Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 79(1): 84-98.e9, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32526163

RESUMEN

Rett syndrome (RTT), mainly caused by mutations in methyl-CpG binding protein 2 (MeCP2), is one of the most prevalent intellectual disorders without effective therapies. Here, we used 2D and 3D human brain cultures to investigate MeCP2 function. We found that MeCP2 mutations cause severe abnormalities in human interneurons (INs). Surprisingly, treatment with a BET inhibitor, JQ1, rescued the molecular and functional phenotypes of MeCP2 mutant INs. We uncovered that abnormal increases in chromatin binding of BRD4 and enhancer-promoter interactions underlie the abnormal transcription in MeCP2 mutant INs, which were recovered to normal levels by JQ1. We revealed cell-type-specific transcriptome impairment in MeCP2 mutant region-specific human brain organoids that were rescued by JQ1. Finally, JQ1 ameliorated RTT-like phenotypes in mice. These data demonstrate that BRD4 dysregulation is a critical driver for RTT etiology and suggest that targeting BRD4 could be a potential therapeutic opportunity for RTT.


Asunto(s)
Azepinas/farmacología , Encéfalo/patología , Proteínas de Ciclo Celular/metabolismo , Interneuronas/patología , Proteína 2 de Unión a Metil-CpG/fisiología , Síndrome de Rett/patología , Factores de Transcripción/metabolismo , Transcriptoma/efectos de los fármacos , Triazoles/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Proteínas de Ciclo Celular/genética , Femenino , Células Madre Embrionarias Humanas/efectos de los fármacos , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/patología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Interneuronas/efectos de los fármacos , Interneuronas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Fenotipo , Síndrome de Rett/tratamiento farmacológico , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Factores de Transcripción/genética
2.
Nat Methods ; 16(11): 1169-1175, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31591580

RESUMEN

Human cortical organoids (hCOs), derived from human embryonic stem cells (hESCs), provide a platform to study human brain development and diseases in complex three-dimensional tissue. However, current hCOs lack microvasculature, resulting in limited oxygen and nutrient delivery to the inner-most parts of hCOs. We engineered hESCs to ectopically express human ETS variant 2 (ETV2). ETV2-expressing cells in hCOs contributed to forming a complex vascular-like network in hCOs. Importantly, the presence of vasculature-like structures resulted in enhanced functional maturation of organoids. We found that vascularized hCOs (vhCOs) acquired several blood-brain barrier characteristics, including an increase in the expression of tight junctions, nutrient transporters and trans-endothelial electrical resistance. Finally, ETV2-induced endothelium supported the formation of perfused blood vessels in vivo. These vhCOs form vasculature-like structures that resemble the vasculature in early prenatal brain, and they present a robust model to study brain disease in vitro.


Asunto(s)
Encéfalo/irrigación sanguínea , Células Madre Embrionarias Humanas/citología , Organoides/irrigación sanguínea , Ingeniería de Tejidos/métodos , Animales , Barrera Hematoencefálica , Células Cultivadas , Humanos , Ratones , Análisis de la Célula Individual , Factores de Transcripción/fisiología
3.
Nucleic Acids Res ; 45(10): e77, 2017 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-28126923

RESUMEN

Conventional DNA bisulfite sequencing has been extended to single cell level, but the coverage consistency is insufficient for parallel comparison. Here we report a novel method for genome-wide CpG island (CGI) methylation sequencing for single cells (scCGI-seq), combining methylation-sensitive restriction enzyme digestion and multiple displacement amplification for selective detection of methylated CGIs. We applied this method to analyzing single cells from two types of hematopoietic cells, K562 and GM12878 and small populations of fibroblasts and induced pluripotent stem cells. The method detected 21 798 CGIs (76% of all CGIs) per cell, and the number of CGIs consistently detected from all 16 profiled single cells was 20 864 (72.7%), with 12 961 promoters covered. This coverage represents a substantial improvement over results obtained using single cell reduced representation bisulfite sequencing, with a 66-fold increase in the fraction of consistently profiled CGIs across individual cells. Single cells of the same type were more similar to each other than to other types, but also displayed epigenetic heterogeneity. The method was further validated by comparing the CpG methylation pattern, methylation profile of CGIs/promoters and repeat regions and 41 classes of known regulatory markers to the ENCODE data. Although not every minor methylation differences between cells are detectable, scCGI-seq provides a solid tool for unsupervised stratification of a heterogeneous cell population.


