Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Vet Res ; 55(1): 103, 2024 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-39155369

RESUMEN

Porcine reproductive and respiratory syndrome virus (PRRSV) is a severe disease with substantial economic consequences for the swine industry. The DEAD-box helicase 3 (DDX3X) is an RNA helicase that plays a crucial role in regulating RNA metabolism, immunological response, and even RNA virus infection. However, it is unclear whether it contributes to PRRSV infection. Recent studies have found that the expression of DDX3X considerably increases in Marc-145 cells when infected with live PRRSV strains Ch-1R and SD16; however, it was observed that inactivated viruses did not lead to any changes. By using the RK-33 inhibitor or DDX3X-specific siRNAs to reduce DDX3X expression, there was a significant decrease in the production of PRRSV progenies. In contrast, the overexpression of DDX3X in host cells substantially increased the proliferation of PRRSV. A combination of transcriptomics and metabolomics investigations revealed that in PRRSV-infected cells, DDX3X gene silencing severely affected biological processes such as ferroptosis, the FoxO signalling pathway, and glutathione metabolism. The subsequent transmission electron microscopy (TEM) imaging displayed the typical ferroptosis features in PRRSV-infected cells, such as mitochondrial shrinkage, reduction or disappearance of mitochondrial cristae, and cytoplasmic membrane rupture. Conversely, the mitochondrial morphology was unchanged in DDX3X-inhibited cells. Furthermore, silencing of the DDX3X gene changed the expression of ferroptosis-related genes and inhibited the virus proliferation, while the drug-induced ferroptosis inversely promoted PRRSV replication. In summary, these results present an updated perspective of how PRRSV infection uses DDX3X for self-replication, potentially leading to ferroptosis via various mechanisms that promote PRRSV replication.


Asunto(s)
ARN Helicasas DEAD-box , Ferroptosis , Virus del Síndrome Respiratorio y Reproductivo Porcino , Replicación Viral , Virus del Síndrome Respiratorio y Reproductivo Porcino/fisiología , Animales , ARN Helicasas DEAD-box/metabolismo , ARN Helicasas DEAD-box/genética , Ferroptosis/fisiología , Porcinos , Síndrome Respiratorio y de la Reproducción Porcina/virología , Síndrome Respiratorio y de la Reproducción Porcina/metabolismo , Línea Celular
2.
Am J Physiol Cell Physiol ; 325(1): C286-C299, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37306391

RESUMEN

Primordial germ cell 7 (PGC7)(Dppa3 or Stella) is a small inherently disordered protein that is mainly expressed in oocytes and plays a vital role in the regulation of DNA methylation reprogramming in imprinted loci through interaction with other proteins. Most of PGC7-deficient zygotes are blocked at two-cell stage with an increased tri-methylation at lysine 27 of histone H3 (H3K27me3) level in the nucleus. Our previous work has indicated that PGC7 interacts with yin-yang1 (YY1) that is essential for the recruitment of enhancer of zeste homolog 2 (EZH2)-containing Polycomb repressive complex 2 (PRC2) to H3K27me3 modification sites. Here, we found that the presence of PGC7 weakened the interaction between YY1 and PRC2 without disrupting the assembly of core subunits of the PRC2 complex. In addition, PGC7 promoted AKT to phosphorylate serine 21 of EZH2, resulting in inhibition of EZH2 activity and the dissociation of EZH2 from YY1, thereby decreasing H3K27me3 level. In zygotes, the PGC7-deficient and AKT inhibitor MK2206 both promoted EZH2 to enter the pronuclei but without disturbing the subcellular localization of YY1 and caused an increase in the level of H3K27me3 in the pronuclei, as well as inhibition of the expression of zygote-activating genes regulated by H3K27me3 in two-cell embryos. In summary, PGC7 could affect zygotic genome activation during early embryonic development by regulating the level of H3K27me3 through regulation of PRC2 recruitment, EZH2 activity, and subcellular localization.NEW & NOTEWORTHY PGC7 and YY1 interaction inhibits recruitment of PRC2 by YY1. PGC7 promotes AKT and EZH2 interaction to increase pEZH2-S21 level, which weakens YY1 and EZH2 interaction, thereby decreasing H3K27me3 level. In zygotes, the PGC7-deficient and AKT inhibitor MK2206 promote EZH2 to enter the pronuclei, and increase H3K27me3 level in the pronuclei, as well as inhibition of the expression of zygote-activating genes regulated by H3K27me3 in two-cell embryos, which ultimately affects early embryo development.


Asunto(s)
Histonas , Complejo Represivo Polycomb 2 , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Histonas/genética , Histonas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Metilación de ADN , Células Germinativas/metabolismo
3.
Front Oncol ; 13: 1106249, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37205188

RESUMEN

Background: Laryngeal squamous cell carcinoma (LSCC) is one of the most frequent head and neck cancers worldwide. Long non-coding RNAs (lncRNAs) play a critical role in tumorigenesis. However, the clinical significance of lncRNAs in LSCC remains largely unknown. Methods: In this study, transcriptome sequencing was performed on 107 LSCC and paired adjacent normal mucosa (ANM) tissues. Furthermore, RNA expression and clinical data of 111 LSCC samples were obtained from The Cancer Genome Atlas (TCGA) database. Bioinformatics analysis were performed to construct a model for predicting the overall survival (OS) of LSCC patients. Moreover, we investigated the roles of lncRNAs in LSCC cells through loss-of-function experiments. Results: A seven-lncRNAs panel including ENSG00000233397, BARX1-DT, LSAMP-AS1, HOXB-AS4, MNX1-AS1, LINC01385, and LINC02893 was identified. The Kaplan-Meier analysis demonstrated that the seven-lncRNAs panel was significantly associated with OS (HR:6.21 [3.27-11.81], p-value<0.0001), disease-specific survival (DSS) (HR:4.34 [1.83-10.26], p-value=0.0008), and progression-free interval (PFI) (HR:3.78 [1.92-7.43], p-value=0.0001). ROC curves showed the seven-lncRNAs panel predicts OS with good specificity and sensitivity. Separately silencing the seven lncRNAs inhibited the proliferation, migration, and invasion capacity of LSCC cells. Conclusion: Collectively, this seven-lncRNAs panel is a promising signature for predicting the prognosis of LSCC patients, and these lncRNAs could serve as potential targets for LSCC treatment.

4.
Int J Mol Sci ; 24(4)2023 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-36834503

RESUMEN

DNA methylation is an epigenetic modification that plays a vital role in a variety of biological processes, including the regulation of gene expression, cell differentiation, early embryonic development, genomic imprinting, and X chromosome inactivation. PGC7 is a maternal factor that maintains DNA methylation during early embryonic development. One mechanism of action has been identified by analyzing the interactions between PGC7 and UHRF1, H3K9 me2, or TET2/TET3, which reveals how PGC7 regulates DNA methylation in oocytes or fertilized embryos. However, the mechanism by which PGC7 regulates the post-translational modification of methylation-related enzymes remains to be elucidated. This study focused on F9 cells (embryonic cancer cells), which display high levels of PGC7 expression. We found that both knockdown of Pgc7 and inhibition of ERK activity resulted in increased genome-wide DNA methylation levels. Mechanistic experiments confirmed that inhibition of ERK activity led to the accumulation of DNMT1 in the nucleus, ERK phosphorylated DNMT1 at ser717, and DNMT1 Ser717-Ala mutation promoted the nuclear localization of DNMT1. Moreover, knockdown of Pgc7 also caused downregulation of ERK phosphorylation and promoted the accumulation of DNMT1 in the nucleus. In conclusion, we reveal a new mechanism by which PGC7 regulates genome-wide DNA methylation via phosphorylation of DNMT1 at ser717 by ERK. These findings may provide new insights into treatments for DNA methylation-related diseases.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Núcleo Celular/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Impresión Genómica , Procesamiento Proteico-Postraduccional , Proteínas Cromosómicas no Histona
5.
Acta Biochim Biophys Sin (Shanghai) ; 54(7): 917-930, 2022 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-35866604

RESUMEN

The gene dosage at the imprinted Dlk1-Dio3 locus is critical for cell growth and development. A relatively high gene expression within the Dlk1-Dio3 region, especially the active expression of Gtl2, has been identified as the only reliable marker for cell pluripotency. The DNA methylation state of the IG-DNA methylated regions (DMR), which is located upstream of the Gtl2 gene, dominantly contributes to the control of gene expression in the Dlk1-Dio3 locus. However, the precise mechanism underlying the regulation of DNA methylation in the IG-DMR remains largely unknown. Here, we use the F9 embryonal carcinoma cell line, a low pluripotent cell model, to identify the mechanism responsible for DNA methylation in the IG-DMR, and find that the interaction of PGC7 with UHRF1 is involved in maintaining DNA methylation and inducing DNA hypermethylation in the IG-DMR region. PGC7 and UHRF1 cooperatively bind in the IG-DMR to regulate the methylation of DNA and histones in this imprinted region. PGC7 promotes the recruitment of DNMT1 by UHRF1 to maintain DNA methylation in the IG-DMR locus. The interaction between PGC7 and UHRF1 strengthens their binding to H3K9me3 and leads to further enrichment of H3K9me3 in the IG-DMR by recruiting the specific histone methyltransferase SETDB1. Consequently, the abundance of H3K9me3 promotes DNMT3A to bind to the IG-DMR and increases DNA methylation level in this region. In summary, we propose a new mechanism of DNA methylation regulation in the IG-DMR locus and provide further insight into the understanding of the difference in Gtl2 expression levels between high and low pluripotent cells.


Asunto(s)
Metilación de ADN , ARN Largo no Codificante , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , ADN/metabolismo , Impresión Genómica , Histona Metiltransferasas/genética , Histona Metiltransferasas/metabolismo , Histonas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
6.
Sci China Life Sci ; 65(11): 2257-2268, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35524909

RESUMEN

Animal cloning can be achieved by somatic cell nuclear transfer (SCNT), but the resulting live birth rate is relatively low. We previously improved the efficiency of bovine SCNT by exogenous melatonin treatment or by overexpression of lysine-specific demethylase 4D (KDM4D) and 4E (KDM4E). In this study, we revealed abundant alternative splicing (AS) transitions during fertilization and embryonic genome activation, and demonstrated abnormal AS in bovine SCNT embryos compared with in vitro fertilized embryos. We used the CRISPR-Cas13d RNA-targeting system to target cis-elements of ABI2 and ZNF106 pre-mRNA to modify AS, thus reducing the ratio of abnormal-isoform SCNT embryos by nearly 50% and achieving a high survival rate (11%-19%). These results indicate that this system may provide an efficient method for bovine cloning, while also paving the way for further improvements in the efficiency of SCNT.


Asunto(s)
Empalme Alternativo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Bovinos , Animales , Desarrollo Embrionario/genética , Técnicas de Transferencia Nuclear , Clonación de Organismos
7.
Int J Mol Sci ; 22(5)2021 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-33668324

RESUMEN

FOXC1, a transcription factor involved in cell differentiation and embryogenesis, is demonstrated to be a negative regulator of Nanog in this study. FOXC1 is up-regulated in retinoic acid-induced differentiation of F9 Embryonal Carcinoma (EC) cells; furthermore, FOXC1 specifically inhibits the core pluripotency factor Nanog by binding to the proximal promoter. Overexpression of FOXC1 in F9 or knockdown in 3T3 results in the down-regulation or up-regulation of Nanog mRNA and proteins, respectively. In order to explain the mechanism by which FOXC1 inhibits Nanog expression, we identified the co-repressor HDAC2 from the FOXC1 interactome. FOXC1 recruits HDAC2 to Nanog promoter to decrease H3K27ac enrichment, resulting in transcription inhibition of Nanog. To the best of our knowledge, this is the first report that FOXC1 is involved in the epigenetic regulation of gene expression.


Asunto(s)
Células Madre de Carcinoma Embrionario/metabolismo , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Histona Desacetilasa 2/metabolismo , Proteína Homeótica Nanog/genética , Regiones Promotoras Genéticas , Tretinoina/farmacología , Animales , Antineoplásicos/farmacología , Células Madre de Carcinoma Embrionario/efectos de los fármacos , Células Madre de Carcinoma Embrionario/patología , Epigénesis Genética , Factores de Transcripción Forkhead/genética , Células HEK293 , Histona Desacetilasa 2/genética , Humanos , Ratones , Células 3T3 NIH , Proteína Homeótica Nanog/metabolismo
8.
Int J Biol Macromol ; 173: 379-398, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33484802

RESUMEN

Vitamin C (VitC) is a requisite nutrient for humans and other primates. Extensive research continuously illustrates the applications of VitC in promoting cell reprogramming, fine-tuning embryonic stem cell function, and fighting diseases. Given its chemical reduction property, VitC predominantly acts as an antioxidant to reduce reactive oxygen species (ROS) and as a cofactor for certain dioxygenases involved in epigenetic regulation. Here, we propose that VitC is also a bio-signaling molecule based on the finding that sodium-dependent VitC transporter (SVCT) 2 is a novel receptor-like transporter of VitC that possesses dual activities in mediating VitC uptake and Janus kinase (JAK) 2/signal transducer and activator of transcription (STAT) 2 signaling pathway. Through interaction, SVCT2 induces JAK2 phosphorylation while transporting VitC into cells. Activated JAK2 phosphorylates the C-terminus of SVCT2, resulting in the recruitment and activation of STAT2. As a highlight, our results suggest that the activation of JAK2 synergistically promotes regulation of VitC in ROS scavenging and epigenetic modifications through phosphorylating pyruvate dehydrogenase kinase 1, ten-eleven translocation enzyme 3, and histone H3 Tyr41. Furthermore, VitC-activated JAK2 exhibits bidirectional effects in regulating cell pluripotency and differentiation. Our results thus reveal that the SVCT2-mediated JAK2 activation facilitates VitC functions in a previously unknown manner.


Asunto(s)
Ácido Ascórbico/metabolismo , Janus Quinasa 2/metabolismo , Transportadores de Sodio Acoplados a la Vitamina C/genética , Transportadores de Sodio Acoplados a la Vitamina C/metabolismo , Animales , Ácido Ascórbico/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Dioxigenasas/genética , Epigénesis Genética/efectos de los fármacos , Células HEK293 , Histonas/metabolismo , Humanos , Ratones , Células 3T3 NIH , Fosforilación , Dominios Proteicos , Factor de Transcripción STAT2/genética , Transducción de Señal/efectos de los fármacos , Transportadores de Sodio Acoplados a la Vitamina C/química
9.
Int J Biol Macromol ; 150: 492-500, 2020 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-32035150

RESUMEN

Somatic cell nuclear transfer (SCNT) has potential applications in agriculture and biomedicine, but the efficiency of cloning is still low. In this study, the transcriptional profiles in cloned and fertilized embryos were measured and compared by RNA sequencing. The 2-cell embryos were detected to identify the earliest transcriptional differences between embryos derived through IVF and SCNT. As a result, 364 genes showed decreased expression in cloned 2-cell embryos and were enriched in "intracellular protein transport" and "ubiquitin mediated proteolysis". In blastocysts, 593 genes showed decreased expression in cloned blastocysts and were enriched in "RNA binding", "nucleotide binding", "embryo development", and "adherens junction". We identified 14 development related genes that were not activated in the cloned embryos. Then, 68 and 245 long non-coding RNAs were recognized abnormally expressed in cloned 2-cell embryos and cloned blastocysts, respectively. Furthermore, we found that incomplete RNA-editing occurred in cloned embryos and might be caused by decreased ADAR expression. In conclusion, our study revealed the abnormal transcripts and deficient RNA-editing sites in cloned embryos and provided new data for further mechanistic studies of somatic nuclear reprogramming.


Asunto(s)
Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/genética , Perfilación de la Expresión Génica , Análisis de Secuencia de ARN , Transcriptoma , Animales , Blastocisto/citología , Blastocisto/metabolismo , Bovinos , Embrión de Mamíferos/citología , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Edición de ARN , ARN no Traducido
10.
Int J Mol Sci ; 20(11)2019 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-31142052

RESUMEN

Somatic cell nuclear transfer (SCNT) is a very powerful technique used to produce genetically identical or modified animals. However, the cloning efficiency in mammals remains low. In this study, we aimed to explore the effects of vitamin C (Vc)-treated donor cells on cloned embryos. As a result, Vc treatment relaxed the chromatin of donor cells and improved cloned embryo development. RNA sequencing was adopted to investigate the changes in the transcriptional profiles in early embryos. We found that Vc treatment increased the expression of genes involved in the cell-substrate adherens junction. Gene ontology (GO) analysis revealed that Vc treatment facilitated the activation of autophagy, which was deficient in cloned two-cell embryos. Rapamycin, an effective autophagy activator, increased the formation of cloned blastocysts (36.0% vs. 25.6%, p < 0.05). Abnormal expression of some coding genes and long non-coding RNAs in cloned embryos was restored by Vc treatment, including the zinc-finger protein 641 (ZNF641). ZNF641 compensation by means of mRNA microinjection improved the developmental potential of cloned embryos. Moreover, Vc treatment rescued some deficient RNA-editing sites in cloned two-cell embryos. Collectively, Vc-treated donor cells improved the development of the cloned embryo by affecting embryonic transcription. This study provided useful resources for future work to promote the reprogramming process in SCNT embryos.


Asunto(s)
Ácido Ascórbico/farmacología , Blastocisto/efectos de los fármacos , Clonación de Organismos/métodos , Oocitos/efectos de los fármacos , Transcriptoma , Vitaminas/farmacología , Animales , Autofagia , Blastocisto/metabolismo , Bovinos/genética , Células Cultivadas , Femenino , Masculino , Técnicas de Transferencia Nuclear , Oocitos/metabolismo
11.
Int J Mol Sci ; 20(9)2019 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-31035455

RESUMEN

Retinoic acid (RA) plays a key role in pluripotent cell differentiation. In F9 embryonic carcinoma cells, RA can induce differentiation towards somatic lineages via the Ras-extracellular signal-regulated kinase (Ras/Erk) pathway, but the mechanism through which it induces the Erk1/2 phosphorylation is unclear. Here, we show that miR-485 is a positive regulator that targets α/ß-hydrolase domain-containing protein 2 (Abhd2), which can result in Erk1/2 phosphorylation and triggers differentiation. RA up-regulates miR-485 and concurrently down-regulates Abhd2. We verified that Abhd2 is targeted by miR-485 and they both can influence the phosphorylation of Erk1/2. In summary, RA can mediate cell differentiation by phosphorylating Erk1/2 via miR-485 and Abhd2.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Hidrolasas/genética , MicroARNs/genética , Interferencia de ARN , Tretinoina/farmacología , Animales , Biomarcadores , Células Madre de Carcinoma Embrionario , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Fosforilación
12.
Nanomaterials (Basel) ; 9(4)2019 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-30987178

RESUMEN

Inorganic boron-based nanostructures have great potential for field emission (FE), flexible displays, superconductors, and energy storage because of their high melting point, low density, extreme hardness, and good chemical stability. Until now, most researchers have been focused on one-dimensional (1D) boron-based nanostructures (rare-earth boride (REB6) nanowires, boron nanowires, and nanotubes). Currently, two-dimensional (2D) borophene attracts most of the attention, due to its unique physical and chemical properties, which make it quite different from its corresponding bulk counterpart. Here, we offer a comprehensive review on the synthesis methods and optoelectronics properties of inorganic boron-based nanostructures, which are mainly concentrated on 1D rare-earth boride nanowires, boron monoelement nanowires, and nanotubes, as well as 2D borophene and borophane. This review paper is organized as follows. In Section I, the synthesis methods of inorganic boron-based nanostructures are systematically introduced. In Section II, we classify their optical and electrical transport properties (field emission, optical absorption, and photoconductive properties). In the last section, we evaluate the optoelectronic behaviors of the known inorganic boron-based nanostructures and propose their future applications.

13.
RSC Adv ; 9(52): 30503-30508, 2019 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-35530248

RESUMEN

Liquid crystal (LC) smart windows with adjustable reflectivity have been gradually applied in green and intelligent building materials for energy saving needs, but their applications are limited by their fundamental defects. In this study, we developed local photo-induced in situ polymerization to rapidly fabricate the infrared reflection microsheets of a cholesteric LC polymer as functional units. With the exception of the LC formula, the photo mask, liquid crystal cell, polymerization inhibitor, and the preparation conditions were specifically managed to control the extent of in situ polymerization, namely the microsheet morphology. The circular, triangular and oval-shaped microsheets were precisely obtained and were slightly bigger than the light hole. This easy, controllable, continuous and recyclable technology is expected to promote the industrialization of a high quality LC smart window with an adjustable reflection band and state.

14.
Acta Biochim Biophys Sin (Shanghai) ; 50(8): 793-799, 2018 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-29945210

RESUMEN

The ability to self-renew is one of the most important properties of embryonic stem (ES) cells. Pluripotin (SC1), a small molecule with high activity and low toxicity, promotes self-renewal in mouse ES cells. SC1 can noticeably change the morphology of retinoic acid (RA)-induced F9 embryonic carcinoma cells (F9 cells). However, in the long term, RA and SC1 together cause cell apoptosis. When being added after 18-24 h of RA-induced F9 cell differentiation, SC1 transitorily activated Nanog and Oct4. Both Nanog and Oct4 were downregulated when SC1 and RA were added simultaneously. On the other hand, Klf4 was continually activated when SC1 was added between 6 and 24 h. Phosphorylated Erk1/2 protein levels were reduced from 6 to 24 h, whereas unphosphorylated Erk1 protein levels remained unchanged. A higher concentration of SC1 promoted cell self-renewal by strengthening the inhibition of Erk1/2 protein phosphorylation in F9 cells. Furthermore, SC1 and RA affect global DNA methylation by influencing the expressions of methylation-associated proteins, including Dnmt3b, Dnmt3l, Tet1, Tet2, and Tet3. In conclusion, SC1 inhibits the differentiation of RA-induced F9 cells mainly by reducing the levels of phosphorylated Erk1/2 and enhancing the expression of Klf4, although it also reduces DNA methylation, which may have an additional effect on ES cell differentiation.


Asunto(s)
Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Pirazoles/farmacología , Pirimidinas/farmacología , Tretinoina/farmacología , Animales , Antineoplásicos/farmacología , Apoptosis/genética , Carcinoma Embrionario/genética , Carcinoma Embrionario/metabolismo , Carcinoma Embrionario/patología , Diferenciación Celular/genética , Línea Celular Tumoral , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Factor 4 Similar a Kruppel , Ratones , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo
15.
Cell Physiol Biochem ; 44(5): 2057-2072, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29241165

RESUMEN

BACKGROUND/AIMS: Self-renewal is one of the most important features of embryonic stem (ES) cells. SC1 is a small molecule modulator that effectively maintains the self-renewal of mouse ES cells in the absence of leukemia inhibitory factor (LIF), serum and feeder cells. However, the mechanism by which SC1 maintains the undifferentiated state of mouse ES cells remains unclear. METHODS: In this study, microarray and small RNA deep-sequencing experiments were performed on mouse ES cells treated with or without SC1 to identify the key genes and microRNAs that contributed to self-renewal. RESULTS: SC1 regulates the expressions of pluripotency and differentiation factors, and antagonizes the retinoic acid (RA)-induced differentiation in the presence or absence of LIF. SC1 inhibits the MEK/ERK pathway through Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis and pathway reporting experiments. Small RNA deep-sequencing revealed that SC1 significantly modulates the expression of multiple microRNAs with crucial functions in ES cells. The expression of miR124-3p is upregulated in SC1-treated ES cells, which significantly inhibits the MEK/ERK pathway by targeting Grb2, Sos2 and Egr1. CONCLUSION: SC1 enhances the self-renewal capacity of mouse ES cells by modulating the expression of key regulatory genes and pluripotency-associated microRNAs. SC1 significantly upregulates miR124-3p expression to further inhibit the MEK/ ERK pathway by targeting Grb2, Sos2 and Egr1.


Asunto(s)
Autorrenovación de las Células/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , MicroARNs/metabolismo , Células Madre Embrionarias de Ratones/citología , Pirazoles/farmacología , Pirimidinas/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Proteína 1 de la Respuesta de Crecimiento Precoz/antagonistas & inhibidores , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteína Adaptadora GRB2/antagonistas & inhibidores , Proteína Adaptadora GRB2/metabolismo , Factor Inhibidor de Leucemia/química , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , MicroARNs/química , MicroARNs/genética , Células Madre Embrionarias de Ratones/metabolismo , Análisis de Secuencia de ARN , Proteínas Son Of Sevenless/antagonistas & inhibidores , Proteínas Son Of Sevenless/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Tretinoina/farmacología , Regulación hacia Arriba/efectos de los fármacos
16.
Oncotarget ; 8(38): 64050-64065, 2017 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-28969051

RESUMEN

Tuberculosis remains a leading health problem worldwide and still accounts for about 1.3 million deaths annually. Expression of the mouse Sp110 nuclear body protein (Sp110) upregulates the apoptotic pathway, which plays an essential role in enhancing host immunity to Mycobacterium tuberculosis (Mtb). However, the mechanism of this upregulation is unclear. Here, we have identified 253 proteins in mouse macrophages that interact with Sp110, of which 251 proteins were previously uncharacterized. The results showed that Sp110 interacts with heat shock protein 5 (Hspa5) to activate endoplasmic reticulum (ER) stress-induced apoptosis, and that this is essential for Sp110 enhanced macrophage resistance to Mtb. Inhibition of the ER stress pathway abolishing the Sp110-enhanced macrophage apoptosis and resulted in increased intracellular survival of Mtb in macrophages overexpressing Sp110 Further studies revealed that Sp110 also interacts with the RNA binding protein, Ncl to promote its degradation. Consequently, the expression of Bcl2, usually stabilized by Ncl, was downregulated in Sp110 overexpressing macrophages. Moreover, overexpression of Sp110 promotes degradation of ribosomal protein Rps3a, resulting in upregulation of the activity of the pro-apoptotic poly (ADP-ribose) polymerase (PARP). In addition, macrophages from transgenic cattle with increased Sp110 expression confirmed that activation of the ER stress response is the main pathway through which Sp110-enhanced macrophages impart resistance to Mtb. This work has revealed the mechanism of Sp110 enhanced macrophage apoptosis in response to Mtb infection, and provides new insights into the study of host-pathogen interactions.

17.
Int J Mol Sci ; 18(9)2017 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-28869544

RESUMEN

DNA methylation is an important epigenetic modification that needs to be carefully controlled as a prerequisite for normal early embryogenesis. Compelling evidence now suggests that four maternal-effect proteins, primordial germ cell 7 (PGC7), zinc finger protein 57 (ZFP57), tripartite motif-containing 28 (TRIM28) and DNA methyltransferase (cytosine-5) 1 (DNMT1) are involved in the maintenance of DNA methylation. However, it is still not fully understood how these maternal-effect proteins maintain the DNA methylation imprint. We noticed that a feature common to these proteins is the presence of significant levels of intrinsic disorder so in this study we started from an intrinsic disorder perspective to try to understand these maternal-effect proteins. To do this, we firstly analysed the intrinsic disorder predispositions of PGC7, ZFP57, TRIM28 and DNMT1 by using a set of currently available computational tools and secondly conducted an intensive literature search to collect information on their interacting partners and structural characterization. Finally, we discuss the potential effect of intrinsic disorder on the function of these proteins in maintaining DNA methylation.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética , Proteínas Intrínsecamente Desordenadas/metabolismo , Animales , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Humanos , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/genética , Ratones , Modelos Moleculares , Mutación , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas/métodos , Mapas de Interacción de Proteínas
18.
J Proteome Res ; 16(9): 3113-3123, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28712289

RESUMEN

Primordial germ cell 7 (PGC7), a maternal factor essential for early development, plays a critical role in the regulation of DNA methylation, transcriptional repression, chromatin condensation, and cell division and the maintenance of cell pluripotentiality. Despite the fundamental roles of PGC7 in these cellular processes, only a few molecular and functional interactions of PGC7 have been reported. Here, a streptavidin-biotin affinity purification technique combined with LC-MS/MS was used to analyze potential proteins that interact with PGC7. In total, 291 potential PGC7-interacting proteins were identified. Through an in-depth bioinformatic analysis of potential interactors, we linked PGC7 to critical cellular processes including translation, RNA processing, cell cycle, and regulation of heterochromatin structure. To better understand the functional interactions of PGC7 with its potential interactors, we constructed a protein-protein interaction network using the STRING database. In addition, we discussed in detail the interactions between PGC7 and some of its newly validated partners. The identification of these potential interactors of PGC7 expands our knowledge on the PGC7 interactome and provides a valuable resource for understanding the diverse functions of this protein.


Asunto(s)
Redes Reguladoras de Genes , Heterocromatina/metabolismo , Mapeo de Interacción de Proteínas , Proteómica/métodos , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/metabolismo , Animales , Ligasas de Carbono-Nitrógeno/genética , Ligasas de Carbono-Nitrógeno/metabolismo , Ciclo Celular/genética , Cromatografía Liquida , Proteínas Cromosómicas no Histona , Clonación Molecular , Metilación de ADN , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Heterocromatina/química , Humanos , Ratones , Anotación de Secuencia Molecular , Oligopéptidos/genética , Oligopéptidos/metabolismo , Plásmidos/química , Plásmidos/metabolismo , Unión Proteica , Biosíntesis de Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas Represoras/genética , Espectrometría de Masas en Tándem
19.
Tuberculosis (Edinb) ; 101: 85-94, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27865405

RESUMEN

SP110 has previously shown to be a genetic determinant of host resistance to the intracellular pathogen infection in mouse and human. However, its relevant biological information in large non-primate animals still remains unknown. Here we report the novel discovery and characterization of three transcript variants of horse SP110. The transcript variant 1 (Tv1) of horse SP110 with the longest open reading frame has four domains (Sp100, SAND, PHD and Bromo domain). Tv2 and Tv3 share the same N-terminal sequence as Tv1, which contains Sp100 and SAND. We show that Tv2 is generated from alternative splicing and deletion of Exon17-Exon18 segment, while Tv3 is generated by pre-mature transcriptional termination at Exon 16. Furthermore, we demonstrate that the heterologous expression of horse SP110 variants stimulate macrophages into an activation-like phenotype. The macrophages underwent a shift in enhancing the secretion of cytokines (interleukin-1 (IL-1) and TNF-α) and accelerating inducible nitric oxide synthase (iNOS) activity, and eventually went into apoptotic cell death. Intriguingly, horse SP110 Tv1 showed more capability to trigger the immune activities compared to Tv2 and Tv3. To our knowledge, the identification of SP110 transcript variants from horse is the first report on biological function of SP110 in perissodactyla animals.


Asunto(s)
Empalme Alternativo/genética , Activación de Macrófagos/genética , Macrófagos/inmunología , Antígenos de Histocompatibilidad Menor/genética , Proteínas Nucleares/genética , Secuencia de Aminoácidos , Animales , Apoptosis/genética , Células Cultivadas , Citocinas/biosíntesis , ADN Complementario/genética , Predisposición Genética a la Enfermedad , Variación Genética , Caballos , Macrófagos/microbiología , Antígenos de Histocompatibilidad Menor/inmunología , Mycobacterium tuberculosis , Proteínas Nucleares/inmunología , Sistemas de Lectura Abierta , Alineación de Secuencia , Análisis de Secuencia de ADN/métodos , Especificidad de la Especie , Tuberculosis/genética , Tuberculosis/inmunología
20.
PLoS One ; 11(3): e0150936, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26938105

RESUMEN

Understanding the mechanisms that regulate pluripotency of embryonic stem cells (ESCs) is important to ensure their safe clinical use. CHIR99021 (CHIR)-induced activation of Wnt/ß-catenin signaling promotes self-renewal in mouse ESCs (mESCs). ß-catenin functions individually or cooperates with transcription factors to activate stemness factors such as c-Myc, Esrrb, Pou5f1, and Nanog. However the relationship between the core pluripotent factor, Kruppel-like factor 4 (also known as GKLF or EZF) and Wnt/ß-catenin signaling, remains ambiguous in J1 mESCs. DNA microarray analysis revealed that CHIR-treatment promoted pluripotency-maintaining transcription factors and repressed germ layer specification markers. CHIR also promoted genes related to the development of extracellular regions and the plasma membrane to maintain pluripotency of J1 mESCs. Among the CHIR-regulated genes, Klf4 has not been reported previously. We identified a novel cis element in the Klf4 gene that was activated by ß-catenin in J1 mESCs. We determined that ß-catenin interacted with this cis element, identifying Klf4 as a ß-catenin target gene in this context. Moreover, several microRNAs that targeted the 3'-UTR of Klf4 mRNA were identified, with miR-7a being down-regulated by CHIR in a ß-catenin-independent manner in J1 mESCs. These data collectively suggest that CHIR enhances Klf4 expression by repressing miR-7a expression or canonical Wnt pathway activation.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/genética , MicroARNs/genética , Células Madre Embrionarias de Ratones/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Pirimidinas/farmacología , beta Catenina/genética , Regiones no Traducidas 3' , Animales , Secuencia de Bases , Línea Celular , Regulación de la Expresión Génica , Ontología de Genes , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/agonistas , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , MicroARNs/metabolismo , Anotación de Secuencia Molecular , Datos de Secuencia Molecular , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA