Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Life Sci ; 254: 116866, 2020 Aug 01.
Article in English | MEDLINE | ID: mdl-31518606

ABSTRACT

Neural stem cells (NSCs) are pluripotent cells that are capable of differentiating into neurons and considered as the most promising cell source for cell replacement therapy. However, the difficulty in inducing neuronal differentiation and maturation from NSCs is a major challenge for their clinical application. Clarifying the molecular mechanisms underlying the neuronal differentiation of NSCs can provide a basis for expanding their uses. Brain 4 (Brn4) is a member of the POU domain family of transcription factors and can induce the neuronal differentiation of NSCs, but its precise function in NSCs is unclear. To address this question, in this study we isolated and expanded radial glial cells (RGCs), a type of NSC, from the cerebral cortex of 14-day embryonic rats and used lentivirus carrying the human Brn4 gene to overexpress Brn4 in these cells. This induced the differentiation of RGCs into neurons and inhibited the expression of C-terminal binding protein 2 (CtBP2), a transcriptional co-repressor. CtBP2 overexpression in RGCs suppressed their differentiation into neurons, whereas CtBP2 knockdown had the opposite effect. These results indicated that Brn4 promoted the neuronal differentiation of NSCs via inhibition of CtBP2 and is a potential tool for generating neurons in cell replacement therapy of neurodegenerative diseases and brain injury.


Subject(s)
Cell Differentiation/physiology , Eye Proteins/antagonists & inhibitors , Nerve Tissue Proteins/physiology , Neuroglia/cytology , Neurons/cytology , POU Domain Factors/physiology , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Eye Proteins/metabolism , Female , Pregnancy , Rats , Rats, Sprague-Dawley
2.
Biochem Biophys Res Commun ; 523(2): 411-415, 2020 03 05.
Article in English | MEDLINE | ID: mdl-31870549

ABSTRACT

Pou3f2/Brn2 is a transcription factor that helps to determine the cellular identity of neocortical or hypothalamic neurons. Mammalian Pou3f2 contains three homopolymeric amino acids that are not present in amphibian Pou3f2. These amino acids contribute to monoamine function, which may play specific roles in mammalian development and behavior. Previous work has indicated that Pou3f2⊿ mice, which lack the homopolymeric amino acids, exhibited declined maternal activity and impaired object and spatial recognition. The current study, analyzed weight gain, brain development, home cage activity, social interaction, and response to novel objects in Pou3f2⊿ mice to determine which aspects of behavior were affected by monoamine dysregulation. Compared to their wild type counterparts, Pou3f2⊿ mice showed decreased social interaction and reduced home cage activity during their active phase. However, they showed normal weight gain, brain development, and responses to novelty. These results indicate that monoamine dysregulation in Pou3f2⊿ mice may specifically affect basal activity and social development, without altering non-social motivation.


Subject(s)
Behavior, Animal/physiology , Nerve Tissue Proteins/physiology , POU Domain Factors/physiology , Social Behavior , Animals , Biogenic Monoamines/physiology , Brain/growth & development , Exploratory Behavior/physiology , Hypothalamus/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neocortex/physiology , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurons/physiology , POU Domain Factors/chemistry , POU Domain Factors/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Weight Gain
3.
Dev Dyn ; 248(2): 173-188, 2019 02.
Article in English | MEDLINE | ID: mdl-30444277

ABSTRACT

BACKGROUND: Many human gene mutations have been linked to congenital heart disease (CHD), yet CHD remains a major health issue worldwide due in part to an incomplete understanding of the molecular basis for cardiac malformation. RESULTS: Here we identify the orthologous mouse Pou6f1 and zebrafish pouC as POU homeodomain transcription factors enriched in the developing heart. We find that pouC is a multi-functional transcriptional regulator containing separable activation, repression, protein-protein interaction, and DNA binding domains. Using zebrafish heart development as a model system, we demonstrate that pouC knockdown impairs cardiac morphogenesis and affects cardiovascular function. We also find that levels of pouC expression must be fine-tuned to enable proper heart formation. At the cellular level, we demonstrate that pouC knockdown disrupts atrioventricular canal (AVC) cardiomyocyte maintenance, although chamber myocyte specification remains intact. Mechanistically, we show that pouC binds a bmp4 intronic regulatory element to mediate transcriptional activation. CONCLUSIONS: Taken together, our study establishes pouC as a novel transcriptional input into the regulatory hierarchy that drives AVC morphogenesis in zebrafish. We anticipate that these findings will inform future efforts to explore functional conservation in mammals and potential association with atrioventricular septal defects in humans. Developmental Dynamics 248:173-188, 2019. © 2018 Wiley Periodicals, Inc.


Subject(s)
Bone Morphogenetic Protein 4/genetics , Gene Expression Regulation, Developmental , Heart Septum/growth & development , POU Domain Factors/physiology , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology , Animals , Bone Morphogenetic Protein 4/metabolism , Heart/embryology , Heart/growth & development , Heart Septal Defects , Heart Septum/embryology , Mice , POU Domain Factors/metabolism , Protein Binding , Transcription Factors , Zebrafish , Zebrafish Proteins/metabolism
4.
Dev Biol ; 444 Suppl 1: S297-S307, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29571612

ABSTRACT

Mutations that disrupt the inwardly rectifying potassium channel Kir2.1 lead to Andersen-Tawil syndrome that includes periodic paralysis, cardiac arrhythmia, cognitive deficits, craniofacial dysmorphologies and limb defects. The molecular mechanism that underlies the developmental consequences of inhibition of these channels has remained a mystery. We show that while loss of Kir2.1 function does not affect expression of several early facial patterning genes, the domain in which Pou3f3 is expressed in the maxillary arch is reduced. Pou3f3 is important for development of the jugal and squamosal bones. The reduced expression domain of Pou3f3 is consistent with the reduction in the size of the squamosal and jugal bones in Kcnj2KO/KO animals, however it does not account for the diverse craniofacial defects observed in Kcnj2KO/KO animals. We show that Kir2.1 function is required in the cranial neural crest for morphogenesis of several craniofacial structures including palate closure. We find that while the palatal shelves of Kir2.1-null embryos elevate properly, they are reduced in size due to decreased proliferation of the palatal mesenchyme. While we find no reduction in expression of BMP ligands, receptors, and associated Smads in this setting, loss of Kir2.1 reduces the efficacy of BMP signaling as shown by the reduction of phosphorylated Smad 1/5/8 and reduced expression of BMP targets Smad6 and Satb2.


Subject(s)
Face/embryology , Potassium Channels, Inwardly Rectifying/physiology , Animals , Body Patterning/genetics , Body Patterning/physiology , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/physiology , Craniofacial Abnormalities/embryology , Gene Expression Regulation/genetics , Mice , Mice, Knockout , Mutation , Nerve Tissue Proteins/physiology , Neural Crest/metabolism , Neural Crest/physiology , POU Domain Factors/physiology , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Signal Transduction , Skull/embryology , Transcription Factors/metabolism
5.
Mol Psychiatry ; 21(10): 1417-33, 2016 10.
Article in English | MEDLINE | ID: mdl-26830142

ABSTRACT

Social interaction is a fundamental behavior in all animal species, but the developmental timing of the social neural circuit formation and the cellular and molecular mechanisms governing its formation are poorly understood. We generated a mouse model with mutations in two Disheveled genes, Dvl1 and Dvl3, that displays adult social and repetitive behavioral abnormalities associated with transient embryonic brain enlargement during deep layer cortical neuron formation. These phenotypes were mediated by the embryonic expansion of basal neural progenitor cells (NPCs) via deregulation of a ß-catenin/Brn2/Tbr2 transcriptional cascade. Transient pharmacological activation of the canonical Wnt pathway during this period of early corticogenesis rescued the ß-catenin/Brn2/Tbr2 transcriptional cascade and the embryonic brain phenotypes. Remarkably, this embryonic treatment prevented adult behavioral deficits and partially rescued abnormal brain structure in Dvl mutant mice. Our findings define a mechanism that links fetal brain development and adult behavior, demonstrating a fetal origin for social and repetitive behavior deficits seen in disorders such as autism.


Subject(s)
Stereotypic Movement Disorder/genetics , Stereotypic Movement Disorder/physiopathology , Adaptor Proteins, Signal Transducing/genetics , Animals , Behavior, Animal , Brain/embryology , Brain/metabolism , Brain/physiology , Dishevelled Proteins/genetics , Dishevelled Proteins/metabolism , Humans , Mice , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Neural Stem Cells/metabolism , Neurons/metabolism , POU Domain Factors/metabolism , POU Domain Factors/physiology , Phosphoproteins/genetics , Signal Transduction/physiology , Stereotyped Behavior/physiology , T-Box Domain Proteins/metabolism , T-Box Domain Proteins/physiology , Wnt Proteins/metabolism , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , beta Catenin/physiology
6.
Dev Growth Differ ; 57(9): 591-600, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26660397

ABSTRACT

POU class V (POU-V) transcription factors play the important role in maintenance of pluripotency and cell differentiation. Pou5f3.2 (Oct25), one of Xenopus POU-V transcription factors, shows the zygotic expression prior to gastrulation. In order to know the molecular mechanism of pou5f3.2 expression at gastrula stage, we examined a responsiveness of pou5f3.2 to Nodal signaling. Animal cap assay demonstrated that Xnr2 activates the gene expression of pou5f3.2. In comparative analysis of the 5'-flanking region of pou5f3.2 between Xenopus laevis and X. tropicalis, two conserved regions were detected within the flanking region. Reporter analyses showed that one of the conserved regions contained an enhancer region, which had several Smad2/3 and FoxH1 binding motifs. ChIP assay demonstrated that Smad2 binds to the enhancer region. These results suggest that Nodal signaling induces zygotic expression of pou5f3.2 at gastrula stage. To understand a role of pou5f3.2 in gastrula embryos, morpholino oligo DNA of pou5f3.2 was injected into the lateral side of one blastomere at the 2-cell stage. The morphant embryos showed diminution of Xbra1 expression and gastrulation defect in the injection side, suggesting the essential role of pou5f3.2 at the gastrula stage. Xbra1 expression and gastrulation were also inhibited by injecting with the synthesized RNAs of pou5f3.2. Furthermore, in the pou5f3.2-injected embryo, gene expression of p27Xic1 was drastically suppressed, and the number of dividing cells increased in the injection side. These results suggest that one role of pou5f3.2 is to keep the embryonic cells in undifferentiated and proliferative state during gastrulation.


Subject(s)
Cell Proliferation/physiology , Gastrulation , POU Domain Factors/physiology , Xenopus laevis/embryology , Animals , Gene Expression Regulation, Developmental , POU Domain Factors/genetics
7.
Am J Pathol ; 185(12): 3164-77, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26482608

ABSTRACT

Insulinoma-associated protein 1 (INSM1) is expressed exclusively in embryonic developing neuroendocrine (NE) tissues. INSM1 gene expression is specific for small-cell lung cancer (SCLC), along with achaete-scute homolog-like 1 (ASCL1) and several NE molecules, such as chromogranin A, synaptophysin, and neural cell adhesion molecule 1. However, the underlying biological role of INSM1 in lung cancer remains largely unknown. We first showed that surgically resected SCLC samples specifically expressed INSM1. Forced expression of the INSM1 gene in adenocarcinoma cell lines (H358 and H1975) induced the expression of ASCL1, brain-2 (BRN2), chromogranin A, synaptophysin, and neural cell adhesion molecule 1; in contrast, knockdown of the INSM1 gene by siRNA in SCLC (H69 and H889) decreased their expression. However, forced/knockdown expression of ASCL1 and BRN2 did not affect INSM1 expression. A chromatin immunoprecipitation study revealed that INSM1 bound to the promoter region of the ASCL1 gene. A xenotransplantation assay using tet-on INSM1 gene-transfected adenocarcinoma cell lines demonstrated that INSM1 induced NE differentiation and growth inhibition. Furthermore, we found that INSM1 was not expressed in non-small-cell lung cancer and some SCLC cell lines expressing Notch1-Hes1. By forced/knockdown expression of Notch1 or Hes1 genes, we revealed that Notch1-Hes1 signaling suppressed INSM1, as well as ASCL1 and BRN2. INSM1, expressed exclusively in SCLC, is a crucial regulator of NE differentiation in SCLCs, and is regulated by the Notch1-Hes1 signaling pathway.


Subject(s)
Lung Neoplasms/metabolism , Neuroendocrine Cells/pathology , Repressor Proteins/physiology , Small Cell Lung Carcinoma/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Apoptosis/physiology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Differentiation/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Gene Knockdown Techniques , Heterografts , Homeodomain Proteins/metabolism , Homeodomain Proteins/physiology , Humans , Lung Neoplasms/pathology , Mice, Inbred Strains , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Proteins/physiology , Neoplasm Transplantation , Neuroendocrine Cells/metabolism , POU Domain Factors/metabolism , POU Domain Factors/physiology , Receptor, Notch1/physiology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/physiology , Small Cell Lung Carcinoma/pathology , Transcription Factor HES-1
8.
Neurosurgery ; 75(4): 472-82; discussion 482, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24887289

ABSTRACT

BACKGROUND: Mixed lineage leukemia-1 (Mll1) epigenetically regulates gene expression patterns that specify cellular identity in both embryonic development and adult stem cell populations. In the adult mouse brain, multipotent neural stem cells (NSCs) in the subventricular zone generate new neurons throughout life, and Mll1 is required for this postnatal neurogenesis but not for glial cell differentiation. Analysis of Mll1-dependent transcription may identify neurogenic genes useful for the direct reprogramming of astrocytes into neurons. OBJECTIVE: To identify Mll1-dependent transcriptional modules and to determine whether genes in the neurogenic modules can be used to directly reprogram astrocytes into neurons. METHODS: We performed gene coexpression module analysis on microarray data from differentiating wild-type and Mll1-deleted subventricular zone NSCs. Key developmental regulators belonging to the neurogenic modules were overexpressed in Mll1-deleted cells and cultured cortical astrocytes, and cell phenotypes were analyzed by immunocytochemistry and electrophysiology. RESULTS: Transcriptional modules that correspond to neurogenesis were identified in wild-type NSCs. Modules related to astrocytes and oligodendrocytes were enriched in Mll1-deleted NSCs, consistent with their gliogenic potential. Overexpression of genes selected from the neurogenic modules enhanced the production of neurons from Mll1-deleted cells, and overexpression of Brn4 (Pou3f4) in nonneurogenic cortical astroglia induced their transdifferentiation into electrophysiologically active neurons. CONCLUSION: Our results demonstrate that Mll1 is required for the expression of neurogenic but not gliogenic transcriptional modules in a multipotent NSC population and further indicate that specific Mll1-dependent genes may be useful for direct reprogramming strategies.


Subject(s)
Astrocytes/physiology , Cell Transdifferentiation/physiology , Histone-Lysine N-Methyltransferase/physiology , Myeloid-Lymphoid Leukemia Protein/physiology , Nerve Tissue Proteins/physiology , Neural Stem Cells/physiology , Neurons/physiology , POU Domain Factors/physiology , Animals , Histone-Lysine N-Methyltransferase/deficiency , Mice , Microarray Analysis , Myeloid-Lymphoid Leukemia Protein/deficiency , Neurogenesis/physiology
9.
PLoS One ; 7(7): e41506, 2012.
Article in English | MEDLINE | ID: mdl-22859994

ABSTRACT

Recent findings have demonstrated that the overexpression of lineage-specific transcription factors induces cell fate changes among diverse cell types. For example, neurons can be generated from mouse and human fibroblasts. It is well known that neurons are terminally differentiated cells that do not divide. Therefore, we consider how to induce glioma cells to become neurons by introducing transcription factors. Here, we describe the efficient generation of induced neuronal (iN) cells from glioma cells by the infection with three transcription factors: Ascl1, Brn2 and Ngn2 (ABN). iN cells expressed multiple neuronal markers and fired action potentials, similar to the properties of authentic neurons. Importantly, the proliferation of glioma cells following ABN overexpression was dramatically inhibited in both in vitro and in vivo experiments. In addition, iN cells that originated from human glioma cells did not continue to grow when they were sorted and cultured in vitro. The strategies by which glioma cells are induced to become neurons may be used to clinically study methods for inhibiting tumor growth.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Transformation, Neoplastic , Homeodomain Proteins/physiology , Nerve Tissue Proteins/physiology , Neurons/physiology , POU Domain Factors/physiology , Action Potentials , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Proliferation , G1 Phase Cell Cycle Checkpoints , Glioma , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurons/metabolism , POU Domain Factors/biosynthesis , POU Domain Factors/genetics , Transduction, Genetic
10.
Dev Neurobiol ; 72(11): 1422-32, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22190420

ABSTRACT

The ability of axons to project correctly to the target is essential for the formation of complex neural networks. The intrinsic regulation of this process is still unclear. Here, we show that POU domain motif 3 (Pdm3) is required in ring (R) neurons to control precise axon targeting to the Drosophila ellipsoid body (EB). Pdm3 is expressed in neurons of the central nervous system in larvae and adults and required for the normal development of the EB of the central complex in the adult brain. The normal EB structure is abolished in pdm3 mutants, and this phenotype is rescued by pdm3 expression in R neurons, suggesting that the defect in axonal targeting of R neurons is the major cause in EB malformation in pdm3 mutants. We show that cell fate determination, dendritic arborization, and initial axon projection of R neurons are normal while the axonal targeting to the EB is defective in pdm3 mutants.


