Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Physiol Rev ; 99(1): 79-114, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30328784

RESUMEN

The discovery of somatic cell nuclear transfer proved that somatic cells can carry the same genetic code as the zygote, and that activating parts of this code are sufficient to reprogram the cell to an early developmental state. The discovery of induced pluripotent stem cells (iPSCs) nearly half a century later provided a molecular mechanism for the reprogramming. The initial creation of iPSCs was accomplished by the ectopic expression of four specific genes (OCT4, KLF4, SOX2, and c-Myc; OSKM). iPSCs have since been acquired from a wide range of cell types and a wide range of species, suggesting a universal molecular mechanism. Furthermore, cells have been reprogrammed to iPSCs using a myriad of methods, although OSKM remains the gold standard. The sources for iPSCs are abundant compared with those for other pluripotent stem cells; thus the use of iPSCs to model the development of tissues, organs, and other systems of the body is increasing. iPSCs also, through the reprogramming of patient samples, are being used to model diseases. Moreover, in the 10 years since the first report, human iPSCs are already the basis for new cell therapies and drug discovery that have reached clinical application. In this review, we examine the generation of iPSCs and their application to disease and development.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Pluripotentes Inducidas/citología , Modelos Biológicos , Células Madre Pluripotentes/clasificación , Animales , Tratamiento Basado en Trasplante de Células y Tejidos , Células Cultivadas , Humanos , Factor 4 Similar a Kruppel
2.
BMC Bioinformatics ; 24(1): 252, 2023 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-37322439

RESUMEN

BACKGROUND: Bioinformatics capability to analyze spatio-temporal dynamics of gene expression is essential in understanding animal development. Animal cells are spatially organized as functional tissues where cellular gene expression data contain information that governs morphogenesis during the developmental process. Although several computational tissue reconstruction methods using transcriptomics data have been proposed, those methods have been ineffective in arranging cells in their correct positions in tissues or organs unless spatial information is explicitly provided. RESULTS: This study demonstrates stochastic self-organizing map clustering with Markov chain Monte Carlo calculations for optimizing informative genes effectively reconstruct any spatio-temporal topology of cells from their transcriptome profiles with only a coarse topological guideline. The method, eSPRESSO (enhanced SPatial REconstruction by Stochastic Self-Organizing Map), provides a powerful in silico spatio-temporal tissue reconstruction capability, as confirmed by using human embryonic heart and mouse embryo, brain, embryonic heart, and liver lobule with generally high reproducibility (average max. accuracy = 92.0%), while revealing topologically informative genes, or spatial discriminator genes. Furthermore, eSPRESSO was used for temporal analysis of human pancreatic organoids to infer rational developmental trajectories with several candidate 'temporal' discriminator genes responsible for various cell type differentiations. CONCLUSIONS: eSPRESSO provides a novel strategy for analyzing mechanisms underlying the spatio-temporal formation of cellular organizations.


Asunto(s)
Perfilación de la Expresión Génica , Transcriptoma , Humanos , Animales , Ratones , Reproducibilidad de los Resultados , Encéfalo , Análisis por Conglomerados , Análisis Espacio-Temporal
3.
Arch Biochem Biophys ; 749: 109791, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37858665

RESUMEN

Cardiomyocyte differentiation and proliferation are essential processes for the regeneration of an injured heart. In recent years, there have been several reports highlighting the involvement of extracellular vesicles (EVs) in cardiomyocyte differentiation and proliferation. These EVs originate from mesenchymal stem cells, pluripotent stem cells, and heart constituting cells (cardiomyocytes, cardiac fibroblasts, cardiac progenitor cells, epicardium). Numerous reports also indicate the involvement of microRNAs (miRNAs) in cardiomyocyte differentiation and proliferation. Among them, miRNA-1, miRNA-133, and miRNA-499, recently demonstrated to promote cardiomyocyte differentiation, and miRNA-199, shown to promote cardiomyocyte proliferation, were found effective in various studies. MiRNA-132 and miRNA-133 have been identified as cargo in EVs and are reported to induce cardiomyocyte differentiation. Similarly, miRNA-30a, miRNA-100, miRNA-27a, miRNA-30e, miRNA-294 and miRNA-590 have also been identified as cargo in EVs and are shown to have a role in the promotion of cardiomyocyte proliferation. Regeneration of the heart by EVs or artificial nanoparticles containing functional miRNAs is expected in the future. In this review, we outline recent advancements in understanding the roles of EVs and miRNAs in cardiomyocyte differentiation and proliferation. Additionally, we explore the related challenges when utilizing EVs and miRNAs as a less risky approach to cardiac regeneration compared to cell transplantation.


Asunto(s)
Vesículas Extracelulares , MicroARNs , MicroARNs/genética , Miocitos Cardíacos , Diferenciación Celular , Proliferación Celular
4.
EMBO Rep ; 21(8): e50984, 2020 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-32715543

RESUMEN

New techniques to generate and culture embryo-like structures from stem cells require a more fine-grained distinction of potential to define the moral status of these structures.


Asunto(s)
Comienzo de la Vida Humana , Investigaciones con Embriones , Embrión de Mamíferos , Humanos , Obligaciones Morales , Condición Moral
5.
Biochem Biophys Res Commun ; 524(1): 57-63, 2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-31980180

RESUMEN

In normal development, the rate of cell differentiation is tightly controlled and critical for normal development and stem cell differentiation. However, the underlying mechanisms regulating the rate of the differentiation are unknown, and manipulation of the rate of the stem cell differentiation is currently difficult. Here we show that activation of protein kinase A (PKA) accelerates the rate of mouse embryonic stem cell (ESC) differentiation through an early loss of ESC pluripotency markers and early appearance of mesodermal and other germ layer cells. The activation of PKA hastened differentiation by increasing the expression of a histone H3 lysine 9 (H3K9) dimethyltransferase, G9a protein, and the level of a negative epigenetic histone mark, H3K9 dimethylation (H3K9me2), in the promoter regions of the pluripotency markers Nanog and Oct4. These results elucidate a novel role of PKA on ESC differentiation and offer an experimental model for controlling the rate of ESC differentiation.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Pluripotentes/metabolismo , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Estratos Germinativos/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Metilación , Ratones , Proteína Homeótica Nanog/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Transducción de Señal
6.
Biochem Biophys Res Commun ; 509(1): 24-31, 2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30573360

RESUMEN

Pluripotent stem cells retain the property to self-renew and differentiate into all cell types under defined conditions. Among mouse embryonic stem cells (ESCs), which are pluripotent but heterogenous in gene expression and morphology, an ESC population cultured in small molecule inhibitors of two kinases, MAPK/ERK kinase (Mek) and Glycogen synthase kinase 3 (Gsk3), and leukemia inhibitory factor (Lif) (2i/L) is considered to be naïve pluripotent with uniform pluripotent machinery operation. Though the gene regulatory mechanism for the naïve pluripotency has been investigated in recent years, it is still not fully elucidated. Here we show a novel signaling involved in the maintenance of naïve pluripotency. An AMP-activated protein kinase (AMPK) activator, AICAR (5-Aminoimidazole-4-carboxamied-1-ß-riboside) blocked the differentiation of mouse naïve ESCs in the absence of 2i/L and maintained the naïve state. AICAR with Lif condition induced an almost comparable level of naïve pluripotent gene expression in mouse ESCs. Another AMPK activator, A769662, also showed similar effects. A p38 inhibitor, SB203580, blocked the AMPK activation-elicited naïve state maintenance. On the other hand, p38 activation partially mimicked the maintenance effects of AMPK activators, suggesting that p38 is one of the functional downstream molecules to conduct the AMPK effects. Thus, AMPK pathway should be involved in the molecular circuitry of naïve pluripotency in mouse ESCs. These findings would be a valuable clue to further elucidate the molecular machinery of naïve pluripotency.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Activadores de Enzimas/farmacología , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/efectos de los fármacos , Proteínas Quinasas/metabolismo , Ribonucleótidos/farmacología , Quinasas de la Proteína-Quinasa Activada por el AMP , Aminoimidazol Carboxamida/farmacología , Animales , Línea Celular , Autorrenovación de las Células/efectos de los fármacos , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Transducción de Señal/efectos de los fármacos
7.
Nucleic Acids Res ; 45(8): 4344-4358, 2017 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-28334937

RESUMEN

Although studies of the differentiation from mouse embryonic stem (ES) cells to vascular endothelial cells (ECs) provide an excellent model for investigating the molecular mechanisms underlying vascular development, temporal dynamics of gene expression and chromatin modifications have not been well studied. Herein, using transcriptomic and epigenomic analyses based on H3K4me3 and H3K27me3 modifications at a genome-wide scale, we analysed the EC differentiation steps from ES cells and crucial epigenetic modifications unique to ECs. We determined that Gata2, Fli1, Sox7 and Sox18 are master regulators of EC that are induced following expression of the haemangioblast commitment pioneer factor, Etv2. These master regulator gene loci were repressed by H3K27me3 throughout the mesoderm period but rapidly transitioned to histone modification switching from H3K27me3 to H3K4me3 after treatment with vascular endothelial growth factor. SiRNA knockdown experiments indicated that these regulators are indispensable not only for proper EC differentiation but also for blocking the commitment to other closely aligned lineages. Collectively, our detailed epigenetic analysis may provide an advanced model for understanding temporal regulation of chromatin signatures and resulting gene expression profiles during EC commitment. These studies may inform the future development of methods to stimulate the vascular endothelium for regenerative medicine.


Asunto(s)
Células Endoteliales/metabolismo , Epigénesis Genética , Factor de Transcripción GATA2/genética , Histonas/genética , Células Madre Embrionarias de Ratones/metabolismo , Proteína Proto-Oncogénica c-ets-1/genética , Factores de Transcripción SOXF/genética , Animales , Diferenciación Celular , Linaje de la Célula/genética , Células Endoteliales/citología , Factor de Transcripción GATA2/antagonistas & inhibidores , Factor de Transcripción GATA2/metabolismo , Histonas/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Análisis de Secuencia por Matrices de Oligonucleótidos , Cultivo Primario de Células , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteína Proto-Oncogénica c-ets-1/antagonistas & inhibidores , Proteína Proto-Oncogénica c-ets-1/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Factores de Transcripción SOXF/antagonistas & inhibidores , Factores de Transcripción SOXF/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Pediatr Res ; 84(5): 733-742, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30188508

RESUMEN

BACKGROUND: Left ventricular noncompaction (LVNC) is a primary cardiomyopathy with heterogeneous genetic origins. The aim of this study was to elucidate the role of sarcomere gene variants in the pathogenesis and prognosis of LVNC. METHODS AND RESULTS: We screened 82 Japanese patients (0-35 years old), with a diagnosis of LVNC, for mutations in seven genes encoding sarcomere proteins, by direct DNA sequencing. We identified variants in a significant proportion of cases (27%), which were associated with poor prognosis (p = 0.012), particularly variants in TPM1, TNNC1, and ACTC1 (p = 0.012). To elucidate the pathological role, we developed and studied human-induced pluripotent stem cells (hiPSCs) from a patient carrying a TPM1 p.Arg178His mutation, who underwent heart transplantation. These cells displayed pathological changes, with mislocalization of tropomyosin 1, causing disruption of the sarcomere structure in cardiomyocytes, and impaired calcium handling. Microarray analysis indicated that the TPM1 mutation resulted in the down-regulation of the expression of numerous genes involved in heart development, and positive regulation of cellular process, especially the calcium signaling pathway. CONCLUSIONS: Sarcomere genes are implicated as genetic triggers in the development of LVNC, regulating the expression of numerous genes involved in heart development, or modifying the severity of disease.


