Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 143
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 54(3): 526-35, 2014 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-24703953

RESUMEN

The discreteness of cell fates is an inherent and fundamental feature of multicellular organisms. Here we show that cross-antagonistic mechanisms of actions of MyoD and PPARγ, which are the master regulators of muscle and adipose differentiation, respectively, confer robustness to the integrity of cell differentiation. Simultaneous expression of MyoD and PPARγ in mesenchymal stem/stromal cells led to the generation of a mixture of multinucleated myotubes and lipid-filled adipocytes. Interestingly, hybrid cells (i.e., lipid-filled myotubes) were not generated, suggesting that these differentiation programs are mutually exclusive. Mechanistically, although exogenously expressed MyoD was rapidly degraded in adipocytes through ubiquitin-proteasome pathways, exogenously expressed PPARγ was not downregulated in myotubes. In PPARγ-expressing myotubes, PPARγ-dependent histone hyperacetylation was inhibited in a subset of adipogenic gene loci, including that of C/EBPα, an essential effector of PPARγ. Thus, the cross-repressive interactions between MyoD- and PPARγ-induced differentiation programs ensure discrete cell-fate decisions.


Asunto(s)
Diferenciación Celular , Células Madre Mesenquimatosas/fisiología , Proteína MioD/metabolismo , PPAR gamma/metabolismo , Acetilación , Adipocitos/metabolismo , Animales , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Línea Celular , Regulación hacia Abajo , Epigénesis Genética , Células HEK293 , Humanos , Ratones , Fibras Musculares Esqueléticas/metabolismo , Proteolisis , Iniciación de la Transcripción Genética
2.
Genes Cells ; 25(2): 86-99, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31788928

RESUMEN

Neurotrophic signaling regulates neural cell behaviors in development and physiology, although its role in regeneration has not been fully investigated. Here, we examined the role of neurotrophic signaling in Xenopus laevis tadpole tail regeneration. After the tadpole tails were amputated, the expression of neurotrophin ligand family genes, especially ngf and bdnf, was up-regulated as regeneration proceeded. Moreover, notochordal expression of the NGF receptor gene TrkA, but not that of other neurotrophin receptor genes TrkB and TrkC, became prominent in the regeneration bud, a structure arising from the tail stump after tail amputation. The regenerated tail length was significantly shortened by the pan-Trk inhibitor K252a or the TrkA inhibitor GW-441756, but not by the TrkB inhibitor ANA-12, suggesting that TrkA signaling is involved in elongation of regenerating tails. Furthermore, during Xenopus laevis embryonic development, TrkA expression was detected in the dorsal mesoderm at the gastrula stage and in the notochord at the neurula stage, and its knockdown led to gastrulation defects with subsequent shortening of the body axis length. These results suggest that Xenopus laevis TrkA signaling, which can act in the mesoderm/notochord, plays a key role in body axis elongation during embryogenesis as well as tail elongation during tadpole regeneration.


Asunto(s)
Desarrollo Embrionario/genética , Larva/genética , Receptor trkA/genética , Receptor trkA/metabolismo , Regeneración/genética , Transducción de Señal , Cola (estructura animal)/fisiología , Xenopus laevis/anomalías , Xenopus laevis/genética , Animales , Azepinas/farmacología , Benzamidas/farmacología , Carbazoles/farmacología , Regulación del Desarrollo de la Expresión Génica , Alcaloides Indólicos/farmacología , Factor de Crecimiento Nervioso/genética , Proteínas del Tejido Nervioso/genética , Receptor trkA/antagonistas & inhibidores , Receptor trkC/genética , Receptores de Factor de Crecimiento Nervioso/genética , Transducción de Señal/efectos de los fármacos , Cola (estructura animal)/anatomía & histología
3.
Genes Dev ; 27(18): 1949-58, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-24029916