Subject(s)
Axons/physiology , Brain/growth & development , Drosophila Proteins/physiology , Neurons/physiology , POU Domain Factors/physiology , Animals , Animals, Genetically Modified , Brain/physiology , Cell Differentiation/physiology , Drosophila/genetics
11.
Dev Biol ; 360(1): 11-29, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21958745

ABSTRACT

The formation of the dorsal-ventral (DV) and anterior-posterior (AP) axes, fundamental to the body plan of animals, is regulated by several groups of polypeptide growth factors including the TGF-ß, FGF, and Wnt families. In order to ensure the establishment of the body plan, the processes of DV and AP axis formation must be linked and coordinately regulated. However, the molecular mechanisms responsible for these interactions remain unclear. Here, we demonstrate that the forkhead box transcription factor FoxB1, which is upregulated by the neuralizing factor Oct-25, plays an important role in the formation of the DV and AP axes. Overexpression of FoxB1 promoted neural induction and inhibited BMP-dependent epidermal differentiation in ectodermal explants, thereby regulating the DV patterning of the ectoderm. In addition, FoxB1 was also found to promote the formation of posterior neural tissue in both ectodermal explants and whole embryos, suggesting its involvement in embryonic AP patterning. Using knockdown analysis, we found that FoxB1 is required for the formation of posterior neural tissues, acting in concert with the Wnt and FGF pathways. Consistent with this, FoxB1 suppressed the formation of anterior structures via a process requiring the function of XWnt-8 and eFGF. Interestingly, while downregulation of FoxB1 had little effect on neural induction, we found that it functionally interacted with its upstream factor Oct-25 and plays a supportive role in the induction and/or maintenance of neural tissue. Our results suggest that FoxB1 is part of a mechanism that fine-tunes, and leads to the coordinated formation of, the DV and AP axes during early development.


Subject(s)
Body Patterning/physiology , Forkhead Transcription Factors/physiology , Xenopus Proteins/physiology , Xenopus laevis/embryology , Xenopus laevis/physiology , Animals , Base Sequence , Body Patterning/genetics , Bone Morphogenetic Proteins/metabolism , Ectoderm/embryology , Ectoderm/metabolism , Fibroblast Growth Factors/physiology , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , HeLa Cells , Humans , Morpholinos/genetics , Neurogenesis/genetics , Neurogenesis/physiology , Oligonucleotides, Antisense/genetics , POU Domain Factors/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Transfection , Up-Regulation , Wnt Proteins/physiology , Wnt Signaling Pathway , Xenopus Proteins/deficiency , Xenopus Proteins/genetics , Xenopus laevis/genetics
12.
Development ; 138(12): 2429-39, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21610024

ABSTRACT

The evolutionary origin of stem cell pluripotency is an unresolved question. In mammals, pluripotency is limited to early embryos and is induced and maintained by a small number of key transcription factors, of which the POU domain protein Oct4 is considered central. Clonal invertebrates, by contrast, possess pluripotent stem cells throughout their life, but the molecular mechanisms that control their pluripotency are poorly defined. To address this problem, we analyzed the expression pattern and function of Polynem (Pln), a POU domain gene from the marine cnidarian Hydractinia echinata. We show that Pln is expressed in the embryo and adult stem cells of the animal and that ectopic expression in epithelial cells induces stem cell neoplasms and loss of epithelial tissue. Neoplasm cells downregulated the transgene but expressed the endogenous Pln gene and also Nanos, Vasa, Piwi and Myc, which are all known cnidarian stem cell markers. Retinoic acid treatment caused downregulation of Pln and the differentiation of neoplasm cells to neurosensory and epithelial cells. Pln downregulation by RNAi led to differentiation. Collectively, our results suggest an ancient role of POU proteins as key regulators of animal stem cells.


Subject(s)
Cnidaria/cytology , Neoplastic Stem Cells/cytology , POU Domain Factors/physiology , Pluripotent Stem Cells/cytology , Animals , Octamer Transcription Factor-3/physiology , Stem Cells , Tretinoin/pharmacology
13.
PLoS One ; 5(10): e13216, 2010 Oct 12.
Article in English | MEDLINE | ID: mdl-20967260

ABSTRACT

Terminal differentiation of skin keratinocytes is a vertically directed multi-step process that is tightly controlled by the sequential expression of a variety of genes. In this study, we investigated the role of the POU domain-containing transcription factor Brn2 in keratinocyte differentiation. Immunohistochemical analysis showed that Brn2 is expressed primarily in the upper granular layer. Consistent with its epidermal localization, Brn2 expression was highly induced at 14 days after calcium treatment of cultured normal human epidermal keratinocytes. When Brn2 was overexpressed by adenoviral transduction, Brn2 led to increased expression of the differentiation-related genes involucrin, filaggrin, and loricrin in addition to inhibition of their proliferation. Chromatin immunoprecipitation demonstrated that Brn2 bound to the promoter regions of these differentiation-related genes. We injected the purified Brn2 adenovirus into rat skin, which led to a thickened epidermis with increased amounts of differentiation related markers. The histopathologic features of adenovirus-Brn2 injected skin tissues looked similar to the features of lichen planus, a human skin disease showing chronic inflammation and well-differentiated epidermal changes. Moreover, Brn2 is shown to be expressed in almost all cell nuclei of the thickened epidermis of lichen planus, and Brn2 also attracts T lymphocytes. Our results demonstrate that Brn2 is probably a transcriptional factor playing an important role in keratinocyte differentiation and probably also in the pathogenesis of lichen planus lesions.


Subject(s)
Cell Differentiation , Homeodomain Proteins/physiology , Keratinocytes/cytology , POU Domain Factors/physiology , Animals , Female , Filaggrin Proteins , Humans , Rats , Rats, Sprague-Dawley
14.
J Neurosci ; 30(14): 5028-36, 2010 Apr 07.
Article in English | MEDLINE | ID: mdl-20371823

ABSTRACT

Individual olfactory receptor neurons (ORNs) selectively express one or a small number of odor receptors from among a large receptor repertoire. The expression of an odor receptor dictates the odor response spectrum of the ORN. The process of receptor gene choice relies in part on a combinatorial code of transcription factors. In Drosophila, the POU domain transcription factor Acj6 is one element of the transcription factor code. In acj6 null mutants, many ORNs do not express an appropriate odor receptor gene and thus are not correctly specified. We find that acj6 is alternatively spliced to yield many structurally distinct transcripts in the olfactory organs. We generate flies that express single splice forms of acj6 in an acj6(-) background. We find that different splice forms are functionally distinct; they differ in their abilities to specify ORN identities. Some individual splice forms can fully rescue the specification of some ORNs. Individual splice forms can function both positively and negatively in receptor gene regulation. ORNs differ in their requirements for splice forms; some are not fully rescued by any single splice form tested, suggesting that some ORNs may require the combinatorial action of multiple splice forms. Late expression of some acj6 splice forms is sufficient to rescue some ORN classes, consistent with a direct role for Acj6 isoforms in receptor gene expression. The results indicate that alternative splicing may add another level of richness to the regulatory code that underlies the process of odor receptor gene choice.


Subject(s)
Alternative Splicing/genetics , Drosophila Proteins/genetics , Nerve Tissue Proteins/genetics , Olfactory Receptor Neurons/physiology , POU Domain Factors/genetics , Protein Isoforms/genetics , Receptors, Odorant/genetics , Alternative Splicing/physiology , Amino Acid Sequence , Animals , Animals, Genetically Modified , Drosophila Proteins/biosynthesis , Drosophila Proteins/deficiency , Drosophila Proteins/physiology , Drosophila melanogaster/genetics , Gene Knockout Techniques , Larva/genetics , Larva/physiology , Molecular Sequence Data , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/physiology , POU Domain Factors/deficiency , POU Domain Factors/physiology , Protein Isoforms/deficiency , Protein Isoforms/physiology , Receptors, Odorant/biosynthesis , Receptors, Odorant/deficiency
15.
Neurosci Res ; 67(1): 8-17, 2010 May.
Article in English | MEDLINE | ID: mdl-20105446

ABSTRACT

Brn-4, a member of the homeobox family of transcription factors, has previously been implicated in the regeneration and repair of denervated striatum. We investigated the effects of Brn-4 on the differentiation and development of neural stem cells (NSCs) from E16 rat hippocampus. Immunocytochemistry revealed that extracts of deafferented hippocampus promoted neuronal differentiation to a greater extent than extracts from normal hippocampus. Deafferented extracts also promoted maturation of newborn neurons as reflected in changes in cell areas and perimeters, and enhanced Brn-4 expression in MAP-2 positive neurons. Suppression or overexpression of Brn-4 in NSCs markedly decreased or increased neuronal differentiation and maturation of newborn neurons, respectively. These results suggest that Brn-4 expression is required both for neuronal differentiation of NSCs and maturation of newborn neurons, and that there may be some regulatory factors in deafferented hippocampus that can regulate Brn-4 expression in neuronal progenitors. Brn-4 is therefore a potential research target for the development of new therapeutics to promote brain repair.


Subject(s)
Nerve Tissue Proteins/physiology , Neurons/cytology , POU Domain Factors/physiology , Stem Cells/cytology , Afferent Pathways , Animals , Cell Differentiation , Embryo, Mammalian , Female , Hippocampus/cytology , Nerve Tissue Proteins/biosynthesis , Neurons/metabolism , POU Domain Factors/biosynthesis , RNA Interference , Rats , Rats, Sprague-Dawley , Stem Cells/metabolism , Tissue Extracts/pharmacology
16.
Development ; 137(1): 43-51, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20023159

ABSTRACT

Numb can antagonize Notch signaling to diversify the fates of sister cells. We report here that paired sister cells acquire different fates in all three Drosophila neuronal lineages that make diverse types of antennal lobe projection neurons (PNs). Only one in each pair of postmitotic neurons survives into the adult stage in both anterodorsal (ad) and ventral (v) PN lineages. Notably, Notch signaling specifies the PN fate in the vPN lineage but promotes programmed cell death in the missing siblings in the adPN lineage. In addition, Notch/Numb-mediated binary sibling fates underlie the production of PNs and local interneurons from common precursors in the lAL lineage. Furthermore, Numb is needed in the lateral but not adPN or vPN lineages to prevent the appearance of ectopic neuroblasts and to ensure proper self-renewal of neural progenitors. These lineage-specific outputs of Notch/Numb signaling show that a universal mechanism of binary fate decision can be utilized to govern diverse neural sibling differentiations.


Subject(s)
Brain/cytology , Brain/metabolism , Drosophila Proteins/physiology , Juvenile Hormones/physiology , Receptors, Notch/physiology , Signal Transduction , Animals , Apoptosis/genetics , Apoptosis/physiology , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Immunohistochemistry , Juvenile Hormones/genetics , Models, Biological , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Neurons/cytology , Neurons/metabolism , POU Domain Factors/genetics , POU Domain Factors/metabolism , POU Domain Factors/physiology , Receptors, Notch/genetics
17.
Dev Biol ; 337(1): 162-70, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19850032

ABSTRACT

POU5F1 (OCT4) encodes a master regulator of pluripotency that is present in all mammals. A paralogue, POU2, is also present in the genomes of marsupials and monotremes and is an orthologue of zebrafish pou2 and chicken POUV. We explored the evolution of class V POU domain transcription factors and show that POU5F1 arose by gene duplication of pou2 early in the evolution of tetrapods and is not mammal-specific, as previously thought. Instead, either POU5F1 or POU2/POUV has become extinct independently in various lineages, although all gnathostomes appear to possess at least one or the other. In the tammar wallaby, POU5F1 expression is limited to pluripotent cell types (embryonic tissues and germ cells). POU2 is similarly expressed in pluripotent tissues but is also expressed in a broad range of adult tissues. Thus, unlike POU5F1, the role of POU2 may not be restricted to pluripotent cell types but could have a related function such as maintaining multipotency in adult stem cells.


Subject(s)
Marsupialia/embryology , POU Domain Factors/genetics , Amino Acid Sequence , Animals , Base Sequence , Conserved Sequence , Embryonic Development , Evolution, Molecular , Exons , Female , Gene Expression Regulation, Developmental , Macropodidae/embryology , Molecular Sequence Data , Octamer Transcription Factor-3/genetics , Opossums/embryology , POU Domain Factors/chemistry , POU Domain Factors/physiology , Pluripotent Stem Cells/metabolism , Zebrafish Proteins/genetics
18.
J Neurosci ; 29(41): 12940-7, 2009 Oct 14.
Article in English | MEDLINE | ID: mdl-19828808

ABSTRACT

Little is known about how individual olfactory receptor neurons (ORNs) select, from among many odor receptor genes, which genes to express. Abnormal chemosensory jump 6 (Acj6) is a POU domain transcription factor essential for the specification of ORN identity and odor receptor (Or) gene expression in the Drosophila maxillary palp, one of the two adult olfactory organs. However, the mechanism by which Acj6 functions in this process has not been investigated. Here, we systematically examine the role of Acj6 in the maxillary palp and in a major subset of antennal ORNs. We define an Acj6 binding site by a reiterative in vitro selection process. The site is found upstream of Or genes regulated by Acj6, and Acj6 binds to the site in Or promoters. Mutational analysis shows that the site is essential for Or regulation in vivo. Surprisingly, a novel ORN class in acj6 adults is found to arise from ectopic expression of a larval Or gene, which is repressed in wild type via an Acj6 binding site. Thus, Acj6 acts directly in the process of receptor gene choice; it plays a dual role, positive and negative, in the logic of the process, and acts in partitioning the larval and adult receptor repertoires.


Subject(s)
Drosophila Proteins/physiology , Gene Expression Regulation, Developmental/physiology , Nerve Tissue Proteins/physiology , Odorants , Olfactory Receptor Neurons/metabolism , POU Domain Factors/physiology , Receptors, Odorant/metabolism , Sense Organs/cytology , Animals , Animals, Genetically Modified , Binding Sites/genetics , Drosophila/genetics , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Larva , Olfactory Pathways/cytology , Olfactory Pathways/metabolism , Protein Binding/genetics , Receptors, Odorant/genetics , Sense Organs/metabolism
19.
Hum Cell ; 22(4): 94-100, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19874398

ABSTRACT

Clear cell adenocarcinoma of the ovary often shows resistance to anticancer agents. We investigated new molecules to use when developing molecular-targeting therapy for clear cell adenocarcinoma of the ovary. RMG-I cells without invasive potential and RMG-V cells with invasive potential (derived from clear cell adenocarcinoma of the ovary) were subjected to complementary deoxyribonucleic acid microarray analysis. Caveolin-1, a molecule involved in cellular motility and invasion, showed differing expression between the two cell lines. An RNA interference experiment using the published siRNA for caveolin-1 was carried out. The results showed suppression of RMG-V cell infiltration by siRNA, but proliferation of the cancer cells was also suppressed. In other words, RMG-V cell infiltration may have been suppressed simply because cell proliferation was suppressed by RNA interference. These findings suggested that POU6F1 might be a transcription factor involved in the proliferation of ovarian cancer cells. Clear cell adenocarcinoma of the ovary shows little response to standard therapy. The results of the present study suggest that the transcription factor POU6F1 could be a new molecular target for treatment of this cancer.


Subject(s)
Adenocarcinoma, Clear Cell/pathology , Cell Proliferation , Ovarian Neoplasms/pathology , POU Domain Factors/physiology , Transcription Factors/physiology , Adenocarcinoma, Clear Cell/genetics , Caveolin 1/metabolism , Caveolin 1/physiology , Cell Line, Tumor , Female , Humans , Neoplasm Invasiveness , Ovarian Neoplasms/genetics , Reverse Transcriptase Polymerase Chain Reaction
20.
J Invest Dermatol ; 129(4): 945-53, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18923447

ABSTRACT

Nestin is an intermediate filament protein and a marker of neuroectodermal stem cells indicating multipotentiality and regenerative capability. In melanoma tissues, nestin re-expression was correlated with tumor progression. Activation of the nestin neural enhancer was shown to be dependent on the binding of class III POU transcription factors, with brain-2 (BRN2) suggested to play a key role. We found both nestin and BRN2 mRNA in almost all of 13 analyzed melanoma cell lines of different progression stages, but expression levels did not correlate. Nestin protein was detected in 11 of 13 and BRN2 protein in 7 of 13 melanoma cell lines independent of progression stage. Downregulation of BRN2 by small-interfering RNA did not alter nestin expression in melanoma cells. However, POU proteins, such as BRN2, commonly cooperate with transcription factors of the Sry-box (SOX) family by binding to a nearby DNA site necessary for their action. SOX9 and SOX10 have been shown to be expressed in melanocyte precursors, with SOX10 downregulated upon differentiation. We now demonstrate SOX9 and SOX10 protein expression in melanoma tissues and cell lines. Downregulation of SOX9 and of SOX10 markedly decreased nestin levels in melanoma cells in a cooperative manner. Thus, SOX9 and SOX10 but not BRN2 seem to be required for nestin expression in human melanoma.


Subject(s)
Homeodomain Proteins/physiology , Intermediate Filament Proteins/genetics , Melanoma/metabolism , Nerve Tissue Proteins/genetics , POU Domain Factors/physiology , SOX9 Transcription Factor/physiology , SOXE Transcription Factors/physiology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/analysis , Homeodomain Proteins/genetics , Humans , Intermediate Filament Proteins/analysis , Melanoma/pathology , Nerve Tissue Proteins/analysis , Nestin , POU Domain Factors/analysis , POU Domain Factors/genetics , RNA, Messenger/analysis , SOX9 Transcription Factor/analysis , SOXE Transcription Factors/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...