291-304, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8645597

ABSTRACT

The mammalian POU-domain factor Brn-3.0 (Brn-3, Brn-3a) is a member of the POU-IV class of transcription factors which resemble the C. elegans factor unc-86 in structure, DNA-binding properties and expression in subsets of sensory neurons. Using specific antisera, we have explored the expression of Brn-3.0 in the early development of the mouse nervous system. Brn-3.0 expression begins at embryonic day 8.5 (E8.5) in a specific set of midbrain tectal neurons whose time and place of appearance are consistent with the earliest CNS neurons previously identified using non-specific markers of neural differentiation. By E9.5, Brn-3.0 immunoreactivity also identifies early CNS neurons in the hindbrain and spinal cord. In the peripheral sensory ganglia, Brn-3.0 expression is first observed at E9.0 in migrating precursors of the trigeminal ganglion, followed by the other sensory cranial and dorsal root ganglia, in a rostral to caudal sequence. Double-label immunofluorescence with Brn-3.0 and the markers of cell division PCNA and BrdU demonstrate that Brn-3.0 is restricted to the post-mitotic phase of CNS development. In the sensory cranial and dorsal root ganglia, however, Brn-3.0 is expressed in dividing neural precursors, suggesting that the nature or timing of developmental events controlled by Brn-3.0 are distinct in the CNS and peripheral neurons. Restriction of Brn-3.0 expression to post-mitotic CNS neurons demonstrates that Brn-3.0 is not required for neurogenesis or patterning of the neuroepithelium in the CNS, but suggests a role in specification of mature neuronal phenotypes.


Subject(s)
Central Nervous System/embryology , DNA-Binding Proteins/analysis , Mitosis/physiology , Nerve Tissue Proteins/analysis , Neurons, Afferent/chemistry , Stem Cells/chemistry , Transcription Factors/analysis , Animals , Base Sequence , Cell Differentiation/physiology , Central Nervous System/chemistry , Central Nervous System/cytology , Ganglia, Sensory/chemistry , Ganglia, Sensory/embryology , Gestational Age , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Neurons, Afferent/cytology , Phenotype , Proliferating Cell Nuclear Antigen/analysis , Stem Cells/cytology , Transcription Factor Brn-3 , Transcription Factor Brn-3A
15.
Neuron ; 12(1): 205-18, 1994 Jan.
Article in English | MEDLINE | ID: mdl-7904822

ABSTRACT

The identification and molecular characterization of Brn-3.2 has revealed a family of Brn-3-related mammalian POU proteins that share homology with the C. elegans developmental regulator Unc-86 and extended similarity with the Drosophila neurodevelopmental gene I-POU, which defines a novel POU-IV box. Brn-3.2 exhibits DNA binding properties similar to those of Brn-3.0, but its expression is uniquely regulated by retinoic acid in teratocarcinoma and neuroblastoma cells. In the developing PNS and retina, the expression pattern of Brn-3.2 is similar to that of Brn-3.0. In the caudal CNS (spinal cord, hindbrain, and midbrain) Brn-3.2 and Brn-3.0 are initially coexpressed, but diverge later in development. Rostral to the midbrain, Brn-3.2 and Brn-3.0 exhibit nonoverlapping patterns of expression, suggesting divergence of gene function in more recently evolved structures. Our analysis suggests that in the CNS Brn-3.2 is selectively expressed in postmitotic neurons, implying a role in specifying terminally differentiated neuronal phenotypes.


Subject(s)
Brain/metabolism , DNA-Binding Proteins/biosynthesis , Gene Expression Regulation/physiology , Homeodomain Proteins , Neurons/metabolism , Spinal Cord/metabolism , Transcription Factors/biosynthesis , Tretinoin/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Cell Differentiation , Cell Line , DNA, Complementary/metabolism , Drosophila/physiology , Embryo, Mammalian , Embryo, Nonmammalian , Gene Expression/drug effects , Gene Expression Regulation/drug effects , Gene Library , Genes, Homeobox , In Situ Hybridization , Mesencephalon/metabolism , Mice , Mitosis , Molecular Sequence Data , Neuroblastoma , Neurons/cytology , Neurons/drug effects , Organ Specificity , Rhombencephalon/metabolism , Sequence Homology, Amino Acid , Teratoma , Transcription Factor Brn-3 , Transcription Factor Brn-3A , Transcription Factor Brn-3B , Transfection , Tumor Cells, Cultured
16.
Cell ; 68(3): 491-505, 1992 Feb 07.
Article in English | MEDLINE | ID: mdl-1346754

ABSTRACT

I-POU, a POU domain nuclear protein that lacks two conserved basic amino acids of the POU homeodomain is coexpressed in the developing Drosophila nervous system with a second POU domain transcription factor, Cf1-a. I-POU does not bind to DNA but forms a POU domain-mediated, high affinity heterodimer with Cf1-a, inhibiting its ability to bind and activate the dopa decarboxylase gene. The I-POU/Cf1-a dimerization interface encompasses only the N-terminal basic region and helices 1 and 2 of the POU homeodomains with precise amino acid and alpha-helical requirements. twin of I-POU, an alternatively spliced transcript of the I-POU gene, encodes a protein containing the two basic amino acid residues absent in I-POU. Twin of I-POU is incapable of dimerizing with Cf1-a, but can act as a positive transcription factor on targets distinct from those regulated by Cf1-a. These findings suggest that the I-POU genomic locus simultaneously generates both a specific activator and inhibitor of gene transcription, capable of modulating two distinct regulatory programs during neural development.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins , Drosophila/genetics , Genes, Homeobox/genetics , Nerve Tissue Proteins/genetics , Repressor Proteins/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Molecular Sequence Data , POU Domain Factors , Sequence Alignment , Transcription, Genetic/genetics
17.
J Biol Chem ; 259(21): 13525-33, 1984 Nov 10.
Article in English | MEDLINE | ID: mdl-6490663

ABSTRACT

The ovoperoxidase from the egg of the sea urchin, Strongylocentrotus purpuratus, has been purified to apparent homogeneity. Ovoperoxidase is secreted from the egg at fertilization and is responsible, in vivo, for hardening of the fertilization membrane by forming cross-links between protein tyrosyl residues. Purification was accomplished by activation of cortical granule exocytosis with acetic acid, followed by NH4SO4 precipitation, DEAE-Sephacel chromatography in the absence of divalent cations, and CM-Sephadex chromatography. The purified enzyme is a glycoprotein of Mr 70,000, based on sodium dodecyl sulfate-polyacrylamide gel electrophoresis. The enzyme exhibits a UV-visible spectrum typical of heme peroxidases (epsilon 412 = 1.19 X 10(5) M-1 cm-1). Ovoperoxidase catalyzes the oxidation of tyrosine, guaiacol, iodide, and bromide, but not chloride, and can employ either H2O2 or, with 8% relative efficiency, ethyl peroxide as an oxidative substrate. Phenylhydrazine, 3-amino-1,2,4-triazole, azide, and sulfite all inhibit purified ovoperoxidase at concentrations similar to those that inhibit hardening in vivo. Inhibition by 3-amino-1,2,4-triazole is reversible, requires H2O2, and is slow relative to substrate turnover. The purified enzyme is sensitive to protease cleavage in the native state, yielding an active product of Mr approximately 50,000 which varies slightly depending upon the protease employed. Ovoperoxidase should provide a useful tool for the study of fertilization membrane formation as a paradigm of macromolecular assembly and modification.


Subject(s)
Extraembryonic Membranes/physiology , Fertilization , Ovum/enzymology , Peroxidases/isolation & purification , Amino Acids/analysis , Animals , Carbohydrates/analysis , Cytoplasmic Granules/physiology , Exocytosis , Female , Kinetics , Lactoperoxidase/metabolism , Molecular Weight , Ovum/physiology , Peptide Fragments/analysis , Peroxidases/metabolism , Sea Urchins , Spectrophotometry
18.
Biochemistry ; 23(3): 492-7, 1984 Jan 31.
Article in English | MEDLINE | ID: mdl-6422984

ABSTRACT

The sex steroid binding protein (SBP) of Macaca mulatta and Macaca nemestrina sera has been purified to homogeneity and chemically characterized. The native protein is a glycoprotein having a molecular weight of approximately 88 000 and is composed of two similar subunits of molecular weight 47 000 as estimated by sodium dodecyl sulfate gel electrophoresis. One molecule of 5 alpha-dihydrotestosterone is bound per dimer with a KD equal to 1.6 nM at 11 degrees C. Isoelectric focusing patterns reveal the presence of at least 12 different forms of dimeric SBP molecules probably resulting from the presence of different amounts or types of carbohydrate side chains. The data indicate a very close similarity in molecular and steroid-binding properties to human SBP and establish the macaque monkey as a valuable animal model to study the physiological role of SBP in humans.


Subject(s)
Carrier Proteins/isolation & purification , Macaca mulatta/blood , Macaca nemestrina/blood , Macaca/blood , Amino Acids/analysis , Animals , Carrier Proteins/metabolism , Dihydrotestosterone/blood , Humans , Macromolecular Substances , Molecular Weight , Sex Hormone-Binding Globulin , Species Specificity , Spectrophotometry, Ultraviolet
SELECTION OF CITATIONS
SEARCH DETAIL
...