RESUMEN

Spermatogonial stem cells (SSCs) present the potential to acquire pluripotency under specific culture conditions. However, the frequency of pluripotent cell derivation is low, and the mechanism of SSC reprogramming remains unknown. In this study, we report that induction of global DNA hypomethylation in germline stem (GS) cells (cultured SSCs) induces pluripotent cell derivation. When DNA demethylation was triggered by Dnmt1 depletion, GS cells underwent apoptosis. However, GS cells were converted into embryonic stem (ES)-like cells by double knockdown of Dnmt1 and p53. This treatment down-regulated Dmrt1, a gene involved in sexual differentiation, meiosis, and pluripotency. Dmrt1 depletion caused apoptosis of GS cells, but a combination of Dmrt1 and p53 depletion also induced pluripotency. Functional screening of putative Dmrt1 target genes revealed that Dmrt1 depletion up-regulates Sox2. Sox2 transfection up-regulated Oct4 and produced pluripotent cells. This conversion was enhanced by Oct1 depletion, suggesting that the balance of Oct proteins maintains SSC identity. These results suggest that spontaneous SSC reprogramming is caused by unstable DNA methylation and that a Dmrt1-Sox2 cascade is critical for regulating pluripotency in SSCs.


Asunto(s)
Células Madre Pluripotentes/fisiología , Factores de Transcripción/metabolismo , Animales , Línea Celular , Reprogramación Celular/genética , Metilación de ADN , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Factor 1 de Transcripción de Unión a Octámeros/genética , Factor 1 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Espermatogonias/metabolismo , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
4.
J Biol Chem ; 294(1): 195-209, 2019 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-30409903

RESUMEN

Lineage specification of the three germ layers occurs during early embryogenesis and is critical for normal development. The nucleosome remodeling and deacetylase (NuRD) complex is a repressive chromatin modifier that plays a role in lineage commitment. However, the role of chromodomain helicase DNA-binding protein 4 (CHD4), one of the core subunits of the NuRD complex, in neural lineage commitment is poorly understood. Here, we report that the CHD4/NuRD complex plays a critical role in neural differentiation of mouse embryonic stem cells (ESCs). We found that RNAi-mediated Chd4 knockdown suppresses neural differentiation, as did knockdown of methyl-CpG-binding domain protein Mbd3, another NuRD subunit. Chd4 and Mbd3 knockdowns similarly affected changes in global gene expression during neural differentiation and up-regulated several mesendodermal genes. However, inhibition of mesendodermal genes by knocking out the master regulators of mesendodermal lineages, Brachyury and Eomes, through a CRISPR/Cas9 approach could not restore the impaired neural differentiation caused by the Chd4 knockdown, suggesting that CHD4 controls neural differentiation by not repressing other lineage differentiation processes. Notably, Chd4 knockdown increased the acetylation levels of p53, resulting in increased protein levels of p53. Double knockdown of Chd4 and p53 restored the neural differentiation rate. Furthermore, overexpression of BCL2, a downstream factor of p53, partially rescued the impaired neural differentiation caused by the Chd4 knockdown. Our findings reveal that the CHD4/NuRD complex regulates neural differentiation of ESCs by down-regulating p53.


Asunto(s)
Diferenciación Celular , ADN Helicasas/metabolismo , Regulación hacia Abajo , Neuronas/metabolismo , Nucleosomas/metabolismo , Proteína p53 Supresora de Tumor/biosíntesis , Animales , Línea Celular , ADN Helicasas/genética , Técnicas de Silenciamiento del Gen , Ratones , Células Madre Embrionarias de Ratones , Neuronas/citología , Nucleosomas/genética , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteína p53 Supresora de Tumor/genética
5.
J Biol Chem ; 293(22): 8342-8361, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29674317

RESUMEN

Epithelia contribute to physical barriers that protect internal tissues from the external environment and also support organ structure. Accordingly, establishment and maintenance of epithelial architecture are essential for both embryonic development and adult physiology. Here, using gene knockout and knockdown techniques along with gene profiling, we show that extracellular signal-regulated kinase 3 (ERK3), a poorly characterized atypical mitogen-activated protein kinase (MAPK), regulates the epithelial architecture in vertebrates. We found that in Xenopus embryonic epidermal epithelia, ERK3 knockdown impairs adherens and tight-junction protein distribution, as well as tight-junction barrier function, resulting in epidermal breakdown. Moreover, in human epithelial breast cancer cells, inhibition of ERK3 expression induced thickened epithelia with aberrant adherens and tight junctions. Results from microarray analyses suggested that transcription factor AP-2α (TFAP2A), a transcriptional regulator important for epithelial gene expression, is involved in ERK3-dependent changes in gene expression. Of note, TFAP2A knockdown phenocopied ERK3 knockdown in both Xenopus embryos and human cells, and ERK3 was required for full activation of TFAP2A-dependent transcription. Our findings reveal that ERK3 regulates epithelial architecture, possibly together with TFAP2A.


Asunto(s)
Neoplasias de la Mama/patología , Embrión no Mamífero/enzimología , Células Epiteliales/química , Proteína Quinasa 6 Activada por Mitógenos/metabolismo , Factor de Transcripción AP-2/metabolismo , Xenopus laevis/fisiología , Animales , Neoplasias de la Mama/enzimología , Sistemas CRISPR-Cas , Adhesión Celular , Membrana Celular , Células Cultivadas , Embrión no Mamífero/citología , Células Epiteliales/enzimología , Células Epiteliales/patología , Femenino , Células Hep G2 , Humanos , Proteína Quinasa 6 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 6 Activada por Mitógenos/genética , Uniones Estrechas , Factor de Transcripción AP-2/antagonistas & inhibidores , Factor de Transcripción AP-2/genética , Xenopus laevis/embriología
6.
EMBO Rep ; 2017 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-28794203

RESUMEN

The well-known link between longevity and the Sir2 histone deacetylase family suggests that histone deacetylation, a modification associated with repressed chromatin, is beneficial to longevity. However, the molecular links between histone acetylation and longevity remain unclear. Here, we report an unexpected finding that the MYST family histone acetyltransferase complex (MYS-1/TRR-1 complex) promotes rather than inhibits stress resistance and longevity in Caenorhabditis elegans Our results show that these beneficial effects are largely mediated through transcriptional up-regulation of the FOXO transcription factor DAF-16. MYS-1 and TRR-1 are recruited to the promoter regions of the daf-16 gene, where they play a role in histone acetylation, including H4K16 acetylation. Remarkably, we also find that the human MYST family Tip60/TRRAP complex promotes oxidative stress resistance by up-regulating the expression of FOXO transcription factors in human cells. Tip60 is recruited to the promoter regions of the foxo1 gene, where it increases H4K16 acetylation levels. Our results thus identify the evolutionarily conserved role of the MYST family acetyltransferase as a key epigenetic regulator of DAF-16/FOXO transcription factors.

7.
J Biol Chem ; 292(27): 11300-11309, 2017 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-28507100

RESUMEN

Intermittent fasting (IF) is a dietary restriction regimen that extends the lifespans of Caenorhabditis elegans and mammals by inducing changes in gene expression. However, how IF induces these changes and promotes longevity remains unclear. One proposed mechanism involves gene regulation by microRNAs (miRNAs), small non-coding RNAs (∼22 nucleotides) that repress gene expression and whose expression can be altered by fasting. To test this proposition, we examined the role of the miRNA machinery in fasting-induced transcriptional changes and longevity in C. elegans We revealed that fasting up-regulated the expression of the miRNA-induced silencing complex (miRISC) components, including Argonaute and GW182, and the miRNA-processing enzyme DRSH-1 (the ortholog of the Drosophila Drosha enzyme). Our lifespan measurements demonstrated that IF-induced longevity was suppressed by knock-out or knockdown of miRISC components and was completely inhibited by drsh-1 ablation. Remarkably, drsh-1 ablation inhibited the fasting-induced changes in the expression of the target genes of DAF-16, the insulin/IGF-1 signaling effector in C. elegans Fasting-induced transcriptome alterations were substantially and modestly suppressed in the drsh-1 null mutant and the null mutant of ain-1, a gene encoding GW182, respectively. Moreover, miRNA array analyses revealed that the expression levels of numerous miRNAs changed after 2 days of fasting. These results indicate that components of the miRNA machinery, especially the miRNA-processing enzyme DRSH-1, play an important role in mediating IF-induced longevity via the regulation of fasting-induced changes in gene expression.


Asunto(s)
Proteínas de Caenorhabditis elegans/biosíntesis , Caenorhabditis elegans/metabolismo , Proteínas Portadoras/biosíntesis , Ayuno , Regulación de la Expresión Génica , Longevidad/fisiología , MicroARNs/metabolismo , Ribonucleasa III/biosíntesis , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas Portadoras/genética , MicroARNs/genética , Ribonucleasa III/genética
8.
Genes Cells ; 22(2): 189-202, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28097777

RESUMEN

The Activin/Nodal/TGF-ß signaling pathway plays a major role in maintaining mouse epiblast stem cells (EpiSCs). The EpiSC-maintaining medium, which contains Activin A and bFGF, induces differentiation of mouse embryonic stem cells (ESCs) to EpiSCs. Here, we show that Activin A also has an ability to efficiently propagate ESCs without differentiation to EpiSCs when combined with a MEK inhibitor PD0325901. ESCs cultured in Activin+PD retained high-level expression of naive pluripotency-related transcription factors. Genomewide analysis showed that the gene expression profile of ESCs cultured in Activin+PD resembles that of ESCs cultured in 2i. ESCs cultured in Activin+PD also showed features common to the naive pluripotency of ESCs, including the preferential usage of the Oct4 distal enhancer and the self-renewal response to Wnt pathway activation. Our finding shows a role of Activin/Nodal/TGF-ß signaling in stabilizing self-renewal gene regulatory networks in ESCs.


Asunto(s)
Activinas/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/metabolismo , Activinas/química , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Estratos Germinativos/citología , Estratos Germinativos/metabolismo , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Células Madre Embrionarias de Ratones/citología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Vía de Señalización Wnt
9.
Genes Cells ; 22(2): 210-219, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28105749

RESUMEN

Dietary restriction regimens lead to enhanced stress resistance and extended life span in many species through the regulation of fasting and/or diet-responsive mechanisms. The fasting stimulus is perceived by sensory neurons and causes behavioral and metabolic adaptations. Octopamine (OA), one of the Caenorhabditis elegans neurotransmitters, is involved in behavioral adaptations, and its levels are increased under fasting conditions. However, it remains largely unknown how OA contributes to the fasting responses. In this study, we found that OA administration enhanced organismal resistance to oxidative stress. This enhanced resistance was suppressed by a mutation of the OA receptors, SER-3 and SER-6. Moreover, we found that OA administration promoted the nuclear translocation of DAF-16, the key transcription factor in fasting responses, and that the OA-induced enhancement of stress resistance required DAF-16. Altogether, our results suggest that OA signaling, which is triggered by the absence of food, shifts the organismal state to a more protective one to prepare for environmental stresses.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Factores de Transcripción Forkhead/metabolismo , Octopamina/farmacología , Estrés Oxidativo/efectos de los fármacos , Animales , Caenorhabditis elegans , Ayuno/fisiología , Longevidad , Mutación , Estrés Oxidativo/fisiología , Transducción de Señal
10.
Genes Cells ; 21(4): 302-10, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26810578

RESUMEN

Brown adipocytes and beige adipocytes can expend energy, generate heat, and increase whole-body energy expenditure. The detailed mechanisms of adipogenesis and thermogenesis of these cells are still obscure. Here, we show that Src family kinases (SFKs) regulate both brown adipogenesis and browning of white adipocytes. To identify factors involved in brown adipogenesis, we first examined the effect of several chemical inhibitors on the differentiation of brown preadipocytes isolated from mouse brown adipose tissue (BAT) and found that treatment with PP2, the specific inhibitor of SFKs, promoted the differentiation. Another inhibitor of SFKs, PP1, also promoted the brown adipogenesis, whereas an inactive analogue of PP2, PP3, did not. Moreover, over-expression of C-terminal Src kinase (CSK), the negative regulator of SFKs, also promoted brown adipogenesis. Next, we examined the effect of inhibition of SFKs on the differentiation of white preadipocytes isolated from white adipose tissue (WAT). Our results showed that either PP2 treatment or CSK-over-expression generated Ucp1-positive beige adipocytes, thus inducing browning of white adipocytes. Finally, our analysis showed that the expression levels and activity of SFKs in WAT were much higher than in BAT. These results taken together suggest that SFKs regulate differentiation and browning of fat cells in vivo.


Asunto(s)
Adipocitos Blancos/enzimología , Adipogénesis , Familia-src Quinasas/metabolismo , Adipocitos Marrones/citología , Adipocitos Marrones/enzimología , Adipocitos Marrones/metabolismo , Adipocitos Blancos/citología , Adipocitos Blancos/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL
11.
J Biol Chem ; 290(52): 31173-88, 2015 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-26553868

RESUMEN

It remains controversial whether the routes from somatic cells to induced pluripotent stem cells (iPSCs) are related to the reverse order of normal developmental processes. Specifically, it remains unaddressed whether or not the differentiated cells become iPSCs through their original tissue stem cell-like state. Previous studies analyzing the reprogramming process mostly used fibroblasts; however, the stem cell characteristics of fibroblasts made it difficult to address this. Here, we generated iPSCs from mouse astrocytes, a type of glial cells, by three (OCT3/4, KLF4, and SOX2), two (OCT3/4 and KLF4), or four (OCT3/4, KLF4, and SOX2 plus c-MYC) factors. Sox1, a neural stem cell (NSC)-specific transcription factor, is transiently up-regulated during reprogramming, and Sox1-positive cells become iPSCs. The up-regulation of Sox1 is essential for OCT3/4- and KLF4-induced reprogramming. Genome-wide analysis revealed that the gene expression profile of Sox1-expressing intermediate-state cells resembles that of NSCs. Furthermore, the intermediate-state cells are able to generate neurospheres, which can differentiate into both neurons and glial cells. Remarkably, during fibroblast reprogramming, neither Sox1 up-regulation nor an increase in neurogenic potential occurs. Our results thus demonstrate that astrocytes are reprogrammed through an NSC-like state.


Asunto(s)
Astrocitos/metabolismo , Técnicas de Reprogramación Celular , Reprogramación Celular , Células-Madre Neurales/metabolismo , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/biosíntesis , Animales , Astrocitos/citología , Células Cultivadas , Factor 4 Similar a Kruppel , Ratones , Células-Madre Neurales/citología , Células Madre Pluripotentes/citología , Factores de Transcripción/genética
12.
BMC Dev Biol ; 16(1): 31, 2016 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-27653971

RESUMEN

BACKGROUND: HECT domain and ankyrin repeat containing E3 ubiquitin protein ligase 1 (HACE1) regulates a wide variety of cellular processes. It has been shown that one of the targets of HACE1 is the GTP-bound form of the small GTPase Rac1. However, the role of HACE1 in early development remains unknown. RESULTS: In situ hybridization revealed that Xenopus laevis hace1 is specifically expressed in the ectoderm at the blastula and gastrula stages and in the epidermis, branchial arch, kidney, and central nervous system at the tailbud stage. Knockdown of hace1 in Xenopus laevis embryos via antisense morpholino oligonucleotides led to defects in body axis elongation, pigment formation, and eye formation at the tadpole stage. Experiments with Keller sandwich explants showed that hace1 knockdown inhibited convergent extension, a morphogenetic movement known to be crucial for body axis elongation. In addition, time lapse imaging of whole embryos during the neurula stage indicated that hace1 knockdown also delayed neural tube closure. The defects caused by hace1 knockdown were partly rescued by knockdown of rac1. Moreover, embryos expressing a constitutively active form of Rac1 displayed phenotypes similar to those of embryos with hace1 knocked down. CONCLUSIONS: Our results suggest that Xenopus laevis hace1 plays an important role in early embryonic development, possibly via regulation of Rac1 activity.

13.
EMBO J ; 31(5): 1109-22, 2012 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-22234184

RESUMEN

The Hippo signalling pathway has emerged as a key regulator of organ size, tissue homeostasis, and patterning. Recent studies have shown that two effectors in this pathway, YAP/TAZ, modulate Wnt/ß-catenin signalling through their interaction with ß-catenin or Dishevelled, depending on biological contexts. Here, we identify a novel mechanism through which Hippo signalling inhibits Wnt/ß-catenin signalling. We show that YAP and TAZ, the transcriptional co-activators in the Hippo pathway, suppress Wnt signalling without suppressing the stability of ß-catenin but through preventing its nuclear translocation. Our results show that YAP/TAZ binds to ß-catenin, thereby suppressing Wnt-target gene expression, and that the Hippo pathway-stimulated phosphorylation of YAP, which induces cytoplasmic translocation of YAP, is required for the YAP-mediated inhibition of Wnt/ß-catenin signalling. We also find that downregulation of Hippo signalling correlates with upregulation of ß-catenin signalling in colorectal cancers. Remarkably, our analysis demonstrates that phosphorylated YAP suppresses nuclear translocation of ß-catenin by directly binding to it in the cytoplasm. These results provide a novel mechanism, in which Hippo signalling antagonizes Wnt signalling by regulating nuclear translocation of ß-catenin.


Asunto(s)
Regulación de la Expresión Génica , Proteínas Nucleares/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Aciltransferasas , Proteínas de Ciclo Celular , Línea Celular , Humanos , Modelos Biológicos
14.
Genes Cells ; 20(4): 324-39, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25652037

RESUMEN

Cannabinoid receptor interacting protein 1 (CNRIP1), which has been originally identified as the binding partner of cannabinoid receptor 1 (CNR1), is evolutionarily conserved throughout vertebrates, but its physiological function has been unknown. Here, we identify a developmental role of CNRIP1 using Xenopus laevis embryos. During early embryogenesis, expression of Xenopus laevis cnrip1 is highly restricted to the animal region of gastrulae where neural and eye induction occur, and afterward it is seen in neural and other tissues with a temporally and spatially regulated pattern. Morpholino-mediated knockdown experiments indicate that cnrip1 has an essential role in early eye and neural development by regulating the onset of expression of key transcription factor genes, sox2, otx2, pax6 and rax. Also, over-expression experiments suggest that cnrip1 has a potential to expand sox2, otx2, pax6 and rax expression. These results suggest an instructive role of Xenopus laevis cnrip1 in early eye and neural development. Furthermore, Xenopus laevis cnr1 knockdown leads to eye defects, which are partly similar to, but milder than, those caused by cnrip1 knockdown, suggesting a possible functional similarity between CNRIP1 and CNR1. This study is the first characterization of an in vivo role of CNRIP1 in the context of whole organisms.


Asunto(s)
Proteínas Portadoras/metabolismo , Ojo/embriología , Gástrula/metabolismo , Neurogénesis , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Xenopus laevis/metabolismo , Animales , Proteínas Portadoras/genética , Ojo/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Xenopus/genética
15.
Proc Natl Acad Sci U S A ; 110(49): 19884-9, 2013 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-24248350

RESUMEN

A major function of innate immune receptors is to recognize pathogen-associated molecular patterns and then evoke immune responses appropriate to the nature of the invading pathogen(s). Because innate immune cells express various types of these receptors, distinct combinations of signaling pathways are activated in response to a given pathogen. Although the conventional wisdom is that these signaling pathways cooperate with one another to ensure an effective host response, a more nuanced view recognizes antagonism between the individual pathways, where the attenuation of a signaling pathway(s) by others may shape the immune response. In this study, we show that, on Listeria monocytogenes infection, Toll-like receptor-triggered MyD88 signaling pathways suppress type I IFN gene induction, which is detrimental to macrophage bactericidal activity. These pathways target and suppress the IFN regulatory factor 3 (IRF3) transcription factor that is activated by the stimulator of IFN genes-TANK-binding kinase-1 kinase pathway. We also provide evidence for the involvement of the MAPK phosphatase family members, which renders IRF3 hypophosphorylated on Toll-like receptor signaling by enhancing the formation of an MAPK phosphatase-IRF3-TANK-binding kinase-1 ternary complex. This study, therefore, reveals a hitherto unrecognized and important contribution of a beneficial innate signaling interference against bacterial infections.


Asunto(s)
Inmunidad Innata/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Complejos Multiproteicos/inmunología , Transducción de Señal/inmunología , Receptores Toll-Like/metabolismo , Animales , Ensayo de Unidades Formadoras de Colonias , Fosfatasa 1 de Especificidad Dual/metabolismo , Immunoblotting , Inmunoprecipitación , Factor 3 Regulador del Interferón/antagonistas & inhibidores , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multiproteicos/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
16.
Nihon Rinsho ; 74(9): 1565-1571, 2016 Sep.
Artículo en Japonés | MEDLINE | ID: mdl-30557494

RESUMEN

Aging is inevitable for almost all the organisms. Aging processes vary from one organism to another. It has been shown that many organisms have lifespan-regulation mechanisms in common. Dietary restriction is the most robust non-genetic interventions to ameliorate aging and aging-associated diseases in many organisms. Reducing overall calorie intake was thought to be a crucial factor for dietary restriction -induced longevity. However, recent studies indicate that both the meal timing and lowered intake of specific nutrients rather than overall calorie intake are important for lifespan regulation. These findings shed light on new aspects of mechanisms underlying dietary restriction -induced longevity. The genetic factors are also shown to be involved in dietary restriction-induced longevity. This review summarizes the dietary interventions and the genetic factors that mediate lifespan extension by dietary interventions.


Asunto(s)
Envejecimiento , Restricción Calórica , Longevidad , Envejecimiento/fisiología , Animales , Humanos , Longevidad/genética , Longevidad/fisiología , Transducción de Señal
17.
J Biol Chem ; 289(48): 33320-32, 2014 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-25342745

RESUMEN

Trp-Asp (WD) repeat protein 68 (WDR68) is an evolutionarily conserved WD40 repeat protein that binds to several proteins, including dual specificity tyrosine phosphorylation-regulated protein kinase (DYRK1A), MAPK/ERK kinase kinase 1 (MEKK1), and Cullin4-damage-specific DNA-binding protein 1 (CUL4-DDB1). WDR68 affects multiple and diverse physiological functions, such as controlling anthocyanin synthesis in plants, tissue growth in insects, and craniofacial development in vertebrates. However, the biochemical basis and the regulatory mechanism of WDR68 activity remain largely unknown. To better understand the cellular function of WDR68, here we have isolated and identified cellular WDR68 binding partners using a phosphoproteomic approach. More than 200 cellular proteins with wide varieties of biochemical functions were identified as WDR68-binding protein candidates. Eight T-complex protein 1 (TCP1) subunits comprising the molecular chaperone TCP1 ring complex/chaperonin-containing TCP1 (TRiC/CCT) were identified as major WDR68-binding proteins, and phosphorylation sites in both WDR68 and TRiC/CCT were identified. Co-immunoprecipitation experiments confirmed the binding between TRiC/CCT and WDR68. Computer-aided structural analysis suggested that WDR68 forms a seven-bladed ß-propeller ring. Experiments with a series of deletion mutants in combination with the structural modeling showed that three of the seven ß-propeller blades of WDR68 are essential and sufficient for TRiC/CCT binding. Knockdown of cellular TRiC/CCT by siRNA caused an abnormal WDR68 structure and led to reduction of its DYRK1A-binding activity. Concomitantly, nuclear accumulation of WDR68 was suppressed by the knockdown of TRiC/CCT, and WDR68 formed cellular aggregates when overexpressed in the TRiC/CCT-deficient cells. Altogether, our results demonstrate that the molecular chaperone TRiC/CCT is essential for correct protein folding, DYRK1A binding, and nuclear accumulation of WDR68.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Núcleo Celular/metabolismo , Chaperonina con TCP-1/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células COS , Núcleo Celular/genética , Chaperonina con TCP-1/genética , Chlorocebus aethiops , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Quinasa 1 de Quinasa de Quinasa MAP/genética , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Modelos Moleculares , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Estructura Secundaria de Proteína , Proteínas Tirosina Quinasas/genética , Relación Estructura-Actividad , Quinasas DyrK
18.
J Biol Chem ; 289(46): 32064-32072, 2014 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-25271155

RESUMEN

The circadian transcription factor CLOCK exhibits a circadian oscillation in its phosphorylation levels. Although it remains unclear whether this phosphorylation contributes to circadian rhythm generation, it has been suggested to be involved in transcriptional activity, intracellular localization, and degradative turnover of CLOCK. Here, we obtained direct evidence that CLOCK phosphorylation may be essential for autonomous circadian oscillation in clock gene expression. Importantly, we found that the circadian transcriptional repressors Cryptochrome (CRY) and Period (PER) showed an opposite effect on CLOCK phosphorylation; CRY impaired BMAL1-dependent CLOCK phosphorylation, whereas PER protected the phosphorylation against CRY. Interestingly, unlike PER1 and PER2, PER3 did not exert a protective action, which correlates with the phenotypic differences among mice lacking the Per genes. Further studies on the regulatory mechanism of CLOCK phosphorylation would thus lead to elucidation of the mechanism of CRY-mediated transcriptional repression and an understanding of the true role of PER in the negative feedback system.


Asunto(s)
Proteínas CLOCK/metabolismo , Criptocromos/metabolismo , Proteínas Circadianas Period/metabolismo , Factores de Transcripción ARNTL/metabolismo , Animales , Encéfalo/metabolismo , Núcleo Celular/metabolismo , Ratones , Ratones Transgénicos , Modelos Teóricos , Células 3T3 NIH , Oscilometría , Fosforilación , Regiones Promotoras Genéticas , Núcleo Supraquiasmático/metabolismo
19.
Development ; 139(16): 2988-98, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22764049

RESUMEN

Sox9 acts together with Sox5 or Sox6 as a master regulator for chondrogenesis; however, the inter-relationship among these transcription factors remains unclear. Here, we show that the protein kinase MLTK plays an essential role in the onset of chondrogenesis through triggering the induction of Sox6 expression by Sox9. We find that knockdown of MLTK in Xenopus embryos results in drastic loss of craniofacial cartilages without defects in neural crest development. We also find that Sox6 is specifically induced during the onset of chondrogenesis, and that the Sox6 induction is inhibited by MLTK knockdown. Remarkably, Sox6 knockdown phenocopies MLTK knockdown. Moreover, we find that ectopic expression of MLTK induces Sox6 expression in a Sox9-dependent manner. Our data suggest that p38 and JNK pathways function downstream of MLTK during chondrogenesis. These results identify MLTK as a novel key regulator of chondrogenesis, and reveal its action mechanism in chondrocyte differentiation during embryonic development.


Asunto(s)
Condrogénesis/fisiología , Quinasas Quinasa Quinasa PAM/metabolismo , Factores de Transcripción SOXD/biosíntesis , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Xenopus laevis/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Movimiento Celular/genética , Movimiento Celular/fisiología , Condrocitos/citología , Condrocitos/metabolismo , Condrogénesis/genética , Cartilla de ADN/genética , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Hibridación in Situ , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/genética , Sistema de Señalización de MAP Quinasas , Cresta Neural/citología , Cresta Neural/embriología , Cresta Neural/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Factores de Transcripción SOXD/antagonistas & inhibidores , Factores de Transcripción SOXD/genética , Proteínas de Xenopus/genética , Xenopus laevis/genética
20.
Genes Cells ; 19(8): 637-49, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24962468

RESUMEN

Retinoic acid (RA) signaling regulates a variety of developmental processes through controlling the expression of numerous genes. Here, we have identified and characterized RA-responsive genes in mouse kidney development. Analysis of isolated embryonic kidneys cultured in the presence and absence of RA identified 33 candidates of RA-responsive genes. Most of these candidate genes were down-regulated by treatment with the RA receptor antagonist. Many of them have potential binding sites for Elf5, one of the RA-responsive genes, in their promoter region. Whole-mount in situ hybridization showed that specific expression of Elf5 in the ureteric trunk depends on RA. RA-dependent expression in the ureteric trunk was also showed for the sodium channel subunit Scnn1b, which has been shown to be the marker gene of the collecting duct. In contrast, the expression of Ecm1, Tnfsf13b and IL-33 was detected in the stromal mesenchymal cells. Both Tnfsf13b and IL-33 were previously shown to cause nuclear factor κB (NF-κB) activation. We have showed that the inhibition of NF-κB signaling with specific inhibitors suppresses branching morphogenesis of the ureteric bud. Our study thus identifies and characterizes RA-dependent up-regulated genes in kidney development, and suggests an involvement of NF-κB signaling in the branching morphogenesis.


Asunto(s)
Riñón/metabolismo , Organogénesis/genética , Tretinoina/metabolismo , Animales , Células Cultivadas , Proteínas de Unión al ADN/genética , Genes del Desarrollo , Riñón/embriología , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Elementos de Respuesta , Factores de Transcripción/genética , Transcriptoma , Uréter/embriología , Uréter/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA