Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Am J Surg Pathol ; 48(9): 1164-1176, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38963187

RESUMEN

We investigated the frequency and associated pathology of embryoid bodies in ovarian tumors by evaluating neoplasms in which they are known to occur: 100 immature teratomas, 125 malignant mixed germ cell tumors, and 6 polyembryomas. Three immature teratomas contained a single relatively well-formed embryoid body, whereas these and 11 others showed foci we categorized as embryoid body remnants consisting of microscopic aggregates of embryonal or yolk sac-type epithelium associated with spaces consistent with yolk sac or amniotic cavity but lacking a classic embryoid body structure. Teratomas with these foci were all high grade. A well-formed embryoid body was found in only 1 malignant mixed tumor, but embryoid body remnants were present in 25%, invariably associated with foci of immature teratoma (100%) and often with yolk sac tumor (97%), embryonal carcinoma (35%), or both (32%). These foci usually took the form of round to oval aggregates, often well-circumscribed, for which the term "polyembryoma background" has been proposed. The polyembryomas were typically grossly hemorrhagic and occurred in patients from 9 to 43 years of age. The embryoid bodies in them generally grew in lobules within an edematous to occasionally myxoid stroma. Four tumors contained liver-like cells, 4 numerous glands likely recapitulating the allantois, 3 syncytiotrophoblast cells, 2 prominent cysts, and 2 striking vascular proliferations. This study indicates that (1) typical embryoid bodies are rare in immature teratomas but about 14% of them have embryoid body remnants. (2) Embryoid body remnants are seen in 25% of malignant mixed germ cell tumors with a teratomatous component and often proliferate to form yolk sac tumor and embryonal carcinoma. (3) Well-formed embryoid bodies growing in a confluent manner (polyembryoma) are rare, and minor foci of teratoma, yolk sac tumor, or embryonal carcinoma are almost always present, indicating that these are fundamentally malignant mixed germ cell tumors but the polyembryoma component is dominant and distinctive which, in our opinion, justifies its own nomenclature. (4) Embryoid bodies are not a feature of other germ cell tumors.


Asunto(s)
Proliferación Celular , Cuerpos Embrioides , Neoplasias de Células Germinales y Embrionarias , Neoplasias Ováricas , Teratoma , Neoplasias Ováricas/patología , Femenino , Humanos , Neoplasias de Células Germinales y Embrionarias/patología , Cuerpos Embrioides/patología , Adolescente , Adulto , Niño , Teratoma/patología , Adulto Joven , Persona de Mediana Edad , Tumor del Seno Endodérmico/patología , Anciano
2.
Int J Gynecol Pathol ; 42(4): 426-431, 2023 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-36044302

RESUMEN

Ovarian malignant mixed germ cell tumors are rare tumors occurring in young women. The presence of prominent embryoid bodies in these tumors is extremely uncommon. Herein, we report such a case, with a histomorphologic description and immunohistochemical and fluorescence in situ hybridization analyses.


Asunto(s)
Neoplasias de Células Germinales y Embrionarias , Neoplasias Ováricas , Humanos , Femenino , Hibridación Fluorescente in Situ , Cuerpos Embrioides/patología , Neoplasias de Células Germinales y Embrionarias/diagnóstico , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología
3.
Cancer Genomics Proteomics ; 19(2): 178-193, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35181587

