Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
1.
Neuron ; 112(4): 523-525, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38387437

ABSTRACT

In this issue of Neuron, Choi and colleagues1 uncover the direct role of the transcription factor Pou3f1 in regulating dominance hierarchy in mice. Pou3f1 accomplishes this role via its action in specific prefrontal projection neurons that regulate behaviors associated with low social status.


Subject(s)
Hierarchy, Social , Interneurons , Animals , Mice , Neurons , Transcription Factors , Octamer Transcription Factor-6
2.
Commun Biol ; 6(1): 771, 2023 07 24.
Article in English | MEDLINE | ID: mdl-37488353

ABSTRACT

The Zfp296 gene encodes a zinc finger-type protein. Its expression is high in mouse embryonic stem cells (ESCs) but rapidly decreases following differentiation. Zfp296-knockout (KO) ESCs grew as flat colonies, which were reverted to rounded colonies by exogenous expression of Zfp296. KO ESCs could not form teratomas when transplanted into mice but could efficiently contribute to germline-competent chimeric mice following blastocyst injection. Transcriptome analysis revealed that Zfp296 deficiency up- and down-regulates a distinct group of genes, among which Dppa3, Otx2, and Pou3f1 were markedly downregulated. Chromatin immunoprecipitation sequencing demonstrated that ZFP296 binding is predominantly seen in the vicinity of the transcription start sites (TSSs) of a number of genes, and ZFP296 was suggested to negatively regulate transcription. Consistently, chromatin accessibility assay clearly showed that ZFP296 binding reduces the accessibility of the TSS regions of target genes. Zfp296-KO ESCs showed increased histone H3K9 di- and trimethylation. Co-immunoprecipitation analyses revealed interaction of ZFP296 with G9a and GLP. These results show that ZFP296 plays essential roles in maintaining the global epigenetic state of ESCs through multiple mechanisms including activation of Dppa3, attenuation of chromatin accessibility, and repression of H3K9 methylation, but that Zfp296-KO ESCs retain a unique state of pluripotency while lacking the teratoma-forming ability.


Subject(s)
Chromatin , Teratoma , Animals , Mice , Embryonic Stem Cells , Histones , Mouse Embryonic Stem Cells , Biological Assay , Chromosomal Proteins, Non-Histone , Octamer Transcription Factor-6
3.
Cell Mol Biol Lett ; 27(1): 75, 2022 Sep 05.
Article in English | MEDLINE | ID: mdl-36064319

ABSTRACT

BACKGROUND: Ulcerative colitis-associated colorectal cancer (UC-CRC) is an important complication of ulcerative colitis. Pou3f1 (POU class 3 homeobox 1) is a critical regulator for developmental events and cellular biological processes. However, the role of Pou3f1 in the development of UC-CRC is unclear. METHODS: In vivo, a UC-CRC mouse model was induced by azoxymethane (AOM) and dextran sulfate sodium (DSS). Body weight, colon length, mucosal damage, tumor formation, and survival rate were assessed to determine the progression of UC-CRC. Western blot, quantitative real-time PCR, ELISA, immunohistochemistry, immunofluorescence and TUNEL were performed to examine the severity of inflammation and tumorigenesis. In vitro, LPS-treated mouse bone marrow-derived macrophages (BMDMs) and RAW264.7 cells were used to study the role of Pou3f1 in inflammation. ChIP and luciferase reporter assays were used to confirm the interaction between Nfatc3 and Pou3f1. RESULTS: Pou3f1 expression was increased in the colons of UC-CRC mice, and its inhibition attenuated mucosal injury, reduced colon tumorigenesis and increased survival ratio. Knockdown of Pou3f1 suppressed cell proliferation and increased cell death in colon tumors. Both the in vivo and in vitro results showed that Pou3f1 depletion reduced the production of proinflammation mediators. In addition, ChIP and luciferase reporter assays demonstrated that Nfatc3 directly bound with the Pou3f1 promoter to induce its expression. The effect of Nfatc3 on the inflammatory response in macrophages was suppressed by Pou3f1 knockdown. CONCLUSION: Overall, it outlines that Pou3f1 mediates the role of Nfatc3 in regulating macrophage inflammation and carcinogenesis in UC-CRC development.


Subject(s)
Colitis-Associated Neoplasms , Octamer Transcription Factor-6/metabolism , Animals , Carcinogenesis , Dextran Sulfate/toxicity , Inflammation , Mice , NFATC Transcription Factors
4.
Nature ; 602(7895): 129-134, 2022 02.
Article in English | MEDLINE | ID: mdl-35082446

ABSTRACT

Differentiation proceeds along a continuum of increasingly fate-restricted intermediates, referred to as canalization1,2. Canalization is essential for stabilizing cell fate, but the mechanisms that underlie robust canalization are unclear. Here we show that the BRG1/BRM-associated factor (BAF) chromatin-remodelling complex ATPase gene Brm safeguards cell identity during directed cardiogenesis of mouse embryonic stem cells. Despite the establishment of a well-differentiated precardiac mesoderm, Brm-/- cells predominantly became neural precursors, violating germ layer assignment. Trajectory inference showed a sudden acquisition of a non-mesodermal identity in Brm-/- cells. Mechanistically, the loss of Brm prevented de novo accessibility of primed cardiac enhancers while increasing the expression of neurogenic factor POU3F1, preventing the binding of the neural suppressor REST and shifting the composition of BRG1 complexes. The identity switch caused by the Brm mutation was overcome by increasing BMP4 levels during mesoderm induction. Mathematical modelling supports these observations and demonstrates that Brm deletion affects cell fate trajectory by modifying saddle-node bifurcations2. In the mouse embryo, Brm deletion exacerbated mesoderm-deleted Brg1-mutant phenotypes, severely compromising cardiogenesis, and reveals an in vivo role for Brm. Our results show that Brm is a compensable safeguard of the fidelity of mesoderm chromatin states, and support a model in which developmental canalization is not a rigid irreversible path, but a highly plastic trajectory.


Subject(s)
Cell Differentiation , Cell Lineage , Mesoderm/cytology , Mesoderm/metabolism , Myocytes, Cardiac/cytology , Transcription Factors/metabolism , Animals , Bone Morphogenetic Protein 4/metabolism , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly , DNA Helicases/metabolism , Embryo, Mammalian , Epigenesis, Genetic , Female , Gene Expression Regulation , Male , Mice , Myocardium/metabolism , Neurogenesis , Neurons/cytology , Neurons/metabolism , Nuclear Proteins/metabolism , Octamer Transcription Factor-6/metabolism , Phenotype , Repressor Proteins/metabolism , Stem Cells/cytology , Time Factors , Transcription Factors/deficiency , Transcription Factors/genetics
5.
Diagn Microbiol Infect Dis ; 99(2): 115232, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33130505

ABSTRACT

Leprosy is an infectious disease caused by Mycobacterium leprae that affects the skin and nerves. The nerve damage in leprosy may be related to alterations in transcriptional factors, such as Krox-20, Oct-6, Sox-10. Thirty skin biopsies in leprosy patients and 15 non-leprosy skin biopsies were evaluated using RT-qPCR to assess Krox-20, Oct-6, and Sox-10 and these data was related with S-100 immunohistochemistry. Changes in gene expression were observed in the skin and dermal nerves of leprosy patients in Oct-6 and Sox-10. When comparing Oct-6 with S-100 IHC as diagnostic tests for leprosy, Oct-6 showed a sensitivity of 73.3%, and specificity of 100%, while S-100 IHC showed a sensitivity of 96.6% and specificity of 100%. Our data suggest Oct-6 could be an auxiliary biomarker specific to detecting changes in dermal nerves in leprosy and thus useful to health workers and pathologists with no expertise to observe nerve injuries in leprosy.


Subject(s)
Leprosy/diagnosis , Octamer Transcription Factor-6/genetics , Adult , Aged , Antibodies, Bacterial/blood , Bacterial Load , Biomarkers/metabolism , Biopsy , Cross-Sectional Studies , Early Growth Response Protein 2/genetics , Female , Humans , Immunoglobulin G/blood , Immunohistochemistry , Leprosy/genetics , Leprosy/metabolism , Leprosy/pathology , Male , Middle Aged , Mycobacterium leprae/immunology , S100 Proteins/metabolism , SOXE Transcription Factors/genetics , Sensitivity and Specificity , Skin/innervation , Skin/metabolism , Skin/pathology , Transcription, Genetic
6.
J Bacteriol ; 203(1)2020 12 07.
Article in English | MEDLINE | ID: mdl-33020223

ABSTRACT

During their synthesis, the C-tailed membrane proteins expose the membrane-spanning segment late from the ribosome and consequently can insert into the membrane only posttranslationally. However, the C-tailed type 6 secretion system (T6SS) component SciP uses the bacterial signal recognition particle (SRP) system for membrane targeting, which operates cotranslationally. Analysis of possible sequence regions in the amino-terminal part of the protein revealed two candidates that were then tested for whether they function as SRP signal peptides. Both sequences were tested positive as synthetic peptides for binding to SRP. In addition, purified ribosomes with stalled nascent chains exposing either sequence were capable of binding to SRP and SRP-FtsY complexes with high affinity. Together, the data suggest that both peptides can serve as an SRP signal sequence promoting an early membrane targeting of SciP during its synthesis. Like observed for multispanning membrane proteins, the two cytoplasmic SRP signal sequences of SciP may also facilitate a retargeting event, making the targeting more efficient.IMPORTANCE C-tail proteins are anchored in the inner membrane with a transmembrane segment at the C terminus in an N-in/C-out topology. Due to this topology, membrane insertion occurs only posttranslationally. Nevertheless, the C-tail-anchored protein SciP is targeted cotranslationally by SRP. We report here that two amino-terminal hydrophobic stretches in SciP are individually recognized by SRP and target the nascent protein to FtsY. The presence of two signal sequences may enable a retargeting mechanism, as already observed for multispanning membrane proteins, to make the posttranslational insertion of SciP by YidC more efficient.


Subject(s)
Octamer Transcription Factor-6/chemistry , Signal Recognition Particle/chemistry , Amino Acid Sequence , Escherichia coli/classification , Escherichia coli/genetics , Escherichia coli/growth & development , Hydrophobic and Hydrophilic Interactions , Mutation , Octamer Transcription Factor-6/genetics , Octamer Transcription Factor-6/metabolism , Signal Recognition Particle/genetics
7.
Nat Commun ; 10(1): 3477, 2019 08 02.
Article in English | MEDLINE | ID: mdl-31375664

ABSTRACT

Oct4, along with Sox2 and Klf4 (SK), can induce pluripotency but structurally similar factors like Oct6 cannot. To decode why Oct4 has this unique ability, we compare Oct4-binding, accessibility patterns and transcriptional waves with Oct6 and an Oct4 mutant defective in the dimerization with Sox2 (Oct4defSox2). We find that initial silencing of the somatic program proceeds indistinguishably with or without Oct4. Oct6 mitigates the mesenchymal-to-epithelial transition and derails reprogramming. These effects are a consequence of differences in genome-wide binding, as the early binding profile of Oct4defSox2 resembles Oct4, whilst Oct6 does not bind pluripotency enhancers. Nevertheless, in the Oct6-SK condition many otherwise Oct4-bound locations become accessible but chromatin opening is compromised when Oct4defSox2 occupies these sites. We find that Sox2 predominantly facilitates chromatin opening, whilst Oct4 serves an accessory role. Formation of Oct4/Sox2 heterodimers is essential for pluripotency establishment; however, reliance on Oct4/Sox2 heterodimers declines during pluripotency maintenance.


Subject(s)
Cellular Reprogramming/genetics , Chromatin/metabolism , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors/metabolism , Animals , Cells, Cultured , Embryo, Mammalian , Epithelial-Mesenchymal Transition/genetics , Fibroblasts , Induced Pluripotent Stem Cells/physiology , Kruppel-Like Factor 4 , Mice, Transgenic , Mutation , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-6/metabolism , Primary Cell Culture , Protein Multimerization/genetics , SOXB1 Transcription Factors/genetics , Time Factors
8.
Stem Cell Reports ; 9(2): 571-586, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28757169

ABSTRACT

During gastrulation, the neuroectoderm cells form the neural tube and neural crest. The nervous system contains significantly more microRNAs than other tissues, but the role of microRNAs in controlling the differentiation of neuroectodermal cells into neural tube epithelial (NTE) cells and neural crest cells (NCCs) remains unknown. Using embryonic stem cell (ESC) neural differentiation systems, we found that miR-29b was upregulated in NTE cells and downregulated in NCCs. MiR-29b promoted the differentiation of ESCs into NTE cells and inhibited their differentiation into NCCs. Accordingly, the inhibition of miR-29b significantly inhibited the differentiation of NTE cells. A mechanistic study revealed that miR-29b targets DNA methyltransferase 3a (Dnmt3a) to regulate neural differentiation. Moreover, miR-29b mediated the function of Pou3f1, a critical neural transcription factor. Therefore, our study showed that the Pou3f1-miR-29b-Dnmt3a regulatory axis was active at the initial stage of neural differentiation and regulated the determination of cell fate.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , MicroRNAs/genetics , Neural Crest/embryology , Neural Crest/metabolism , Neural Tube/embryology , Neural Tube/metabolism , Animals , Biomarkers , Cell Line , Cell Lineage/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Gene Expression Regulation , Humans , Mice , Octamer Transcription Factor-6/genetics , RNA Interference
9.
Stem Cell Reports ; 8(5): 1270-1286, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28434941

ABSTRACT

Environmental stresses are increasingly acknowledged as core causes of abnormal neural induction leading to neural tube defects (NTDs). However, the mechanism responsible for environmental stress-triggered neural induction defects remains unknown. Here, we report that a spectrum of environmental stresses, including oxidative stress, starvation, and DNA damage, profoundly activate SIRT1, an NAD+-dependent lysine deacetylase. Both mouse embryos and in vitro differentiated embryonic stem cells (ESCs) demonstrated a negative correlation between the expression of SIRT1 and that of OCT6, a key neural fate inducer. Activated SIRT1 radically deacetylates OCT6, triggers an OCT6 ubiquitination/degradation cascade, and consequently increases the incidence of NTD-like phenotypes in mice or hinders neural induction in both human and mouse ESCs. Together, our results suggest that early exposure to environmental stresses results in the dysregulation of the SIRT1/OCT6 axis and increases the risk of NTDs.


Subject(s)
Environmental Exposure , Neural Tube Defects/metabolism , Octamer Transcription Factor-6/metabolism , Oxidative Stress , Sirtuin 1/metabolism , Animals , Cells, Cultured , DNA Damage , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Tube Defects/etiology , Neural Tube Defects/genetics , Octamer Transcription Factor-6/genetics , Proteolysis , Sirtuin 1/genetics , Ubiquitination
10.
Biosci Trends ; 11(1): 95-104, 2017 Mar 22.
Article in English | MEDLINE | ID: mdl-28154340

ABSTRACT

As is similar to glial cell line-derived neurotrophic factor (GDNF), the Yangjing Capsule (YC) extract could also lead to proliferation of spermatogonial stem cells (SSCs) by stimulating the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway; however, the regulatory effect of YC extract on the expression of POU3F1 still remains unknown. The objective of this study is to determine whether the transcription factor POU3F1 is up-regulated by YC extract through the PI3K/AKT signaling pathway to regulate SSCs survival and proliferation. Cultured GC-1 spermatogonial (spg) cells were treated with 0.01, 0.1, and 1 mg/mL YC extract for 48 h. Cell viability was analyzed using MTT assay, while POU3F1 expression was quantitatively detected using real time-polymerase chain reaction and Western blot analysis. POU3F1, GDNF family receptor alpha1 (GFRα1) short interfering ribonucleic acid (siRNA), and LY294002 (PI3K inhibitor) were applied as blockers to explore the underlying pathway. After 48 h treatment with YC extract, GC-1 spg cells proliferated and POU3F1 expression was significantly increased in a dose-dependent manner. POU3F1 siRNA partially blocked those effects of YC extract. Both GFRα1 siRNA and LY294002, as upstream blockers, reduced POU3F1 expression induced by YC extract. The conclusion is that YC extract promotes proliferation of GC-1 spg cells via up-regulation of POU3F1. The potential mechanism is that YC extract triggers the activation of the PI3K/AKT pathway and then up-regulates POU3F1 expression.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Octamer Transcription Factor-6/metabolism , Signal Transduction/drug effects , Spermatogonia/cytology , Spermatogonia/metabolism , Up-Regulation/drug effects , Animals , Capsules , Cell Line , Cell Proliferation/drug effects , Gene Knockdown Techniques , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Male , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spermatogonia/drug effects
11.
Brain Struct Funct ; 221(8): 4187-4202, 2016 11.
Article in English | MEDLINE | ID: mdl-26687503

ABSTRACT

Somatic motor neurons in the hypoglossal nucleus innervate tongue muscles controlling vital functions such as chewing, swallowing and respiration. Formation of functional hypoglossal nerve circuits depends on the establishment of precise hypoglossal motor neuron maps correlating with specific tongue muscle innervations. Little is known about the molecular mechanisms controlling mammalian hypoglossal motor neuron topographic map formation. Here we show that combinatorial expression of transcription factors Runx1, SCIP and FoxP1 defines separate mouse hypoglossal motor neuron groups with different topological, neurotransmitter and calcium-buffering phenotypes. Runx1 and SCIP are coexpressed in ventromedial hypoglossal motor neurons involved in control of tongue protrusion whereas FoxP1 is expressed in dorsomedial motor neurons associated with tongue retraction. Establishment of separate hypoglossal motor neuron maps depends in part on Runx1-mediated suppression of ventrolateral and dorsomedial motor neuron phenotypes and regulation of FoxP1 expression pattern. These findings suggest that combinatorial actions of Runx1, SCIP and FoxP1 are important for mouse hypoglossal nucleus somatotopic map formation.


Subject(s)
Gene Expression Regulation, Developmental , Hypoglossal Nerve/embryology , Hypoglossal Nerve/metabolism , Motor Neurons/metabolism , Motor Neurons/physiology , Animals , Core Binding Factor Alpha 2 Subunit/metabolism , Forkhead Transcription Factors/metabolism , Mice , Mice, Transgenic , Octamer Transcription Factor-6/metabolism , Repressor Proteins/metabolism , Tongue/embryology , Tongue/innervation
12.
J Comp Neurol ; 522(18): 4057-73, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25048219

ABSTRACT

Among sodium channel isoforms, Nav 1.6 is selectively expressed at nodes of Ranvier in both the CNS and the PNS. However, non-Nav 1.6 isoforms such as Nav 1.2 are also present at the CNS nodes in early development but gradually diminish later. It has been proposed that myelination is part of a glia-neuron signaling mechanism that produces this change in nodal isoform expression. The present study used isoform-specific antibodies to demonstrate that, in the PNS, four other neuronal sodium channel isoforms were also clustered at nodes in early development but eventually disappeared during maturation. To study possible roles of myelination in such transitions, we investigated the nodal expression of selected isoforms in the sciatic nerve of the transgenic mouse Oct6(ΔSCE/ßgeo) , whose PNS myelination is delayed in the first postnatal week but eventually resumes. We found that delayed myelination retarded the formation of nodal channel clusters and altered the expression-elimination patterns of sodium channel isoforms, resulting in significantly reduced expression levels of non-Nav 1.6 isoforms in such delayed nodes. However, delayed myelination did not significantly affect the gene expression, protein synthesis, or axonal trafficking of any isoform studied. Rather, we found evidence for a developmentally programmed increase in neuronal Nav 1.6 expression with constant or decreasing neuronal expression of other isoforms that were unaffected by delayed myelination. Thus our results suggest that, in the developmental isoform switch of the PNS, myelination does not play a signaling role as that proposed for the CNS but rather serves only to form nodal clusters from existing isoform pools.


Subject(s)
Ranvier's Nodes/metabolism , Sciatic Nerve/growth & development , Sciatic Nerve/metabolism , Sodium Channels/metabolism , Animals , Ganglia, Spinal/growth & development , Ganglia, Spinal/metabolism , Immunoblotting , Immunohistochemistry , Lumbar Vertebrae , Mice, Transgenic , Microarray Analysis , Mutation , Myelin Sheath/metabolism , NAV1.2 Voltage-Gated Sodium Channel/metabolism , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Neurons/metabolism , Octamer Transcription Factor-6/genetics , Octamer Transcription Factor-6/metabolism , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
13.
Cell Mol Neurobiol ; 34(7): 1023-36, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24962097

ABSTRACT

SYF2 is a putative homolog of human p29 in Saccharomyces cerevisiae. It seems to be involved in pre-mRNA splicing and cell cycle progression. Disruption of SYF2 leads to reduced α-tubulin expression and delayed nerve system development in zebrafish. Due to the potential of SYF2 in modulating microtubule dynamics in nervous system, we investigated the spatiotemporal expression of SYF2 in a rat sciatic nerve crush (SNC) model. We found that SNC resulted in a significant upregulation of SYF2 from 3 days to 1 week and subsequently returned to the normal level at 4 weeks. At its peak expression, SYF2 distributed predominantly in Schwann cells. In addition, upregulation of SYF2 was approximately in parallel with Oct-6, and numerous Schwann cells expressing SYF2 were Oct-6 positive. In vitro, we observed enhanced expression of SYF2 during the process of cyclic adenosine monophosphate (cAMP)-induced Schwann cell differentiation. SYF2-specific siRNA-transfected Schwann cells did not show significant morphological change in the process of Schwann cell differentiation. Also, we found shorter and disorganized microtubule structure and a decreased migration in SYF2-specific siRNA-transfected Schwann cells. Together, these findings indicated that the upregulation of SYF2 was associated with Schwann cell differentiation and migration following sciatic nerve crush.


Subject(s)
Cell Differentiation , Cell Movement , Nerve Crush , Nuclear Proteins/metabolism , Schwann Cells/pathology , Sciatic Nerve/pathology , Up-Regulation , Animals , Biomarkers/metabolism , Blotting, Western , CD11b Antigen/metabolism , Cell Differentiation/drug effects , Cell Movement/drug effects , Cyclic AMP/pharmacology , Immunohistochemistry , Male , Models, Biological , Octamer Transcription Factor-6/metabolism , Phenotype , RNA, Small Interfering/metabolism , Rats, Sprague-Dawley , Schwann Cells/drug effects , Schwann Cells/metabolism , Tubulin/metabolism , Up-Regulation/drug effects
14.
Elife ; 32014 Jun 14.
Article in English | MEDLINE | ID: mdl-24929964

ABSTRACT

The neural fate commitment of pluripotent stem cells requires the repression of extrinsic inhibitory signals and the activation of intrinsic positive transcription factors. However, how these two events are integrated to ensure appropriate neural conversion remains unclear. In this study, we showed that Pou3f1 is essential for the neural differentiation of mouse embryonic stem cells (ESCs), specifically during the transition from epiblast stem cells (EpiSCs) to neural progenitor cells (NPCs). Chimeric analysis showed that Pou3f1 knockdown leads to a markedly decreased incorporation of ESCs in the neuroectoderm. By contrast, Pou3f1-overexpressing ESC derivatives preferentially contribute to the neuroectoderm. Genome-wide ChIP-seq and RNA-seq analyses indicated that Pou3f1 is an upstream activator of neural lineage genes, and also is a repressor of BMP and Wnt signaling. Our results established that Pou3f1 promotes the neural fate commitment of pluripotent stem cells through a dual role, activating internal neural induction programs and antagonizing extrinsic neural inhibitory signals.


Subject(s)
Embryonic Stem Cells/cytology , Germ Layers/metabolism , Neural Stem Cells/cytology , Octamer Transcription Factor-6/metabolism , Signal Transduction , Animals , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Cell Lineage , Chick Embryo , Chromatin Immunoprecipitation , Green Fluorescent Proteins/metabolism , Mice , Neural Plate/cytology , Sequence Analysis, RNA , Wnt Proteins/metabolism
15.
Development ; 141(4): 784-94, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24496616

ABSTRACT

Air breathing is an essential motor function for vertebrates living on land. The rhythm that drives breathing is generated within the central nervous system and relayed via specialised subsets of spinal motor neurons to muscles that regulate lung volume. In mammals, a key respiratory muscle is the diaphragm, which is innervated by motor neurons in the phrenic nucleus. Remarkably, relatively little is known about how this crucial subtype of motor neuron is generated during embryogenesis. Here, we used direct differentiation of motor neurons from mouse embryonic stem cells as a tool to identify genes that direct phrenic neuron identity. We find that three determinants, Pou3f1, Hoxa5 and Notch, act in combination to promote a phrenic neuron molecular identity. We show that Notch signalling induces Pou3f1 in developing motor neurons in vitro and in vivo. This suggests that the phrenic neuron lineage is established through a local source of Notch ligand at mid-cervical levels. Furthermore, we find that the cadherins Pcdh10, which is regulated by Pou3f1 and Hoxa5, and Cdh10, which is controlled by Pou3f1, are both mediators of like-like clustering of motor neuron cell bodies. This specific Pcdh10/Cdh10 activity might provide the means by which phrenic neurons are assembled into a distinct nucleus. Our study provides a framework for understanding how phrenic neuron identity is conferred and will help to generate this rare and inaccessible yet vital neuronal subtype directly from pluripotent stem cells, thus facilitating subsequent functional investigations.


Subject(s)
Cadherins/metabolism , Embryonic Stem Cells/cytology , Motor Neurons/cytology , Octamer Transcription Factor-6/metabolism , Phrenic Nerve/embryology , Signal Transduction/physiology , Animals , Cell Differentiation/physiology , Diaphragm/innervation , Flow Cytometry , Homeodomain Proteins/metabolism , Mice , Motor Neurons/physiology , Phosphoproteins/metabolism , Phrenic Nerve/cytology , Protocadherins , Real-Time Polymerase Chain Reaction , Receptors, Notch/metabolism , Signal Transduction/genetics , Transcription Factors , Transcriptome
16.
Dev Cell ; 26(2): 123-35, 2013 Jul 29.
Article in English | MEDLINE | ID: mdl-23906064

ABSTRACT

We recently demonstrated that the expression of the importin α subtype is switched from α2 to α1 during neural differentiation in mouse embryonic stem cells (ESCs) and that this switching has a major impact on cell differentiation. In this study, we report a cell-fate determination mechanism in which importin α2 negatively regulates the nuclear import of certain transcription factors to maintain ESC properties. The nuclear import of Oct6 and Brn2 was inhibited via the formation of a transport-incompetent complex of the cargo bound to a nuclear localization signal binding site in importin α2. Unless this dominant-negative effect was downregulated upon ESC differentiation, inappropriate cell death was induced. We propose that although certain transcription factors are necessary for differentiation in ESCs, these factors are retained in the cytoplasm by importin α2, thereby preventing transcription factor activity in the nucleus until the cells undergo differentiation.


Subject(s)
Cell Nucleus/metabolism , Embryonic Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Octamer Transcription Factor-6/metabolism , POU Domain Factors/metabolism , Active Transport, Cell Nucleus , Animals , Cell Differentiation , Cell Line , Mice , Nuclear Localization Signals/metabolism , Octamer Transcription Factor-3/metabolism , Protein Binding , Signal Transduction , alpha Karyopherins , beta Karyopherins/metabolism
17.
Muscle Nerve ; 48(3): 423-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23824709

ABSTRACT

INTRODUCTION: Severe lesions in the facial nerve may have extensive axonal loss and leave isolated stumps that impose technical difficulties for nerve grafting. METHODS: We evaluated bone marrow stem cells (BMSC) in a silicone conduit for rat facial nerve regeneration from isolated stumps. Group A utilized empty silicone tubes; in groups B-D, the tube was filled with acellular gel; and, in groups C and D, undifferentiated BMSC (uBMSC) or Schwann-like cells differentiated from BMSC (dBMSC) were added, respectively. Compound muscle action potentials (CMAPs) were measured, and histology was evaluated. RESULTS: Groups C and D had the highest CMAP amplitudes. Group C had shorter CMAP durations than groups A, B, and D. Distal axonal number and density were increased in group C compared with groups A and B. CONCLUSIONS: Regeneration of the facial nerve was improved by both uBMSC and dBMSC in rats, yet uBMSC was associated with superior functional results.


Subject(s)
Amputation Stumps/surgery , Bone Marrow Transplantation/methods , Facial Nerve/cytology , Mesenchymal Stem Cells/physiology , Muscle, Skeletal/physiopathology , Nerve Regeneration/physiology , Action Potentials/physiology , Animals , Axons/pathology , Cells, Cultured , Electromyography , Follow-Up Studies , Male , Octamer Transcription Factor-6/metabolism , Organ Culture Techniques , Rats , Rats, Wistar , Receptor, Nerve Growth Factor/metabolism , S100 Proteins/metabolism , Statistics, Nonparametric , Transduction, Genetic , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
18.
Mol Microbiol ; 87(6): 1277-89, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23368090

ABSTRACT

Cell cycle transitions are often triggered by the proteolysis of key regulatory proteins. In Caulobacter crescentus, the G1-S transition involves the degradation of an essential DNA-binding response regulator, CtrA, by the ClpXP protease. Here, we show that another critical cell cycle regulator, SciP, is also degraded during the G1-S transition, but by the Lon protease. SciP is a small protein that binds directly to CtrA and prevents it from activating target genes during G1. We demonstrate that SciP must be degraded during the G1-S transition so that cells can properly activate CtrA-dependent genes following DNA replication initiation and the reaccumulation of CtrA. These results indicate that like CtrA, SciP levels are tightly regulated during the Caulobacter cell cycle. In addition, we show that formation of a complex between CtrA and SciP at target promoters protects both proteins from their respective proteases. Degradation of either protein thus helps trigger the destruction of the other, facilitating a cooperative disassembly of the complex. Collectively, our results indicate that ClpXP and Lon each degrade an important cell cycle regulator, helping to trigger the onset of S phase and prepare cells for the subsequent programmes of gene expression critical to polar morphogenesis and cell division.


Subject(s)
Caulobacter crescentus/physiology , Cell Cycle , Gene Expression Regulation, Bacterial , Octamer Transcription Factor-6/metabolism , Protease La/metabolism , Caulobacter crescentus/cytology , Caulobacter crescentus/genetics , Caulobacter crescentus/growth & development , Proteolysis
19.
Arch Dermatol Res ; 305(5): 371-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23341029

ABSTRACT

Loricrin is a major component of the epidermal cornified cell envelope, and is expressed only in terminally differentiated keratinocytes. This cell differentiation-specific expression pattern suggests specific regulatory mechanisms for activation and suppression of loricrin gene transcription in differentiated keratinocytes. Here, we identified a regulatory element in the proximal promoter region of the loricrin gene involved in suppression of its expression in keratinocytes. A database search indicated that this sequence contained a POU transcription factor binding motif. Electrophoretic mobility shift assay revealed that Oct-1, Oct-6, and Oct-11 actually bind to the motif. Constructs with point mutations in the POU-binding motif showed increased reporter activity, indicating that the POU factors negatively regulate loricrin gene transcription. Cotransfection experiments suggested that Oct-6 and Oct-11 suppress loricrin gene transcription in a cooperative manner with AP-1 and Sp1. Furthermore, in vitro experiments indicated that the Oct-6 and Oct-11 can physically associate with both AP-1 factors and Sp1/Sp3. These findings indicate that Oct-6 and Oct-11 contribute to the regulation of loricrin gene transcription via interaction with AP-1 factors and Sp1/Sp3.


Subject(s)
Keratinocytes/metabolism , Membrane Proteins/metabolism , Octamer Transcription Factor-6/metabolism , Octamer Transcription Factors/metabolism , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Transcription Factor AP-1/metabolism , Animals , Binding Sites , Cells, Cultured , Databases, Genetic , Down-Regulation , Electrophoretic Mobility Shift Assay , Genes, Reporter , Membrane Proteins/genetics , Mice , Point Mutation , Promoter Regions, Genetic , Regulatory Elements, Transcriptional , Sequence Analysis, DNA , Transcription, Genetic , Transfection
20.
J Mol Neurosci ; 49(3): 491-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22869340

ABSTRACT

Transcription Initiation Factor IIB (TFIIB), as a general transcription factor, plays an essential role in preinitiation complex assembly and transcription initiation by recruiting RNA polymerase II to the promoter. However, its distribution and function in peripheral system lesion and repair were still unknown. Here, we investigated the spatiotemporal expression of TFIIB in an acute sciatic nerve crush model in adult rats. Western blot analysis revealed that TFIIB was expressed in normal sciatic nerve. It gradually increased, reached a peak at the seventh day after crush, and then returned to the normal level at 4 weeks. We observed that TFIIB expressed mainly increased in Schwann cells and co-localized with Oct-6. In vitro, we induced Schwann cell differentiation with cyclic adenosine monophosphate (cAMP) and found that TFIIB expression was increased in the differentiated process. TFIIB-specific siRNA inhibited cAMP-induced Schwann cell morphological change and the expression of P0. Collectively, we hypothesized peripheral nerve crush-induced upregulation of TFIIB in the sciatic nerve was associated with Schwann cell differentiation.


Subject(s)
Nerve Crush , Nerve Regeneration , Nerve Tissue Proteins/physiology , Schwann Cells/metabolism , Sciatic Nerve/injuries , Transcription Factor TFIIB/physiology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Cyclic AMP/pharmacology , Gene Expression Regulation , Male , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Octamer Transcription Factor-6/biosynthesis , Octamer Transcription Factor-6/genetics , Primary Cell Culture , RNA Interference , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Schwann Cells/pathology , Sciatic Nerve/physiology , Transcription Factor TFIIB/antagonists & inhibitors , Transcription Factor TFIIB/biosynthesis , Transcription Factor TFIIB/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...