Asunto(s)
Islas de CpG , Metilación de ADN , Epigénesis Genética , Regiones Promotoras Genéticas , Análisis de la Célula Individual/métodos , Línea Celular , Línea Celular Tumoral , Mapeo Cromosómico , Enzimas de Restricción del ADN/química , Fibroblastos/citología , Fibroblastos/metabolismo , Variación Genética , Genoma Humano , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Células K562 , Linfocitos/citología , Linfocitos/metabolismo
4.
Hum Mol Genet ; 23(4): 1045-55, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24129406

RESUMEN

Rett syndrome (RTT) is one of the most prevalent female mental disorders. De novo mutations in methyl CpG-binding protein 2 (MeCP2) are a major cause of RTT. MeCP2 regulates gene expression as a transcription regulator as well as through long-range chromatin interaction. Because MeCP2 is present on the X chromosome, RTT is manifested in an X-linked dominant manner. Investigation using murine MeCP2 null models and post-mortem human brain tissues has contributed to understanding the molecular and physiological function of MeCP2. In addition, RTT models using human induced pluripotent stem cells derived from RTT patients (RTT-iPSCs) provide novel resources to elucidate the regulatory mechanism of MeCP2. Previously, we obtained clones of female RTT-iPSCs that express either wild-type or mutant MECP2 due to the inactivation of one X chromosome. Reactivation of the X chromosome also allowed us to have RTT-iPSCs that express both wild-type and mutant MECP2. Using these unique pluripotent stem cells, we investigated the regulation of gene expression by MeCP2 in pluripotent stem cells by transcriptome analysis. We found that MeCP2 regulates genes encoding mitochondrial membrane proteins. In addition, loss of function in MeCP2 results in de-repression of genes on the inactive X chromosome. Furthermore, we showed that each mutation in MECP2 affects a partly different set of genes. These studies suggest that fundamental cellular physiology is affected by mutations in MECP2 from early development, and that a therapeutic approach targeting to unique forms of mutant MeCP2 is needed.


Asunto(s)
Regulación de la Expresión Génica , Células Madre Pluripotentes Inducidas/metabolismo , Proteína 2 de Unión a Metil-CpG/fisiología , Transcripción Genética , Células Cultivadas , Células Madre Embrionarias/metabolismo , Femenino , Ontología de Genes , Humanos , Mutación , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Síndrome de Rett/patología , Transcriptoma
5.
Proc Natl Acad Sci U S A ; 108(34): 14169-74, 2011 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-21807996

RESUMEN

Rett syndrome (RTT) is one of the most prevalent female neurodevelopmental disorders that cause severe mental retardation. Mutations in methyl CpG binding protein 2 (MeCP2) are mainly responsible for RTT. Patients with classical RTT exhibit normal development until age 6-18 mo, at which point they become symptomatic and display loss of language and motor skills, purposeful hand movements, and normal head growth. Murine genetic models and postmortem human brains have been used to study the disease and enable the molecular dissection of RTT. In this work, we applied a recently developed reprogramming approach to generate a novel in vitro human RTT model. Induced pluripotent stem cells (iPSCs) were derived from RTT fibroblasts by overexpressing the reprogramming factors OCT4, SOX2, KLF4, and MYC. Intriguingly, whereas some iPSCs maintained X chromosome inactivation, in others the X chromosome was reactivated. Thus, iPSCs were isolated that retained a single active X chromosome expressing either mutant or WT MeCP2, as well as iPSCs with reactivated X chromosomes expressing both mutant and WT MeCP2. When these cells underwent neuronal differentiation, the mutant monoallelic or biallelelic RTT-iPSCs displayed a defect in neuronal maturation consistent with RTT phenotypes. Our in vitro model of RTT is an important tool allowing the further investigation of the pathophysiology of RTT and the development of the curative therapeutics.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas/patología , Neuronas/patología , Síndrome de Rett/patología , Adulto , Secuencia de Aminoácidos , Secuencia de Bases , Biomarcadores/metabolismo , Diferenciación Celular/genética , Niño , Preescolar , Cromosomas Humanos X/genética , Células Madre Embrionarias/metabolismo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Proteína 2 de Unión a Metil-CpG/química , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Datos de Secuencia Molecular , Neuronas/metabolismo , Síndrome de Rett/genética , Inactivación del Cromosoma X/genética
6.
Circulation ; 126(14): 1695-704, 2012 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-22914687

RESUMEN

BACKGROUND: Supravalvular aortic stenosis (SVAS) is caused by mutations in the elastin (ELN) gene and is characterized by abnormal proliferation of vascular smooth muscle cells (SMCs) that can lead to narrowing or blockage of the ascending aorta and other arterial vessels. Having patient-specific SMCs available may facilitate the study of disease mechanisms and development of novel therapeutic interventions. METHODS AND RESULTS: Here, we report the development of a human induced pluripotent stem cell (iPSC) line from a patient with SVAS caused by the premature termination in exon 10 of the ELN gene resulting from an exon 9 four-nucleotide insertion. We showed that SVAS iPSC-derived SMCs (iPSC-SMCs) had significantly fewer organized networks of smooth muscle α-actin filament bundles, a hallmark of mature contractile SMCs, compared with control iPSC-SMCs. The addition of elastin recombinant protein or enhancement of small GTPase RhoA signaling was able to rescue the formation of smooth muscle α-actin filament bundles in SVAS iPSC-SMCs. Cell counts and BrdU analysis revealed a significantly higher proliferation rate in SVAS iPSC-SMCs than control iPSC-SMCs. Furthermore, SVAS iPSC-SMCs migrated at a markedly higher rate to the chemotactic agent platelet-derived growth factor compared with the control iPSC-SMCs. We also provided evidence that elevated activity of extracellular signal-regulated kinase 1/2 is required for hyperproliferation of SVAS iPSC-SMCs. The phenotype was confirmed in iPSC-SMCs generated from a patient with deletion of elastin owing to Williams-Beuren syndrome. CONCLUSIONS: SVAS iPSC-SMCs recapitulate key pathological features of patients with SVAS and may provide a promising strategy to study disease mechanisms and to develop novel therapies.


Asunto(s)
Estenosis Aórtica Supravalvular/patología , Células Madre Pluripotentes Inducidas/patología , Síndrome de Williams/patología , Adulto , Animales , Células Cultivadas , Niño , Humanos , Masculino , Ratones
7.
Sci Adv ; 9(31): eadf2245, 2023 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-37540754

RESUMEN

Three-dimensional (3D) genomics shows immense promise for studying X chromosome inactivation (XCI) by interrogating changes to the X chromosomes' 3D states. Here, we sought to characterize the 3D state of the X chromosome in naïve and primed human pluripotent stem cells (hPSCs). Using chromatin tracing, we analyzed X chromosome folding conformations in these cells with megabase genomic resolution. X chromosomes in female naïve hPSCs exhibit folding conformations similar to the active X chromosome (Xa) and the inactive X chromosome (Xi) in somatic cells. However, naïve X chromosomes do not exhibit the chromatin compaction typically associated with these somatic X chromosome states. In H7 naïve human embryonic stem cells, XIST accumulation observed on damaged X chromosomes demonstrates the potential for naïve hPSCs to activate XCI-related mechanisms. Overall, our findings provide insight into the X chromosome status of naïve hPSCs with a single-chromosome resolution and are critical in understanding the unique epigenetic regulation in early embryonic cells.


Asunto(s)
Células Madre Pluripotentes , ARN Largo no Codificante , Humanos , Femenino , Epigénesis Genética , Cromosomas Humanos X/genética , ARN Largo no Codificante/genética , Cromatina/genética
8.
Curr Opin Neurol ; 25(2): 125-30, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22357218

RESUMEN

PURPOSE OF REVIEW: The lack of effective treatments for various neurodegenerative disorders has placed huge burdens on society. We review the current status in applying induced pluripotent stem cell (iPSC) technology for the cellular therapy, drug screening, and in-vitro modeling of neurodegenerative diseases. RECENT FINDINGS: iPSCs are generated from somatic cells by overexpressing four reprogramming factors (Oct4, Sox2, Klf4, and Myc). Like human embryonic stem cells, iPSCs have features of self-renewal and pluripotency, and allow in-vitro disease modeling, drug screening, and cell replacement therapy. Disease-specific iPSCs were derived from patients of several neurodegenerative diseases, including Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, and spinal muscular atrophy. Neurons differentiated from these iPSCs recapitulated the in-vivo phenotypes, providing platforms for drug screening. In the case of Parkinson's disease, iPSC-derived dopaminergic neurons gave positive therapeutic effect on a rodent Parkinson's disease model as a proof of principle in using iPSCs as sources of cell replacement therapy. Beyond iPSC technology, much effort is being made to generate neurons directly from dermal fibroblasts with neuron-specific transcription factors, which does not require making iPSCs as an intermediate cell type. SUMMARY: We summarize recent progress in using iPSCs for modeling the progress and treatment of neurodegenerative diseases and provide evidence for future perspectives in this field.


Asunto(s)
Enfermedades Neurodegenerativas/cirugía , Células Madre Pluripotentes/fisiología , Trasplante de Células Madre/métodos , Diferenciación Celular , Proliferación Celular , Humanos , Factor 4 Similar a Kruppel
9.
FASEB J ; 22(5): 1502-11, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18171693

RESUMEN

Complement-C1q TNF-related protein 1 (CTRP1), a member of the CTRP superfamily, is expressed at high levels in adipose tissues of obese Zucker diabetic fatty (fa/fa) rats, and CTRP1 expression is induced by proinflammatory cytokines, including TNF-alpha and IL-1beta. In the present study, we investigated stimulation of aldosterone production by CTRP1, since it was observed that CTRP1 was specifically expressed in the zona glomerulosa of the adrenal cortex, where aldosterone is produced. Increased aldosterone production by CTRP1 in cells of the human adrenal cortical cell line H295R was dose-dependent. Expression levels of aldosterone synthase CYP11B2 were examined to investigate the molecular mechanisms by which CTRP1 enhances the production of aldosterone. The expression of CYP11B2 was greatly increased by treatment with CTRP1, as was the expression of the transcription factors NGFIB and NURR1, which play critical roles in stimulation of CYP11B2 gene expression. It was also revealed that angiotensin II-induced aldosterone production is, at least in part, mediated by the stimulation of CTRP1 secretion, not by the increase of CTRP1 mRNA transcription. In addition, the levels of CTRP1 were significantly up-regulated in hypertensive patients' serum. As CTRP1 was highly expressed in obese subjects as well as up-regulated in hypertensive patients, CTRP1 may be a newly identified molecular link between obesity and hypertension.


Asunto(s)
Adipoquinas/fisiología , Aldosterona/biosíntesis , Proteínas/fisiología , Angiotensina II/farmacología , Animales , Calcio/metabolismo , Citocromo P-450 CYP11B2/genética , Proteínas de Unión al ADN/biosíntesis , Humanos , Hipertensión/sangre , Losartán/farmacología , Masculino , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/biosíntesis , Receptores de Esteroides/biosíntesis , Factores de Transcripción/biosíntesis , Células Tumorales Cultivadas
10.
J Cell Biol ; 218(8): 2564-2582, 2019 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-31308215