RESUMEN

BACKGROUND: Testicular germ cell tumours (TGCTs) are the most frequent tumour type among young, adult men. TGCTs can be efficiently treated, but metastases of the teratoma subtype, for which there are no circulating biomarkers, represent a challenge. MATERIALS AND METHODS: Global microRNA expression in teratoma tissue and embryoid bodies was assessed using next-generation sequencing. Levels of microRNAs identified as potential biomarkers were obtained from serum of patients with teratoma and matched healthy men. RESULTS: We identified miR-222-5p, miR-200a-5p, miR-196b-3p and miR-454-5p as biomarker candidates from the tumour tissue and embryoid body screening but the expression of these microRNAs was very low in serum and not statistically different between patients and controls. miR-375-3p was highly expressed, being highest in patients with teratoma (p=0.012) but the levels of expression in serum from these patients and healthy controls overlapped. miR-371a-3p was not expressed in serum from patients with pure teratoma, only in patients with mixed tumours. CONCLUSION: The microRNA profiles of the teratoma subtype of TGCT and embryoid bodies were obtained and assessed for candidate circulating biomarkers, but none with high sensitivity and specificity for teratoma were identified in our study. We conclude that neither the proposed teratoma marker miR-375-3p nor miR-371a-3p are suitable as circulating teratoma markers.


Asunto(s)
MicroARNs , Neoplasias de Células Germinales y Embrionarias , Teratoma , Neoplasias Testiculares , Adulto , Biomarcadores de Tumor/genética , Cuerpos Embrioides/metabolismo , Cuerpos Embrioides/patología , Humanos , Masculino , MicroARNs/metabolismo , Neoplasias de Células Germinales y Embrionarias/diagnóstico , Neoplasias de Células Germinales y Embrionarias/genética , Teratoma/genética , Neoplasias Testiculares/genética , Neoplasias Testiculares/patología
4.
Toxicol Appl Pharmacol ; 433: 115792, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34742744

RESUMEN

Concurrent with the '3R' principle, the embryonic stem cell test (EST) using mouse embryonic stem cells, developed in 2000, remains the solely accepted in vitro method for embryotoxicity testing. However, the scope and implementation of EST for embryotoxicity screening, compliant with regulatory requirements, are limited. This is due to its technical complexity, long testing period, labor-intensive methodology, and limited endpoint data, leading to misclassification of embryotoxic potential. In this study, we used human induced pluripotent stem cell (hiPSC)-derived embryoid bodies (EB) as an in vitro model to investigate the embryotoxic effects of a carefully selected set of pharmacological compounds. Morphology, viability, and differentiation potential were investigated after exposing EBs to folic acid, all-trans-retinoic acid, dexamethasone, and valproic acid for 15 days. The results showed that the compounds differentially repressed cell growth, compromised morphology, and triggered apoptosis in the EBs. Further, transcriptomics was employed to compare subtle temporal changes between treated and untreated cultures. Gene ontology and pathway analysis revealed that dysregulation of a large number of genes strongly correlated with impaired neuroectoderm and cardiac mesoderm formation. This aberrant gene expression pattern was associated with several disorders of the brain like mental retardation, multiple sclerosis, stroke and of the heart like dilated cardiomyopathy, ventricular tachycardia, and ventricular arrhythmia. Lastly, these in vitro findings were validated using in ovo chick embryo model. Taken together, pharmacological compound or drug-induced defective EB development from hiPSCs could potentially be used as a suitable in vitro platform for embryotoxicity screening.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Cuerpos Embrioides/efectos de los fármacos , Perfilación de la Expresión Génica , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Teratógenos/toxicidad , Pruebas de Toxicidad , Transcriptoma/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Linaje de la Célula , Embrión de Pollo , Dexametasona/toxicidad , Relación Dosis-Respuesta a Droga , Cuerpos Embrioides/metabolismo , Cuerpos Embrioides/patología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Concentración 50 Inhibidora , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Neurogénesis/efectos de los fármacos , Medición de Riesgo , Tretinoina/toxicidad , Ácido Valproico/toxicidad
5.
APMIS ; 129 Suppl 142: 1-30, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34399444

