Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34504013

RESUMEN

Islet transplantation for type 1 diabetes treatment has been limited by the need for lifelong immunosuppression regimens. This challenge has prompted the development of macroencapsulation devices (MEDs) to immunoprotect the transplanted islets. While promising, conventional MEDs are faced with insufficient transport of oxygen, glucose, and insulin because of the reliance on passive diffusion. Hence, these devices are constrained to two-dimensional, wafer-like geometries with limited loading capacity to maintain cells within a distance of passive diffusion. We hypothesized that convective nutrient transport could extend the loading capacity while also promoting cell viability, rapid glucose equilibration, and the physiological levels of insulin secretion. Here, we showed that convective transport improves nutrient delivery throughout the device and affords a three-dimensional capsule geometry that encapsulates 9.7-fold-more cells than conventional MEDs. Transplantation of a convection-enhanced MED (ceMED) containing insulin-secreting ß cells into immunocompetent, hyperglycemic rats demonstrated a rapid, vascular-independent, and glucose-stimulated insulin response, resulting in early amelioration of hyperglycemia, improved glucose tolerance, and reduced fibrosis. Finally, to address potential translational barriers, we outlined future steps necessary to optimize the ceMED design for long-term efficacy and clinical utility.


Asunto(s)
Encapsulación Celular/métodos , Sistemas de Liberación de Medicamentos/métodos , Células Secretoras de Insulina/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Convección , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Sistemas de Liberación de Medicamentos/instrumentación , Insulina/metabolismo , Secreción de Insulina/efectos de los fármacos , Secreción de Insulina/fisiología , Células Secretoras de Insulina/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Trasplante de Islotes Pancreáticos/métodos , Masculino , Ratas
2.
Nature ; 518(7539): 355-359, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-25533951

RESUMEN

Models derived from human pluripotent stem cells that accurately recapitulate neural development in vitro and allow for the generation of specific neuronal subtypes are of major interest to the stem cell and biomedical community. Notch signalling, particularly through the Notch effector HES5, is a major pathway critical for the onset and maintenance of neural progenitor cells in the embryonic and adult nervous system. Here we report the transcriptional and epigenomic analysis of six consecutive neural progenitor cell stages derived from a HES5::eGFP reporter human embryonic stem cell line. Using this system, we aimed to model cell-fate decisions including specification, expansion and patterning during the ontogeny of cortical neural stem and progenitor cells. In order to dissect regulatory mechanisms that orchestrate the stage-specific differentiation process, we developed a computational framework to infer key regulators of each cell-state transition based on the progressive remodelling of the epigenetic landscape and then validated these through a pooled short hairpin RNA screen. We were also able to refine our previous observations on epigenetic priming at transcription factor binding sites and suggest here that they are mediated by combinations of core and stage-specific factors. Taken together, we demonstrate the utility of our system and outline a general framework, not limited to the context of the neural lineage, to dissect regulatory circuits of differentiation.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias/citología , Epigénesis Genética/genética , Epigenómica/métodos , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Sitios de Unión , Linaje de la Célula/genética , Células Madre Embrionarias/metabolismo , Humanos , ARN Interferente Pequeño/análisis , ARN Interferente Pequeño/genética , Reproducibilidad de los Resultados , Factores de Transcripción/metabolismo , Transcripción Genética/genética
3.
PLoS Biol ; 8(9)2010 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-20877475

RESUMEN

Hematopoietic progenitors undergo differentiation while navigating several cell division cycles, but it is unknown whether these two processes are coupled. We addressed this question by studying erythropoiesis in mouse fetal liver in vivo. We found that the initial upregulation of cell surface CD71 identifies developmentally matched erythroblasts that are tightly synchronized in S-phase. We show that DNA replication within this but not subsequent cycles is required for a differentiation switch comprising rapid and simultaneous committal transitions whose precise timing was previously unknown. These include the onset of erythropoietin dependence, activation of the erythroid master transcriptional regulator GATA-1, and a switch to an active chromatin conformation at the ß-globin locus. Specifically, S-phase progression is required for the formation of DNase I hypersensitive sites and for DNA demethylation at this locus. Mechanistically, we show that S-phase progression during this key committal step is dependent on downregulation of the cyclin-dependent kinase p57(KIP2) and in turn causes the downregulation of PU.1, an antagonist of GATA-1 function. These findings therefore highlight a novel role for a cyclin-dependent kinase inhibitor in differentiation, distinct to their known function in cell cycle exit. Furthermore, we show that a novel, mutual inhibition between PU.1 expression and S-phase progression provides a "synchromesh" mechanism that "locks" the erythroid differentiation program to the cell cycle clock, ensuring precise coordination of critical differentiation events.