Asunto(s)
Ventrículos Cardíacos/patología , Sarcómeros/genética , Adolescente , Adulto , Pueblo Asiatico/genética , Señalización del Calcio , Niño , Preescolar , Femenino , Ventrículos Cardíacos/metabolismo , Humanos , Lactante , Recién Nacido , Japón , Masculino , Mutación , Pronóstico , Sarcómeros/metabolismo , Adulto Joven
9.
Bioorg Med Chem ; 26(13): 3852-3857, 2018 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-29983284

RESUMEN

Sameuramide A (1), a new cyclic depsipeptide encompassing one each of alanine, N-methyl alanine, N-methyl dehydroalanine, N,O-dimethyl threonine, phenyllactic acid, three ß-hydroxy leucines, and two propionates, was isolated from a didemnid ascidian collected at the northern part of Japan. The planar structure was established based on the interpretation of MS and NMR data. The absolute configuration of the subunits was determined by the advanced Marfey's method and the chiral LC-MS analysis. Compound 1 exhibited the activity of maintaining colony formation of murine embryonic stem (mES) cells without leukemia inhibitory factor (LIF). Down regulation of the gene expression of Krüppel-like transcription factor 4 (Klf4) indicated that 1 itself was not able to maintain the undifferentiated state of the mES cells. However, the expression levels of the marker genes (Nestin, T, Sox17) for three germ layers were upregulated in embryoid bodies (EBs) after treatment of 1 together with LIF, suggesting that 1 plays a supportive role for LIF in maintaining the multipotency of mES cells.


Asunto(s)
Depsipéptidos/química , Urocordados/química , Animales , Diferenciación Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Depsipéptidos/aislamiento & purificación , Depsipéptidos/farmacología , Regulación hacia Abajo/efectos de los fármacos , Cuerpos Embrioides/citología , Cuerpos Embrioides/efectos de los fármacos , Cuerpos Embrioides/metabolismo , Células Madre Embrionarias , Proteínas HMGB/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/metabolismo , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Ratones , Conformación Molecular , Factores de Transcripción SOXF/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Urocordados/metabolismo
10.
Biochem Biophys Res Commun ; 444(2): 158-63, 2014 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-24462858

RESUMEN

Specific gene knockout and rescue experiments are powerful tools in developmental and stem cell biology. Nevertheless, the experiments require multiple steps of molecular manipulation for gene knockout and subsequent rescue procedures. Here we report an efficient and single step strategy to generate gene knockout-rescue system in pluripotent stem cells by promoter insertion with CRISPR/Cas9 genome editing technology. We inserted a tetracycline-regulated inducible gene promoter (tet-OFF/TRE-CMV) upstream of the endogenous promoter region of vascular endothelial growth factor receptor 2 (VEGFR2/Flk1) gene, an essential gene for endothelial cell (EC) differentiation, in mouse embryonic stem cells (ESCs) with homologous recombination. Both homo- and hetero-inserted clones were efficiently obtained through a simple selection with a drug-resistant gene. The insertion of TRE-CMV promoter disrupted endogenous Flk1 expression, resulting in null mutation in homo-inserted clones. When the inserted TRE-CMV promoter was activated with doxycycline (Dox) depletion, Flk1 expression was sufficiently recovered from the downstream genomic Flk1 gene. Whereas EC differentiation was almost completely perturbed in homo-inserted clones, Flk1 rescue with TRE-CMV promoter activation restored EC appearance, indicating that phenotypic changes in EC differentiation can be successfully reproduced with this knockout-rescue system. Thus, this promoter insertion strategy with CRISPR/Cas9 would be a novel attractive method for knockout-rescue experiments.


Asunto(s)
Proteínas Asociadas a CRISPR/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Técnicas de Inactivación de Genes/métodos , Células Madre Pluripotentes/metabolismo , Regiones Promotoras Genéticas/genética , Animales , Proteínas Asociadas a CRISPR/metabolismo , Diferenciación Celular/genética , Línea Celular , Citomegalovirus/genética , Doxiciclina/farmacología , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Expresión Génica/efectos de los fármacos , Immunoblotting , Ratones , Modelos Genéticos , Mutagénesis Insercional , Células Madre Pluripotentes/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tetraciclina/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
11.
Inflamm Regen ; 44(1): 4, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38225584

RESUMEN

In recent years, extracellular vesicles (EVs) have attracted significant attention as carriers in intercellular communication. The vast array of information contained within EVs is critical for various cellular activities, such as proliferation and differentiation of multiple cell types. Moreover, EVs are being employed in disease diagnostics, implicated in disease etiology, and have shown promise in tissue repair. Recently, a phenomenon has been discovered in which cellular phenotypes, including the progression of differentiation, are synchronized among cells via EVs. This synchronization could be prevalent in widespread different situations in embryogenesis and tissue organization and maintenance. Given the increasing research on multi-cellular tissues and organoids, the role of EV-mediated intercellular communication has become increasingly crucial. This review begins with fundamental knowledge of EVs and then discusses recent findings, various modes of information transfer via EVs, and synchronization of cellular phenotypes.

12.
Sci Rep ; 14(1): 2586, 2024 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-38297132

RESUMEN

Disease modeling using human induced pluripotent stem cells (hiPSCs) from patients with genetic disease is a powerful approach for dissecting pathophysiology and drug discovery. Nevertheless, isogenic controls are required to precisely compare phenotypic outcomes from presumed causative mutations rather than differences in genetic backgrounds. Moreover, 2D cellular models often fail to exhibit authentic disease phenotypes resulting in poor validation in vitro. Here we show that a combination of precision gene editing and bioengineered 3D tissue models can establish advanced isogenic hiPSC-derived cardiac disease models, overcoming these drawbacks. To model inherited cardiac arrhythmias we selected representative N588D and N588K missense mutations affecting the same codon in the hERG potassium channel gene KCNH2, which are reported to cause long (LQTS) and short (SQTS) QT syndromes, respectively. We generated compound heterozygous variants in normal hiPSCs, and differentiated cardiomyocytes (CMs) and mesenchymal cells (MCs) to form 3D cardiac tissue sheets (CTSs). In hiPSC-derived CM monolayers and 3D CTSs, electrophysiological analysis with multielectrode arrays showed prolonged and shortened repolarization, respectively, compared to the isogenic controls. When pharmacologically inhibiting the hERG channels, mutant 3D CTSs were differentially susceptible to arrhythmic events than the isogenic controls. Thus, this strategy offers advanced disease models that can reproduce clinically relevant phenotypes and provide solid validation of gene mutations in vitro.


Asunto(s)
Células Madre Pluripotentes Inducidas , Síndrome de QT Prolongado , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Síndrome de QT Prolongado/genética , Canal de Potasio ERG1/genética , Arritmias Cardíacas/genética , Mutación , Miocitos Cardíacos/fisiología , Fenotipo , Potenciales de Acción/genética
13.
Blood ; 118(3): 775-85, 2011 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-21460241

RESUMEN

The opioid system (opioid peptides and receptors) regulates a variety of neurophysiologic functions, including pain control. Here we show novel roles of the κ opioid system in vascular development. Previously, we revealed that cAMP/protein kinase A (PKA) signaling enhanced differentiation of vascular progenitors expressing VEGF receptor-2 (fetal liver kinase 1; Flk1) into endothelial cells (ECs) through dual up-regulation of Flk1 and Neuropilin1 (NRP1), which form a selective and sensitive VEGF(164) receptor. Kappa opioid receptor (KOR), an inhibitory G protein-coupled receptor, was highly expressed in embryonic stem cell-derived Flk1(+) vascular progenitors. The addition of KOR agonists to Flk1(+) vascular progenitors inhibited EC differentiation and 3-dimensional vascular formation. Activation of KOR decreased expression of Flk1 and NRP1 in vascular progenitors. The inhibitory effects of KOR were reversed by 8-bromoadenosine-3',5'-cAMP or a PKA agonist, N(6)-benzoyl-cAMP, indicating that KOR inhibits cAMP/PKA signaling. Furthermore, KOR-null or dynorphin (an endogenous KOR agonist)-null mice showed a significant increase in overall vascular formation and ectopic vascular invasion into somites at embryonic day -10.5. ECs in these null mice showed significant increase in Flk1 and NRP1, along with reciprocal decrease in plexinD1, which regulates vascular pathfinding. The opioid system is, thus, a new regulator of vascular development that simultaneously modifies 2 distinct vascular properties, EC differentiation and vascular pathfinding.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Células Endoteliales/citología , Células Endoteliales/fisiología , Receptores Opioides kappa/fisiología , Transducción de Señal/fisiología , Animales , Diferenciación Celular/fisiología , Línea Celular , Movimiento Celular/fisiología , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Dinorfinas/genética , Dinorfinas/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Fisiológica/fisiología , Neuropilina-1/metabolismo , Embarazo , Receptores Opioides kappa/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
14.
Stem Cells ; 30(4): 687-96, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22267325

RESUMEN

Ets family protein Etv2 (also called ER71 or Etsrp) is a key factor for initiation of vascular and blood development from mesodermal cells. However, regulatory mechanisms and inducing signals for Etv2 expression have been largely unknown. Previously, we revealed that cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) signaling enhanced differentiation of vascular progenitors into endothelial cells (ECs) and hematopoietic cells (HPCs) using an embryonic stem cell (ESC) differentiation system. Here, we show that PKA activation in an earlier differentiation stage can trigger EC/HPC differentiation through Etv2 induction. We found Etv2 was markedly upregulated by PKA activation preceding EC and HPC differentiation. We identified two cAMP response element (CRE) sequences in the Etv2 promoter and 5'-untranslated region and confirmed that CRE-binding protein (CREB) directly binds to the CRE sites and activates Etv2 transcription. Expression of a dominant negative form of CREB completely inhibited PKA-elicited Etv2 expression and induction of EC/HPCs from ESCs. Furthermore, blockade of PKA significantly inhibited Etv2 expression in ex vivo whole-embryo culture using Etv2-Venus knockin mice. These data indicated that PKA/CREB pathway is a critical regulator for the initiation of EC/HPC differentiation via Etv2 transcription. This early-stage molecular linkage between a triggering signal and transcriptional cascades for differentiation would provide novel insights in vascular and blood development and cell fate determination.


Asunto(s)
Diferenciación Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células Endoteliales/citología , Células Madre Hematopoyéticas/citología , Proteína Proto-Oncogénica c-ets-1/metabolismo , Transducción de Señal , Regiones no Traducidas 5'/genética , Animales , Secuencia de Bases , Línea Celular , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/enzimología , Células Endoteliales/metabolismo , Activación Enzimática , Células Madre Hematopoyéticas/metabolismo , Ratones , Modelos Biológicos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/genética , Proteína Proto-Oncogénica c-ets-1/genética , ARN Interferente Pequeño/metabolismo , Técnicas de Cultivo de Tejidos , Transcripción Genética
15.
Stem Cells ; 30(6): 1196-205, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22438013

RESUMEN

Although stem cell therapy is a promising strategy for cardiac restoration, the heterogeneity of transplanted cells has been hampering the precise understanding of the cellular and molecular mechanisms. Previously, we established a cardiovascular cell differentiation system from mouse pluripotent stem cells, in which cardiomyocytes (CMs), endothelial cells (ECs), and mural cells (MCs) can be systematically induced and purified. Combining this with cell sheet technology, we generated cardiac tissue sheets reassembled with defined cardiovascular populations. Here, we show the potentials and mechanisms of cardiac tissue sheet transplantation in cardiac function after myocardial infarction (MI). Transplantation of the cardiac tissue sheet to a rat MI model showed significant and sustained improvement of systolic function accompanied by neovascularization. Reduction of the infarct wall thinning and fibrotic length indicated the attenuation of left ventricular remodeling. Cell tracing with species-specific fluorescent in situ hybridization after transplantation revealed a relatively early loss of transplanted cells and an increase in endogenous neovascularization in the proximity of the graft, suggesting an indirect angiogenic effect of cardiac tissue sheets rather than direct CM contributions. We prospectively dissected the functional mechanisms with cell type-controlled sheet analyses. Sheet CMs were the main source of vascular endothelial growth factor. Transplantation of sheets lacking CMs resulted in the disappearance of neovascularization and subsequent functional improvement, indicating that the beneficial effects of the sheet were achieved by sheet CMs. ECs and MCs enhanced the sheet functions and structural integration. Supplying CMs to ischemic regions with cellular interaction could be a strategic key in future cardiac cell therapy.


Asunto(s)
Infarto del Miocardio/cirugía , Miocitos Cardíacos/trasplante , Células Madre Pluripotentes/trasplante , Ingeniería de Tejidos/métodos , Animales , Vasos Coronarios/crecimiento & desarrollo , Modelos Animales de Enfermedad , Masculino , Ratones , Infarto del Miocardio/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Neovascularización Fisiológica , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología , Estudios Prospectivos , Ratas , Ratas Desnudas , Trasplante de Células Madre/métodos
16.
Artículo en Inglés | MEDLINE | ID: mdl-37981100

RESUMEN

OBJECTIVE: This study aimed to explore the therapeutic potential of human induced pluripotent stem cell (hiPSC)-derived cardiac tissues (HiCTs) in the emerging approach of bridge to recovery for severe heart failure with ventricular assist devices. We used a rat model of heterotopic heart transplantation (HTx) to mimic ventricular assist device support and heart unloading. METHODS: HiCTs were created by inserting gelatin hydrogel microspheres between cell sheets made from hiPSC-derived cardiovascular cells. Male athymic nude rats underwent myocardial infarction (MI) and were divided into the following groups: MI (loaded, untreated control), MI + HTx (unloaded, untreated control), MI + HTx + HiCT (unloaded, treated), and MI + HiCT (loaded, treated). HiCTs were placed on the epicardium of the heart in treated groups. We evaluated HiCT engraftment, fibrosis, and neovascularization using histologic analysis. RESULTS: After 4 weeks, HiCTs successfully engrafted in 5 of 6 rats in the MI + HTx + HiCT group (83.3%). The engrafted HiCT area was greater under unloaded conditions (MI + HTx + HiCT) than loaded conditions (MI + HiCT) (P < .05). MI + HTx + HiCT had a significantly smaller infarct area compared with MI and MI + HTx. The MI + HTx + MiCT group exhibited greater vascular density in the border zone than MI and MI + HTx. HiCT treatment suppressed cardiomyocyte atrophy due to left ventricular unloading (P = .001). The protein level of muscle-specific RING finger 1, an atrophy-related ubiquitin ligase, was lower in the MI + HTx + HiCT group than in MI + HTx (P = .036). CONCLUSIONS: Transplanting HiCTs into ischemic hearts under unloaded conditions promoted engraftment, neovascularization, attenuated infarct remodeling, and suppressed myocyte atrophy. These results suggest that HiCT treatment could contribute to future advancements in bridge to recovery.

17.
iScience ; 25(7): 104538, 2022 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35754715

RESUMEN

An alternative model that reliably predicts human-specific toxicity is necessary because the translatability of effects on animal models for human disease is limited to context. Previously, we developed a method that accurately predicts developmental toxicity based on the gene networks of undifferentiated human embryonic stem (ES) cells. Here, we advanced this method to predict adult toxicities of 24 chemicals in six categories (neurotoxins, cardiotoxins, hepatotoxins, two types of nephrotoxins, and non-genotoxic carcinogens) and achieved high predictability (AUC = 0.90-1.00) in all categories. Moreover, we screened for an induced pluripotent stem (iPS) cell line to predict the toxicities based on the gene networks of iPS cells using transfer learning of the gene networks of ES cells, and predicted toxicities in four categories (neurotoxins, hepatotoxins, glomerular nephrotoxins, and non-genotoxic carcinogens) with high performance (AUC = 0.82-0.99). This method holds promise for tailor-made safety evaluations using personalized iPS cells.

18.
Blood ; 114(17): 3707-16, 2009 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-19706882

RESUMEN

Fine tuning of vascular endothelial growth factor (VEGF) signaling is critical in endothelial cell (EC) differentiation and vascular development. Nevertheless, the system for regulating the sensitivity of VEGF signaling has remained unclear. Previously, we established an embryonic stem cell culture reproducing early vascular development using Flk1 (VEGF receptor-2)+ cells as common progenitors, and demonstrated that cyclic adenosine monophosphate (cAMP) enhanced VEGF-induced EC differentiation. Here we show that protein kinase A (PKA) regulates sensitivity of Flk1+ vascular progenitors to VEGF signaling for efficient EC differentiation. Blockade of PKA perturbed EC differentiation and vascular formation in vitro and ex vivo. Overexpression of constitutive active form of PKA (CA-PKA) potently induced EC differentiation and vascular formation. Expression of Flk1 and Neuropilin-1 (NRP1), which form a selective and sensitive receptor for VEGF(165), was increased only in CA-PKA-expressing progenitors, enhancing the sensitivity of the progenitors to VEGF(165) by more than 10 times. PKA activation induced the formation of a VEGF(165), Flk1, and NRP1 protein complex in vascular progenitors. These data indicate that PKA regulates differentiation potential of vascular progenitors to be endothelial competent via the dual induction of Flk1 and NRP1. This new-mode mechanism regulating "progenitor sensitivity" would provide a novel understanding in vascular development and regeneration.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células Madre Embrionarias/metabolismo , Endotelio Vascular/citología , Neovascularización Fisiológica , Neuropilina-1/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Western Blotting , Técnicas de Cultivo de Célula , Diferenciación Celular , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Endotelio Vascular/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnicas para Inmunoenzimas , Inmunoprecipitación , Ratones , Neuropilina-1/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
19.
Circ J ; 75(2): 253-60, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21178292

RESUMEN

Cyclic adenosine monophosphate (cAMP) is an important second messenger mediating physiological functions, including metabolism, gene expression, cell growth and differentiation. Recently, we demonstrated novel roles of cAMP pathway in endothelial cell (EC) differentiation and arterial-venous specification using an embryonic stem cell differentiation system. These studies offered a concept that vascular formation is accomplished by a 2-layered mechanism: (1) a basal mechanism for common EC differentiation, whereby vascular endothelial growth factor (VEGF) signaling plays a central role in the basal mechanism, and (2) a vascular diversification mechanism working on the basis of common EC differentiation. Vascular diversification, such as artery and vein formation, can be only achieved by enacting specific machineries in the presence of the basal EC machinery. cAMP/protein kinase A signaling contributes to common EC differentiation through upregulation of the VEGF-A receptors, Flk1 and neuropilin1. On the other hand, cAMP can activate phosphatidylinositol-3 kinase, which induces an arterial fate in vascular progenitors via dual activation of Notch and ß-catenin signaling as an arterial-specific machinery. cAMP signaling thus plays a pivotal role in both the basal and diversification machinery during vascular development.


Asunto(s)
Arterias/embriología , AMP Cíclico/fisiología , Células Endoteliales/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Neovascularización Fisiológica/fisiología , Sistemas de Mensajero Secundario/fisiología , Venas/embriología , Animales , Diferenciación Celular/fisiología , Hipoxia de la Célula/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Efrinas/fisiología , Humanos , Mamíferos , Modelos Biológicos , Fosfatidilinositol 3-Quinasas/fisiología , Receptores Notch/fisiología , Receptores de Factores de Crecimiento Endotelial Vascular/biosíntesis , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Pez Cebra , beta Catenina/fisiología
20.
Exp Cell Res ; 316(16): 2555-9, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20385126

RESUMEN

Embryonic stem (ES) cells and induced pluripotent stem (iPS) cells, which are ES-like stem cells induced from adult tissues, are twin stem cells with currently (with the exception of fertilized eggs) the broadest differentiation potentials. These two stem cells show various similarities in appearance, maintenance methods, growth and differentiation potentials, i.e. theoretically, those cells can give rise to all kinds of cells including germ-line cells. Generation of human ES and iPS cells is further facilitating the researches towards the realization of regenerative medicine. The following three issues are important purposes of ES and iPS cell researches for regenerative medicine: (1) dissection of differentiation mechanisms, (2) application to cell transplantation, and (3) drug discovery. In this review, the current status of cardiovascular regenerative trials using ES and iPS cells is briefly discussed.


Asunto(s)
Sistema Cardiovascular/fisiopatología , Células Madre Embrionarias/citología , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Adulto , Investigación Biomédica , Humanos , Medicina Regenerativa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA