Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Mol Biol Lett ; 27(1): 75, 2022 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-36064319

RESUMO

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.


Assuntos
Neoplasias Associadas a Colite , Fator 6 de Transcrição de Octâmero/metabolismo , Animais , Carcinogênese , Sulfato de Dextrana/toxicidade , Inflamação , Camundongos , Fatores de Transcrição NFATC
2.
Nature ; 602(7895): 129-134, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35082446

RESUMO

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.


Assuntos
Diferenciação Celular , Linhagem da Célula , Mesoderma/citologia , Mesoderma/metabolismo , Miócitos Cardíacos/citologia , Fatores de Transcrição/metabolismo , Animais , Proteína Morfogenética Óssea 4/metabolismo , Cromatina/genética , Cromatina/metabolismo , Montagem e Desmontagem da Cromatina , DNA Helicases/metabolismo , Embrião de Mamíferos , Epigênese Genética , Feminino , Regulação da Expressão Gênica , Masculino , Camundongos , Miocárdio/metabolismo , Neurogênese , Neurônios/citologia , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Fator 6 de Transcrição de Octâmero/metabolismo , Fenótipo , Proteínas Repressoras/metabolismo , Células-Tronco/citologia , Fatores de Tempo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
3.
J Bacteriol ; 203(1)2020 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-33020223

RESUMO

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.


Assuntos
Fator 6 de Transcrição de Octâmero/química , Partícula de Reconhecimento de Sinal/química , Sequência de Aminoácidos , Escherichia coli/classificação , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Interações Hidrofóbicas e Hidrofílicas , Mutação , Fator 6 de Transcrição de Octâmero/genética , Fator 6 de Transcrição de Octâmero/metabolismo , Partícula de Reconhecimento de Sinal/genética
4.
Nat Commun ; 10(1): 3477, 2019 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-31375664

RESUMO

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.


Assuntos
Reprogramação Celular/genética , Cromatina/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Animais , Células Cultivadas , Embrião de Mamíferos , Transição Epitelial-Mesenquimal/genética , Fibroblastos , Células-Tronco Pluripotentes Induzidas/fisiologia , Fator 4 Semelhante a Kruppel , Camundongos Transgênicos , Mutação , Fator 3 de Transcrição de Octâmero/genética , Fator 6 de Transcrição de Octâmero/metabolismo , Cultura Primária de Células , Multimerização Proteica/genética , Fatores de Transcrição SOXB1/genética , Fatores de Tempo
5.
Stem Cell Reports ; 8(5): 1270-1286, 2017 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-28434941

RESUMO

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.


Assuntos
Exposição Ambiental , Defeitos do Tubo Neural/metabolismo , Fator 6 de Transcrição de Octâmero/metabolismo , Estresse Oxidativo , Sirtuína 1/metabolismo , Animais , Células Cultivadas , Dano ao DNA , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Humanos , Camundongos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Defeitos do Tubo Neural/etiologia , Defeitos do Tubo Neural/genética , Fator 6 de Transcrição de Octâmero/genética , Proteólise , Sirtuína 1/genética , Ubiquitinação
6.
Biosci Trends ; 11(1): 95-104, 2017 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-28154340

RESUMO

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.


Assuntos
Medicamentos de Ervas Chinesas/farmacologia , Fator 6 de Transcrição de Octâmero/metabolismo , Transdução de Sinais/efeitos dos fármacos , Espermatogônias/citologia , Espermatogônias/metabolismo , Regulação para Cima/efeitos dos fármacos , Animais , Cápsulas , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Masculino , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espermatogônias/efeitos dos fármacos
7.
Brain Struct Funct ; 221(8): 4187-4202, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-26687503

RESUMO

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.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Nervo Hipoglosso/embriologia , Nervo Hipoglosso/metabolismo , Neurônios Motores/metabolismo , Neurônios Motores/fisiologia , Animais , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Camundongos , Camundongos Transgênicos , Fator 6 de Transcrição de Octâmero/metabolismo , Proteínas Repressoras/metabolismo , Língua/embriologia , Língua/inervação
8.
J Comp Neurol ; 522(18): 4057-73, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25048219

RESUMO

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.


Assuntos
Nós Neurofibrosos/metabolismo , Nervo Isquiático/crescimento & desenvolvimento , Nervo Isquiático/metabolismo , Canais de Sódio/metabolismo , Animais , Gânglios Espinais/crescimento & desenvolvimento , Gânglios Espinais/metabolismo , Immunoblotting , Imuno-Histoquímica , Vértebras Lombares , Camundongos Transgênicos , Análise em Microsséries , Mutação , Bainha de Mielina/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.2/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Neurônios/metabolismo , Fator 6 de Transcrição de Octâmero/genética , Fator 6 de Transcrição de Octâmero/metabolismo , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real
9.
Elife ; 32014 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-24929964

RESUMO

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.


Assuntos
Células-Tronco Embrionárias/citologia , Camadas Germinativas/metabolismo , Células-Tronco Neurais/citologia , Fator 6 de Transcrição de Octâmero/metabolismo , Transdução de Sinais , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular , Linhagem da Célula , Embrião de Galinha , Imunoprecipitação da Cromatina , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Placa Neural/citologia , Análise de Sequência de RNA , Proteínas Wnt/metabolismo
10.
Cell Mol Neurobiol ; 34(7): 1023-36, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24962097

RESUMO

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.


Assuntos
Diferenciação Celular , Movimento Celular , Compressão Nervosa , Proteínas Nucleares/metabolismo , Células de Schwann/patologia , Nervo Isquiático/patologia , Regulação para Cima , Animais , Biomarcadores/metabolismo , Western Blotting , Antígeno CD11b/metabolismo , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , AMP Cíclico/farmacologia , Imuno-Histoquímica , Masculino , Modelos Biológicos , Fator 6 de Transcrição de Octâmero/metabolismo , Fenótipo , RNA Interferente Pequeno/metabolismo , Ratos Sprague-Dawley , Células de Schwann/efeitos dos fármacos , Células de Schwann/metabolismo , Tubulina (Proteína)/metabolismo , Regulação para Cima/efeitos dos fármacos
11.
Development ; 141(4): 784-94, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24496616

RESUMO

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.


Assuntos
Caderinas/metabolismo , Células-Tronco Embrionárias/citologia , Neurônios Motores/citologia , Fator 6 de Transcrição de Octâmero/metabolismo , Nervo Frênico/embriologia , Transdução de Sinais/fisiologia , Animais , Diferenciação Celular/fisiologia , Diafragma/inervação , Citometria de Fluxo , Proteínas de Homeodomínio/metabolismo , Camundongos , Neurônios Motores/fisiologia , Fosfoproteínas/metabolismo , Nervo Frênico/citologia , Protocaderinas , Reação em Cadeia da Polimerase em Tempo Real , Receptores Notch/metabolismo , Transdução de Sinais/genética , Fatores de Transcrição , Transcriptoma
12.
Dev Cell ; 26(2): 123-35, 2013 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-23906064

RESUMO

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.


Assuntos
Núcleo Celular/metabolismo , Células-Tronco Embrionárias/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Fator 6 de Transcrição de Octâmero/metabolismo , Fatores do Domínio POU/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Diferenciação Celular , Linhagem Celular , Camundongos , Sinais de Localização Nuclear/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Ligação Proteica , Transdução de Sinais , alfa Carioferinas , beta Carioferinas/metabolismo
13.
Muscle Nerve ; 48(3): 423-9, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23824709

RESUMO

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.


Assuntos
Cotos de Amputação/cirurgia , Transplante de Medula Óssea/métodos , Nervo Facial/citologia , Células-Tronco Mesenquimais/fisiologia , Músculo Esquelético/fisiopatologia , Regeneração Nervosa/fisiologia , Potenciais de Ação/fisiologia , Animais , Axônios/patologia , Células Cultivadas , Eletromiografia , Seguimentos , Masculino , Fator 6 de Transcrição de Octâmero/metabolismo , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar , Receptor de Fator de Crescimento Neural/metabolismo , Proteínas S100/metabolismo , Estatísticas não Paramétricas , Transdução Genética , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
14.
Mol Microbiol ; 87(6): 1277-89, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23368090

RESUMO

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.


Assuntos
Caulobacter crescentus/fisiologia , Ciclo Celular , Regulação Bacteriana da Expressão Gênica , Fator 6 de Transcrição de Octâmero/metabolismo , Protease La/metabolismo , Caulobacter crescentus/citologia , Caulobacter crescentus/genética , Caulobacter crescentus/crescimento & desenvolvimento , Proteólise
15.
Arch Dermatol Res ; 305(5): 371-8, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23341029

RESUMO

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.


Assuntos
Queratinócitos/metabolismo , Proteínas de Membrana/metabolismo , Fator 6 de Transcrição de Octâmero/metabolismo , Fatores de Transcrição de Octâmero/metabolismo , Fator de Transcrição Sp1/metabolismo , Fator de Transcrição Sp3/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Sítios de Ligação , Células Cultivadas , Bases de Dados Genéticas , Regulação para Baixo , Ensaio de Desvio de Mobilidade Eletroforética , Genes Reporter , Proteínas de Membrana/genética , Camundongos , Mutação Puntual , Regiões Promotoras Genéticas , Elementos Reguladores de Transcrição , Análise de Sequência de DNA , Transcrição Gênica , Transfecção
16.
Cereb Cortex ; 23(11): 2632-43, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22892427

RESUMO

The upper layers (II-IV) are the most prominent distinguishing feature of mammalian neocortex compared with avian or reptilian dorsal cortex, and are vastly expanded in primates. Although the time-dependent embryonic generation of upper-layer cells is genetically instructed within their parental progenitors, mechanisms governing cell-intrinsic fate transitions remain obscure. POU-homeodomain transcription factors Pou3f3 and Pou3f2 (Brn1 and Brn2) are known to label postmitotic upper-layer cells, and are redundantly required for their production. We find that the onset of Pou3f3/2 expression actually occurs in ventricular zone (VZ) progenitors, and that Pou3f3/2 subsequently label neural progeny switching from deep-layer Ctip2(+) identity to Satb2(+) upper-layer fate as they migrate to proper superficial positions. By using an Engrailed dominant-negative repressor, we show that sustained neurogenesis after the deep- to upper-layer transition requires the proneual action of Pou3fs in VZ progenitors. Conversely, single-gene overexpression of any Pou3f in early neural progenitors is sufficient to specify the precocious birth of Satb2(+) daughter neurons that extend axons to the contralateral hemisphere, as well as exhibit robust pia-directed migration that is characteristic of upper-layer cells. Finally, we demonstrate that Pou3fs influence multiple stages of neurogenesis by suppressing Notch effector Hes5, and promoting the expression of proneural transcription factors Tbr2 and Tbr1.


Assuntos
Movimento Celular , Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Neurogênese , Neurônios/metabolismo , Fatores do Domínio POU/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Córtex Cerebral/citologia , Macaca mulatta , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Fator 6 de Transcrição de Octâmero/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo
17.
Nat Neurosci ; 16(1): 48-54, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23222914

RESUMO

After peripheral nerve injury, axons regenerate and become remyelinated by resident Schwann cells. However, myelin repair never results in the original myelin thickness, suggesting insufficient stimulation by neuronal growth factors. Upon testing this hypothesis, we found that axonal neuregulin-1 (NRG1) type III and, unexpectedly, also NRG1 type I restored normal myelination when overexpressed in transgenic mice. This led to the observation that Wallerian degeneration induced de novo NRG1 type I expression in Schwann cells themselves. Mutant mice lacking a functional Nrg1 gene in Schwann cells are fully myelinated but exhibit impaired remyelination in adult life. We suggest a model in which loss of axonal contact triggers denervated Schwann cells to transiently express NRG1 as an autocrine/paracrine signal that promotes Schwann cell differentiation and remyelination.


Assuntos
Doenças Desmielinizantes/metabolismo , Neuregulina-1/metabolismo , Recuperação de Função Fisiológica/genética , Células de Schwann/metabolismo , Neuropatia Ciática/patologia , Animais , Animais Recém-Nascidos , Axônios/efeitos dos fármacos , Axônios/patologia , Axônios/ultraestrutura , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Doenças Desmielinizantes/etiologia , Modelos Animais de Doenças , Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Estimulação Elétrica , Inibidores Enzimáticos/farmacologia , Potencial Evocado Motor/efeitos dos fármacos , Potencial Evocado Motor/fisiologia , Gânglios Espinais/citologia , Regulação da Expressão Gênica/genética , Proteínas Hedgehog/genética , Antígeno Ki-67/metabolismo , Locomoção/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Proteína Básica da Mielina/metabolismo , Proteína P0 da Mielina/metabolismo , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Neuregulina-1/genética , Neurônios/química , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fator 6 de Transcrição de Octâmero/metabolismo , RNA Mensageiro/metabolismo , Ratos , Recuperação de Função Fisiológica/efeitos dos fármacos , Proteínas S100/metabolismo , Células de Schwann/efeitos dos fármacos , Células de Schwann/ultraestrutura , Nervo Isquiático/citologia , Neuropatia Ciática/fisiopatologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Estatísticas não Paramétricas , Fatores de Tempo
18.
Brain ; 135(Pt 12): 3551-66, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23250879

RESUMO

Charcot-Marie-Tooth disease type 1B is caused by mutations in myelin protein zero. R98C mice, an authentic model of early onset Charcot-Marie-Tooth disease type 1B, develop neuropathy in part because the misfolded mutant myelin protein zero is retained in the endoplasmic reticulum where it activates the unfolded protein response. Because oral curcumin, a component of the spice turmeric, has been shown to relieve endoplasmic reticulum stress and decrease the activation of the unfolded protein response, we treated R98C mutant mice with daily gastric lavage of curcumin or curcumin derivatives starting at 4 days of age and analysed them for clinical disability, electrophysiological parameters and peripheral nerve morphology. Heterozygous R98C mice treated with curcumin dissolved in sesame oil or phosphatidylcholine curcumin performed as well as wild-type littermates on a rotarod test and had increased numbers of large-diameter axons in their sciatic nerves. Treatment with the latter two compounds also increased compound muscle action potential amplitudes and the innervation of neuromuscular junctions in both heterozygous and homozygous R98C animals, but it did not improve nerve conduction velocity, myelin thickness, G-ratios or myelin period. The expression of c-Jun and suppressed cAMP-inducible POU (SCIP)-transcription factors that inhibit myelination when overexpressed-was also decreased by treatment. Consistent with its role in reducing endoplasmic reticulum stress, treatment with curcumin dissolved in sesame oil or phosphatidylcholine curcumin was associated with decreased X-box binding protein (XBP1) splicing. Taken together, these data demonstrate that treatment with curcumin dissolved in sesame oil or phosphatidylcholine curcumin improves the peripheral neuropathy of R98C mice by alleviating endoplasmic reticulum stress, by reducing the activation of unfolded protein response and by promoting Schwann cell differentiation.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Diferenciação Celular/efeitos dos fármacos , Doença de Charcot-Marie-Tooth , Curcumina/uso terapêutico , Proteína P0 da Mielina/genética , Células de Schwann/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Fatores Etários , Análise de Variância , Animais , Animais Recém-Nascidos , Arginina/genética , Células COS/efeitos dos fármacos , Células Cultivadas , Doença de Charcot-Marie-Tooth/tratamento farmacológico , Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/patologia , Chlorocebus aethiops , Cisteína/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Estimulação Elétrica/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Força Muscular/efeitos dos fármacos , Força Muscular/genética , Mutação/genética , Proteína P0 da Mielina/metabolismo , Junção Neuromuscular/efeitos dos fármacos , Junção Neuromuscular/genética , Fator 6 de Transcrição de Octâmero/metabolismo , Dobramento de Proteína/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-jun/metabolismo , Fatores de Transcrição de Fator Regulador X , Teste de Desempenho do Rota-Rod , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção , Proteína 1 de Ligação a X-Box
19.
J Comp Neurol ; 520(18): 4184-203, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22592645

RESUMO

Myelination is a cellular adaptation allowing rapid conduction along axons. We have investigated peripheral axons of the zebrafish maxillary barbel (ZMB), an optically clear sensory appendage. Each barbel carries taste buds, solitary chemosensory cells, and epithelial nerve endings, all of which regenerate after amputation (LeClair and Topczewski [2010] PLoS One 5:e8737). The ZMB contains axons from the facial nerve; however, myelination within the barbel itself has not been established. Transcripts of myelin basic protein (mbp) are expressed in normal and regenerating adult barbels, indicating activity in both maintenance and repair. Myelin was confirmed in situ by using toluidine blue, an anti-MBP antibody, and transmission electron microscopy (TEM). The adult ZMB contains ∼180 small-diameter axons (<2 µm), approximately 60% of which are myelinated. Developmental myelination was observed via whole-mount immunohistochemistry 4-6 weeks postfertilization, showing myelin sheaths lagging behind growing axons. Early-regenerating axons (10 days postsurgery), having no or few myelin layers, were disorganized within a fibroblast-rich collagenous scar. Twenty-eight days postsurgery, barbel axons had grown out several millimeters and were organized with compact myelin sheaths. Fiber types and axon areas were similar between normal and regenerated tissue; within 4 weeks, regenerating axons restored ∼85% of normal myelin thickness. Regenerating barbels express multiple promyelinating transcription factors (sox10, oct6 = pou3f1; krox20a/b = egr2a/b) typical of Schwann cells. These observations extend our understanding of the zebrafish peripheral nervous system within a little-studied sensory appendage. The accessible ZMB provides a novel context for studying axon regeneration, Schwann cell migration, and remyelination in a model vertebrate.


Assuntos
Axônios/fisiologia , Regulação da Expressão Gênica/fisiologia , Proteína Básica da Mielina/metabolismo , Regeneração Nervosa/fisiologia , Traumatismos dos Nervos Periféricos/metabolismo , Traumatismos dos Nervos Periféricos/fisiopatologia , Animais , Axônios/ultraestrutura , Proteína 2 de Resposta de Crescimento Precoce/genética , Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Larva , Microscopia Eletrônica de Transmissão , Proteína Básica da Mielina/genética , Fator 6 de Transcrição de Octâmero/genética , Fator 6 de Transcrição de Octâmero/metabolismo , Fatores de Transcrição SOXE/genética , Fatores de Transcrição SOXE/metabolismo , Células de Schwann/metabolismo , Células de Schwann/ultraestrutura , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
20.
Mol Cell Biol ; 32(13): 2618-27, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22566684

RESUMO

Otx2 plays essential roles in rostral brain development, and its counteraction with Gbx2 has been suggested to determine the midbrain-hindbrain boundary (MHB) in vertebrates. We previously identified the FM enhancer that is conserved among vertebrates and drives Otx2 transcription in forebrain/midbrain from the early somite stage. In this study, we found that the POU homeodomain of class III POU factors (Brn1, Brn2, Brn4, and Oct6) associates with noncanonical target sequence TAATTA in the FM enhancer. MicroRNA-mediated knockdown of Brn2 and Oct6 diminished the FM enhancer activity in anterior neural progenitor cells (NPCs) differentiated from P19 cells. The class III POU factors associate with the FM enhancer in forebrain and midbrain but not in hindbrain. We also demonstrated that the Gbx2 homeodomain recognizes the same target TAATTA in the FM enhancer, and Gbx2 associates with the FM enhancer in hindbrain. Gbx2 misexpression in the anterior NPCs repressed the FM enhancer activity and inhibited Brn2 association with the enhancer, whereas Gbx2 knockdown caused ectopic Brn2 association in the posterior NPCs. These results suggest that class III POU factors and Gbx2 share the same target site, TAATTA, in the FM enhancer and that their region-specific binding restricts Otx2 expression at the MHB.


Assuntos
Proteínas de Homeodomínio/metabolismo , Mesencéfalo/metabolismo , Fatores de Transcrição Otx/genética , Fatores do Domínio POU/metabolismo , Prosencéfalo/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação/genética , Ligação Competitiva , Linhagem Celular , Sequência Conservada , Primers do DNA/genética , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Proteínas de Homeodomínio/antagonistas & inibidores , Proteínas de Homeodomínio/genética , Glicoproteínas de Membrana/genética , Mesencéfalo/embriologia , Camundongos , Camundongos Transgênicos , Modelos Neurológicos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/metabolismo , Fator 6 de Transcrição de Octâmero/antagonistas & inibidores , Fator 6 de Transcrição de Octâmero/genética , Fator 6 de Transcrição de Octâmero/metabolismo , Fatores do Domínio POU/antagonistas & inibidores , Fatores do Domínio POU/classificação , Fatores do Domínio POU/genética , Prosencéfalo/embriologia , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...