Asunto(s)
Ciclo Celular , Eritropoyesis , Proteínas Proto-Oncogénicas/metabolismo , Fase S , Transactivadores/metabolismo , Animales , Antígenos CD/metabolismo , Cromatina/metabolismo , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Metilación de ADN , Replicación del ADN , Regulación hacia Abajo , Ratones , Receptores de Transferrina/metabolismo
4.
J Biol Chem ; 286(19): 16758-67, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21454573

RESUMEN

Survivin is a multifunctional protein with essential roles in cell division and inhibition of apoptosis, but the molecular underpinnings of its cytoprotective properties are poorly understood. Here we show that homozygous deletion of the aryl hydrocarbon receptor-interacting protein (AIP), a survivin-associated immunophilin, causes embryonic lethality in mice by embryonic day 13.5-14, increased apoptosis of Ter119(-)/CD71(-) early erythropoietic progenitors, and loss of survivin expression in its cytosolic and mitochondrial compartments in vivo. In import assays using recombinant proteins, AIP directly mediated the import of survivin to mitochondria, thus enabling its anti-apoptotic function, whereas a survivin 1-141 mutant that does not bind AIP was not imported to mitochondria and failed to inhibit apoptosis. AIP-directed mitochondrial import of survivin did not affect cell division, was independent of the organelle transmembrane potential, did not require the chaperone Heat Shock Protein 90 (Hsp90), and was inhibited by cytosolic factor(s) present in normal cells. shRNA knockdown of the mitochondrial import receptor Tom20 abolished mitochondrial import of survivin and sensitized tumor cells to apoptosis, whereas silencing of Tom70 had no effect. Therefore, an AIP-Tom20 recognition contributes to cell survival in development and cancer by mediating the mitochondrial import of survivin.


Asunto(s)
Apoptosis , Proteínas Inhibidoras de la Apoptosis/metabolismo , Péptidos y Proteínas de Señalización Intracelular/química , Mitocondrias/metabolismo , Proteínas Represoras/metabolismo , Animales , Transporte Biológico , Citosol/metabolismo , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Proteínas HSP90 de Choque Térmico/metabolismo , Células HeLa , Homocigoto , Humanos , Ratones , Ratones Noqueados , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Survivin , Factores de Tiempo
5.
Nat Commun ; 12(1): 7334, 2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34921133

RESUMEN

The erythroid terminal differentiation program couples sequential cell divisions with progressive reductions in cell size. The erythropoietin receptor (EpoR) is essential for erythroblast survival, but its other functions are not well characterized. Here we use Epor-/- mouse erythroblasts endowed with survival signaling to identify novel non-redundant EpoR functions. We find that, paradoxically, EpoR signaling increases red cell size while also increasing the number and speed of erythroblast cell cycles. EpoR-regulation of cell size is independent of established red cell size regulation by iron. High erythropoietin (Epo) increases red cell size in wild-type mice and in human volunteers. The increase in mean corpuscular volume (MCV) outlasts the duration of Epo treatment and is not the result of increased reticulocyte number. Our work shows that EpoR signaling alters the relationship between cycling and cell size. Further, diagnostic interpretations of increased MCV should now include high Epo levels and hypoxic stress.


Asunto(s)
Ciclo Celular , Tamaño de la Célula , Eritrocitos/citología , Eritrocitos/metabolismo , Eritropoyesis , Receptores de Eritropoyetina/metabolismo , Adulto , Animales , Antígenos CD/metabolismo , Antígenos CD4/metabolismo , Diferenciación Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Supervivencia Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Embrión de Mamíferos/metabolismo , Eritroblastos/citología , Eritroblastos/efectos de los fármacos , Eritroblastos/metabolismo , Eritropoyetina/administración & dosificación , Eritropoyetina/farmacología , Femenino , Feto/metabolismo , Voluntarios Sanos , Humanos , Hierro/metabolismo , Hígado/embriología , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Modelos Biológicos , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Transferrina/metabolismo , Reticulocitos/citología , Reticulocitos/efectos de los fármacos , Reticulocitos/metabolismo , Transducción de Señal , Proteína bcl-X/metabolismo
6.
PLoS Biol ; 5(10): e252, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17896863

RESUMEN

Tissue development is regulated by signaling networks that control developmental rate and determine ultimate tissue mass. Here we present a novel computational algorithm used to identify regulatory feedback and feedforward interactions between progenitors in developing erythroid tissue. The algorithm makes use of dynamic measurements of red cell progenitors between embryonic days 12 and 15 in the mouse. It selects for intercellular interactions that reproduce the erythroid developmental process and endow it with robustness to external perturbations. This analysis predicts that negative autoregulatory interactions arise between early erythroblasts of similar maturation stage. By studying embryos mutant for the death receptor FAS, or for its ligand, FASL, and by measuring the rate of FAS-mediated apoptosis in vivo, we show that FAS and FASL are pivotal negative regulators of fetal erythropoiesis, in the manner predicted by the computational model. We suggest that apoptosis in erythroid development mediates robust homeostasis regulating the number of red blood cells reaching maturity.


Asunto(s)
Eritrocitos/citología , Eritropoyesis/fisiología , Proteína Ligando Fas/metabolismo , Feto/metabolismo , Homeostasis/fisiología , Receptor fas/metabolismo , Algoritmos , Animales , Apoptosis/fisiología , Diferenciación Celular , Separación Celular , Eritrocitos/metabolismo , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Retroalimentación Fisiológica , Feto/embriología , Citometría de Flujo , Hígado/embriología , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción de Señal
7.
Sci Adv ; 6(47)2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33208361

RESUMEN

Advances in treating ß cell loss include islet replacement therapies or increasing cell proliferation rate in type 1 and type 2 diabetes, respectively. We propose developing multiple proliferation-inducing prodrugs that target high concentration of zinc ions in ß cells. Unfortunately, typical two-dimensional (2D) cell cultures do not mimic in vivo conditions, displaying a markedly lowered zinc content, while 3D culture systems are laborious and expensive. Therefore, we developed the Disque Platform (DP)-a high-fidelity culture system where stem cell-derived ß cells are reaggregated into thin, 3D discs within 2D 96-well plates. We validated the DP against standard 2D and 3D cultures and interrogated our zinc-activated prodrugs, which release their cargo upon zinc chelation-so preferentially in ß cells. Through developing a reliable screening platform that bridges the advantages of 2D and 3D culture systems, we identified an effective hit that exhibits 2.4-fold increase in ß cell proliferation compared to harmine.


Asunto(s)
Diabetes Mellitus Tipo 2 , Profármacos , Técnicas de Cultivo de Célula/métodos , Proliferación Celular , Humanos , Profármacos/farmacología , Zinc
8.
Elife ; 72018 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-30468428

RESUMEN

DNA methylation plays an essential role in mammalian genomes and expression of the responsible enzymes is tightly controlled. Deregulation of the de novo DNA methyltransferase DNMT3B is frequently observed across cancer types, yet little is known about its ectopic genomic targets. Here, we used an inducible transgenic mouse model to delineate rules for abnormal DNMT3B targeting, as well as the constraints of its activity across different cell types. Our results explain the preferential susceptibility of certain CpG islands to aberrant methylation and point to transcriptional state and the associated chromatin landscape as the strongest predictors. Although DNA methylation and H3K27me3 are usually non-overlapping at CpG islands, H3K27me3 can transiently co-occur with DNMT3B-induced DNA methylation. Our genome-wide data combined with ultra-deep locus-specific bisulfite sequencing suggest a distributive activity of ectopically expressed Dnmt3b that leads to discordant CpG island hypermethylation and provides new insights for interpreting the cancer methylome.


Asunto(s)
Islas de CpG , ADN (Citosina-5-)-Metiltransferasas/biosíntesis , Metilación de ADN , Expresión Génica , Proteínas Recombinantes/biosíntesis , Animales , ADN (Citosina-5-)-Metiltransferasas/genética , Células Madre Embrionarias/fisiología , Regulación de la Expresión Génica , Humanos , Ratones Transgénicos , Neoplasias/patología , Proteínas Recombinantes/genética , ADN Metiltransferasa 3B
9.
Nat Struct Mol Biol ; 25(4): 327-332, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29531288

RESUMEN

Cytosine methylation is widespread among organisms and essential for mammalian development. In line with early postulations of an epigenetic role in gene regulation, symmetric CpG methylation can be mitotically propagated over many generations with extraordinarily high fidelity. Here, we combine BrdU labeling and immunoprecipitation with genome-wide bisulfite sequencing to explore the inheritance of cytosine methylation onto newly replicated DNA in human cells. Globally, we observe a pronounced lag between the copying of genetic and epigenetic information in embryonic stem cells that is reconsolidated within hours to accomplish faithful mitotic transmission. Populations of arrested cells show a global reduction of lag-induced intermediate CpG methylation when compared to proliferating cells, whereas sites of transcription factor engagement appear cell-cycle invariant. Alternatively, the cancer cell line HCT116 preserves global epigenetic heterogeneity independently of cell-cycle arrest. Taken together, our data suggest that heterogeneous methylation largely reflects asynchronous proliferation, but is intrinsic to actively engaged cis-regulatory elements and cancer.


Asunto(s)
Citosina/química , Metilación de ADN , Ciclo Celular , Proliferación Celular , Islas de CpG , ADN/química , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Replicación del ADN , Células Madre Embrionarias/citología , Epigénesis Genética , Regulación de la Expresión Génica , Genoma Humano , Células HCT116 , Humanos , Masculino , Metilación , Mitosis , Neuronas Motoras/metabolismo , Neoplasias/genética , Análisis de Secuencia de ARN , Factores de Transcripción/metabolismo , ADN Metiltransferasa 3B
10.
Nat Struct Mol Biol ; 25(4): 355, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29581568

RESUMEN

Following online publication of this article, the Gene Expression Omnibus records corresponding to accession codes GSM2406773, MN-d6, and GSM2406772, MN-d14, listed in the data availability statement were deleted. The data are now available under accession codes GSM3039355, WGBS_hESC_WT_D6_R4 (MN day 6), and GSM3039351, WGBS_hESC_WT_D14_R4 (MN day 14), and the data availability statement has been updated with the new accession codes in the HTML and PDF versions of the article.

11.
Nat Genet ; 50(2): 250-258, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29358654

RESUMEN

Transcription factors (TFs) direct developmental transitions by binding to target DNA sequences, influencing gene expression and establishing complex gene-regultory networks. To systematically determine the molecular components that enable or constrain TF activity, we investigated the genomic occupancy of FOXA2, GATA4 and OCT4 in several cell types. Despite their classification as pioneer factors, all three TFs exhibit cell-type-specific binding, even when supraphysiologically and ectopically expressed. However, FOXA2 and GATA4 can be distinguished by low enrichment at loci that are highly occupied by these factors in alternative cell types. We find that expression of additional cofactors increases enrichment at a subset of these sites. Finally, FOXA2 occupancy and changes to DNA accessibility can occur in G1-arrested cells, but subsequent loss of DNA methylation requires DNA replication.


Asunto(s)
ADN/metabolismo , Epigénesis Genética/fisiología , Redes Reguladoras de Genes/fisiología , Factores de Transcripción/metabolismo , Células A549 , Sitios de Unión/genética , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/genética , Células Cultivadas , Biología Computacional , ADN/genética , Epistasis Genética/fisiología , Factor de Transcripción GATA4/metabolismo , Regulación de la Expresión Génica , Genes de Cambio , Células HEK293 , Células Hep G2 , Factor Nuclear 3-beta del Hepatocito/metabolismo , Humanos , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Unión Proteica
12.
Cell Stem Cell ; 22(4): 559-574.e9, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29551301

RESUMEN

The somatic DNA methylation (DNAme) landscape is established early in development but remains highly dynamic within focal regions that overlap with gene regulatory elements. The significance of these dynamic changes, particularly in the central nervous system, remains unresolved. Here, we utilize a powerful human embryonic stem cell differentiation model for the generation of motor neurons (MNs) in combination with genetic mutations in the de novo DNAme machinery. We quantitatively dissect the role of DNAme in directing somatic cell fate with high-resolution genome-wide bisulfite-, bulk-, and single-cell-RNA sequencing. We find defects in neuralization and MN differentiation in DNMT3A knockouts (KO) that can be rescued by the targeting of DNAme to key developmental loci using catalytically inactive dCas9. We also find decreased dendritic arborization and altered electrophysiological properties in DNMT3A KO MNs. Our work provides a list of DNMT3A-regulated targets and a mechanistic link between de novo DNAme, cellular differentiation, and human MN function.


Asunto(s)
Diferenciación Celular , Metilación de ADN , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Biocatálisis , Diferenciación Celular/genética , ADN (Citosina-5-)-Metiltransferasas/deficiencia , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , ADN Metiltransferasa 3A , Humanos
13.
Sci Adv ; 3(5): e1700298, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28560351

RESUMEN

Cell cycle regulators are increasingly implicated in cell fate decisions, such as the acquisition or loss of pluripotency and self-renewal potential. The cell cycle mechanisms that regulate these cell fate decisions are largely unknown. We studied an S phase-dependent cell fate switch, in which murine early erythroid progenitors transition in vivo from a self-renewal state into a phase of active erythroid gene transcription and concurrent maturational cell divisions. We found that progenitors are dependent on p57KIP2-mediated slowing of replication forks for self-renewal, a novel function for cyclin-dependent kinase inhibitors. The switch to differentiation entails rapid down-regulation of p57KIP2 with a consequent global increase in replication fork speed and an abruptly shorter S phase. Our work suggests that cell cycles with specialized global DNA replication dynamics are integral to the maintenance of specific cell states and to cell fate decisions.


Asunto(s)
Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Replicación del ADN/fisiología , Células Eritroides/metabolismo , Fase S/fisiología , Animales , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Células Eritroides/citología , Femenino , Ratones , Ratones Mutantes , Transcripción Genética/fisiología
14.
Mol Genet Genomic Med ; 5(3): 261-268, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28546996

RESUMEN

BACKGROUND: Campomelic dysplasia (CD) is a semilethal developmental disorder caused by mutations in and around SOX9. CD is characterized by multiple skeletal malformations including bending (campomelia) of long bones. Surviving patients frequently have the acampomelic form of CD (ACD). METHODS: This is a single case report on a patient with clinical and radiological features of ACD who has no mutation in the SOX9 protein-coding sequence nor a translocation with breakpoint in the SOX9 regulatory domain. We include functional studies of the novel mutant protein in vitro and in cultured cells. RESULTS: The patient was found to have a de novo heterozygous mutation c.-185G>A in the SOX9 5'UTR. The mutation creates an upstream translation start codon, uAUG, with a much better fit of its flanking sequence to the Kozak consensus than the wild-type AUG. By in vitro transcription-translation and transient transfection into COS-7 cells, we show that the uAUG leads to translation of a short peptide from a reading frame that terminates just after the wild-type AUG start codon. This results in reduced translation of the wild-type protein, compatible with the milder phenotype of the patient. CONCLUSION: Findings support the notion that more mildly affected, surviving CD/ACD patients carry mutant SOX9 alleles with residual expression of SOX9 wild-type protein. Although rarely described in human genetic disease and for the first time here for CD, mutations creating upstream AUG codons may be more common than generally assumed.

15.
Stem Cell Reports ; 7(5): 983-997, 2016 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-27773702

RESUMEN

Following injury, adult skeletal muscle undergoes a well-coordinated sequence of molecular and physiological events to promote repair and regeneration. However, a thorough understanding of the in vivo epigenomic and transcriptional mechanisms that control these reparative events is lacking. To address this, we monitored the in vivo dynamics of three histone modifications and coding and noncoding RNA expression throughout the regenerative process in a mouse model of traumatic muscle injury. We first illustrate how both coding and noncoding RNAs in tissues and sorted satellite cells are modified and regulated during various stages after trauma. Next, we use chromatin immunoprecipitation followed by sequencing to evaluate the chromatin state of cis-regulatory elements (promoters and enhancers) and view how these elements evolve and influence various muscle repair and regeneration transcriptional programs. These results provide a comprehensive view of the central factors that regulate muscle regeneration and underscore the multiple levels through which both transcriptional and epigenetic patterns are regulated to enact appropriate repair and regeneration.


Asunto(s)
Ensamble y Desensamble de Cromatina , Músculo Esquelético/lesiones , Músculo Esquelético/fisiología , Regeneración/genética , Transcripción Genética , Animales , Masculino , Ratones , MicroARNs/genética , ARN Mensajero/genética , Cicatrización de Heridas/genética
16.
Am J Cardiol ; 95(1): 113-6, 2005 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-15619406

RESUMEN

The purpose of this study was to report the angiographic findings of the first human evaluation of the everolimus-eluting stent (EES) for the treatment of noncomplex coronary lesions. Forty-two patients with de novo coronary lesions (2.75 to 4.00 mm vessels; lesion length, <18 mm) were prospectively randomized in a 2:1 ratio to receive either the EES (n = 27) or a metallic stent (n = 15). Baseline clinical and angiographic characteristics were similar among both groups. At 6-month follow-up, EES had a lower in-stent late lumen loss (0.10 +/- 0.22 vs 0.85 +/- 0.32 mm, p <0.0001) and in-segment diameter stenoses (20.7 +/- 12.3% vs 37.0 +/- 15.8%, p = 0.002). There was no in-stent restenosis with EES; however, 1 focal distal edge restenosis was present. There was 1 in-stent and 1 in-segment (proximal edge) restenosis in the metallic stent group. There was no stent thrombosis or aneurysm formation at follow-up in either group.


Asunto(s)
Angiografía Coronaria , Estenosis Coronaria/diagnóstico por imagen , Estenosis Coronaria/terapia , Sistemas de Liberación de Medicamentos , Sirolimus/análogos & derivados , Sirolimus/administración & dosificación , Stents , Anciano , Terapia Combinada , Everolimus , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos
17.
Nat Biotechnol ; 33(11): 1182-92, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26501952

RESUMEN

Research on human pluripotent stem cells has been hampered by the lack of a standardized, quantitative, scalable assay of pluripotency. We previously described an assay called ScoreCard that used gene expression signatures to quantify differentiation efficiency. Here we report an improved version of the assay based on qPCR that enables faster, more quantitative assessment of functional pluripotency. We provide an in-depth characterization of the revised signature panel (commercially available as the TaqMan hPSC Scorecard Assay) through embryoid body and directed differentiation experiments as well as a detailed comparison to the teratoma assay. We further show that the improved ScoreCard enables a wider range of applications, such as screening of small molecules, genetic perturbations and assessment of culture conditions. Our approach can be extended beyond stem cell applications to characterize and assess the utility of other cell types and lineages.


Asunto(s)
Diferenciación Celular/genética , Biología Computacional/métodos , Cuerpos Embrioides/fisiología , Células Madre Pluripotentes/fisiología , Reacción en Cadena de la Polimerasa/métodos , Animales , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Humanos , Ratones , Neoplasias Experimentales/patología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Teratoma/patología
18.
Sci Rep ; 5: 13885, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26381351

RESUMEN

Traumatic lower-limb musculoskeletal injuries are pervasive amongst athletes and the military and typically an individual returns to activity prior to fully healing, increasing a predisposition for additional injuries and chronic pain. Monitoring healing progression after a musculoskeletal injury typically involves different types of imaging but these approaches suffer from several disadvantages. Isolating and profiling transcripts from the injured site would abrogate these shortcomings and provide enumerative insights into the regenerative potential of an individual's muscle after injury. In this study, a traumatic injury was administered to a mouse model and healing progression was examined from 3 hours to 1 month using high-throughput RNA-Sequencing (RNA-Seq). Comprehensive dissection of the genome-wide datasets revealed the injured site to be a dynamic, heterogeneous environment composed of multiple cell types and thousands of genes undergoing significant expression changes in highly regulated networks. Four independent approaches were used to determine the set of genes, isoforms, and genetic pathways most characteristic of different time points post-injury and two novel approaches were developed to classify injured tissues at different time points. These results highlight the possibility to quantitatively track healing progression in situ via transcript profiling using high- throughput sequencing.


Asunto(s)
Perfilación de la Expresión Génica , Extremidad Inferior , Músculo Esquelético/lesiones , Músculo Esquelético/metabolismo , Transcriptoma , Cicatrización de Heridas/genética , Animales , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Biología Computacional/métodos , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Masculino , Ratones , Anotación de Secuencia Molecular , Músculo Esquelético/patología , Fenotipo , Receptores Notch/metabolismo , Reproducibilidad de los Resultados , Transducción de Señal , Máquina de Vectores de Soporte , Proteínas Wnt/metabolismo
19.
Nat Biotechnol ; 33(11): 1173-81, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26501951

RESUMEN

The equivalence of human induced pluripotent stem cells (hiPSCs) and human embryonic stem cells (hESCs) remains controversial. Here we use genetically matched hESC and hiPSC lines to assess the contribution of cellular origin (hESC vs. hiPSC), the Sendai virus (SeV) reprogramming method and genetic background to transcriptional and DNA methylation patterns while controlling for cell line clonality and sex. We find that transcriptional and epigenetic variation originating from genetic background dominates over variation due to cellular origin or SeV infection. Moreover, the 49 differentially expressed genes we detect between genetically matched hESCs and hiPSCs neither predict functional outcome nor distinguish an independently derived, larger set of unmatched hESC and hiPSC lines. We conclude that hESCs and hiPSCs are molecularly and functionally equivalent and cannot be distinguished by a consistent gene expression signature. Our data further imply that genetic background variation is a major confounding factor for transcriptional and epigenetic comparisons of pluripotent cell lines, explaining some of the previously observed differences between genetically unmatched hESCs and hiPSCs.


Asunto(s)
Células Madre Embrionarias/metabolismo , Genes/genética , Células Madre Pluripotentes Inducidas/metabolismo , Análisis por Conglomerados , Biología Computacional , Células Madre Embrionarias/fisiología , Perfilación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Masculino
20.
Nat Genet ; 47(5): 469-78, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25822089

RESUMEN

DNA methylation is a key epigenetic modification involved in regulating gene expression and maintaining genomic integrity. Here we inactivated all three catalytically active DNA methyltransferases (DNMTs) in human embryonic stem cells (ESCs) using CRISPR/Cas9 genome editing to further investigate the roles and genomic targets of these enzymes. Disruption of DNMT3A or DNMT3B individually as well as of both enzymes in tandem results in viable, pluripotent cell lines with distinct effects on the DNA methylation landscape, as assessed by whole-genome bisulfite sequencing. Surprisingly, in contrast to findings in mouse, deletion of DNMT1 resulted in rapid cell death in human ESCs. To overcome this immediate lethality, we generated a doxycycline-responsive tTA-DNMT1* rescue line and readily obtained homozygous DNMT1-mutant lines. However, doxycycline-mediated repression of exogenous DNMT1* initiates rapid, global loss of DNA methylation, followed by extensive cell death. Our data provide a comprehensive characterization of DNMT-mutant ESCs, including single-base genome-wide maps of the targets of these enzymes.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , Células Madre Embrionarias/enzimología , Animales , Apoptosis , Secuencia de Bases , Dominio Catalítico , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Técnicas de Cocultivo , Islas de CpG , ADN (Citosina-5-)-Metiltransferasa 1 , ADN Metiltransferasa 3A , Células Madre Embrionarias/fisiología , Epigénesis Genética , Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Ratones , ADN Metiltransferasa 3B
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA