Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Cell Rep ; 42(6): 112645, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37314926

RESUMEN

Various culture conditions by small molecules have been explored to extend pluripotency of stem cells, but their impacts on cell fate in vivo remain elusive. We systematically compared the effects of various culture conditions on the pluripotency and cell fate in vivo of mouse embryonic stem cells (ESCs) by tetraploid embryo complementation assay. Conventional ESC cultures in serum/LIF-based condition produced complete ESC mice and also the survival to adulthood at the highest rates of all other chemical-based cultures. Moreover, long-term examination of the survived ESC mice demonstrated that conventional ESC cultures did not lead to visible abnormality for up to 1.5-2 years, whereas the prolonged chemical-based cultures developed retroperitoneal atypical teratomas or leiomyomas. The chemical-based cultures exhibited transcriptomes and epigenomes that typically differed from those of conventional ESC cultures. Our results warrant further refinement of culture conditions in promoting the pluripotency and safety of ESCs in future applications.


Asunto(s)
Células Madre Pluripotentes , Teratoma , Ratones , Animales , Células Madre Embrionarias de Ratones/patología , Células Cultivadas , Células Madre Embrionarias , Teratoma/patología , Diferenciación Celular
2.
Int J Mol Sci ; 22(23)2021 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-34884750

RESUMEN

Cyclic siloxane octamethylcyclotetrasiloxane (D4) has raised concerns as an endocrine-disrupting chemical (EDC). D4 is widely used in detergent products, cosmetics, and personal care products. Recently, robust toxicological data for D4 has been reported, but the adverse effects of D4 on brain development are unknown. Here, pregnant mice on gestational day 9.5 were treated daily with D4 to postnatal day 28, and the offspring mice were studied. The prenatal D4-treated mice exhibited cognitive dysfunction, limited memory, and motor learning defect. Moreover, prenatal D4 exposure reduced the proliferation of neuronal progenitors in the offspring mouse brain. Next, the mechanisms through which D4 regulated the cell cycle were investigated. Aberrant gene expression, such as cyclin-dependent kinases CDK6 and cyclin-dependent kinase inhibitor p27, were found in the prenatal D4-treated mice. Furthermore, the estrogen receptors ERa and ERb were increased in the brain of prenatal D4-treated mice. Overall, these findings suggest that D4 exerts estrogen activity that affects the cell cycle progression of neuronal progenitor cells during neurodevelopment, which may be associated with cognitive deficits in offspring.


Asunto(s)
Disruptores Endocrinos/toxicidad , Células-Madre Neurales/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/patología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Siloxanos/toxicidad , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Proliferación Celular , Cognición/efectos de los fármacos , Disruptores Endocrinos/administración & dosificación , Femenino , Técnicas de Sustitución del Gen , Proteínas Fluorescentes Verdes/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Neurológicos , Actividad Motora/efectos de los fármacos , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Embrionarias de Ratones/patología , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Neurogénesis/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal/psicología , Factores de Transcripción SOXB1/genética , Siloxanos/administración & dosificación , Conducta Social
3.
Toxicol Appl Pharmacol ; 433: 115781, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34737147

RESUMEN

The cardiac embryonic stem cell test (ESTc) is an in vitro embryotoxicity screen which uses cardiomyocyte formation as the main differentiation route. Studies are ongoing into whether an improved specification of the biological domain can broaden the applicability of the test, e.g. to discriminate between structurally similar chemicals by measuring expression of dedicated gene transcript biomarkers. We explored this with two chemical classes: morpholines (tridemorph; fenpropimorph) and piperidines (fenpropidin; spiroxamine). These compounds cause embryotoxicity in rat such as cleft palate. This malformation can be linked to interference with retinoic acid balance, neural crest (NC) cell migration, or cholesterol biosynthesis. Also neural differentiation within the ESTc was explored in relation to these compounds. Gene transcript expression of related biomarkers were measured at low and high concentrations on differentiation day 4 (DD4) and DD10. All compounds showed stimulating effects on the cholesterol biosynthesis related marker Msmo1 after 24 h exposure and tridemorph showed inhibition of Cyp26a1 which codes for one of the enzymes that metabolises retinoic acid. A longer exposure duration enhanced expression levels for differentiation markers for cardiomyocytes (Nkx2-5; Myh6) and neural cells (Tubb3) on DD10. This readout gave additional mechanistic insight which enabled previously unavailable in vitro discrimination between the compounds, showing the practical utility of specifying the biological domain of the ESTc.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Morfolinas/toxicidad , Células Madre Embrionarias de Ratones/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Piperidinas/toxicidad , Pruebas de Toxicidad , Animales , Células Cultivadas , Redes Reguladoras de Genes , Proteína Homeótica Nkx-2.5/genética , Proteína Homeótica Nkx-2.5/metabolismo , Ratones , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Embrionarias de Ratones/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Ácido Retinoico 4-Hidroxilasa/genética , Ácido Retinoico 4-Hidroxilasa/metabolismo , Medición de Riesgo , Compuestos de Espiro/toxicidad , Factores de Tiempo , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
4.
Cell Rep ; 37(3): 109851, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34686339

RESUMEN

Early embryogenesis depends on proper control of intracellular homeostasis of ions including Ca2+ and Mg2+. Deletion of the Ca2+ and Mg2+ conducting the TRPM7 channel is embryonically lethal in mice but leaves compaction, blastomere polarization, blastocoel formation, and correct specification of the lineages of the trophectoderm and inner cell mass unaltered despite that free cytoplasmic Ca2+ and Mg2+ is reduced at the two-cell stage. Although Trpm7-/- embryos are able to hatch from the zona pellucida, no expansion of Trpm7-/- trophoblast cells can be observed, and Trpm7-/- embryos are not identifiable in utero at E6.5 or later. Given the proliferation and adhesion defect of Trpm7-/- trophoblast stem cells and the ability of Trpm7-/- ESCs to develop to embryos in tetraploid embryo complementation assays, we postulate a critical role of TRPM7 in trophectoderm cells and their failure during implantation as the most likely explanation of the developmental arrest of Trpm7-deficient mouse embryos.


Asunto(s)
Calcio/metabolismo , Adhesión Celular , Proliferación Celular , Magnesio/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Canales Catiónicos TRPM/deficiencia , Trofoblastos/metabolismo , Animales , Muerte Celular , Linaje de la Célula , Células Cultivadas , Implantación del Embrión , Desarrollo Embrionario , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Ratones Noqueados , Células Madre Embrionarias de Ratones/patología , Transducción de Señal , Canales Catiónicos TRPM/genética , Trofoblastos/patología
5.
Cell Death Dis ; 12(7): 637, 2021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-34155199

RESUMEN

Embryonic stem cell (ESC) differentiation and somatic cell reprogramming are biological processes governed by antagonistic expression or repression of a largely common set of genes. Accurate regulation of gene expression is thus essential for both processes, and alterations in RNA processing are predicted to negatively affect both. We show that truncation of the DIDO gene alters RNA splicing and transcription termination in ESC and mouse embryo fibroblasts (MEF), which affects genes involved in both differentiation and reprogramming. We combined transcriptomic, protein interaction, and cellular studies to identify the underlying molecular mechanism. We found that DIDO3 interacts with the helicase DHX9, which is involved in R-loop processing and transcription termination, and that DIDO3-exon16 deletion increases nuclear R-loop content and causes DNA replication stress. Overall, these defects result in failure of ESC to differentiate and of MEF to be reprogrammed. MEF immortalization restored impaired reprogramming capacity. We conclude that DIDO3 has essential functions in ESC differentiation and somatic cell reprogramming by supporting accurate RNA metabolism, with its exon16-encoded domain playing the main role.


Asunto(s)
Diferenciación Celular , Técnicas de Reprogramación Celular , Reprogramación Celular , Proteínas de Unión al ADN/genética , Fibroblastos/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Mutación , Estructuras R-Loop , Empalme del ARN , Factores de Transcripción/genética , Animales , Células Cultivadas , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Fibroblastos/patología , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre Embrionarias de Ratones/patología , Fenotipo , Factores de Transcripción/metabolismo , Terminación de la Transcripción Genética
6.
Methods Mol Biol ; 2311: 39-49, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34033076

RESUMEN

This chapter describes the culture and propagation of murine embryonic stem cells, F9 and P19, and strategies for differentiation of these stem cells into neurons. Additional techniques are described for obtaining enriched populations of mature neurons from P19 cells and differentiation of F9 cells into serotonergic or catecholaminergic neurons. The protocols described herein can be used for dissection of the pathways such as gliogenesis and neurogenesis that are involved in differentiation of pluripotent stem cells such as F9 and P19 into glial cells or terminally differentiated neurons.


Asunto(s)
Células Madre Embrionarias de Ratones/patología , Células-Madre Neurales/patología , Neurogénesis , Neuronas/patología , Teratocarcinoma/patología , Animales , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Proliferación Celular , Ratones , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/metabolismo , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fenotipo , Teratocarcinoma/metabolismo , Tretinoina/farmacología
7.
Cardiovasc Toxicol ; 21(9): 695-709, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33983555

RESUMEN

Hyperglycaemia, a key metabolic abnormality in diabetes mellitus, is implicated in pathological cardiogenesis during embryological development. However, the underlying mechanisms and potential therapeutic targets remain unknown. We, therefore, studied the effect of hyperglycaemia on mouse embryonic stem cell (mESC) cardiac differentiation. The mESCs were differentiated via embryoid body (EB) formation and cultured under conditions with baseline (25 mM) or high (50 mM) glucose. Time-lapse microscopy images of pulsatile mESCs and Ca2+ transients were recorded. Biomarkers of cellular changes were detected using immunocytochemistry, terminal deoxynucleotidyl transferase dUTP nick-end labelling (TUNEL) assay, and Western blot analyses. Differentiated, spontaneously beating mESCs stained positive for cardiac troponin T, α-actinin 2, myosin heavy chain, and connexin 43. Hyperglycaemia decreased the EB diameter and number of beating EBs as well as the cellular amplitude of contraction, the Ca2+ transient, and the contractile response to caffeine (1 mM), but had no effect on the expression of the sarco-endoplasmic reticulum calcium transport ATPase 2 (SERCA 2). Furthermore, hyperglycaemia decreased the expression of B cell lymphoma 2 (Bcl-2) and increased the expression of cytoplasmic cytochrome c and the number of TUNEL-positive cells, but had no effect on the expression of one of the mitochondrial fusion regulatory proteins, optic atrophy protein 1 (OPA1). Overall, hyperglycaemia suppressed the mESC cardiomyocyte-like differentiation and induced contractile dysfunction. The results are consistent with mechanisms involving abnormal Ca2+ handling and mitochondrial-dependent apoptosis, factors which represent potential therapeutic targets in developmental diabetic cardiac disease.


Asunto(s)
Apoptosis/efectos de los fármacos , Glucemia/metabolismo , Diferenciación Celular/efectos de los fármacos , Glucosa/toxicidad , Hiperglucemia/sangre , Células Madre Embrionarias de Ratones/efectos de los fármacos , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Animales , Señalización del Calcio/efectos de los fármacos , Línea Celular , Hiperglucemia/patología , Hiperglucemia/fisiopatología , Ratones , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Embrionarias de Ratones/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología
8.
J Clin Invest ; 131(1)2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33393499

RESUMEN

LMNA mutations in patients are responsible for a dilated cardiomyopathy. Molecular mechanisms underlying the origin and development of the pathology are unknown. Herein, using mouse pluripotent embryonic stem cells (ESCs) and a mouse model both harboring the p.H222P Lmna mutation, we found early defects in cardiac differentiation of mutated ESCs and dilatation of mutated embryonic hearts at E13.5, pointing to a developmental origin of the disease. Using mouse ESCs, we demonstrated that cardiac differentiation of LmnaH222P/+ was impaired at the mesodermal stage. Expression of Mesp1, a mesodermal cardiogenic gene involved in epithelial-to-mesenchymal transition of epiblast cells, as well as Snai1 and Twist expression, was decreased in LmnaH222P/+ cells compared with WT cells in the course of differentiation. In turn, cardiomyocyte differentiation was impaired. ChIP assay of H3K4me1 in differentiating cells revealed a specific decrease of this histone mark on regulatory regions of Mesp1 and Twist in LmnaH222P/+ cells. Downregulation or inhibition of LSD1 that specifically demethylated H3K4me1 rescued the epigenetic landscape of mesodermal LmnaH222P/+ cells and in turn contraction of cardiomyocytes. Inhibition of LSD1 in pregnant mice or neonatal mice prevented cardiomyopathy in E13.5 LmnaH222P/H222P offspring and adults, respectively. Thus, LSD1 appeared to be a therapeutic target to prevent or cure dilated cardiomyopathy associated with a laminopathy.