RESUMEN

A defining feature of embryonic stem cells (ESCs) is the ability to differentiate into all three germ layers. Pluripotency is maintained in part by a unique transcription network that maintains expression of pluripotency-specific transcription factors and represses developmental genes. While the mechanisms that establish this transcription network are well studied, little is known of the posttranscriptional surveillance pathways that degrade differentiation-related RNAs. We report that the surveillance pathway mediated by the RNA exosome nuclease complex represses ESC differentiation. Depletion of the exosome expedites differentiation of human ESCs into all three germ layers. LINE-1 retrotransposons and specific miRNAs, lncRNAs, and mRNAs that encode developmental regulators or affect their expression are all bound by the exosome and increase in level upon exosome depletion. The exosome restrains differentiation in part by degrading transcripts encoding FOXH1, a transcription factor crucial for mesendoderm formation. Our studies establish the exosome as a regulator of human ESC differentiation and reveal the importance of RNA decay in maintaining pluripotency.


Asunto(s)
Diferenciación Celular , Complejo Multienzimático de Ribonucleasas del Exosoma/metabolismo , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Reactivos de Enlaces Cruzados/química , Endodermo/embriología , Endodermo/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Células HeLa , Humanos , Elementos de Nucleótido Esparcido Largo/genética , Mesodermo/embriología , Mesodermo/metabolismo , MicroARNs/genética , Fenotipo , ARN/aislamiento & purificación , ARN Largo no Codificante/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Transcripción Genética , Transgenes
11.
Cell Stem Cell ; 24(3): 487-497.e7, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30799279

RESUMEN

Human brain organoid techniques have rapidly advanced to facilitate investigating human brain development and diseases. These efforts have largely focused on generating telencephalon due to its direct relevance in a variety of forebrain disorders. Despite its importance as a relay hub between cortex and peripheral tissues, the investigation of three-dimensional (3D) organoid models for the human thalamus has not been explored. Here, we describe a method to differentiate human embryonic stem cells (hESCs) to thalamic organoids (hThOs) that specifically recapitulate the development of thalamus. Single-cell RNA sequencing revealed a formation of distinct thalamic lineages, which diverge from telencephalic fate. Importantly, we developed a 3D system to create the reciprocal projections between thalamus and cortex by fusing the two distinct region-specific organoids representing the developing thalamus or cortex. Our study provides a platform for understanding human thalamic development and modeling circuit organizations and related disorders in the brain.


Asunto(s)
Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Células Madre Embrionarias Humanas/citología , Organoides/citología , Organoides/metabolismo , Tálamo/citología , Humanos , Modelos Biológicos
12.
Cancer Lett ; 261(2): 253-62, 2008 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-18164124

RESUMEN

Obesity is one of the potential risk factors in causing breast cancer. As a result, adipose tissue surrounding breast ductal cells may play an important role in the breast cancer development or progression. To identify the genes that are regulated by factors secreted from adipocytes in breast cancer cells, MDA-MB-231 cells were treated with the culture medium of adipocytes. Most of induced genes were related to immune function and wound healing, which share a common gene expression signature with cancer progression. In present study macrophage inhibitory cytokine 1 (MIC-1) gene was studied among the induced genes. It was found that both MIC-1 mRNA and protein were dramatically increased by the culture medium of adipocytes. Furthermore, proteinase K-treated adipocyte culture supernatants also induced MIC-1 expression. These findings indicate that proteins are not major MIC-1 inducing factors in adipocyte culture medium. Consequently, we examined the effect of free fatty acids such as palmitate and oleate on MIC-1 induction and found that palmitate markedly induced MIC-1 gene expression, whereas oleate did not. Adipocyte culture medium- and palmitate-induced MIC-1 gene expression was mediated by the activation of p38 MAPK, but not by the activation of JNK, ERK, and NF-kappaB pathway. In addition, adipocyte-CM-induced MIC-1 also increased invasiveness of MDA-MB-231 cells.


Asunto(s)
Adipocitos/citología , Neoplasias de la Mama/metabolismo , Citocinas/metabolismo , Animales , Western Blotting , Neoplasias de la Mama/patología , Movimiento Celular , Células Cultivadas , Medios de Cultivo/farmacología , Citocinas/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor 15 de Diferenciación de Crecimiento , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Análisis por Micromatrices , FN-kappa B/genética , FN-kappa B/metabolismo , Células 3T3 NIH , Invasividad Neoplásica , Palmitatos/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Cicatrización de Heridas , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
Proc Inst Mech Eng H ; 232(8): 779-786, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29993331

RESUMEN

To ensure accurate glucose readings when dispensing glucose oxidase enzyme solution from a jetting dispenser onto glucose test strips fabricated from an immersion gold-plated printed circuit board, every drop of the enzyme solution needs to have nearly the same weight and to be dispensed on the reaction zone of the test strips. Experimental results in this study show that the filling pressure in the fluid reservoir containing the glucose enzyme solution to dispense onto the test strips significantly affect the glucose test results. A filling pressure of 12 psi produces test strips with lower coefficient of variation and standard deviation than 10 and 14 psi. Proper filling pressure for dispensing glucose enzyme onto glucose test strips needs to be determined for any enzyme compound formulation.


Asunto(s)
Análisis Químico de la Sangre/instrumentación , Glucemia/análisis , Equipos y Suministros Eléctricos , Oro , Presión , Impresión , Tiras Reactivas , Inmersión
14.
Artículo en Inglés | MEDLINE | ID: mdl-30854156

RESUMEN

Three-dimensional (3D) brain organoid culture has become an essential tool for investigating human brain development and modeling neurological disorders during the past few years. Given the specific regionalization during brain development, it is important to produce distinct brain organoids that reproduce different brain regions and their interaction. The authors' laboratory recently established the platform to generate brain organoids resembling the medial ganglionic eminence (MGE), a specific brain region responsible for interneurogenesis, and found when fusing with organoid resembling the cortex, the fused organoids enabled modeling of interneuron migration in the brain. This unit describes four basic protocols that have been successfully applied in the authors' laboratory, covering the generation of embryonic body (EB) with neuroectodermal fate, the production of MGE organoids (hMGEOs) and cortical organoids (hCOs), and the fusion of the two organoids.


Asunto(s)
Encéfalo , Técnicas de Cultivo de Órganos , Organoides , Humanos , Interneuronas/citología , Eminencia Media/citología , Neurogénesis , Células Madre Pluripotentes
15.
Nat Commun ; 9(1): 2583, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29968706

RESUMEN

Embryonic stem cells (ESCs) maintain pluripotency through unique epigenetic states. When ESCs commit to a specific lineage, epigenetic changes in histones and DNA accompany the transition to specialized cell types. Investigating how epigenetic regulation controls lineage specification is critical in order to generate the required cell types for clinical applications. Uhrf1 is a widely known hemi-methylated DNA-binding protein, playing a role in DNA methylation through the recruitment of Dnmt1 and in heterochromatin formation alongside G9a, Trim28, and HDACs. Although Uhrf1 is not essential in ESC self-renewal, it remains elusive how Uhrf1 regulates cell specification. Here we report that Uhrf1 forms a complex with the active trithorax group, the Setd1a/COMPASS complex, to maintain bivalent histone marks, particularly those associated with neuroectoderm and mesoderm specification. Overall, our data demonstrate that Uhrf1 safeguards proper differentiation via bivalent histone modifications.


Asunto(s)
Reprogramación Celular/genética , Código de Histonas/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Proteínas Nucleares/metabolismo , Animales , Proteínas Potenciadoras de Unión a CCAAT , Técnicas de Reprogramación Celular , Quimera , Metilación de ADN/fisiología , Epigénesis Genética , Femenino , Fibroblastos , Técnicas de Inactivación de Genes , Células HEK293 , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/aislamiento & purificación , Histonas/metabolismo , Humanos , Masculino , Mesodermo/citología , Mesodermo/fisiología , Ratones , Células Madre Embrionarias de Ratones , Placa Neural/citología , Placa Neural/fisiología , Proteínas Nucleares/genética , Cultivo Primario de Células , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Ubiquitina-Proteína Ligasas
16.
Cell Stem Cell ; 21(3): 383-398.e7, 2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28757360

RESUMEN

Organoid techniques provide unique platforms to model brain development and neurological disorders. Whereas several methods for recapitulating corticogenesis have been described, a system modeling human medial ganglionic eminence (MGE) development, a critical ventral brain domain producing cortical interneurons and related lineages, has been lacking until recently. Here, we describe the generation of MGE and cortex-specific organoids from human pluripotent stem cells that recapitulate the development of MGE and cortex domains, respectively. Population and single-cell RNA sequencing (RNA-seq) profiling combined with bulk assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) analyses revealed transcriptional and chromatin accessibility dynamics and lineage relationships during MGE and cortical organoid development. Furthermore, MGE and cortical organoids generated physiologically functional neurons and neuronal networks. Finally, fusing region-specific organoids followed by live imaging enabled analysis of human interneuron migration and integration. Together, our study provides a platform for generating domain-specific brain organoids and modeling human interneuron migration and offers deeper insight into molecular dynamics during human brain development.


Asunto(s)
Encéfalo/embriología , Movimiento Celular , Interneuronas/citología , Modelos Biológicos , Organoides/citología , Células Madre Pluripotentes/citología , Encéfalo/citología , Diferenciación Celular , Linaje de la Célula , Corteza Cerebral/citología , Cromatina/metabolismo , Humanos , Interneuronas/metabolismo , Eminencia Media/citología , Células Madre Pluripotentes/metabolismo , Análisis de Secuencia de ARN , Transcriptoma/genética
17.
FEBS Lett ; 580(16): 3953-60, 2006 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-16806199

RESUMEN

CTRP1, a member of the CTRP superfamily, consists of an N-terminal signal peptide sequence followed by a variable region, a collagen repeat domain, and a C-terminal globular domain. CTRP1 is expressed at high levels in adipose tissues of LPS-stimulated Sprague-Dawley rats. The LPS-induced increase in CTRP1 gene expression was found to be mediated by TNF-alpha and IL-1beta. Also, a high level of expression of CTRP1 mRNA was observed in adipose tissues of Zucker diabetic fatty (fa/fa) rats, compared to Sprague-Dawley rats in the absence of LPS stimulation. These findings indicate that CTRP1 expression may be associated with a low-grade chronic inflammation status in adipose tissues.


Asunto(s)
Adipoquinas/metabolismo , Tejido Adiposo/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-1/farmacología , Proteínas/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Adipoquinas/química , Adipoquinas/genética , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Secuencia de Aminoácidos , Animales , Diferenciación Celular , Clonación Molecular , ADN Complementario/genética , Perfilación de la Expresión Génica , Humanos , Lipopolisacáridos/farmacología , Masculino , Ratones , Datos de Secuencia Molecular , Obesidad , Proteínas/química , Proteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Zucker , Alineación de Secuencia
18.
Cancer Res ; 63(15): 4648-55, 2003 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12907645

RESUMEN

In our search for genes associated with gastric cancer progression, we identified macrophage inhibitory cytokine-1 (MIC-1), a member of the transforming growth factor beta superfamily, as an overexpressed gene in gastric tumor tissues. Expression analysis of MIC-1 in gastric tumor tissues revealed a specific expression in gastric cancer cells, and this expression level was well correlated with invasive potential in various human gastric cancer cell lines. Stable transfection of MIC-1 into SNU-216, a human gastric cancer cell line, significantly increased its invasiveness. The overexpression of MIC-1 into SNU-216 cells significantly increased the activity of urokinase-type plasminogen activator (uPA), and the expressions of uPA and urokinase-type plasminogen activator receptor (uPAR). Similarly, the stimulation of gastric cancer cell lines with purified recombinant MIC-1 dose-dependently increased cell invasiveness, uPA activity, and uPA and uPAR expression. However, MIC-1 did not significantly suppress the proliferation of gastric cancer cell lines. We also found that the stimulation of human gastric cell lines with recombinant MIC-1 strongly induced activation of mitogen-activated protein kinase kinase-1/2 and extracellular signal-regulated kinase-1/2. Additional analysis revealed that PD98059, a selective inhibitor of mitogen-activated protein kinase kinase-1/2, suppressed not only gastric cancer cell invasiveness and uPA activity, but also the mRNA expressions of uPA and uPAR, as induced by recombinant MIC-1. Our results indicate that MIC-1 may contribute to the malignant progression of gastric cancer cells by inducing tumor cell invasion through the up-regulation of the uPA activation system via extracellular signal-regulated kinase-1/2-dependent pathway.


Asunto(s)
Citocinas/fisiología , Neoplasias Gástricas/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , División Celular/efectos de los fármacos , División Celular/fisiología , Citocinas/biosíntesis , Citocinas/genética , Citocinas/farmacología , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Factor 15 de Diferenciación de Crecimiento , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Proteínas Recombinantes/farmacología , Neoplasias Gástricas/enzimología , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Transfección , Factor de Crecimiento Transformador beta/farmacología , Células Tumorales Cultivadas , Regulación hacia Arriba
19.
Sci Rep ; 6: 35355, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27752090

RESUMEN

DNA methylation is an important epigenetic mark that regulates gene expression. Dnmt1 plays an important role in maintaining DNA methylation patterns on daughter DNA strands. Studies have shed light into the functional role of Dnmt1 regulation in the hematopoietic and epidermal systems. Here we show that Dnmt1 is required for myogenesis. Loss of Dnmt1 results in reduced expression of myogenic genes and defects in myogenic differentiation. We have utilized a conditional knockout mouse approach to examine the functional consequences of Dnmt1 depletion specifically in the developing muscle. These mice were born runted, with smaller body weights, and reduced ability to form myotubes in vitro. We show that expression of Id-1, a negative regulator of myogenesis, is enhanced in Dnmt1-deficient cultures, leading to enhanced transdifferentiation of myoblasts toward the osteogenic lineage. Thus, these studies demonstrate that Dnmt1 influences cellular identity and determines lineage fidelity.


Asunto(s)
Diferenciación Celular/genética , ADN (Citosina-5-)-Metiltransferasa 1/genética , Proteína 1 Inhibidora de la Diferenciación/genética , Desarrollo de Músculos/genética , Animales , Benzomorfanos , Linaje de la Célula/genética , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Metilación de ADN/genética , Regulación del Desarrollo de la Expresión Génica , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Ratones , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Células Madre/citología , Células Madre/metabolismo
20.
Stem Cell Reports ; 7(1): 43-54, 2016 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-27373925

RESUMEN

Reprogramming to pluripotency after overexpression of OCT4, SOX2, KLF4, and MYC is accompanied by global genomic and epigenomic changes. Histone modification and DNA methylation states in induced pluripotent stem cells (iPSCs) have been shown to be highly similar to embryonic stem cells (ESCs). However, epigenetic differences still exist between iPSCs and ESCs. In particular, aberrant DNA methylation states found in iPSCs are a major concern when using iPSCs in a clinical setting. Thus, it is critical to find factors that regulate DNA methylation states in reprogramming. Here, we found that the miR-29 family is an important epigenetic regulator during human somatic cell reprogramming. Our global DNA methylation and hydroxymethylation analysis shows that DNA demethylation is a major event mediated by miR-29a depletion during early reprogramming, and that iPSCs derived from miR-29a depletion are epigenetically closer to ESCs. Our findings uncover an important miRNA-based approach to generate clinically robust iPSCs.


Asunto(s)
Metilación de ADN/genética , Células Madre Embrionarias Humanas/citología , Células Madre Pluripotentes Inducidas/citología , MicroARNs/genética , Reprogramación Celular/genética , Epigénesis Genética/genética , Células Madre Embrionarias Humanas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , MicroARNs/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA