Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
PLoS One ; 19(4): e0301663, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38603701

RESUMEN

The multikinase inhibitor sorafenib is the standard first-line treatment for advanced hepatocellular carcinoma (HCC), but many patients become sorafenib-resistant (SR). This study investigated the efficacy of another kinase inhibitor, regorafenib (Rego), as a second-line treatment. We produced SR HCC cells, wherein the PI3K-Akt, TNF, cAMP, and TGF-beta signaling pathways were affected. Acute Rego treatment of these cells reversed the expression of genes involved in TGF-beta signaling but further increased the expression of genes involved in PI3K-Akt signaling. Additionally, Rego reversed the expression of genes involved in nucleosome assembly and epigenetic gene expression. Weighted gene co-expression network analysis (WGCNA) revealed four differentially expressed long non-coding RNA (DElncRNA) modules that were associated with the effectiveness of Rego on SR cells. Eleven putative DElncRNAs with distinct expression patterns were identified. We associated each module with DEmRNAs of the same pattern, thus obtaining DElncRNA/DEmRNA co-expression modules. We discuss the potential significance of each module. These findings provide insights and resources for further investigation into the potential mechanisms underlying the response of SR HCC cells to Rego.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Compuestos de Fenilurea , Piridinas , ARN Largo no Codificante , Humanos , Sorafenib/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , ARN Largo no Codificante/genética , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas , ARN Mensajero/metabolismo , Factor de Crecimiento Transformador beta
2.
Stem Cells Dev ; 32(23-24): 731-746, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37823730

RESUMEN

Rat primitive extraembryonic endoderm (pXEN) stem cell lines indefinitely preserve the characteristic features of the early extraembryonic endoderm (ExEn) in vitro, but require unknown serum factors and exhibit a hybrid (mesenchymal-epithelial) phenotype. We report two chemically defined conditions that differ by the addition of the cytokine leukemia inhibitory factor (Lif) and the ß-catenin-stabilizing drug Chir99021, and enable permanent self-renewal as mesenchymal and epithelial morphotypes, respectively. The morphotypes are interconvertible and equipotent, as shown by the formation of well-differentiated organoids. Surprisingly, the proliferation of both morphotypes requires Lif-type Gp130/Stat3 signaling (autocrine in the absence of added Lif) and noncanonical Wnt signaling (autocrine). In addition, the epithelial version requires ß-catenin for proliferation and morphology. Interestingly, the mesenchymal cells also express key epithelial markers, but those are improperly structured and/or not functional, indicating a primed state. These results provide an improved platform for studying the proliferation and plasticity of the early ExEn, which occurs in mesenchymal and epithelial forms in vivo.


Asunto(s)
Endodermo , beta Catenina , Ratas , Animales , Diferenciación Celular , beta Catenina/metabolismo , Línea Celular , Células Madre Embrionarias
3.
BMC Genomics ; 23(1): 853, 2022 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-36575377

RESUMEN

BACKGROUND: Long non-coding RNAs (lncRNAs) are emerging as key modulators of inflammatory gene expression, but their roles in neuroinflammation are poorly understood. Here, we identified the inflammation-related lncRNAs and correlated mRNAs of the lipopolysaccharide (LPS)-treated human microglial cell line HMC3. We explored their potential roles and interactions using bioinformatics tools such as gene ontology (GO), kyoto encyclopedia of genes and genomes (KEGG), and weighted gene co-expression network analysis (WGCNA). RESULTS: We identified 5 differentially expressed (DE) lncRNAs, 4 of which (AC083837.1, IRF1-AS1, LINC02605, and MIR3142HG) are novel for microglia. The DElncRNAs with their correlated DEmRNAs (99 total) fell into two network modules that both were enriched with inflammation-related RNAs. However, treatment with the anti-inflammatory agent JQ1, an inhibitor of the bromodomain and extra-terminal (BET) protein BRD4, neutralized the LPS effect in only one module, showing little or even enhancing effect on the other. CONCLUSIONS: These results provide insight into, and a resource for studying, the regulation of microglia-mediated neuroinflammation and its potential therapy by small-molecule BET inhibitors.


Asunto(s)
Lipopolisacáridos , ARN Largo no Codificante , Humanos , Lipopolisacáridos/farmacología , Microglía/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Enfermedades Neuroinflamatorias , Proteínas Nucleares/genética , Redes Reguladoras de Genes , Factores de Transcripción/genética , Inflamación/genética , Proteínas de Ciclo Celular/genética
4.
Sci Rep ; 12(1): 7779, 2022 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-35546353

RESUMEN

The epigenetic reader, bromodomain-containing 4 (BRD4), is overexpressed in hepatocellular carcinoma (HCC), and BRD4 inhibition is considered as a new therapeutic approach. The BRD inhibitor JQ1 is known to inhibit the enrichment of BRD4 at enhancer sites. Gene network analyses have implicated long non-coding RNAs (lncRNAs) in the effects of JQ1, but the precise molecular events remain unexplored. Here, we report that in HepG2 cells, JQ1 significantly reduced various proliferation-related lncRNAs, but up-regulated the known liver tumor marker, MALAT1. Using ChIP-sequencing data, ChIP-qPCR, luciferase reporter assays, and chromatin conformation capture (3C), we characterized the MALAT1 gene locus. We found that JQ1 elicited a rearrangement of its chromatin looping conformation, which involved the putative enhancers E1, E2, E3, the gene body, and the promoter. We further found that the forkhead box protein A2 (FOXA2) binds to E2 and the promoter; suppression of FOXA2 expression resulted in MALAT1 up-regulation and increased cell proliferation. These results suggest that the inhibition of MALAT1 may improve the effect of BET inhibitors as an anti-cancer therapy and that FOXA2 would be a suitable target for that approach.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , ARN Largo no Codificante , Azepinas/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Cromatina , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Proteínas Nucleares/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/farmacología , Factores de Transcripción/metabolismo , Triazoles/farmacología
5.
PLoS One ; 17(4): e0266966, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35486664

RESUMEN

Hepatocellular carcinoma (HCC) is the most common primary liver cancer and poor prognosis. Emerging evidence suggests that epigenetic alterations play a crucial role in HCC, suggesting epigenetic inhibition as a promising therapeutic approach. Indeed, the bromodomain and extra-terminal (BET) inhibitors inhibit the proliferation and invasion of various cancers but still lack a strong mechanistic rationale. Here, we identified the differentially expressed mRNAs (DEmRNAs) and lncRNAs (DElncRNAs) in human HCC cell line HepG2 treated with the BET inhibitors, JQ1, OTX015, or ABBV-075. We analyzed the correlation between DEmRNAs and DElncRNAs in common for the three inhibitors based on their expression profiles and performed functional annotation pathway enrichment analysis. Most of these shared DEmRNAs and DElncRNAs, including some novel transcripts, were downregulated, indicating decreased proliferation/adhesion and increased apoptosis/inflammation. Our study suggests that BET proteins play a crucial role in regulating cancer progression-related genes and provide a valuable resource for novel putative biomarkers and therapeutic targets in HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , ARN Largo no Codificante , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Perfilación de la Expresión Génica , Células Hep G2 , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , ARN Largo no Codificante/genética
6.
Sci Rep ; 12(1): 855, 2022 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-35039581

RESUMEN

The proto-oncogene MYC is important for development and cell growth, however, its abnormal regulation causes cancer. Recent studies identified distinct enhancers of MYC in various cancers, but any MYC enhancer(s) in hepatocellular carcinoma (HCC) remain(s) elusive. By analyzing H3K27ac enrichment and enhancer RNA (eRNA) expression in cultured HCC cells, we identified six putative MYC enhancer regions. Amongst these, two highly active enhancers, located ~ 800 kb downstream of the MYC gene, were identified by qRT-PCR and reporter assays. We functionally confirmed these enhancers by demonstrating a significantly reduced MYC expression and cell proliferation upon CRISPR/Cas9-based deletion and/or antisense oligonucleotide (ASO)-mediated inhibition. In conclusion, we identified potential MYC enhancers of HCC and propose that the associated eRNAs may be suitable targets for HCC treatment.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Proliferación Celular/genética , Elementos de Facilitación Genéticos/genética , Elementos de Facilitación Genéticos/fisiología , Genes myc/efectos de los fármacos , Genes myc/fisiología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , ARN no Traducido , Línea Celular Tumoral , Humanos
7.
Sci Rep ; 11(1): 11799, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-34083693

RESUMEN

Hepatocellular carcinoma (HCC) is one of the most prevalent and poorly responsive cancers worldwide. Bromodomain and extraterminal (BET) inhibitors, such as JQ1 and OTX-015, inhibit BET protein binding to acetylated residues in histones. However, the physiological mechanisms and regulatory processes of BET inhibition in HCC remain unclear. To explore BET inhibitors' potential role in the molecular mechanisms underlying their anticancer effects in HCC, we analyzed BET inhibitor-treated HCC cells' gene expression profiles with RNA-seq and bioinformatics analysis. BET inhibitor treatment significantly downregulated genes related to bromodomain-containing proteins 4 (BRD4), such as ACSL5, SLC38A5, and ICAM2. Importantly, some cell migration-related genes, including AOC3, CCR6, SSTR5, and SCL7A11, were significantly downregulated. Additionally, bioinformatics analysis using Ingenuity Knowledge Base Ingenuity Pathway Analysis (IPA) revealed that SMARCA4 regulated migration response molecules. Furthermore, knockdown of SMARCA4 gene expression by siRNA treatment significantly reduced cell migration and the expression of migration-related genes. In summary, our results indicated that BET inhibitor treatment in HCC cell lines reduces cell migration through the downregulation of SMARCA4.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Hepatocelular/genética , ADN Helicasas/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Hepáticas/genética , Proteínas Nucleares/genética , Proteínas/antagonistas & inhibidores , Factores de Transcripción/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/mortalidad , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular/efectos de los fármacos , Biología Computacional/métodos , ADN Helicasas/metabolismo , Perfilación de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/mortalidad , Proteínas Nucleares/metabolismo , Pronóstico , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo
8.
Sci Rep ; 11(1): 8828, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33893325

RESUMEN

Microglia, resident macrophages of the brain that act as primary immune cells, play essential roles in innate immunity and neuroinflammatory pathologies. Microglial cells are rapidly activated in response to infection and inflammation/injury, associated with the expression of proinflammatory genes and secretion of cytokines. The bromodomain and extra-terminal (BET) inhibitor JQ1 has been shown to be an epigenetic agent that reduces inflammation. In this study, we investigated the mechanisms underlying the anti-inflammatory and anti-migratory functions of JQ1 and the genes targeted by JQ1 in lipopolysaccharide (LPS)-activated human microglial clone 3 (HMC3) cells using RNA-sequencing (RNA-seq). We analyzed the pattern of inflammation-related genes (chemokines, cytokines, and interferon-stimulated genes) and migration-related genes with JQ1 treatment from differentially expressed genes analysis in HMC3 cells. We found that LPS-induced IRF1 directly regulated inflammation- and migration-related genes and that JQ1 significantly reduced IRF1 and its target genes. Additionally, IRF1 attenuation significantly downregulated target genes and inhibited microglial migration. Our data suggest that the BET inhibitor JQ1 can modulate the inflammatory response and migration through the regulation of LPS-induced IRF1 in human microglia.


Asunto(s)
Azepinas/farmacología , Movimiento Celular/efectos de los fármacos , Inflamación/prevención & control , Microglía/efectos de los fármacos , Proteínas/antagonistas & inhibidores , Triazoles/farmacología , Línea Celular , Movimiento Celular/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/genética , Lipopolisacáridos/farmacología , Microglía/citología , Microglía/metabolismo , ARN Mensajero/metabolismo , Factores de Transcripción/metabolismo
9.
Stem Cell Res Ther ; 11(1): 16, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31910882

RESUMEN

BACKGROUND: Toll-like receptor 4 (TLR4) ligands such as lipopolysaccharide (LPS) activate immunomodulatory functions and the migration of human mesenchymal stromal cells (hMSCs). Here, we study the migration-related gene expression of LPS-stimulated hMSCs and the role and regulation of one of the upregulated genes, encoding the interferon-induced transmembrane protein 1 (IFITM1). METHODS: Gene expression profiles were determined by whole-transcriptome analysis (RNA-seq) and quantitative real-time PCR (qRT-PCR). Bioinformatics approaches were used to perform network and pathway analyses. The cell migration-related genes were identified with an in vitro wound healing assay. RNA interference (RNAi) was used to suppress the IFITM1 gene expression. The IFITM1 gene enhancer was analyzed by chromatin immunoprecipitation (ChIP) sequencing, ChIP-to-PCR, luciferase reporter assays, and qRT-PCR for enhancer RNAs (eRNAs). RESULTS: RNA-seq confirmed IFITM1 as an LPS-stimulated gene, and RNAi demonstrated its importance for the LPS-stimulated migration. LPS treatment increased the eRNA expression in enhancer region R2 (2 kb upstream) of the IFITM1 gene and enriched R2 for H3K27ac. Bioinformatics implicated the transcription factors NF-κB and IRF1, ChIP assays revealed their binding to R2, and chemical inhibition of NF-κB and RNAi directed against IRF1 prevented R2 eRNA and IFITM1 gene expression. CONCLUSIONS: Increased expression of the IFITM1 gene is required for LPS-stimulated hMSC migration. We described several underlying changes in the IFITM1 gene enhancer, most notably the NF-κB-mediated activation of enhancer region R2.


Asunto(s)
Antígenos de Diferenciación/biosíntesis , Lipopolisacáridos/farmacología , Células Madre Mesenquimatosas/metabolismo , Adulto , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Epigénesis Genética , Femenino , Humanos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Adulto Joven
10.
J Cell Sci ; 132(19)2019 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-31492758

RESUMEN

Cultured rat primitive extraembryonic endoderm (pXEN) cells easily form free-floating multicellular vesicles de novo, exemplifying a poorly studied type of morphogenesis. Here, we reveal the underlying mechanism and the identity of the vesicles. We resolve the morphogenesis into vacuolization, vesiculation and maturation, and define the molecular characteristics and requirements of each step. Vacuolization is fueled by macropinocytosis and occurs by default if not blocked by high cell density or matrix proteins. Fine-tuned cell-cell contact then forms nascent three-cell vesicles with vacuole-derived lumina. In maturation, the vesicles complete epithelialization, expand via mitosis and continued fluid uptake, and differentiate further. The mature vesicles consist of a simple squamous epithelium with an apical-outside/basal-inside polarity that we trace back to the single cell stage. The polarity and gene expression pattern of the vesicles are similar to those of the early visceral endoderm. pXEN cells provide a useful in vitro model for study of matrix-independent, basal-type lumenogenesis and the physiology of the visceral endoderm.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Endodermo/metabolismo , Células Madre/metabolismo , Vacuolas/metabolismo , Animales , Calcio/metabolismo , Ciclo Celular/fisiología , Muerte Celular/fisiología , Diferenciación Celular/fisiología , Línea Celular , Biología Computacional , Citocinesis/fisiología , Microscopía Confocal , Microscopía Electrónica de Transmisión , Ratas , Células Madre/ultraestructura
11.
PLoS One ; 14(8): e0220823, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31369650

RESUMEN

This in vitro study examined the sealing ability of different desensitizing agents under a chemo-mechanical stress condition. For the study, a total of 144 extracted, caries-free human third molars were used to produce 1 mm-thick dentin discs. The specimens were divided randomly into four groups: Superseal (SS), Gluma (GL), Gluma Self-etch (GS), and Tooth Coat (TC). For each group, the permeability was measured before and after applying the desensitizer, after being exposed to Coca Cola for 5 minutes, and after 3150 strokes of a brushing abrasion. The decrease in permeability after the erosive and abrasive stress was analyzed by ANOVA and Tukey post hoc test. As a result, the dentin permeability decreased significantly for all desensitizers immediately after application (p < 0.05). SS and GS showed a significant difference in permeability reduction observed immediately after application and after acid action with Coca Cola (p < 0.05). After brushing abrasion, the permeability reduction decreased significantly for all desensitizers tested in this study (p < 0.05). TC showed the largest decrease in dentinal permeability compared to that of the other desensitizers and the differences were significant after brushing abrasion (p < 0.05). All tested desensitizers were effective in reducing dentin permeability. The behavioral characteristics under erosive and abrasive stress varied according to the products used. TC exhibited excellent sealing ability among the other desensitizers.


Asunto(s)
Desensibilizantes Dentinarios/uso terapéutico , Sensibilidad de la Dentina/tratamiento farmacológico , Tercer Molar/efectos de los fármacos , Selladores de Fosas y Fisuras/uso terapéutico , Dentina/efectos de los fármacos , Glutaral/uso terapéutico , Humanos , Técnicas In Vitro , Ácido Oxálico/uso terapéutico , Ácidos Polimetacrílicos/uso terapéutico , Abrasión de los Dientes/metabolismo , Erosión de los Dientes/metabolismo
12.
Front Immunol ; 10: 530, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30949176

RESUMEN

Recent studies have reported an increased incidence of inflammatory bowel disease (IBD) in patients with pulmonary diseases. Despite clinical and epidemiological studies of the interplay between colitis and asthma, the diseases' related underlying mechanisms remain unclear. In this study, we evaluated the development of colitis in a model of allergic airway inflammation. We revealed that intratracheal chronic ovalbumin (OVA) exposure induces colitis and allergic airway inflammation. Interestingly, induction of colitis was largely regulated by Th1, rather than Th2 responses, whereas allergic airway inflammation was primarily mediated by Th2 responses. Experiments in Tbx21 (T-bet) and Ifng (IFN-γ) deficient mice have confirmed that IFN-γ is a major mediator involved in OVA-induced colitis. These findings broaden current understanding of allergen induced colitis pathology and could play a role in the development of novel clinical treatment strategies for asthmatic patients who are at risk of developing colitis.


Asunto(s)
Colitis/inmunología , Interferón gamma/inmunología , Ovalbúmina/toxicidad , Transducción de Señal/inmunología , Células TH1/inmunología , Células Th2/inmunología , Animales , Colitis/inducido químicamente , Colitis/genética , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/inmunología , Interferón gamma/genética , Ratones , Ratones Noqueados , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/inmunología
13.
J Biol Chem ; 294(21): 8424-8437, 2019 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-30944148

RESUMEN

Mesenchymal stromal cells (MSCs) can potently regulate the functions of immune cells and are being investigated for the management of inflammatory diseases. Toll-like receptor 3 (TLR3)-stimulated human MSCs (hMSCs) exhibit increased migration and chemotaxis within and toward damaged tissues. However, the regulatory mechanisms underlying these migratory activities are unclear. Therefore, we analyzed the migration capability and gene expression profiles of TLR3-stimulated hMSCs using RNA-Seq, wound healing, and transwell cell migration assay. Along with increased cell migration, the TLR3 stimulation also increased the expression of cytokines, chemokines, and cell migration-related genes. The promoter regions of the latter showed an enrichment of putative motifs for binding the transcription factors forkhead box O1 (FOXO1), FOXO3, NF-κB (NF-κB1), and RELA proto-oncogene and NF-κB subunit. Of note, FOXO1 inhibition by the FOXO1-selective inhibitor AS1842856 significantly reduced both migration and the expression of migration-related genes. In summary, our results indicate that TLR3 stimulation induces hMSC migration through the expression of FOXO1-activated genes.


Asunto(s)
Movimiento Celular , Proteína Forkhead Box O1/metabolismo , Regulación de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Receptor Toll-Like 3/metabolismo , Adulto , Femenino , Proteína Forkhead Box O1/antagonistas & inhibidores , Proteína Forkhead Box O1/genética , Humanos , Masculino , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/metabolismo , Proto-Oncogenes Mas , Quinolonas/farmacología , Receptor Toll-Like 3/agonistas , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo
14.
Stem Cell Res ; 30: 100-112, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29843002

RESUMEN

Mouse blastocysts contain the committed precursors of the extraembryonic endoderm (ExEn), which express the key transcription factor Oct4, depend on LIF/LIF-like factor-driven Jak/Stat signaling, and initially exhibit lineage plasticity. Previously described rat blastocyst-derived ExEn precursor-like cell lines (XENP cells/HypoSCs) also show these features, but equivalent mouse blastocyst-derived cell lines are lacking. We now present mouse blastocyst-derived cell lines, named primitive XEN (pXEN) cells, which share these and additional characteristics with the XENP cells/HypoSCs, but not with previously known mouse blastocyst-derived XEN cell lines. Otherwise, pXEN cells are highly similar to XEN cells by morphology, lineage-intrinsic differentiation potential, and multi-gene expression profile, although the pXEN cell profile correlates better with the blastocyst stage. Finally, we show that pXEN cells easily convert into XEN-like cells but not vice versa. The findings indicate that (i) pXEN cells are more representative than XEN cells of the blastocyst stage; (ii) mouse pXEN, rather than XEN, cells are homologs of rat XENP cells/HypoSCs, which we propose to call rat pXEN cells.


Asunto(s)
Blastocisto/metabolismo , Endodermo/metabolismo , Animales , Diferenciación Celular , Línea Celular , Células Madre Embrionarias , Endodermo/citología , Ratones , Ratas
15.
Front Immunol ; 9: 22, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29403501

RESUMEN

Macrophages are the prime innate immune cells of the inflammatory response, and the combination of multiple signaling inputs derived from the recognition of host factors [e.g., interferon-g (IFN-γ)] and invading pathogen products (e.g., toll-like receptors (TLRs) agonists) are required to maintain essential macrophage function. The profound effects on biological outcomes of inflammation associated with IFN-γ pretreatment ("priming") and TLR4 ligand bacterial lipopolysaccharide (LPS)-induced macrophage activation (M1 or classical activation) have long been recognized, but the underlying mechanisms are not well defined. Therefore, we analyzed gene expression profiles of macrophages and identified genes, transcription factors (TFs), and transcription co-factors (TcoFs) that are uniquely or highly expressed in IFN-γ-mediated TLR4 ligand LPS-inducible versus only TLR4 ligand LPS-inducible primary macrophages. This macrophage gene expression has not been observed in macrophage cell lines. We also showed that interleukin (IL)-4 and IL-13 (M2 or alternative activation) elicited the induction of a distinct subset of genes related to M2 macrophage polarization. Importantly, this macrophage gene expression was also associated with promoter conservation. In particular, our approach revealed novel roles for the TFs and TcoFs in response to inflammation. We believe that the systematic approach presented herein is an important framework to better understand the transcriptional machinery of different macrophage subtypes.


Asunto(s)
Células de la Médula Ósea/inmunología , Lipopolisacáridos/inmunología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Línea Celular , Perfilación de la Expresión Génica , Interferón gamma/inmunología , Interleucina-13/inmunología , Ratones , Ratones Endogámicos C57BL , Células RAW 264.7 , Receptor Toll-Like 4/inmunología
16.
J Appl Toxicol ; 38(1): 100-107, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28677831

RESUMEN

Alcohol is a teratogenic agent that can cause a wide range of developmental disorders, and sometimes, the effects persist throughout an individual's lifetime. Researchers have shown the involvement of epigenetic mechanisms in alcohol-mediated disorders. Non-coding RNAs are one of the major sources of epigenetic modifications, especially microRNAs. The association of microRNAs with alcohol consumption leads to a new focus on finding the molecular mechanisms of alcohol toxicity. It has been suggested that alcohol alters the relative expression of microRNAs and regulates target mRNA expression in both in vitro and in vivo models. Currently, we lack information regarding the relationship between altered microRNA expression and disease phenotypes in alcohol-mediated disorders. In this review, we tried to gather all of the available information about the alcohol-mediated dysregulation of microRNA expression in utero. We hope that our efforts will help future researchers identify major microRNAs in the field of prenatal alcohol toxicity and related therapeutics.


Asunto(s)
Consumo de Bebidas Alcohólicas/efectos adversos , Etanol/toxicidad , Desarrollo Fetal/efectos de los fármacos , Exposición Materna/efectos adversos , MicroARNs/biosíntesis , Animales , Encéfalo/efectos de los fármacos , Encéfalo/embriología , Encéfalo/metabolismo , Etanol/farmacocinética , Femenino , Desarrollo Fetal/genética , Humanos , Intercambio Materno-Fetal/efectos de los fármacos , MicroARNs/genética , Organogénesis/efectos de los fármacos , Organogénesis/genética , Embarazo
17.
Toxicol In Vitro ; 46: 66-76, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28986285

RESUMEN

During the differentiation process, various epigenetic factors regulate the precise expression of important genes and control cellular fate. During this stage, the differentiating cells become vulnerable to external stimuli. Here, we used an early neural differentiation model to observe ethanol-mediated transcriptional alterations. Our objective was to identify important molecular regulators of ethanol-related alterations in the genome during differentiation. A transcriptomic analysis was performed to profile the mRNA expression in differentiating embryoid bodies with or without ethanol treatment. In total, 147 differentially expressed genes were identified in response to 50mM ethanol. Of these differentially expressed genes, 78 genes were up-regulated and 69 genes were down-regulated. Our analysis revealed a strong association among the transcript signatures of the important modulators which were involved in protein modification, protein synthesis and gene expression. Additionally, ethanol-mediated activation of DNA transcription was observed. We also profiled ethanol-responsive transcription factors (TFs), upstream transcriptional regulators and TF-binding motifs in the differentiating embryoid bodies. In this study, we established a platform that we hope will help other researchers determine the ethanol-mediated changes that occur during cellular differentiation.


Asunto(s)
Cuerpos Embrioides/efectos de los fármacos , Etanol/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Transcriptoma/efectos de los fármacos , ADN/genética , Cuerpos Embrioides/metabolismo , Perfilación de la Expresión Génica , Humanos , Procesamiento Proteico-Postraduccional , ARN/genética , ARN/metabolismo , Análisis de Secuencia de ARN , Factores de Transcripción
18.
Scanning ; 2017: 6074253, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29109820

RESUMEN

This study examined the effects of different application times of sodium ascorbate (SA) on the bond strength of composite resin to bleached dentin. Specimens with an exposed dentin surface were divided into 3 groups according to the type of bleaching agent used: Group A, mixture of sodium perborate (SP) and distilled water (DW); Group B, mixture of SP and hydrogen peroxide (HP); control group, no bleaching. Each group was classified into 10 subgroups. Subgroups IB and DB underwent immediate bonding and delayed bonding, respectively. 10% SA was applied to 3, 5, 10, and 30 minutes and 1, 24, 48, and 72 hours, respectively. Microtensile bond strength (µTBS) was measured after restoration, and the data was analyzed by one-way ANOVA and Scheffé's test. Before restoration, the dentin surfaces were examined by scanning electron microscopy (SEM) and energy dispersive X-ray spectroscopy (EDS). SEM showed that most dentin surfaces were filled with crystals when SA was applied to more than 24 hours. EDS revealed peaks of calcium, carbon, oxygen, and sodium. The application of SA for 5 minutes to 48 hours or for 30 minutes to 24 hours is suggested when a mixture of SP and DW or HP is used, respectively.


Asunto(s)
Ácido Ascórbico/metabolismo , Resinas Compuestas/metabolismo , Dentina/metabolismo , Resistencia a la Tracción , Humanos , Microscopía Electrónica de Rastreo , Espectrometría por Rayos X , Propiedades de Superficie , Factores de Tiempo , Resultado del Tratamiento
19.
Int J Biol Sci ; 13(9): 1100-1108, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29104501

RESUMEN

Ethanol is well known for its teratogenic effects during fetal development. Maternal alcohol consumption allows the developing fetus to experience the detrimental effects of alcohol exposure. Alcohol-mediated teratogenic effects can vary based on the dosage and the length of exposure. The specific mechanism of action behind this teratogenic effect is still unknown. Previous reports demonstrated that alcohol participates in epigenetic alterations, especially histone modifications during fetal development. Additional research is necessary to understand the correlation between major epigenetic events and alcohol-mediated teratogenesis such as that observed in fetal alcohol spectrum disorder (FASD). Here, we attempted to collect all the available information concerning alcohol-mediated histone modifications during gestational fetal development. We hope that this review will aid researchers to further examine the issues associated with ethanol exposure.


Asunto(s)
Consumo de Bebidas Alcohólicas/efectos adversos , Epigénesis Genética/genética , Código de Histonas/efectos de los fármacos , Código de Histonas/genética , Exposición Materna/efectos adversos , Animales , Modelos Animales de Enfermedad , Femenino , Trastornos del Espectro Alcohólico Fetal/genética , Trastornos del Espectro Alcohólico Fetal/patología , Feto/metabolismo , Humanos , Masculino , Ratones , Ratas , Factores de Riesgo
20.
Front Immunol ; 8: 1344, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29163466

RESUMEN

The increased incidence of Crohn's disease in smokers has been recently reported, suggesting a strong association of cigarette smoke (CS) with colitis. However, the mechanism of the action of CS on colitis has not yet been explored. Here, we demonstrate that CS exposure is sufficient to induce colitis in mice. Interestingly, the colitis is mainly mediated by Th1, but not Th17, responses. CD4+ T-cell depletion or T-bet/IFN-γ deficiency protects against the development of colitis induced by CS. Additionally, IFN-γ-producing CD4+ T cells play a substantial role in CS-induced colitis. The adoptive transfer (AT) of effector T cells from CS-exposed WT mice into colitis-prone mice caused these mice to develop colitis, while the AT of effector T cells from IFN-γ knock-out mice did not. These findings have implications for broadening our understanding of CS-induced pathology and for the development of novel therapeutic strategies to treat Crohn's disease.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA