Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Appl Toxicol ; 42(10): 1553-1569, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35023172

RESUMEN

Epigenetic modifications by toxic heavy metals are one of the intensively investigated fields of modern genomic research. Among a diverse group of heavy metals, lead (Pb) is an extensively distributed toxicant causing an immense number of abnormalities in the developing fetus via a wide variety of epigenetic changes. As a divalent cation, Pb can readily cross the placental membrane and the fetal blood brain barrier leading to far-reaching alterations in DNA methylation patterns, histone protein modifications, and micro-RNA expression. Over recent years, several human cohorts and animal model studies have documented hypermethylation and hypomethylation of developmental genes along with altered DNA methyl-transferase expression by in utero Pb exposure in a dose-, duration-, and sex-dependent manner. Modifications in the expression of specific histone acetyltransferase enzymes along with histone acetylation and methylation levels have been reported in rodent and murine models. Apart from these, down-regulation and up-regulation of certain microRNAs crucial for fetal development have been shown to be associated with in utero Pb exposure in human placenta samples. All these modifications in the developing fetus during the prenatal and perinatal stages reportedly caused severe abnormalities in early or adult age, such as impaired growth, obesity, autism, diabetes, cardiovascular diseases, risks of cancer development, and Alzheimer's disease. In this review, currently available information on Pb-mediated alterations in the fetal epigenome is summarized. Further research on Pb-induced epigenome modification will help to understand the mechanisms in detail and will enable us to formulate safety guidelines for pregnant women and developing children.


Asunto(s)
Epigénesis Genética , Plomo , Efectos Tardíos de la Exposición Prenatal , Adulto , Animales , Niño , Metilación de ADN , Femenino , Histonas/genética , Humanos , Plomo/toxicidad , Ratones , Placenta/metabolismo , Embarazo
2.
Curr Mol Med ; 22(2): 151-164, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33602084

RESUMEN

Liver fibrosis is one of the leading causes of cirrhotic liver disease, and the lack of therapies to treat fibrotic liver is a major concern. Liver fibrosis is mainly occurred by activation of hepatic stellate cells, and some stem cell therapies had previously reported for treatment. However, due to some problems with cell-based treatment, a safe therapeutic agent is vehemently sought by the researchers. Extracellular vesicles are cell-derived nanoparticles that are employed in several therapeutic approaches, including fibrosis, for their ability to transfer specific molecules in the target cells. In this review, the possibilities of extracellular vesicles to inactivate stellate cells are summarized and discussed. According to several studies, extracellular vesicles from different sources can either have beneficial or detrimental effects by regulating the activation of stellate cells. Therefore, targeting extracellular vesicles for maximizing or inhibiting their production is a potential approach for fibrotic liver treatment. Extracellular vesicles from different cells can also inactivate stellate cells by carrying out the paracrine effects of those cells, working as the agents. They are also implicated as a smart carrier of anti-fibrotic molecules when their respective parent cells are engineered to produce specific stellate cell-regulating substances. A number of studies showed stellate cell activation can be regulated by up/downregulation of specific proteins, and extracellular vesicle-based therapies can be an effective move to exploit these mechanisms. In conclusion, EVs are advantageous nano-carriers with the potential to treat fibrotic liver by inactivating activated stellate cells by various mechanisms.


Asunto(s)
Vesículas Extracelulares , Hepatopatías , Vesículas Extracelulares/metabolismo , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Humanos , Hígado/metabolismo , Cirrosis Hepática/metabolismo
3.
Am J Pathol ; 191(12): 2219-2230, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34428424

RESUMEN

Transdifferentiation (or activation) of hepatic stellate cells (HSCs) to myofibroblasts is a key event in liver fibrosis. Activated HSCs in the tumor microenvironment reportedly promote tumor progression. This study analyzed the effect of an inhibitor of HSC activation, retinol-binding protein-albumin domain III fusion protein (R-III), on protumorigenic functions of HSCs. Although conditioned medium collected from activated HSCs enhanced the migration, invasion, and proliferation of the hepatocellular carcinoma cell line Hepa-1c1c7, this effect was not observed in Hepa-1c1c7 cells treated with conditioned medium from R-III-exposed HSCs. In a subcutaneous tumor model, larger tumors with increased vascular density were formed in mice transplanted with Hepa-1c1c7+HSC than in mice transplanted with Hepa-1c1c7 cells alone. Intriguingly, when Hepa-1c1c7+HSC-transplanted mice were injected intravenously with R-III, a reduction in vascular density and extended tumor necrosis were observed. In an orthotopic tumor model, co-transplantation of HSCs enhanced tumor growth, angiogenesis, and regional metastasis accompanied by increased peritumoral lymphatic vessel density, which was abolished by R-III. In vitro study showed that R-III treatment affected the synthesis of pro-angiogenic and anti-angiogenic factors in activated HSCs, which might be the potential mechanism underlying the R-III effect. These findings suggest that the inhibition of HSC activation abrogates HSC-induced tumor angiogenesis and growth, which represents an attractive therapeutic strategy.


Asunto(s)
Carcinoma Hepatocelular/patología , Células Estrelladas Hepáticas/efectos de los fármacos , Neoplasias Hepáticas/patología , Proteínas Recombinantes de Fusión/farmacología , Albúminas/química , Albúminas/farmacología , Albúminas/uso terapéutico , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/irrigación sanguínea , Carcinoma Hepatocelular/terapia , Transformación Celular Neoplásica/efectos de los fármacos , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Femenino , Células Estrelladas Hepáticas/fisiología , Neoplasias Hepáticas/irrigación sanguínea , Neoplasias Hepáticas/terapia , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/genética , Neovascularización Patológica/prevención & control , Dominios y Motivos de Interacción de Proteínas/fisiología , Proteínas Recombinantes de Fusión/uso terapéutico , Proteínas de Unión al Retinol/farmacología , Proteínas de Unión al Retinol/uso terapéutico , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Biomedicines ; 8(10)2020 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-33086608

RESUMEN

Interstitial fibrosis is a common feature of chronic kidney disease, and platelet-derived growth factor receptor-ß (PDGFR-ß)-positive mesenchymal cells are reportedly the major source of scar-producing myofibroblasts. We had previously demonstrated that albumin and its derivative R-III (a retinol-binding protein-albumin domain III fusion protein) inhibited the transdifferentiation/activation of hepatic stellate cells (HSCs) to myofibroblasts and that R-III administration reduced liver fibrosis. In this study, we isolated cells (referred to as renal stellate cells, RSCs) from rat kidney tissues using the HSC isolation protocol and compared their morphological and biochemical characteristics with those of HSCs. RSCs shared many characteristics with HSCs, such as storage of vitamin A-containing lipid droplets and expression of HSC markers as well as pericyte markers. RSCs underwent spontaneous transdifferentiation into myofibroblasts in in vitro culture, which was inhibited by albumin expression or R-III treatment. We also evaluated the therapeutic effects of R-III in unilateral ureteral obstruction (UUO)-induced renal fibrosis in mice. Injected R-III localized predominantly in cytoglobin/stellate cell activation-associated protein (Cygb/STAP)-positive cells in the kidney and reduced renal fibrosis. These findings suggest that RSCs can be recognized as the renal counterparts of HSCs and that RSCs represent an attractive therapeutic target for anti-fibrotic therapy.

5.
World J Stem Cells ; 12(9): 938-951, 2020 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-33033556

RESUMEN

In recent years, several studies have reported positive outcomes of cell-based therapies despite insufficient engraftment of transplanted cells. These findings have created a huge interest in the regenerative potential of paracrine factors released from transplanted stem or progenitor cells. Interestingly, this notion has also led scientists to question the role of proteins in the secretome produced by cells, tissues or organisms under certain conditions or at a particular time of regenerative therapy. Further studies have revealed that the secretomes derived from different cell types contain paracrine factors that could help to prevent apoptosis and induce proliferation of cells residing within the tissues of affected organs. This could also facilitate the migration of immune, progenitor and stem cells within the body to the site of inflammation. Of these different paracrine factors present within the secretome, researchers have given proper consideration to stromal cell-derived factor-1 (SDF1) that plays a vital role in tissue-specific migration of the cells needed for regeneration. Recently researchers recognized that SDF1 could facilitate site-specific migration of cells by regulating SDF1-CXCR4 and/or HMGB1-SDF1-CXCR4 pathways which is vital for tissue regeneration. Hence in this study, we have attempted to describe the role of different types of cells within the body in facilitating regeneration while emphasizing the HMGB1-SDF1-CXCR4 pathway that orchestrates the migration of cells to the site where regeneration is needed.

6.
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
7.
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
8.
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
9.
Mol Cells ; 40(10): 737-751, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29047260

RESUMEN

Histone-modifying enzymes are key players in the field of cellular differentiation. Here, we used GSK-J4 to profile important target genes that are responsible for neural differentiation. Embryoid bodies were treated with retinoic acid (10 µM) to induce neural differentiation in the presence or absence of GSK-J4. To profile GSKJ4-target genes, we performed RNA sequencing for both normal and demethylase-inhibited cells. A total of 47 and 58 genes were up- and down-regulated, respectively, after GSK-J4 exposure at a log2-fold-change cut-off value of 1.2 (p-value < 0.05). Functional annotations of all of the differentially expressed genes revealed that a significant number of genes were associated with the suppression of cellular proliferation, cell cycle progression and induction of cell death. We also identified an enrichment of potent motifs in selected genes that were differentially expressed. Additionally, we listed upstream transcriptional regulators of all of the differentially expressed genes. Our data indicate that GSK-J4 affects cellular biology by inhibiting cellular proliferation through cell cycle suppression and induction of cell death. These findings will expand the current understanding of the biology of histone-modifying enzymes, thereby promoting further investigations to elucidate the underlying mechanisms.


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular/genética , Cuerpos Embrioides/metabolismo , Transcriptoma/genética , Benzazepinas/administración & dosificación , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Pirimidinas/administración & dosificación , Transcriptoma/efectos de los fármacos , Tretinoina/administración & dosificación
10.
Int J Mol Sci ; 18(7)2017 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-28657590

RESUMEN

Ethanol is well known as a teratogenic factor that is capable of inducing a wide range of developmental abnormalities if the developing fetus is exposed to it. Duration and dose are the critical parameters of exposure that affect teratogenic variation to the developing fetus. It is suggested that ethanol interferes with epigenetic processes especially DNA methylation. We aimed to organize all of the available information on the alteration of DNA methylation by ethanol in utero. Thus, we have summarized all published information regarding alcohol-mediated alterations in DNA methylation during gestation. We tried to arrange information in a way that anyone can easily find the alcohol exposure time, doses, sampling time, and major changes in genomic level. Manuscript texts will also represent the correlation between ethanol metabolites and subsequent changes in methylome patterns. We hope that this review will help future researchers to further examine the issues associated with ethanol exposure.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Etanol/toxicidad , Animales , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/genética , Femenino , Feto/efectos de los fármacos , Feto/embriología , Feto/metabolismo , Embarazo
11.
PLoS One ; 11(3): e0149976, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26930486

RESUMEN

Fetal alcohol spectrum disorder is a collective term representing fetal abnormalities associated with maternal alcohol consumption. Prenatal alcohol exposure and related anomalies are well characterized, but the molecular mechanism behind this phenomenon is not well characterized. In this present study, our aim is to profile important genes that regulate cellular development during fetal development. Human embryonic carcinoma cells (NCCIT) are cultured to form embryoid bodies and then treated in the presence and absence of ethanol (50 mM). We employed RNA sequencing to profile differentially expressed genes in the ethanol-treated embryoid bodies from NCCIT vs. EB, NCCIT vs. EB+EtOH and EB vs. EB+EtOH data sets. A total of 632, 205 and 517 differentially expressed genes were identified from NCCIT vs. EB, NCCIT vs. EB+EtOH and EB vs. EB+EtOH, respectively. Functional annotation using bioinformatics tools reveal significant enrichment of differential cellular development and developmental disorders. Furthermore, a group of 42, 15 and 35 transcription factor-encoding genes are screened from all of the differentially expressed genes obtained from NCCIT vs. EB, NCCIT vs. EB+EtOH and EB vs. EB+EtOH, respectively. We validated relative gene expression levels of several transcription factors from these lists by quantitative real-time PCR. We hope that our study substantially contributes to the understanding of the molecular mechanism underlying the pathology of alcohol-mediated anomalies and ease further research.


Asunto(s)
Cuerpos Embrioides/metabolismo , Etanol/farmacología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Análisis de Secuencia de ARN/métodos , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Depresores del Sistema Nervioso Central/farmacología , Células Madre de Carcinoma Embrionario/metabolismo , Femenino , Trastornos del Espectro Alcohólico Fetal/genética , Humanos , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Gene ; 576(1 Pt 1): 119-25, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26456191

RESUMEN

Fetal alcohol spectrum disorder is a collective term that represents fetal abnormalities associated with maternal alcohol consumption. Prenatal alcohol exposure and related anomalies are well characterized, but the molecular mechanism behind this phenomenon is not yet understood. Few insights have been gained from genetic and epigenetic studies of fetal alcohol spectrum disorder. Our aim was to profile the important molecular regulators of ethanol-related alterations of the genome. For this purpose, we have analyzed the gene expression pattern of human carcinoma cell-derived embryoid bodies in the absence or presence of ethanol. A cDNA microarray analysis was used to profile mRNA expression in embryoid bodies at day 7 with or without ethanol treatment. A total of 493 differentially expressed genes were identified in response to 50 mM ethanol exposure. Of these, 111 genes were up-regulated, and 382 were down-regulated. Gene ontology term enrichment analysis revealed that these genes are involved in important biological processes: neurological system processes, cognition, behavior, sensory perception of smell, taste and chemical stimuli and synaptic transmission. Similarly, the enrichment of disease-related genes included relevant categories such as neurological diseases, developmental disorders, skeletal and muscular disorders, and connective tissue disorders. Furthermore, we have identified a group of 26 genes that encode transcription factors. We validated the relative gene expression of several transcription factors using quantitative real time PCR. We hope that our study substantially contributes to the understanding of the molecular mechanisms underlying the pathology of alcohol-mediated anomalies and facilitates further research.


Asunto(s)
Carcinoma/metabolismo , Cuerpos Embrioides/metabolismo , Etanol/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Neoplasias/biosíntesis , Carcinoma/patología , Línea Celular Tumoral , Cuerpos Embrioides/patología , Trastornos del Espectro Alcohólico Fetal/metabolismo , Trastornos del Espectro Alcohólico Fetal/patología , Humanos
13.
Acta Biochim Biophys Sin (Shanghai) ; 47(8): 581-7, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26063602

RESUMEN

It is well known that consuming alcohol prior to and during pregnancy can cause harm to the developing fetus. Fetal alcohol spectrum disorder is a term commonly used to describe a range of disabilities that may arise from prenatal alcohol exposure such as fetal alcohol syndrome, partial fetal alcohol syndrome, alcohol-related neurodevelopmental disorders, and alcohol-related birth defects. Here, we report that maternal binge alcohol consumption alters several important genes that are involved in nervous system development in the mouse hippocampus at embryonic day 18. Microarray analysis revealed that Nova1, Ntng1, Gal, Neurog2, Neurod2, and Fezf2 gene expressions are altered in the fetal hippocampus. Pathway analysis also revealed the association of the calcium signaling pathway in addition to other pathways with the differentially expressed genes during early brain development. Alteration of such important genes and dynamics of the signaling pathways may cause neurodevelopmental disorders. Our findings offer insight into the molecular mechanism involved in neurodevelopmental disorders associated with alcohol-related defects.


Asunto(s)
Etanol/toxicidad , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Consumo de Bebidas Alcohólicas/efectos adversos , Animales , Señalización del Calcio/efectos de los fármacos , Femenino , Trastornos del Espectro Alcohólico Fetal/etiología , Hipocampo/embriología , Hipocampo/metabolismo , Ratones , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Embarazo , Transcriptoma/efectos de los fármacos
14.
Neurosci Lett ; 598: 73-8, 2015 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-25982323

RESUMEN

The objective of the present study was to investigate the changes in gene expression in the fetal brain (forebrain and hippocampus) caused by maternal binge alcohol consumption. Pregnant C57BL/6J mice were treated intragastrically with distilled phosphate-buffered saline (PBS) or ethanol (2.9 g/kg) from embryonic day (ED) 8-12. Microarray analysis revealed that a significant number of genes were altered at ED 18 in the developing brain. Specifically, in hippocampus, nuclear factor one alpha (Nfia) and three N-methyl-D-aspartate (Nmda) receptors (Nmdar1, Nmdar2b, and Nmdar2d) were down-regulated. The transcription factor Nfia controls gliogenesis, cell proliferation and Nmda-induced neuronal survival by regulating the expression of target genes. Some of the Nfia-target gene (Aldh1a, Folh1, Gjb6, Fgf1, Neurod1, Sept4, and Ntsr2) expressions were also altered as expected. These results suggest that the altered expression of Nfia and Nmda receptors may be associated with the etiology of fetal alcohol syndrome (FAS). The data presented in this report will contribute to the understanding of the molecular mechanisms associated with the effects of alcohol in FASD individuals.


Asunto(s)
Intoxicación Alcohólica/metabolismo , Encéfalo/metabolismo , Exposición Materna , Intercambio Materno-Fetal , Factores de Transcripción NFI/metabolismo , Animales , Encéfalo/embriología , Femenino , Feto/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Ratones Endogámicos C57BL , Embarazo
15.
Mol Biol Rep ; 42(7): 1233-9, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25697417

RESUMEN

Neural stem cells (NSCs) can be differentiated into one of three cell lineages: neurons, astrocytes or, oligodendrocytes. Some neurotoxins have the ability to deregulate this dynamic process. NSC cell fate can be altered by ethanol as reported previously. Our aim was to investigate the alteration of genes by ethanol during NSC differentiation and to explore the molecular mechanism underlying this phenomenon. Here, mouse fetal forebrain derived NSCs were differentiated for 2 days with or without of ethanol (50 mM). We performed a comparative microarray analysis at day two using GeneChip(®) Mouse Genome 430A 2.0 arrays. Microarray analysis showed that the expressions of 496 genes were altered by ethanol (56 and 440 were up- and down-regulated, respectively). Kyoto Encyclopedia of Genes and Genomes pathway analysis revealed the association of the following altered genes in the Wnt signaling pathway: Wnt5a, Csnk2a1, Tcf7l2, Ccnd2, Nlk, Tbl1x, Tbl1xr1, Rac2 and Nfatc3. Quantitative real time PCR analysis also demonstrated the relative expression levels of these genes. As Wnt signaling is a player of brain development, ethanol-induced alterations may contribute to improper development of the brain. Our data could be a useful resource for elucidating the mechanism behind the ethanol neurotoxicity in developing brain.


Asunto(s)
Astrocitos/efectos de los fármacos , Etanol/farmacología , Células-Madre Neurales/efectos de los fármacos , Neuronas/efectos de los fármacos , Transcriptoma , Animales , Astrocitos/citología , Astrocitos/metabolismo , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/genética , Ciclinas/genética , Ciclinas/metabolismo , Feto , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/citología , Neuronas/metabolismo , Oligodendroglía/citología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Cultivo Primario de Células , Prosencéfalo/citología , Prosencéfalo/efectos de los fármacos , Prosencéfalo/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Vía de Señalización Wnt
16.
J Appl Toxicol ; 34(1): 66-75, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23378141

RESUMEN

Fetal alcohol spectrum disorder (FASD) is a set of developmental malformations caused by excess alcohol consumption during pregnancy. Using an in vitro system, we examined the role that chronic ethanol (EtOH) exposure plays in gene expression changes during the early stage of embryonic differentiation. We demonstrated that EtOH affected the cell morphology, cell cycle progression and also delayed the down-regulation of OCT4 and NANOG during differentiation. Gene expression profiling and pathway analysis demonstrated that EtOH deregulates many genes and pathways that are involved in early embryogenesis. Follow-up analyzes revealed that EtOH exposure to embryoid bodies (EBs) induced the expression of an organizer-specific gene, goosecoid (GSC), in comparison to controls. Moreover, EtOH treatment altered several important genes that are involved in embryonic structure formation, nervous system development, and placental and embryonic vascularization, which are all common processes that FASD can disrupt. Specifically, EtOH treatment let to a reduction in ALDOC, ENO2 and CDH1 expression, whereas EtOH treatment induced the expression of PTCH1, EGLN1, VEGFA and DEC2 in treated EBs. We also found that folic acid (FA) treatment was able to correct the expression of the majority of genes deregulated by EtOH exposure during early embryo development. Finally, the present study identified a gene set including GSC, which was deregulated by EtOH exposure that may contribute to the etiology of fetal alcohol syndrome (FAS). We also reported that EtOH-induced GSC expression is mediated by Nodal signaling, which may provide a new avenue for analyzing the molecular mechanisms behind EtOH teratogenicity in FASD individuals.


Asunto(s)
Etanol/efectos adversos , Trastornos del Espectro Alcohólico Fetal/genética , Proteína Goosecoide/metabolismo , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Análisis por Conglomerados , Regulación hacia Abajo , Desarrollo Embrionario/efectos de los fármacos , Femenino , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Proteína Goosecoide/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Análisis por Micromatrices , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Placenta/efectos de los fármacos , Embarazo , Reproducibilidad de los Resultados , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA