Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Cells ; 10(11)2021 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-34831070

RESUMEN

Embryonic cancer stem cells (CSCs) can differentiate into any cancer type. Targeting CSC using natural compounds is a good approach as it suppresses cancer recurrence with fewer adverse effects, and methylsulfonylmethane (MSM) is a sulfur-containing compound with well-known anticancer activities. This study determined the mechanistic aspects of the anticancer activity of MSM. We used Western blotting and real-time qPCR for molecular signaling studies and conducted flow cytometry for analyzing the processes in cells. Our results suggested an inhibition in the expression of CSC markers and Wnt/ß-catenin signaling. MSM induced TRAIL-mediated extrinsic apoptosis in NCCIT and NTERA-2 cells rather than an intrinsic pathway. Inhibition of iron metabolism-dependent reactive oxygen species (ROS) generation takes part in TRAIL-mediated apoptosis induction by MSM. Suppressing iron metabolism by MSM also regulated p38/p53/ERK signaling and microRNA expressions, such as upregulating miR-130a and downregulating miR-221 and miR-222, which resulted in TRAIL induction and thereby extrinsic pathway of apoptosis. Hence, MSM could be a good candidate for neoadjuvant therapy by targeting CSCs by inhibiting iron metabolism.


Asunto(s)
Apoptosis , Dimetilsulfóxido/farmacología , Células Madre de Carcinoma Embrionario/patología , Hierro/metabolismo , Sulfonas/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Daño del ADN , Células Madre de Carcinoma Embrionario/efectos de los fármacos , Células Madre de Carcinoma Embrionario/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Modelos Biológicos , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
2.
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
3.
J Ethnopharmacol ; 246: 112214, 2020 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-31491437

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Medicinal herb Cichorium intybus L. (chicory) has been used traditionally for the treatment of various diseases, including diabetes. One of the promising therapeutic options to treat diabetes is replacing the degenerative pancreatic ß cells by stem cell-derived IPCs (insulin-producing cells). AIM OF THE STUDY: By the combination of cell therapy as a modern approach and traditional medicine, the current study was designed to evaluate the effects of chicory leaf extract (LE) on the differentiation potential of P19 EC cells (an embryonal carcinoma stem cell line) into IPCs. MATERIALS AND METHODS: The plant (voucher no. 4567) were collected and deposited in the herbarium of Shahrekord University. In vitro experiments were designed to compare the effects of various concentrations of LE on the differentiation potential of P19 EC cells. RESULTS: The differentiated cells showed morphological characteristics of pancreatic ß cells. They could also synthesized and secreted insulin when exposed to glucose. Moreover, the cells expressed specific proteins and genes of mature pancreatic ß cells. CONCLUSIONS: In conclusion, LE as a natural herbal extract was efficiently able to induce the differentiation of P19 EC cells into the clusters similar to pancreatic islets with the molecular, cellular and functional characteristics of mature ß cells.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Cichorium intybus , Células Madre de Carcinoma Embrionario/efectos de los fármacos , Insulina/metabolismo , Extractos Vegetales/farmacología , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Madre de Carcinoma Embrionario/fisiología , Células Secretoras de Insulina/metabolismo , Ratones , Fitoquímicos/análisis , Fitoquímicos/farmacología , Extractos Vegetales/química , Hojas de la Planta
4.
Dev Neurobiol ; 79(6): 559-577, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31177638

RESUMEN

A large number of studies have focused on the generation of dopaminergic neurons from pluripotent cells. Differentiation of stem cells into distinct cell types is influenced by tissue-specific microenvironment. Since, central nervous system undergoes further development during postnatal life, in the present study neonatal rat brain tissue extract (NRBE) was applied to direct the differentiation of embryonal carcinoma stem cell line, P19 into dopaminergic (DA) phenotypes. Additionally, a neuroprotective drug, deprenyl was used alone or in combination with the extract. Results from morphological, immunofluorescence, and qPCR analyses showed that during a period of one to three weeks, a large percentage of stem cells were differentiated into neural cells. The results also indicated the greater effect of NRBE on the differentiation of the cells into tyrosine hydroxylase-expressing cells. MS analysis of NRBE showed the enrichment of gene ontology terms related to cell differentiation and neurogenesis. Network analysis of the studied genes and some DA markers resulted in the suggestion of potential regulatory candidates such as AVP, ACHE, LHFPL5, and DLK1 genes. In conclusion, NRBE as a natural native inducer was apparently able to simulate the brain microenvironment and support neural differentiation of P19 cells.


Asunto(s)
Células Madre de Carcinoma Embrionario/efectos de los fármacos , Células Madre de Carcinoma Embrionario/enzimología , Regulación Enzimológica de la Expresión Génica , Selegilina/farmacología , Tirosina 3-Monooxigenasa/biosíntesis , Animales , Animales Recién Nacidos , Células Cultivadas , Inhibidores de la Monoaminooxidasa/farmacología , Ratas , Ratas Wistar , Tirosina 3-Monooxigenasa/genética
5.
Neurotoxicol Teratol ; 72: 58-70, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30776472

RESUMEN

Lead (Pb) is a teratogen that poses health risks after acute and chronic exposure. Lead is deposited in the bones of adults and is continuously leached into the blood for decades. While this chronic lead exposure can have detrimental effects on adults such as high blood pressure and kidney damage, developing fetuses and young children are particularly vulnerable. During pregnancy, bone-deposited lead is released into the blood at increased rates and can cross the placental barrier, exposing the embryo to the toxin. Embryos exposed to lead display serious developmental and cognitive defects throughout life. Although studies have investigated lead's effect on late-stage embryos, few studies have examined how lead affects stem cell determination and differentiation. For example, it is unknown whether lead is more detrimental to neuronal determination or differentiation of stem cells. We sought to determine the effect of lead on the determination and differentiation of pluripotent embryonic testicular carcinoma (P19) cells into neurons. Our data indicate that lead exposure significantly inhibits the determination of P19 cells to the neuronal lineage by alteration of N-cadherin and Sox2 expression. We also observed that lead significantly alters subsequent neuronal and glial differentiation. Consequently, this research emphasizes the need to reduce public exposure to lead.


Asunto(s)
Cadherinas/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Madre de Carcinoma Embrionario/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Plomo/toxicidad , Neuronas/efectos de los fármacos , Factores de Transcripción SOXB1/metabolismo , Teratógenos/toxicidad , Animales , Cadherinas/genética , Técnicas de Cultivo de Célula , Supervivencia Celular/efectos de los fármacos , Células Madre de Carcinoma Embrionario/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Ratones , Neuronas/metabolismo , Factores de Transcripción SOXB1/genética
6.
Biomed Res Int ; 2019: 4378710, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30800669

RESUMEN

Curcumin is a natural polyphenolic compound, isolated from Curcuma longa, and is an important ingredient of Asian foods. Curcumin has revealed its strong activities of anti-inflammatory, antioxidant, and anticancer. The efficient amount of curcumin could induce differentiation of stem cells and promoted the differentiation of glioma-initiating cells; however, the mechanisms underlying neural induction of curcumin have not yet been revealed. In this study, neural-inducing ability of curcumin was explored by using human pluripotent embryonal carcinoma cells, NTERA2 cells. The cells were induced toward neural lineage with curcumin and were compared with a standard neutralizing agent (retinoic acid). It was found that, after 14 days of the induction by curcumin, NTERA2 cells showed neuronal morphology and expressed neural-specific genes, including NeuroD, TUJ1, and PAX6. Importantly, curcumin activated neurogenesis of NTERA2 cells via the activation of autophagy, since autophagy-related genes, such as LC3, LAMP1, and ATG5, were upregulated along with the expression of neural genes. The inhibition of autophagy by chloroquine suppressed both autophagy and neural differentiation, highlighting the positive role of autophagy during neural differentiation. This autophagy-mediated neural differentiation of curcumin was found to be an ROS-dependent manner; curcumin induced ROS generation and suppressed antioxidant gene expression. Altogether, this study proposed the neural-inducing activity of curcumin via the regulation of autophagy within NTERA2 cells and underscored the health beneficial effects of curcumin for neurodegenerative disorders, such as Alzheimer's disease and Parkinson's disease.


Asunto(s)
Autofagia/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Curcumina/farmacología , Células Madre de Carcinoma Embrionario/efectos de los fármacos , Neuronas/efectos de los fármacos , Línea Celular Tumoral , Curcuma/química , Expresión Génica/efectos de los fármacos , Humanos , Neurogénesis/efectos de los fármacos , Extractos Vegetales/farmacología , Células Madre/efectos de los fármacos
7.
Mol Cell Biol ; 38(21)2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30104253

RESUMEN

The involvement of cell division in cellular differentiation has long been accepted. Cell division may be required not only for the expansion of a differentiated cell population but also for the execution of differentiation processes. Nonetheless, knowledge regarding how specific differentiation processes are controlled in a cell division-dependent manner is far from complete. Here, we determined the involvement of cell division in neuronal differentiation. We initially confirmed that cell division is an essential event for the neuronal differentiation of P19 embryonic carcinoma cells. We investigated the induction mechanisms of Tshz1, whose expression is induced by retinoic acid (RA) in a cell division-dependent manner. Promoter analysis of Tshz1 revealed a specific region required for RA-dependent transcription. A series of experiments was used to identify E2F1 as the induction factor for the RA-dependent transcription of Tshz1 We propose that E2F1 mediates neuronal differentiation in a cell division-dependent manner.


Asunto(s)
Diferenciación Celular/genética , División Celular/genética , Factor de Transcripción E2F1/genética , Proteínas Represoras/genética , Transcripción Genética/genética , Tretinoina/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular Tumoral , Células Madre de Carcinoma Embrionario/efectos de los fármacos , Células HeLa , Proteínas de Homeodominio , Humanos , Ratones , Neuronas/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Transcripción Genética/efectos de los fármacos
8.
Cell Death Dis ; 9(2): 246, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29445146

RESUMEN

Oxidative stress generates reactive oxygen species (ROS) that can promote or inhibit cardiac differentiation of stem cells dependent on the intensity of stimuli as well as cellular context in redox and differentiation status. In the current study, we confirmed that suitable intensity of hydrogen peroxide at the formation stage of embryoid bodies (EBs) effectively favored the formation of spontaneously beating cardiomyocytes from P19 embryonal carcinoma cells. Mechanistic studies implicated that extrinsic ROS enhanced the Caspase-mediated degradation of Oct4 and Nanog, two factors that governing pluripotent property. Further experiments suggested that a cohort of Nanog together with histone deacetylase 4 (Hdac4) played a critical role in establishing and maintaining the silent transcriptional status of Gata4 and Nkx2.5 in undifferentiated cells. Thus, an impulse of hydrogen peroxide depleted Nanog and Hdac4 via a caspase-dependent manner to ameliorate the repression on Gata4 and Nkx2.5 promoters, thereby generating a persistent activation on cardiac differentiation program. Meanwhile, we found that excessive ROS-activated JNK cascade to facilitate the ubiquitination and subsequent degradation of Gata4 protein. Overall, our results indicate that suitable ROS promotes the activation of Gata4 in transcription, while excessive ROS targets Gata4 protein for proteasome-dependent degradation. Gata4 is an important modulator balancing the promoting and inhibitory effects of oxidative stress on differentiation program of cardiomyogenesis.


Asunto(s)
Células Madre de Carcinoma Embrionario/efectos de los fármacos , Factor de Transcripción GATA4/genética , Regulación del Desarrollo de la Expresión Génica , Peróxido de Hidrógeno/farmacología , Miocitos Cardíacos/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Animales , Caspasas/genética , Caspasas/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Embrión de Mamíferos , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Células Madre de Carcinoma Embrionario/citología , Células Madre de Carcinoma Embrionario/metabolismo , Factor de Transcripción GATA4/metabolismo , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Proteína Homeótica Nkx-2.5/genética , Proteína Homeótica Nkx-2.5/metabolismo , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , 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 , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Regiones Promotoras Genéticas , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis/efectos de los fármacos , Transducción de Señal , Ubiquitinación/efectos de los fármacos
9.
Cell Cycle ; 17(3): 362-366, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29372665

RESUMEN

The epigenetic mechanisms underlying chemoresistance in cancer cells resulting from drug-induced reversible senescence are poorly understood. Chemoresistant ESC-like embryonal carcinoma PA1 cells treated with etoposide (ETO) were previously found to undergo prolonged G2 arrest with transient p53-dependent upregulation of opposing fate regulators, p21CIP1 (senescence) and OCT4A (self-renewal). Here we report on the analysis of the DNA methylation state of the distal enhancer (DE) and proximal enhancer (PE) of the Oct4A gene during this dual response. When compared to non-treated controls the methylation level increased from 1.3% to 12.5% and from 3% to 19.4%, in the DE and PE respectively. It included CpG and non-CpG methylation, which was not chaotic but presented two patterns in each enhancer. Discorrelating with methylation of enhancers, the transcription of Oct4A increased, however, a strong expression of the splicing form Oct4B was also induced, along with down-regulation of the Oct4A partners of in the pluripotency/self-renewal network Sox2 and Lin28. WB demonstrated disjoining of the OCT4A protein from the chromatin-bound fraction. In survival clones, methylation of the DE was considerably erased, while some remnant of methylation of the PE was still observed. The alternative splicing for Oct4B was reduced, Oct4A level insignificantly decreased, while the expression of Sox2 and Lin28 recovered, all three became proportionally above the control. These findings indicate the involvement of the transient patterned methylation of the Oct4A enhancers and alternative splicing in the adaptive regulation of cell fate choice during the p53-dependant dual state of reversible senescence in ESC-like cancer stem cells.


Asunto(s)
Empalme Alternativo/genética , Senescencia Celular/efectos de los fármacos , Metilación de ADN/genética , Células Madre de Carcinoma Embrionario/metabolismo , Elementos de Facilitación Genéticos/genética , Etopósido/farmacología , Factor 3 de Transcripción de Unión a Octámeros/genética , Células Madre Pluripotentes/metabolismo , Empalme Alternativo/efectos de los fármacos , Secuencia de Bases , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Clonales , Metilación de ADN/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Células Madre de Carcinoma Embrionario/efectos de los fármacos , Humanos , Células Madre Pluripotentes/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
10.
Int J Nanomedicine ; 12: 7529-7549, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29066898

RESUMEN

BACKGROUND: Silver nanoparticles (AgNPs) exhibit strong antibacterial and anticancer activity owing to their large surface-to-volume ratios and crystallographic surface structure. Owing to their various applications, understanding the mechanisms of action, biological interactions, potential toxicity, and beneficial effects of AgNPs is important. Here, we investigated the toxicity and differentiation-inducing effects of AgNPs in teratocarcinoma stem cells. MATERIALS AND METHODS: AgNPs were synthesized and characterized using various analytical techniques such as UV-visible spectroscopy, X-ray diffraction, energy-dispersive X-ray spectroscopy, and transmission electron microscopy. The cellular responses of AgNPs were analyzed by a series of cellular and biochemical assays. Gene and protein expressions were analyzed by reverse transcription-quantitative polymerase chain reaction and western blotting, respectively. RESULTS: The AgNPs showed typical crystalline structures and spherical shapes (average size =20 nm). High concentration of AgNPs induced cytotoxicity in a dose-dependent manner by increasing lactate dehydrogenase leakage and reactive oxygen species. Furthermore, AgNPs caused mitochondrial dysfunction, DNA fragmentation, increased expression of apoptotic genes, and decreased expression of antiapoptotic genes. Lower concentrations of AgNPs induced neuronal differentiation by increasing the expression of differentiation markers and decreasing the expression of stem cell markers. Cisplatin reduced the viability of F9 cells that underwent AgNPs-induced differentiation. CONCLUSION: The results showed that AgNPs caused differentially regulated cytotoxicity and induced neuronal differentiation of F9 cells in a concentration-dependent manner. Therefore, AgNPs can be used for differentiation therapy, along with chemotherapeutic agents, for improving cancer treatment by targeting specific chemotherapy-resistant cells within a tumor. Furthermore, understanding the molecular mechanisms of apoptosis and differentiation in stem cells could also help in developing new strategies for cancer stem cell (CSC) therapies. The findings of this study could significantly contribute to the nanomedicine because this study is the first of its kind, and our results will lead to new strategies for cancer and CSC therapies.


Asunto(s)
Apoptosis , Diferenciación Celular , Células Madre de Carcinoma Embrionario/patología , Nanopartículas del Metal/química , Modelos Biológicos , Plata/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cisplatino/farmacología , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Células Madre de Carcinoma Embrionario/efectos de los fármacos , Matriz Extracelular/metabolismo , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Especies Reactivas de Oxígeno/farmacología , Plata/química , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Difracción de Rayos X
11.
Cell Death Differ ; 24(11): 1975-1986, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28885616

RESUMEN

We have previously reported that myeloid differentiation primary response gene 88 (MyD88) is downregulated during all-trans retinoic acid (RA)-induced differentiation of pluripotent NTera2 human embryonal carcinoma cells (hECCs), whereas its maintained expression is associated with RA differentiation resistance in nullipotent 2102Ep hECCs. MyD88 is the main adapter for toll-like receptor (TLR) signalling, where it determines the secretion of chemokines and cytokines in response to pathogens. In this study, we report that loss of MyD88 is essential for RA-facilitated differentiation of hECCs. Functional analysis using a specific MyD88 peptide inhibitor (PepInh) demonstrated that high MyD88 expression in the self-renewal state inhibits the expression of a specific set of HOX genes. In NTera2 cells, MyD88 is downregulated during RA-induced differentiation, a mechanism that could be broadly replicated by MyD88 PepInh treatment of 2102Ep cells. Notably, MyD88 inhibition transitioned 2102Ep cells into a stable, self-renewing state that appears to be primed for differentiation upon addition of RA. At a molecular level, MyD88 inhibition combined with RA treatment upregulated HOX, RA signalling and TLR signalling genes. These events permit differentiation through a standard downregulation of Oct4-Sox2-Nanog mechanism. In line with its role in regulating secretion of specific proteins, conditioned media experiments demonstrated that differentiated (MyD88 low) NTera2 cell media was sufficient to differentiate NTera2 cells. Protein array analysis indicated that this was owing to secretion of factors known to regulate angiogenesis, neurogenesis and all three branches of TGF-ß Superfamily signalling. Collectively, these data offer new insights into RA controlled differentiation of pluripotent cells, with notable parallels to the ground state model of embryonic stem cell self-renewal. These data may provide insights to facilitate improved differentiation protocols for regenerative medicine and differentiation-therapies in cancer treatment.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre de Carcinoma Embrionario/patología , Factor 88 de Diferenciación Mieloide/metabolismo , Células Madre Pluripotentes/patología , Tretinoina/farmacología , Diferenciación Celular/genética , Autorrenovación de las Células/efectos de los fármacos , Autorrenovación de las Células/genética , Células Madre de Carcinoma Embrionario/efectos de los fármacos , Células Madre de Carcinoma Embrionario/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Mesodermo/patología , Modelos Biológicos , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
12.
Methods Mol Biol ; 1601: 61-70, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28470517

RESUMEN

The incidence of neurological diseases including learning and developmental disorders has increased in recent years. Concurrently, the number and volume of worldwide registered and traded chemicals have also increased. There is a broad consensus that the developing brain is particularly sensitive to damage by chemicals and that evaluation of chemicals for developmental toxicity or neurotoxicity is critical to human health. Human pluripotent embryonal carcinoma (NTERA-2 or NT2) cells are increasingly considered as a suitable model for in vitro developmental toxicity and neurotoxicity (DT/DNT) studies as they undergo neuronal differentiation upon stimulation with retinoic acid (RA) and allow toxicity assessment at different stages of maturation. Here we describe a protocol for cell fitness screening in differentiating NT2 cells based on the analysis of intracellular ATP levels allowing for the identification of chemicals which are potentially harmful to the developing brain. The described method is suitable to be adapted to low-, medium-, and high-throughput screening and allows multiplexing with other cell fitness indicators. While the presented protocol focuses on cell fitness screening in human pluripotent stem cells it may also be applied to other in vitro models.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/embriología , Supervivencia Celular/efectos de los fármacos , Células Madre de Carcinoma Embrionario/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Células Madre Pluripotentes/efectos de los fármacos , Pruebas de Toxicidad , Adenosina Trifosfato/metabolismo , Diferenciación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Humanos , Neuronas/citología , Neuronas/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/toxicidad , Tretinoina/toxicidad
13.
Methods Mol Biol ; 1601: 205-214, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28470528

RESUMEN

Glycine receptor chloride channels (GlyRs) are attractive drug targets for therapeutic intervention and are also more and more recognized in the context of in vitro neurotoxicity and developmental neurotoxicity testing. Assaying the functional properties of GlyR can serve as an indicator of cellular viability and the integrity of the developing and mature central nervous system. Human pluripotent NTERA-2 (NT2) stem cells undergo neuronal differentiation upon stimulation with retinoic acid and express a large variety of neuronal proteins-including GlyR. YFP-I152L, a halide-sensitive variant of yellow fluorescent protein, allows high-throughput fluorescence-based functional analysis of GlyRs in NT2 cells. Here we describe a protocol for phenotyping of cellular viability by functional analysis of GlyR in neuronally differentiated NT2 (NT2-N) cells using YFP-I152L as a reporter of functional integrity of GlyRs. The protocol describes neuronal differentiation of NT2 stem cells, transient transfection of NT2-N cells with YFP-I152L as well as functional imaging and analysis of data from high-content imaging.


Asunto(s)
Supervivencia Celular , Células Madre de Carcinoma Embrionario/citología , Neurogénesis , Neuronas/citología , Células Madre Pluripotentes/citología , Receptores de Glicina/metabolismo , Diferenciación Celular , Evaluación Preclínica de Medicamentos , Células Madre de Carcinoma Embrionario/efectos de los fármacos , Humanos , Proteínas Luminiscentes/metabolismo , Neurogénesis/efectos de los fármacos , Imagen Óptica/métodos , Tretinoina/farmacología
14.
Anticancer Agents Med Chem ; 17(9): 1184-1198, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27306567

RESUMEN

Retinoic acid (RA), especially all-trans retinoic acid is the most potent natural metabolite of vitamin A. RA is involved in a variety of biological functions including embryogenesis, cell differentiation and apoptosis. RA acts through its nuclear receptors to induce transcription of specific target genes. Mouse P19 embryonic carcinoma (EC) stem cells (ES) are one of the most studied in vitro systems for RA-induced differentiation. P19 ES cells can differentiate to endodermal-like, mesodermal-like, and neuronal-like phenotypes in response to specific morphogens including RA and dimethyl sulfoxide (DMSO). At low concentrations, RA directs P19 ES cells to differentiate into cells displaying an endodermal phenotype, whereas at higher concentrations it induces differentiation to neuroectoderm. In the past, many RA---regulated genes have been discovered in EC and ES cells and efforts are ongoing to elucidate the exact mechanisms of RA-induced ES cell differentiation and apoptosis. In the RA-triggered differentiation process of the P19 ES cells, several proteins belonging to different families participate, some being obligatory while others, dispensable. Revealing the mechanisms behind RA-induced effects on ES cells has a bearing on understanding how cells proliferate, differentiate and undergo apoptosis that can provide greater insight into cancer biology and therapy. In addition to summarizing the reports on gene/protein targets of RA in stem cells, the signaling pathways driven by some of the specific class of proteins in the presence or absence of RA in P19 ES cell differentiation, especially to an endodermal phenotype, are the focus of this review.


Asunto(s)
Células Madre de Carcinoma Embrionario/metabolismo , Transducción de Señal , Tretinoina/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Células Madre de Carcinoma Embrionario/efectos de los fármacos , Células Madre de Carcinoma Embrionario/patología , Ratones , Tretinoina/farmacología
15.
Mol Cell Probes ; 32: 46-54, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28013042

RESUMEN

Murine P19 embryonal carcinoma (EC) cells are convenient to differentiate into all germ layer derivatives. One of the advantages of P19 cells is that the exogenous DNA can be easily inserted into them. Here, at the first part of this study, we generated stable GFP-expressing P19 cells (P19-GFP+). FACS and western-blot analysis confirmed stable expression of GFP in the cells. We previously demonstrated the efficient induction of neuronal differentiation from mouse ES and EC cells by application of a neuroprotective drug, selegiline In the second part of this study selegiline was used to induce differentiation of P19-GFP+ into stable GFP-expressing neuron-like cells. Cresyl violet staining confirmed neuronal morphology of the differentiated cells. Furthermore, real-time PCR and immunoflourescence approved the expression of neuron specific markers. P19-GFP+ cells were able to survive, migrate and integrated into host tissues when transplanted to developing chick embryo CNS. The obtained live GFP-expressing cells can be used as an abundant source of developmentally pluripotent material for transplantation studies, investigating the cellular and molecular aspects of early differentiation.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre de Carcinoma Embrionario/patología , Proteínas Fluorescentes Verdes/metabolismo , Neuronas/metabolismo , Neuronas/patología , Animales , Diferenciación Celular/efectos de los fármacos , Pollos , Células Madre de Carcinoma Embrionario/efectos de los fármacos , Células Madre de Carcinoma Embrionario/trasplante , Fluorescencia , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ratones , Neuronas/efectos de los fármacos , Selegilina/farmacología , Transfección
16.
Toxicol In Vitro ; 35: 86-92, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27247146

RESUMEN

Calmidazolium chloride (CMZ) is widely used as a calmodulin (CaM) antagonist, but is also known to induce apoptosis in certain cancer cell lines. However, in spite of the importance of cancer stem cells (CSCs) in cancer therapy, the effects of CMZ on CSCs are not yet well understood. We investigated the effects of CMZ on the F9 embryonal carcinoma cell (ECC) line as a surrogate model of CSCs. To avoid bias due to culture conditions, F9 ECCs and E14 embryonic stem cells (ESCs) were grown in the same culture medium. Results obtained using a cell-counting kit showed that CMZ significantly inhibited growth in F9 ECCs compared with growth in E14 ESCs. CMZ also induced apoptosis of F9 ECCs, but not of E14 ESCs, which was associated with caspase-3 activation and an increased fraction of the sub-G1 cell population. In addition, our data revealed that the expression of stemness-related genes including c-Myc was selectively down regulated in CMZ-treated F9 ECCs. Our results suggest that CMZ can inhibit the growth of ECCs by inducing apoptosis and down regulating stemness-related genes, without causing any harm to normal stem cells. These findings indicate a potential application of CMZ in the development of anti-CSC therapeutics.


Asunto(s)
Antineoplásicos/farmacología , Células Madre de Carcinoma Embrionario/efectos de los fármacos , Imidazoles/farmacología , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Fragmentación del ADN , Regulación hacia Abajo , Células Madre de Carcinoma Embrionario/metabolismo , Ratones , Proteína Homeótica Nanog/genética , Proteínas Proto-Oncogénicas c-myc/genética , Factores de Transcripción SOXB1/genética
17.
Biochem Biophys Res Commun ; 470(2): 300-305, 2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26774337

RESUMEN

Nicotine is considered to contribute to the health risks associated with cigarette smoking. Nicotine exerts its cellular functions by acting on nicotinic acetylcholine receptors (nAChRs), and adversely affects normal embryonic development. However, nicotine toxicity has not been elucidated in human embryonic stage. In the present study, we examined the cytotoxic effects of nicotine in human multipotent embryonal carcinoma cell line NT2/D1. We found that exposure to 10 µM nicotine decreased intracellular ATP levels and inhibited proliferation of NT2/D1 cells. Because nicotine suppressed energy production, which is a critical mitochondrial function, we further assessed the effects of nicotine on mitochondrial dynamics. Staining with MitoTracker revealed that 10 µM nicotine induced mitochondrial fragmentation. The levels of the mitochondrial fusion proteins, mitofusins 1 and 2, were also reduced in cells exposed to nicotine. These nicotine effects were blocked by treatment with mecamylamine, a nonselective nAChR antagonist. These data suggest that nicotine degrades mitofusin in NT2/D1 cells and thus induces mitochondrial dysfunction and cell growth inhibition in a nAChR-dependent manner. Thus, mitochondrial function in embryonic cells could be used to assess the developmental toxicity of chemicals.


Asunto(s)
Células Madre de Carcinoma Embrionario/metabolismo , GTP Fosfohidrolasas/metabolismo , Mitocondrias/metabolismo , Dinámicas Mitocondriales/efectos de los fármacos , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas Mitocondriales/metabolismo , Nicotina/administración & dosificación , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Células Madre de Carcinoma Embrionario/efectos de los fármacos , Células Madre de Carcinoma Embrionario/patología , Células Madre Embrionarias Humanas/efectos de los fármacos , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/patología , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/patología
18.
Toxicol Sci ; 149(1): 15-30, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26385866

RESUMEN

In utero exposure to certain chemicals can impair embryo development, causing embryonic death, growth retardation, or severe birth defects. Establishment of effective in vitro tests is crucial for identifying developmental toxicants and for reducing the financial and ethical burden of animal-based tests. Previously, we created an in vitro morphogenesis model using pluripotent P19C5 mouse embryonal carcinoma stem cells that mimics the process of gastrulation and axial body elongation of embryos. Because many birth defects are caused by dysregulation of cellular behaviors during embryogenesis, the morphogenesis model may serve as a unique tool to investigate the impacts of developmental toxicants. The aim of this study is to evaluate the applicability and limitations of the model using 20 therapeutic drugs, 16 of which are contraindicated in pregnancy and 4 are considered safe. P19C5 embryoid bodies (EBs) were exposed to different concentrations of drugs during 4 days of 3-dimensional culture. The treatment effects on growth and morphogenesis were analyzed using morphometric measurements of EB size and shape, respectively. Viability assays of P19C5 cells and NIH/3T3 fibroblasts were used to determine the drug concentrations that caused general cytotoxicity and those that selectively diminished P19C5 proliferation relative to NIH/3T3 proliferation. Thirteen contraindicated drugs diminished P19C5 cell proliferation, reduced EB growth, or altered morphogenesis at concentrations below generally cytotoxic levels. Two safe drugs also exhibited these impacts at the highest concentration tested. Although additional validation studies are required, this study introduces morphogenesis-based stem cell models as potentially effective in vitro tools for developmental toxicity research.


Asunto(s)
Cuerpos Embrioides/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Morfogénesis/efectos de los fármacos , Animales , Proliferación Celular , Relación Dosis-Respuesta a Droga , Evaluación de Medicamentos , Células Madre de Carcinoma Embrionario/efectos de los fármacos , Femenino , Ratones , Células 3T3 NIH , Embarazo
19.
Toxicology ; 338: 69-76, 2015 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-26498558

RESUMEN

Human pluripotent embryonal carcinoma (NT2) cells are increasingly considered as a suitable model for in vitro toxicity testing, e.g. developmental toxicity and neurotoxicity (DT/DNT) studies, as they undergo neuronal differentiation upon stimulation with retinoic acid (RA) and permit toxicity testing at different stages of maturation. NT2 cells have recently been reported to show specific changes in dielectric resistance profiles during differentiation which can be observed as early as 24h upon RA-stimulation. These observations suggest altered susceptibility to chemicals at an early stage of differentiation. However, chemical susceptibility of early differentiating NT cells has not yet been studied. To address this question, we have established a cell fitness screening assay based on the analysis of intracellular ATP levels and we applied the assay in a large-scale drug screening experiment in NT2 stem cells and early differentiating NT2 cells. Subsequent analysis of ranked fitness phenotypes revealed 19 chemicals with differential toxicity profile in early differentiating NT2 cells. To evaluate whether any of the identified drugs have previously been associated with DT/DNT, we conducted a literature search on the identified molecules and quantified the fraction of chemicals assigned to the FDA (Food and Drug Administration) pregnancy risk categories (PRC) N, A, B, C, D, and X in the hit list and the small molecule library. While the fractions of the categories N and B were decreased (0.81 and 0.35-fold), the classes C, D and X were increased (1.35, 1.47 and 3.27-fold) in the hit list compared to the chemical library. From these data as well as from the literature review, identifying large fractions of chemicals being directly (∼42%) and indirectly associated with DT/DNT (∼32%), we conclude that our method may be beneficial to systematic in vitro-based primary screening for developmental toxicants and neurotoxicants and we propose cell fitness screening in early differentiating NT2 cells as a strategy for evaluating chemical susceptibility at different stages of differentiation to reduce animal testing in the context of the 3Rs.


Asunto(s)
Células Madre de Carcinoma Embrionario/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Síndromes de Neurotoxicidad/etiología , Células Madre Pluripotentes/efectos de los fármacos , Pruebas de Toxicidad/métodos , Tretinoina/farmacología , Adenosina Trifosfato/metabolismo , Biomarcadores/metabolismo , Línea Celular Tumoral , Biología Computacional , Bases de Datos Factuales , Células Madre de Carcinoma Embrionario/metabolismo , Células Madre de Carcinoma Embrionario/patología , Ensayos Analíticos de Alto Rendimiento , Humanos , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Síndromes de Neurotoxicidad/metabolismo , Síndromes de Neurotoxicidad/patología , Fenotipo , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/patología , Medición de Riesgo , Bibliotecas de Moléculas Pequeñas , Factores de Tiempo
20.
Chin J Nat Med ; 13(8): 602-8, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26253493

RESUMEN

The purpose of this study was to establish a drug screening method for small molecules extracted from traditional Chinese medicines (TCM) that have neuronal differentiation promoting effects, using P19 embryonic carcinoma cell as a cell-based model. First, the constructed plasmid (pTα1-Luc) was transfected into P19 cells to establish a screening model. Second, several TCMs were screened using the established model and all-trans-retinoic acid as a positive control. Finally, the underlying molecular mechanism was explored using immunofluorescence staining, qT-PCR, and Western blot analysis. Our results indicated that the drug screen model was established successfully and that both honokiol and hyperoside induced P19 differentiation into neurons, with the possible molecular mechanism being modulating the Wnt signaling pathway. In conclusion, the drug screening model developed in the present study provides a rapid, cell-based screening platform for identifying natural compounds with neuronal differentiation effects.


Asunto(s)
Compuestos de Bifenilo/farmacología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Células Madre de Carcinoma Embrionario/efectos de los fármacos , Lignanos/farmacología , Neuronas/efectos de los fármacos , Quercetina/análogos & derivados , Animales , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos/métodos , Ratones , Quercetina/farmacología , Tretinoina/fisiología , Vía de Señalización Wnt
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...