Asunto(s)
Cardiomiopatías/enzimología , Cardiomiopatías/prevención & control , Histona Demetilasas/metabolismo , Laminopatías/complicaciones , Laminopatías/enzimología , Miocitos Cardíacos/enzimología , Sustitución de Aminoácidos , Animales , Cardiomiopatías/genética , Diferenciación Celular , Modelos Animales de Enfermedad , Histona Demetilasas/genética , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Laminopatías/genética , Ratones , Ratones Mutantes , Células Madre Embrionarias de Ratones/enzimología , Células Madre Embrionarias de Ratones/patología , Mutación Missense , Miocitos Cardíacos/patología
9.
FEBS J ; 288(5): 1533-1545, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32705746

RESUMEN

MicroRNAs (miRNAs) post-transcriptionally repress almost all genes in mammals and thereby form an additional layer of gene regulation. As such, miRNAs impact on nearly every physiological process and have also been associated with cancer. Prominent examples of such miRNAs can be found in the miR-15 family, composed of the bicistronic clusters miR-15a/16-1, miR-15b/16-2, and miR-497/195. In particular, the miR-15a/16-1 cluster is deleted in almost two thirds of all chronic B lymphocytic leukemia (CLL) cases, a phenotype that is also recapitulated by miR-15a/16-1-deficient as well as miR-15b/16-2-deficient mice. Under physiological conditions, those two clusters have been implicated in T-cell function, and B-cell and natural killer (NK) cell development; however, it is unclear whether miR-497 and miR-195 confer similar roles in health and disease. Here, we have generated a conditional mouse model for tissue-specific deletion of miR-497 and miR-195. While mice lacking miR-15a/16-1 in the hematopoietic compartment developed clear signs of CLL over time, aging mice deficient for miR-497/195 did not show such a phenotype. Likewise, loss of miR-15a/16-1 impaired NK and early B-cell development, whereas miR-497/195 was dispensable for these processes. In fact, a detailed analysis of miR-497/195-deficient mice did not reveal any effect on steady-state hematopoiesis or immune cell function. Unexpectedly, even whole-body deletion of the cluster was well-tolerated and had no obvious impact on embryonic development or healthy life span. Therefore, we postulate that the miR-497/195 cluster is redundant to its paralog clusters or that its functional relevance is restricted to certain physiological and pathological conditions.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/inmunología , Leucemia Linfocítica Crónica de Células B/genética , MicroARNs/genética , Animales , Animales Modificados Genéticamente , Linfocitos B/inmunología , Linfocitos B/patología , Médula Ósea/inmunología , Médula Ósea/patología , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Edición Génica/métodos , Homeostasis/genética , Homeostasis/inmunología , Humanos , Inmunofenotipificación , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Leucemia Linfocítica Crónica de Células B/inmunología , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Masculino , Ratones , MicroARNs/inmunología , Células Madre Embrionarias de Ratones/inmunología , Células Madre Embrionarias de Ratones/patología , Eliminación de Secuencia , Transducción de Señal , Análisis de la Célula Individual/métodos , Bazo/inmunología , Bazo/patología , Linfocitos T/inmunología , Linfocitos T/patología
10.
Arch Toxicol ; 95(1): 253-269, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32926198

RESUMEN

Pluripotent stem cells recapitulate in vitro the early developmental stages and are considered promising cell models for predictive developmental toxicity studies. To investigate the consistency between adverse drug effects on early development and the early stages of embryonic stem cell differentiation in three-dimensional (3D) in vitro culture, the toxic responses to 5-hydroxytryptophan (5-HTP; 0.5-2 mM) were evaluated in early mouse embryos and the embryoid body (EB) differentiation model. 3D architectures, developmental and differentiation dynamics and the cell death rates were analyzed in early mouse embryos (E2.5-E5.5) and EBs at 1 and 6 days of differentiation using a combination of confocal immunofluorescence microscopy with high content imaging analysis and quantitative gene expression analysis. Comparative analysis of toxic responses in early embryos and EBs revealed a similar dose- and stage-dependent decrease in the 5-HTP toxic effects during development and differentiation. The integral toxic responses in the early embryos and EBs were significantly dependent on their 3D architecture and cellular composition. Treatment with 5-HTP (1 mM and above) induced developmental arrest, growth inhibition, and increased cell death in the early embryos without the trophoblasts (E2.5) and those with impaired trophoblasts and in early EBs, whereas later embryos and EBs were more resistant due to the protection of the extraembryonic tissues. This study demonstrates that the EB differentiation model is a relevant 3D-model of early mammalian development and can be useful for the predictive evaluation of toxic and teratogenic effects in embryos at the preimplantation and early post-implantation developmental stages.


Asunto(s)
5-Hidroxitriptófano/toxicidad , Diferenciación Celular/efectos de los fármacos , Embrión de Mamíferos/efectos de los fármacos , Cuerpos Embrioides/efectos de los fármacos , Células Madre Embrionarias de Ratones/efectos de los fármacos , Animales , Muerte Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Técnicas de Cultivo de Embriones , Embrión de Mamíferos/patología , Cuerpos Embrioides/patología , Desarrollo Embrionario/efectos de los fármacos , Femenino , Edad Gestacional , Cinética , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Microscopía Fluorescente , Células Madre Embrionarias de Ratones/patología , Embarazo , Medición de Riesgo , Pruebas de Toxicidad
11.
FEBS J ; 288(10): 3317-3329, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33245852

RESUMEN

Differentiation of the embryonic stem cells (ESCs) is regulated by a variety of different signaling pathways. Genetic depletion of murine Pelota gene (Pelo) leads to early embryonic lethality. Here, we aimed at determining the embryonic stage and deciphering the dysregulated signaling pathways affected upon Pelo deletion. We found that development of PELO-null embryos is perturbed between the embryonic days E4.5 and E5.5, at which first differentiation process of ESCs takes place. Molecular analysis revealed enhanced activity of phosphoinositide 3-kinase-protein kinase B/ AKT (PI3K-PKB/AKT) signaling, but nuclear accumulation of forkhead box O1 (FOXO1), and upregulation of the pluripotency-related gene, Oct4, in mutant ESCs cultured under differentiation condition. Despite increased levels of nuclear ß-catenin in PELO-null ESCs as a result of decreased activity of glycogen synthase kinase-3ß, the activity of the canonical wingless (Wnt)/ß-catenin/T-cell factor (TCF) was significantly attenuated as judged by the promoter reporter assay, downregulated Wnt/ß-catenin target genes, and impaired cell proliferation. Interestingly, we demonstrated an increased binding of ß-catenin to FOXO1 in PELO-mutant ESCs cultured under differentiation condition that could explain, on one side, the nuclear accumulation of FOXO1 protein and hence persistent pluripotency of PELO-mutant ESCs, and on the other side, the dysregulated transcriptional activity of ß-catenin/TCF and therefore attenuated PELO-null ESC self-renewal. Taken together, our results strongly suggest that PELO deletion averts ESC differentiation through promoting FOXO1/ß-catenin binding with subsequent dysregulation of FOXO1 and canonical ß-catenin/TCF signaling pathways.


Asunto(s)
Proteínas de Ciclo Celular/genética , Endonucleasas/genética , Proteína Forkhead Box O1/genética , Regulación del Desarrollo de la Expresión Génica , Genes Letales , Células Madre Embrionarias de Ratones/metabolismo , beta Catenina/genética , Animales , Proteínas de Ciclo Celular/deficiencia , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Medios de Cultivo/farmacología , Embrión de Mamíferos , Endonucleasas/deficiencia , Proteína Forkhead Box O1/metabolismo , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Ratones , Ratones Noqueados , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/patología , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Cultivo Primario de Células , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Factores de Transcripción TCF/genética , Factores de Transcripción TCF/metabolismo , beta Catenina/metabolismo
12.
FASEB J ; 34(11): 15062-15079, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32951265

RESUMEN

The transcription factor TBX1 is the major gene implicated in 22q11.2 deletion syndrome (22q11.2DS). The complex clinical phenotype includes vascular anomalies and a recent report presented new cases of primary lymphedema in 22q11.2DS patients. We have previously shown that TBX1 is required for systemic lymphatic vessel development in prenatal mice and it is critical for their survival postnatally. Using loss-of-function genetics and transgenesis in the mouse, we show here a strong genetic interaction between Tbx1 and Vegfr3 in cardiac lymphangiogenesis. Intriguingly, we found that different aspects of the cardiac lymphatic phenotype in Tbx1-Vegfr3 compound heterozygotes were regulated independently by the two genes, with Tbx1 primarily regulating vessel numbers and Vegfr3 vessel morphology. Consistent with this observation, Tbx1Cre -activated expression of a Vegfr3 transgene rescued partially the cardiac lymphatic abnormalities in compound heterozygotes. Through time-controlled genetic experiments, we show that Tbx1 is activated and required in cardiac lymphatic endothelial cell (LEC) progenitors between E10.5 and E11.5. Furthermore, we found that it is also required later in development for the growth of the cardiac lymphatics. Finally, our study revealed a differential sensitivity between ventral and dorsal cardiac lymphatics to the effects of altered Tbx1 and Vegfr3 gene dosage, and we show that this likely results from an earlier requirement for Tbx1 in ventral cardiac LEC progenitors.


Asunto(s)
Corazón/fisiopatología , Linfangiogénesis , Vasos Linfáticos/patología , Células Madre Embrionarias de Ratones/patología , Proteínas de Dominio T Box/fisiología , Receptor 3 de Factores de Crecimiento Endotelial Vascular/fisiología , Animales , Femenino , Heterocigoto , Vasos Linfáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Madre Embrionarias de Ratones/metabolismo
13.
J Physiol Pharmacol ; 71(2)2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32633240

RESUMEN

The embryoid body test (EBT) is a developmental toxicity test method that measures the size of embryoid bodies (EBs) and the viability of mouse embryonic stem cells (mESCs) and fibroblasts (3T3 cells). The previous pre-validation study confirmed the high accuracy (above 80%) of EBT using 26 coded test chemicals. This second-phase validation study assessed the inter-laboratory reproducibility (5 chemicals in common) and predictive capacity (10 chemicals in each laboratory) test using the coded test chemicals at three laboratories. For the prediction model, the accuracy is increased when more data is accumulated. Therefore, we updated the prediction model and analyzed the results of the second year with the newly created-prediction model. Statistical analysis of the inter-laboratory reproducibility test results indicated that accuracy, sensitivity, and specificity were 87%, 78%, and 100%, respectively. The results of the statistical analysis of the predictive capacity test showed an accuracy of 80%, sensitivity of 78%, and specificity of 81%. In conclusion, the EBT can accurately classify various embryotoxicants within a short period and with relatively little effort. Therefore, EBT can be used as a good way to test developmental toxicity.


Asunto(s)
Alternativas a las Pruebas en Animales/métodos , Cuerpos Embrioides/patología , Células Madre Embrionarias de Ratones/patología , Pruebas de Toxicidad/métodos , Alternativas a las Pruebas en Animales/normas , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cuerpos Embrioides/efectos de los fármacos , Ratones , Células Madre Embrionarias de Ratones/efectos de los fármacos , Variaciones Dependientes del Observador , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados
14.
J Appl Toxicol ; 40(3): 330-341, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31808176

RESUMEN

In vitro assays presently used for prenatal developmental toxicity (PDT) testing only assess the embryotoxic potential of parent substances and not that of potentially embryotoxic metabolites. Here we combined a biotransformation system, using hamster liver microsomes, with the ES-D3 cell differentiation assay of the embryonic stem cell test (EST) to compare the in vitro PDT potency of two 5-ring polycyclic aromatic hydrocarbons (PAHs), benzo[a]pyrene (BaP) and dibenz[a,h]anthracene (DBA), and dimethyl sulfoxide extracts from five PAH-containing petroleum substances (PS) and a gas-to-liquid base oil (GTLb), with and without bioactivation. In the absence of bioactivation, DBA, but not BaP, inhibited the differentiation of ES-D3 cells into beating cardiomyocytes in a concentration-dependent manner. Upon bioactivation, BaP induced in vitro PDT, while its major metabolite 3-hydroxybenzo[a]pyrene was shown to be active in the EST as well. This means BaP needs biotransformation to exert its embryotoxic effects. GTLb extracts tested negative in the EST, with and without bioactivation. The PS-induced PDT in the EST was not substantially changed following bioactivation, implying that metabolism may not play a crucial role for the PS extracts under study to exert the in vitro PDT effects. Altogether, these results indicate that although some PAH require bioactivation to induce PDT, some do not and this latter appears to hold for the (majority of) the PS constituents responsible for the in vitro PDT of these complex substances.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Células Madre Embrionarias de Ratones/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Petróleo/toxicidad , Hidrocarburos Policíclicos Aromáticos/toxicidad , Activación Metabólica , Animales , Benzo(a)Antracenos/toxicidad , Benzo(a)pireno/toxicidad , Línea Celular , Relación Dosis-Respuesta a Droga , Masculino , Mesocricetus , Ratones , Células Madre Embrionarias de Ratones/patología , Miocitos Cardíacos/patología , Petróleo/metabolismo , Hidrocarburos Policíclicos Aromáticos/metabolismo , Medición de Riesgo , Pruebas de Toxicidad
15.
Nat Cell Biol ; 21(11): 1449-1461, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31659274

RESUMEN

Development and differentiation are associated with profound changes to histone modifications, yet their in vivo function remains incompletely understood. Here, we generated mouse models expressing inducible histone H3 lysine-to-methionine (K-to-M) mutants, which globally inhibit methylation at specific sites. Mice expressing H3K36M developed severe anaemia with arrested erythropoiesis, a marked haematopoietic stem cell defect, and rapid lethality. By contrast, mice expressing H3K9M survived up to a year and showed expansion of multipotent progenitors, aberrant lymphopoiesis and thrombocytosis. Additionally, some H3K9M mice succumbed to aggressive T cell leukaemia/lymphoma, while H3K36M mice exhibited differentiation defects in testis and intestine. Mechanistically, induction of either mutant reduced corresponding histone trimethylation patterns genome-wide and altered chromatin accessibility as well as gene expression landscapes. Strikingly, discontinuation of transgene expression largely restored differentiation programmes. Our work shows that individual chromatin modifications are required at several specific stages of differentiation and introduces powerful tools to interrogate their roles in vivo.


Asunto(s)
Epigénesis Genética , Histonas/metabolismo , Leucemia de Células T/genética , Lisina/metabolismo , Metionina/metabolismo , Teratoma/genética , Animales , Trasplante de Médula Ósea , Linaje de la Célula/genética , Modelos Animales de Enfermedad , Doxiciclina/farmacología , Células Eritroides/metabolismo , Células Eritroides/patología , Femenino , Granulocitos/metabolismo , Granulocitos/patología , Histonas/genética , Leucemia de Células T/inducido químicamente , Leucemia de Células T/metabolismo , Leucemia de Células T/patología , Masculino , Metilación , Ratones , Ratones Transgénicos , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Embrionarias de Ratones/patología , Mutación , Transducción de Señal , Análisis de Supervivencia , Linfocitos T/metabolismo , Linfocitos T/patología , Teratoma/inducido químicamente , Teratoma/metabolismo , Teratoma/patología
16.
EMBO Mol Med ; 11(8): e9266, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31267692

RESUMEN

Angiogenesis is a hallmark of cancer, promoting growth and metastasis. Anti-angiogenic treatment has limited efficacy due to therapy-induced blood vessel alterations, often followed by local hypoxia, tumor adaptation, progression, and metastasis. It is therefore paramount to overcome therapy-induced resistance. We show that Apelin inhibition potently remodels the tumor microenvironment, reducing angiogenesis, and effectively blunting tumor growth. Functionally, targeting Apelin improves vessel function and reduces polymorphonuclear myeloid-derived suppressor cell infiltration. Importantly, in mammary and lung cancer, Apelin prevents resistance to anti-angiogenic receptor tyrosine kinase (RTK) inhibitor therapy, reducing growth and angiogenesis in lung and breast cancer models without increased hypoxia in the tumor microenvironment. Apelin blockage also prevents RTK inhibitor-induced metastases, and high Apelin levels correlate with poor prognosis of anti-angiogenic therapy patients. These data identify a druggable anti-angiogenic drug target that reduces tumor blood vessel densities and normalizes the tumor vasculature to decrease metastases.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Receptores de Apelina/metabolismo , Apelina/metabolismo , Movimiento Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neovascularización Patológica , Inhibidores de Proteínas Quinasas/farmacología , Sunitinib/farmacología , Animales , Apelina/antagonistas & inhibidores , Apelina/deficiencia , Apelina/genética , Receptores de Apelina/antagonistas & inhibidores , Receptores de Apelina/deficiencia , Receptores de Apelina/genética , Línea Celular Tumoral , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Embrionarias de Ratones/patología , Metástasis de la Neoplasia , Transducción de Señal , Carga Tumoral/efectos de los fármacos , Microambiente Tumoral
17.
Stem Cell Reports ; 12(6): 1366-1379, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31155506

RESUMEN

Zygotic genome activation (ZGA) begins after fertilization and is essential for establishing pluripotency and genome stability. However, it is unclear how ZGA genes prevent mitotic errors. Here we show that knockout of the ZGA gene Zscan5b, which encodes a SCAN domain with C2H2 zinc fingers, causes a high incidence of chromosomal abnormalities in embryonic stem cells (ESCs), and leads to the development of early-stage cancers. After irradiation, Zscan5b-deficient ESCs displayed significantly increased levels of γ-H2AX despite increased expression of the DNA repair genes Rad51l3 and Bard. Re-expression of Zscan5b reduced γ-H2AX content, implying a role for Zscan5b in DNA damage repair processes. A co-immunoprecipitation analysis showed that Zscan5b bound to the linker histone H1, suggesting that Zscan5b may protect chromosomal architecture. Our report demonstrates that the ZGA gene Zscan5b is involved in genomic integrity and acts to promote DNA damage repair and regulate chromatin dynamics during mitosis.


Asunto(s)
Aberraciones Cromosómicas , Cromosomas de los Mamíferos , Daño del ADN , Factores de Transcripción de Tipo Kruppel/deficiencia , Mitosis , Células Madre Embrionarias de Ratones/metabolismo , Animales , Cromosomas de los Mamíferos/genética , Cromosomas de los Mamíferos/metabolismo , Femenino , Histonas/genética , Histonas/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Mutantes , Células Madre Embrionarias de Ratones/patología
18.
J Cell Biochem ; 120(10): 18066-18076, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31148273

RESUMEN

Embryonic developmental stages and regulations have always been one of the most intriguing aspects of science. Since the cancer stem cell discovery, striking for cancer development and recurrence, embryonic stem cells and control mechanisms, as well as cancer cells and cancer stem cell control mechanisms become important research materials. It is necessary to reveal the similarities and differences between somatic and cancer cells which are formed of embryonic stem cells divisions and determinations. For this purpose, mouse embryonic stem cells (mESCs), mouse skin fibroblast cells (MSFs) and mouse lung squamous cancer cells (SqLCCs) were grown in vitro and the differences between these three cell lines signalling regulations of mechanistic target of rapamycin (mTOR) and autophagic pathways were demonstrated by immunofluorescence and real-time polymerase chain reaction. Expressional differences were clearly shown between embryonic, cancer and somatic cells that mESCs displayed higher expressional level of Atg10, Hdac1 and Cln3 which are related with autophagic regulation and Hsp4, Prkca, Rhoa and ribosomal S6 genes related with mTOR activity. LC3 and mTOR protein levels were lower in mESCs than MSFs. Thus, the mechanisms of embryonic stem cell regulation results in the formation of somatic tissues whereas that these cells may be the causative agents of cancer in any deterioration.


Asunto(s)
Autofagia , Fibroblastos/patología , Neoplasias Pulmonares/patología , Células Madre Embrionarias de Ratones/patología , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Animales , Autofagia/genética , Línea Celular , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Neoplasias Pulmonares/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Células Madre Embrionarias de Ratones/metabolismo
19.
Stem Cell Reports ; 12(6): 1329-1341, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31080111

RESUMEN

Oculomotor neurons, which regulate eye movement, are resilient to degeneration in the lethal motor neuron disease amyotrophic lateral sclerosis (ALS). It would be highly advantageous if motor neuron resilience could be modeled in vitro. Toward this goal, we generated a high proportion of oculomotor neurons from mouse embryonic stem cells through temporal overexpression of PHOX2A in neuronal progenitors. We demonstrate, using electrophysiology, immunocytochemistry, and RNA sequencing, that in vitro-generated neurons are bona fide oculomotor neurons based on their cellular properties and similarity to their in vivo counterpart in rodent and man. We also show that in vitro-generated oculomotor neurons display a robust activation of survival-promoting Akt signaling and are more resilient to the ALS-like toxicity of kainic acid than spinal motor neurons. Thus, we can generate bona fide oculomotor neurons in vitro that display a resilience similar to that seen in vivo.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Diferenciación Celular , Neuronas Motoras/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Transducción de Señal , Esclerosis Amiotrófica Lateral/patología , Animales , Supervivencia Celular , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Neuronas Motoras/patología , Células Madre Embrionarias de Ratones/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo
20.
Stem Cell Reports ; 12(6): 1313-1328, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31080114

RESUMEN

Neurogenesis in specific brain regions in adult mammals decreases with age. Progressive reduction in the proliferation of neural stem and progenitor cells (NS/PCs) is a primary cause of this age-associated decline. However, the mechanism responsible for this reduction is poorly understood. We identify p38 MAPK as a key factor in the proliferation of neural progenitor cells (NPCs) in adult neurogenic niches. p38 expression in adult NS/PCs is downregulated during aging. Deletion of p38α in NS/PCs specifically reduces the proliferation of NPCs but not stem cells. Conversely, forced expression of p38α in NS/PCs in the aged mouse subventricular zone (SVZ) restores NPC proliferation and neurogenesis, and prevents age-dependent SVZ atrophy. We also found that p38 is necessary for suppressing the expression of Wnt antagonists DKK1 and SFRP3, which inhibit the proliferation of NPCs. Age-related reduction in p38 thus leads to decreased adult neurogenesis via downregulation of Wnt signaling.


Asunto(s)
Envejecimiento/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis , Vía de Señalización Wnt , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Envejecimiento/patología , Animales , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Ratones Transgénicos , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Células Madre Embrionarias de Ratones/patología , Células-Madre Neurales/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...