RESUMEN

Over the past decade, 3D culture models of human and animal cells have found their way into tissue differentiation, drug development, personalized medicine and tumour behaviour studies. Embryoid bodies (EBs) are in vitro 3D cultures established from murine pluripotential stem cells, whereas tumoroids are patient-derived in vitro 3D cultures. This thesis aims to describe a new implication of an embryoid body model and to characterize the patient-specific microenvironment of the parental tumour in relation to tumoroid growth rate. In this thesis, we described a high-throughput monitoring method, where EBs are used as a dynamic angiogenesis model. In this model, digital image analysis (DIA) is implemented on immunohistochemistry (IHC) stained sections of the cultures over time. Furthermore, we have investigated the correlation between the genetic profile and inflammatory microenvironment of parental tumours on the in vitro growth rate of tumoroids. The EBs were cultured in spinner flasks. The samples were collected at days 4, 6, 9, 14, 18 and 21, dehydrated and embedded in paraffin. The histological sections were IHC stained for the endothelial marker CD31 and digitally scanned. The virtual whole-image slides were digitally analysed by Visiopharm® software. Histological evaluation showed vascular-like structures over time. The quantitative DIA was plausible to monitor significant increase in the total area of the EBs and an increase in endothelial differentiation. The tumoroids were established from 32 colorectal adenocarcinomas. The in vitro growth rate of the tumoroids was followed by automated microscopy over an 11-day period. The parental tumours were analysed by next-generation sequencing for KRAS, TP53, PIK3CA, SMAD4, MAP2K1, BRAF, FGFR3 and FBXW7 status. The tumoroids established from KRAS-mutated parental tumours showed a significantly higher growth rate compared to their wild-type counterparts. The density of CD3+ T lymphocytes and CD68+ macrophages was calculated in the centre of the tumours and at the invasive margin of the tumours. The high density of CD3+ cells and the low density of CD68+ cells showed a significant correlation with a higher growth rate of the tumoroids. In conclusion, a novel approach for histological monitoring of endothelial differentiation is presented in the stem cell-derived EBs. Furthermore, the KRAS status and density of CD3+ T cells and macrophages in the parental tumour influence the growth rate of the tumoroids. Our results indicate that these parameters should be included when tumoroids are to be implemented in personalized medicine.


Asunto(s)
Adenocarcinoma/patología , Técnicas de Cultivo de Célula/métodos , Neoplasias Colorrectales/patología , Animales , Diferenciación Celular/fisiología , Cuerpos Embrioides/patología , Células Endoteliales/patología , Humanos , Macrófagos/patología , Ratones , Células Madre/patología , Linfocitos T/patología , Microambiente Tumoral/fisiología
6.
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
7.
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
8.
Stem Cell Res ; 41: 101604, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31678774

RESUMEN

Mucopolysaccharidosis type I-Hurler (MPS1-H) is the most severe form of inherited metabolic diseases caused by mutations in the IDUA gene. The resulting deficiency of alpha L-iduronidase enzyme leads to a progressive accumulation of glycosaminoglycans in lysosomes which damages multiple organs and highly reduces life expectancy of affected children. Skin fibroblasts of a 2-year-old MPS1-H male, carrying two mutations in each IDUA alleles (H358_T364del; W402X), were reprogrammed into induced pluripotent stem cells (iPSCs) using the CytoTune-iPS Sendai Reprogramming method applying Yamanaka-factors (OCT4, SOX2, KLF4, c-MYC). iPSCs expressed pluripotency transcription factors while iPSC-derived embryoid bodies reveal markers of the three germ layers.


Asunto(s)
Diferenciación Celular , Fibroblastos/patología , Iduronidasa/genética , Células Madre Pluripotentes Inducidas/patología , Mucopolisacaridosis I/genética , Mucopolisacaridosis I/patología , Mutación , Células Cultivadas , Reprogramación Celular , Preescolar , Cuerpos Embrioides/metabolismo , Cuerpos Embrioides/patología , Fibroblastos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Masculino
10.
Epigenomics ; 11(7): 751-766, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31172793

RESUMEN

Aim: To investigate the function of Kdm2a in embryonic stem cells (ESCs). Materials & methods: Expression profile analysis after Kdm2a knockout. Analysis of Kdm2a, H3K4me3 and H3K27me3 ChIP-seq data in ESCs. qPCR analysis and ChIP-qPCR analysis of epigenetic changes after Kdm2a loss. Results:Kdm2a was dispensable for pluripotency maintenance in ESCs. Kdm2a genomic binding profile was positively correlated with that of H3K4me3, Zfx and Tet1. Kdm2a directly regulated germ cell genes in primordial germ cell-like cells. Kdm2a loss led to the reduced expression of endogenous retrovirus IAPEy and resulted in the gain of H3K36me2 and loss of H3K4me3 on IAPEy. Conclusion: Kdm2a regulates germ cell genes and endogenous retroviruses in ESCs possibly through demethylating H3K36me2 and influencing H3K4me3 deposition.


Asunto(s)
Retrovirus Endógenos/fisiología , Histona Demetilasas con Dominio de Jumonji/metabolismo , Animales , Sitios de Unión , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Cuerpos Embrioides/patología , Regulación de la Expresión Génica , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/deficiencia , Histona Demetilasas con Dominio de Jumonji/genética , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Embrionarias de Ratones/virología , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Guía de Kinetoplastida/metabolismo
11.
Cell Death Dis ; 9(11): 1100, 2018 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-30368521

RESUMEN

Spinal Muscular Atrophy (SMA) is caused by genetic mutations in the SMN1 gene, resulting in drastically reduced levels of Survival of Motor Neuron (SMN) protein. Although SMN is ubiquitously expressed, spinal motor neurons are one of the most affected cell types. Previous studies have identified pathways uniquely activated in SMA motor neurons, including a hyperactivated ER stress pathway, neuronal hyperexcitability, and defective spliceosomes. To investigate why motor neurons are more affected than other neural types, we developed a spinal organoid model of SMA. We demonstrate overt motor neuron degeneration in SMA spinal organoids, and this degeneration can be prevented using a small molecule inhibitor of CDK4/6, indicating that spinal organoids are an ideal platform for therapeutic discovery.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/genética , Neuronas Motoras/efectos de los fármacos , Organoides/efectos de los fármacos , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Proteína Quinasa CDC2/antagonistas & inhibidores , Proteína Quinasa CDC2/genética , Proteína Quinasa CDC2/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Diferenciación Celular , Línea Celular , Colágeno/química , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/metabolismo , Combinación de Medicamentos , Cuerpos Embrioides/efectos de los fármacos , Cuerpos Embrioides/metabolismo , Cuerpos Embrioides/patología , Regulación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Laminina/química , Modelos Biológicos , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Atrofia Muscular Espinal/tratamiento farmacológico , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/patología , Organoides/metabolismo , Organoides/patología , Cultivo Primario de Células , Proteoglicanos/química , Transducción de Señal , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo
12.
Mater Sci Eng C Mater Biol Appl ; 91: 303-310, 2018 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-30033259

RESUMEN

Conducting polymers (CP), namely polyaniline (PANI) and polypyrrole (PPy), are promising materials applicable for the use as biointerfaces as they intrinsically combine electronic and ionic conductivity. Although a number of works have employed PANI or PPy in the preparation of copolymers, composites, and blends with other polymers, there is no systematic study dealing with the comparison of their fundamental biological properties. The present study, therefore, compares the biocompatibility of PANI and PPy in terms of cytotoxicity (using NIH/3T3 fibroblasts and embryonic stem cells) and embryotoxicity (their impact on erythropoiesis and cardiomyogenesis within embryonic bodies). The novelty of the study lies not only in the fact that embryotoxicity is presented for the first time for both studied polymers, but also in the elimination of inter-laboratory variations within the testing, such variation making the comparison of previously published works difficult. The results clearly show that there is a bigger difference between the biocompatibility of the respective polymers in their salt and base forms than between PANI and PPy as such. PANI and PPy can, therefore, be similarly applied in biomedicine when solely their biological properties are considered. Impurity content detected by mass spectroscopy is presented. These results can change the generally accepted opinion of the scientific community on better biocompatibility of PPy in comparison with PANI.


Asunto(s)
Compuestos de Anilina , Cuerpos Embrioides/metabolismo , Eritropoyesis/efectos de los fármacos , Ensayo de Materiales , Células Madre Embrionarias de Ratones/metabolismo , Polímeros , Pirroles , Compuestos de Anilina/efectos adversos , Compuestos de Anilina/farmacología , Animales , Cuerpos Embrioides/patología , Ratones , Células Madre Embrionarias de Ratones/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Células 3T3 NIH , Polímeros/efectos adversos , Polímeros/farmacología , Pirroles/efectos adversos , Pirroles/farmacología
13.
Stem Cells Dev ; 27(12): 838-847, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29630478

RESUMEN

Embryonic developmental studies under microgravity conditions in space are very limited. To study the effects of short-term altered gravity on embryonic development processes, we exposed mouse embryonic stem cells (mESCs) to phases of hypergravity and microgravity and studied the differentiation potential of the cells using wide-genome microarray analysis. During the 64th European Space Agency's parabolic flight campaign, mESCs were exposed to 31 parabolas. Each parabola comprised phases lasting 22 s of hypergravity, microgravity, and a repeat of hypergravity. On different parabolas, RNA was isolated for microarray analysis. After exposure to 31 parabolas, mESCs (P31 mESCs) were further differentiated under normal gravity (1 g) conditions for 12 days, producing P31 12-day embryoid bodies (EBs). After analysis of the microarrays, the differentially expressed genes were analyzed using different bioinformatic tools to identify developmental and nondevelopmental biological processes affected by conditions on the parabolic flight experiment. Our results demonstrated that several genes belonging to GOs associated with cell cycle and proliferation were downregulated in undifferentiated mESCs exposed to gravity changes. However, several genes belonging to developmental processes, such as vasculature development, kidney development, skin development, and to the TGF-ß signaling pathway, were upregulated. Interestingly, similar enriched and suppressed GOs were obtained in P31 12-day EBs compared with ground control 12-day EBs. Our results show that undifferentiated mESCs exposed to alternate hypergravity and microgravity phases expressed several genes associated with developmental/differentiation and cell cycle processes, suggesting a transition from the undifferentiated pluripotent to a more differentiated stage of mESCs.


Asunto(s)
Ciclo Celular , Diferenciación Celular , Hipergravedad/efectos adversos , Células Madre Embrionarias de Ratones/metabolismo , Transducción de Señal , Ingravidez/efectos adversos , Animales , Línea Celular , Cuerpos Embrioides/metabolismo , Cuerpos Embrioides/patología , Ratones
14.
Cell Biochem Funct ; 35(7): 392-400, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28895148

RESUMEN

Induced pluripotent stem (iPS) cells are important for clinical application and stem cell research. Although human melanoma-associated antigen A2 (hMAGEA2) expression is known to affect differentiation in embryonic stem cells, its specific role in iPS cells remains unclear. To evaluate the function of hMAGEA2 and its characteristics in iPS cells, we produced hMAGEA2-overexpressing iPS cells from hMAGEA2-overexpressing transgenic mice. Although the iPS cells with overexpressed hMAGEA2 did not differ in morphology, their pluripotency, and self-renewal related genes (Nanog, Oct3/4, Sox2, and Stat3), expression level was significantly upregulated. Moreover, hMAGEA2 contributed to the promotion of cell cycle progression, thereby accelerating cell proliferation. Through embryoid body formation in vitro and teratoma formation in vivo, we demonstrated that hMAGEA2 critically decreases the differentiation ability of iPS cells. These data indicate that hMAGEA2 intensifies the self-renewal, pluripotency, and degree of proliferation of iPS cells, while significantly repressing their differentiation efficiency. Therefore, our findings prove that hMAGEA2 plays key roles in iPS cells.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Células Madre Pluripotentes Inducidas/metabolismo , Antígenos Específicos del Melanoma/metabolismo , Proteínas de Neoplasias/metabolismo , Animales , Puntos de Control del Ciclo Celular , Células Cultivadas , Cuerpos Embrioides/metabolismo , Cuerpos Embrioides/patología , Fibroblastos/citología , Fibroblastos/metabolismo , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Genotipo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/trasplante , Masculino , Antígenos Específicos del Melanoma/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Proteínas de Neoplasias/genética , Retroviridae/genética , Teratoma/metabolismo , Teratoma/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
J Vis Exp ; (125)2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28745632

RESUMEN

In this study, we used peripheral blood cells (PBCs) as seed cells to produce chondrocytes via induced pluripotent stem cells (iPSCs) in an integration-free method. Following embryoid body (EB) formation and fibroblastic cell expansion, the iPSCs are induced for chondrogenic differentiation for 21 days under serum-free and xeno-free conditions. After chondrocyte induction, the phenotypes of the cells are evaluated by morphological, immunohistochemical, and biochemical analyses, as well as by the quantitative real-time PCR examination of chondrogenic differentiation markers. The chondrogenic pellets show positive alcian blue and toluidine blue staining. The immunohistochemistry of collagen II and X staining is also positive. The sulfated glycosaminoglycan (sGAG) content and the chondrogenic differentiation markers COLLAGEN 2 (COL2), COLLAGEN 10 (COL10), SOX9, and AGGRECAN are significantly upregulated in chondrogenic pellets compared to hiPSCs and fibroblastic cells. These results suggest that PBCs can be used as seed cells to generate iPSCs for cartilage repair, which is patient-specific and cost-effective.


Asunto(s)
Células Sanguíneas/citología , Condrocitos/citología , Células Madre Pluripotentes Inducidas/citología , Agrecanos/genética , Agrecanos/metabolismo , Diferenciación Celular , Células Cultivadas , Condrocitos/metabolismo , Condrogénesis , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Cuerpos Embrioides/citología , Cuerpos Embrioides/patología , Humanos , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Regulación hacia Arriba , Grabación en Video
16.
Stem Cell Res ; 20: 10-13, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28395735

RESUMEN

Bernard Soulier Syndrome (BSS) is a rare autosomal platelet disorder characterized by mutations in the von Willebrand factor platelet receptor complex GPIb-V-IX. In this work we have generated an induced pluripotent stem cell (BSS3-PBMC-iPS4F8) from peripheral blood mononuclear cells of a BSS patient with a p.Phe55Ser mutation in the GPIX gene. Characterization of BSS3-PBMC-iPS4F8 showed that these cells maintained the original mutation present in the BSS patient, expressed pluripotent stem cell markers and were able to differentiate into the three germline layers. This new iPSC line will contribute to better understand the biology of BSS disease.


Asunto(s)
Síndrome de Bernard-Soulier/patología , Células Madre Pluripotentes Inducidas/citología , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Secuencia de Bases , Síndrome de Bernard-Soulier/genética , Síndrome de Bernard-Soulier/metabolismo , Diferenciación Celular , Línea Celular , Reprogramación Celular , Análisis Mutacional de ADN , Cuerpos Embrioides/metabolismo , Cuerpos Embrioides/patología , Femenino , Homocigoto , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Cariotipo , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Polimorfismo de Nucleótido Simple , Secuencias Repetidas en Tándem/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
Stem Cell Res ; 20: 34-37, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28395738

RESUMEN

The primary human dermal fibroblasts (PHDFs) from breast cancer patient were obtained to generate the human induced pluripotent stem cell line GPCCi001-A via lentiviral transfection. Thus, a modified EF1a-hSTEMCCA-loxP with tetO operator which regulates transgene expression was used. This method takes advantage of epigenetic regulation of transcription and allows for stable silencing of the reprogramming factors in obtained hiPS cells. To increase the potential utility of hiPSCs for clinical applications, they were adapted to feeder- and xeno-free conditions. The pluripotency of GPCCi001-A cell line and ability to differentiate into three germ layers was confirmed.


Asunto(s)
Reprogramación Celular , Fibroblastos/citología , Células Madre Pluripotentes Inducidas/citología , Factores de Transcripción/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular , Células Cultivadas , Cuerpos Embrioides/metabolismo , Cuerpos Embrioides/patología , Represión Epigenética , Células Nutrientes/citología , Femenino , Fibroblastos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Cariotipo , Microscopía Fluorescente , Factores de Transcripción/genética
18.
Stem Cell Res ; 20: 38-41, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28395739

RESUMEN

Fibroblasts from a female patient carrying a heterozygous variation in GTP cyclohydrolase 1 (GCH1; OMIM: 600225; HGNC: 4193; c.235_240del/p.(L79_S80del)), the rate-limiting enzyme of tetrahydrobiopterin (BH4) synthesis, were reprogrammed to iPSCs using the Cytotune®-iPS 2.0 Sendai Reprogramming Kit (Invitrogen) delivering the four reprogramming factors Oct3/4, Sox2, c-Myc and Klf4. Pluripotency of HDMC0061i-GCH1 was verified using immunohistochemistry and RT-PCR analysis. Cells differentiated spontaneously into the 3 germ layers in vitro and presented a normal karyotype. HDMC0061i-GCH1 represents the first model system to elucidate the pathomechanism underlying this rare metabolic disease and a useful tool for drug testing.


Asunto(s)
Reprogramación Celular , GTP Ciclohidrolasa/genética , Células Madre Pluripotentes Inducidas/citología , Fenilcetonurias/patología , Secuencia de Bases , Diferenciación Celular , Línea Celular , Análisis Mutacional de ADN , Cuerpos Embrioides/metabolismo , Cuerpos Embrioides/patología , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Eliminación de Gen , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Cariotipo , Factor 4 Similar a Kruppel , Microscopía Fluorescente , Fenilcetonurias/genética , Fenilcetonurias/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
Stem Cell Res ; 20: 50-53, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28395740

RESUMEN

Human epicardium-derived cells (EPDC) were reprogrammed to generate two iPSC lines, MCDU1i-EPDC and MCDU2i-EPDC, by nucleofection of episomal-based plasmids expressing the reprogramming factors OCT4, SOX2, KLF4, c-MYC, NANOG and LIN28. Pluripotency was confirmed in vitro by immunofluorescence analysis and embryoid body formation. The iPSC lines and the human embryonic stem cell line H1 show a Pearson correlation co-efficient of 0.951 (MCDU1i-EPDC) and 0.937 (MCDU2i-EPDC) as assessed by comparative transcriptome profiling.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas/citología , Pericardio/citología , Diferenciación Celular , Línea Celular , Dermatoglifia del ADN , Cuerpos Embrioides/metabolismo , Cuerpos Embrioides/patología , Femenino , Perfilación de la Expresión Génica , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Cariotipo , Factor 4 Similar a Kruppel , Microscopía Fluorescente , Pericardio/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
20.
Stem Cell Res ; 20: 67-69, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28395743

RESUMEN

Human fibroblast cells collected from a 3-year old, female Rett Syndrome patient with a 32bp deletion in the X-linked MECP2 gene were obtained from the Coriell Institute. Fibroblasts were reprogrammed to iPSC cells using a Sendai-virus delivery system expressing human KOSM transcription factors. Cell-line pluripotency was demonstrated by gene expression, immunocytochemistry, in-vitro differentiation trilineage capacity and was of normal karyotype. Interestingly, subsequent clones retained the epigenetic memory of the parent fibroblasts allowing for the segregation of wild-type and mutant expressing clones. This MECP2 mutant expressing clone may serve as a model for investigating MECP2 reactivation in Rett's Syndrome.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas/citología , Proteína 2 de Unión a Metil-CpG/genética , Síndrome de Rett/patología , Alelos , Diferenciación Celular , Línea Celular , Preescolar , Cuerpos Embrioides/metabolismo , Cuerpos Embrioides/patología , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Eliminación de Gen , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Genotipo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Cariotipo , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Virus Sendai/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...