Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 34(3): 3570-3582, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31960495

RESUMEN

Biomedical research training has undergone considerable change over the past several years. At its core, the goal of graduate and postdoctoral training is to provide individuals with the skills and knowledge to become outstanding scientists and expand knowledge through the scientific method. Historically, graduate school training has focused on preparation for academic positions. Increasingly, however, a shift toward preparation for a wider range of career options has emerged. This is largely because most biomedical PhD graduates do not become Principal Investigators in academic laboratories. Here we describe an National Institutes of Health Common Fund program with the major goal of culture change for biomedical research training and training that prepares individuals for a broader expanse of careers in the biomedical research enterprise. These "Broadening Experiences in Scientific Training" (BEST) awards, issued in 2012 and 2013, provided support to institutions to develop innovative approaches to achieving these goals, as a complement to traditional training. Awardees were tasked with catalyzing change at their institutions and sharing best practices across the training community. Awardees were required to participate in a cross-site evaluation that assessed the impact of BEST activities on three main areas: (a) trainee confidence and knowledge to make career decisions, (b) influence of this added activity on time in training, and (c) ability of the institutions to sustain activities deemed to be beneficial. Here we present the fundamental approach to the BEST program and early evaluative data.


Asunto(s)
National Institutes of Health (U.S.)/organización & administración , Investigación Biomédica/educación , Humanos , Investigadores/educación , Estados Unidos
2.
Development ; 141(7): 1480-91, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24574008

RESUMEN

Neurovascular alignment is a common anatomical feature of organs, but the mechanisms leading to this arrangement are incompletely understood. Here, we show that vascular endothelial growth factor (VEGF) signaling profoundly affects both vascularization and innervation of the pancreatic islet. In mature islets, nerves are closely associated with capillaries, but the islet vascularization process during embryonic organogenesis significantly precedes islet innervation. Although a simple neuronal meshwork interconnects the developing islet clusters as they begin to form at E14.5, the substantial ingrowth of nerve fibers into islets occurs postnatally, when islet vascularization is already complete. Using genetic mouse models, we demonstrate that VEGF regulates islet innervation indirectly through its effects on intra-islet endothelial cells. Our data indicate that formation of a VEGF-directed, intra-islet vascular plexus is required for development of islet innervation, and that VEGF-induced islet hypervascularization leads to increased nerve fiber ingrowth. Transcriptome analysis of hypervascularized islets revealed an increased expression of extracellular matrix components and axon guidance molecules, with these transcripts being enriched in the islet-derived endothelial cell population. We propose a mechanism for coordinated neurovascular development within pancreatic islets, in which endocrine cell-derived VEGF directs the patterning of intra-islet capillaries during embryogenesis, forming a scaffold for the postnatal ingrowth of essential autonomic nerve fibers.


Asunto(s)
Vasos Sanguíneos/fisiología , Comunicación Celular/genética , Islotes Pancreáticos/irrigación sanguínea , Islotes Pancreáticos/inervación , Neovascularización Fisiológica/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Vasos Sanguíneos/embriología , Células Cultivadas , Embrión de Mamíferos , Endotelio Vascular/embriología , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiología , Femenino , Islotes Pancreáticos/embriología , Ratones , Ratones Transgénicos , Factor A de Crecimiento Endotelial Vascular/genética
3.
FASEB J ; 30(2): 507-14, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26432783

RESUMEN

Recent national reports and commentaries on the current status and needs of the U.S. biomedical research workforce have highlighted the limited career development opportunities for predoctoral and postdoctoral trainees in academia, yet little attention is paid to preparation for career pathways outside of the traditional faculty path. Recognizing this issue, in 2013, the U.S. National Institutes of Health (NIH) Common Fund issued a request for application titled "NIH Director's Biomedical Research Workforce Innovation Award: Broadening Experiences in Scientific Training (BEST)." These 5-yr 1-time grants, awarded to 17 single or partnering institutions, were designed to develop sustainable approaches to broaden graduate and postgraduate training, aimed at creating training programs that reflect the range of career options that trainees may ultimately pursue. These institutions have formed a consortium in order to work together to develop, evaluate, share, and disseminate best practices and challenges. This is a first report on the early experiences of the consortium and the scope of participating BEST programs. In this report, we describe the state of the U.S. biomedical workforce and development of the BEST award, variations of programmatic approaches to assist with program design without BEST funding, and novel approaches to engage faculty in career development programs. To test the effectiveness of these BEST programs, external evaluators will assess their outcomes not only over the 5 yr grant period but also for an additional 10 yr beyond award completion.


Asunto(s)
Disciplinas de las Ciencias Biológicas/educación , Educación de Postgrado/economía , National Institutes of Health (U.S.) , Investigación/educación , Educación de Postgrado/estadística & datos numéricos , Humanos , Estados Unidos
4.
Development ; 140(11): 2269-79, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23615280

RESUMEN

Understanding when and how multipotent progenitors segregate into diverse fates is a key question during embryonic development. The neural crest (NC) is an exemplary model system with which to investigate the dynamics of progenitor cell specification, as it generates a multitude of derivatives. Based on 'in ovo' lineage analysis, we previously suggested an early fate restriction of premigratory trunk NC to generate neural versus melanogenic fates, yet the timing of fate segregation and the underlying mechanisms remained unknown. Analysis of progenitors expressing a Foxd3 reporter reveals that prospective melanoblasts downregulate Foxd3 and have already segregated from neural lineages before emigration. When this downregulation is prevented, late-emigrating avian precursors fail to upregulate the melanogenic markers Mitf and MC/1 and the guidance receptor Ednrb2, generating instead glial cells that express P0 and Fabp. In this context, Foxd3 lies downstream of Snail2 and Sox9, constituting a minimal network upstream of Mitf and Ednrb2 to link melanogenic specification with migration. Consistent with the gain-of-function data in avians, loss of Foxd3 function in mouse NC results in ectopic melanogenesis in the dorsal tube and sensory ganglia. Altogether, Foxd3 is part of a dynamically expressed gene network that is necessary and sufficient to regulate fate decisions in premigratory NC. Their timely downregulation in the dorsal neural tube is thus necessary for the switch between neural and melanocytic phases of NC development.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Melanocitos/metabolismo , Tubo Neural/embriología , Tubo Neural/fisiología , Neuronas/metabolismo , Proteínas Represoras/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Embrión de Pollo , Melaninas/metabolismo , Ratones , Factor de Transcripción Asociado a Microftalmía/metabolismo , Microscopía Fluorescente , Receptor de Endotelina B/metabolismo , Factor de Transcripción SOX9/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Tiempo , Factores de Transcripción/metabolismo
5.
Proc Natl Acad Sci U S A ; 110(31): 12709-14, 2013 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-23858437

RESUMEN

Skin melanocytes arise from two sources: either directly from neural crest progenitors or indirectly from neural crest-derived Schwann cell precursors after colonization of peripheral nerves. The relationship between these two melanocyte populations and the factors controlling their specification remains poorly understood. Direct lineage tracing reveals that neural crest and Schwann cell progenitor-derived melanocytes are differentially restricted to the epaxial and hypaxial body domains, respectively. Furthermore, although both populations are initially part of the Foxd3 lineage, hypaxial melanocytes lose Foxd3 at late stages upon separation from the nerve, whereas we recently found that epaxial melanocytes segregate earlier from Foxd3-positive neural progenitors while still residing in the dorsal neural tube. Gain- and loss-of-function experiments in avians and mice, respectively, reveal that Foxd3 is both sufficient and necessary for regulating the balance between melanocyte and Schwann cell development. In addition, Foxd3 is also sufficient to regulate the switch between neuronal and glial fates in sensory ganglia. Together, we propose that differential fate acquisition of neural crest-derived cells depends on their progressive segregation from the Foxd3-positive lineage.


Asunto(s)
Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Factores de Transcripción Forkhead/metabolismo , Melanocitos/metabolismo , Cresta Neural/embriología , Células-Madre Neurales/metabolismo , Proteínas Represoras/metabolismo , Células de Schwann/metabolismo , Animales , Embrión de Pollo , Pollos , Ganglios Sensoriales/citología , Ganglios Sensoriales/embriología , Melanocitos/citología , Ratones , Cresta Neural/citología , Células-Madre Neurales/citología , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/citología , Neuronas/metabolismo , Células de Schwann/citología
6.
Development ; 139(1): 179-90, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22115757

RESUMEN

The spinal cord contains a diverse array of physiologically distinct interneuron cell types that subserve specialized roles in somatosensory perception and motor control. The mechanisms that generate these specialized interneuronal cell types from multipotential spinal progenitors are not known. In this study, we describe a temporally regulated transcriptional program that controls the differentiation of Renshaw cells (RCs), an anatomically and functionally discrete spinal interneuron subtype. We show that the selective activation of the Onecut transcription factors Oc1 and Oc2 during the first wave of V1 interneuron neurogenesis is a key step in the RC differentiation program. The development of RCs is additionally dependent on the forkhead transcription factor Foxd3, which is more broadly expressed in postmitotic V1 interneurons. Our demonstration that RCs are born, and activate Oc1 and Oc2 expression, in a narrow temporal window leads us to posit that neuronal diversity in the developing spinal cord is established by the composite actions of early spatial and temporal determinants.


Asunto(s)
Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Factor Nuclear 6 del Hepatocito/metabolismo , Proteínas de Homeodominio/metabolismo , Interneuronas/citología , Médula Espinal/citología , Médula Espinal/embriología , Factores de Transcripción/metabolismo , Animales , Bromodesoxiuridina , Cruzamientos Genéticos , Electrofisiología , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica , Interneuronas/metabolismo , Interneuronas/fisiología , Ratones , Factores de Tiempo
7.
J Mol Cell Cardiol ; 69: 88-96, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24508561

RESUMEN

Vascular smooth muscle cells (VSMCs) are derived from distinct embryonic origins. Vessels originating from differing smooth muscle cell populations have distinct vascular and pathological properties involving calcification, atherosclerosis, and structural defects such as aneurysm and coarctation. We hypothesized that domains within a single vessel, such as the aorta, vary in phenotype based on embryonic origin. Gene profiling and myographic analyses demonstrated that embryonic ascending and descending aortic domains exhibited distinct phenotypes. In vitro analyses demonstrated that VSMCs from each region were dissimilar in terms of cytoskeletal and migratory properties, and retention of different gene expression patterns. Using the same analysis, we found that these same two domains are indistinguishable in the adult vessel. Our data demonstrate that VSMCs from different embryonic origins are functionally distinct in the embryonic mouse, but converge to assume a common phenotype in the aorta of healthy adults. These findings have fundamental implications for aortic development, function and disease progression.


Asunto(s)
Aorta/embriología , Diferenciación Celular , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Variación Genética , Músculo Liso Vascular/embriología , Animales , Aorta/metabolismo , Biomarcadores/metabolismo , Western Blotting , Células Cultivadas , Femenino , Perfilación de la Expresión Génica , Masculino , Ratones , Músculo Liso Vascular/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Development ; 138(4): 641-52, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21228004

RESUMEN

Neural crest (NC) progenitors generate a wide array of cell types, yet molecules controlling NC multipotency and self-renewal and factors mediating cell-intrinsic distinctions between multipotent versus fate-restricted progenitors are poorly understood. Our earlier work demonstrated that Foxd3 is required for maintenance of NC progenitors in the embryo. Here, we show that Foxd3 mediates a fate restriction choice for multipotent NC progenitors with loss of Foxd3 biasing NC toward a mesenchymal fate. Neural derivatives of NC were lost in Foxd3 mutant mouse embryos, whereas abnormally fated NC-derived vascular smooth muscle cells were ectopically located in the aorta. Cranial NC defects were associated with precocious differentiation towards osteoblast and chondrocyte cell fates, and individual mutant NC from different anteroposterior regions underwent fate changes, losing neural and increasing myofibroblast potential. Our results demonstrate that neural potential can be separated from NC multipotency by the action of a single gene, and establish novel parallels between NC and other progenitor populations that depend on this functionally conserved stem cell protein to regulate self-renewal and multipotency.


Asunto(s)
Linaje de la Célula , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Factores de Transcripción Forkhead/metabolismo , Células Madre Multipotentes/metabolismo , Cresta Neural/metabolismo , Células-Madre Neurales/metabolismo , Proteínas Represoras/metabolismo , Animales , Diferenciación Celular , Embrión de Mamíferos/inervación , Factores de Transcripción Forkhead/deficiencia , Factores de Transcripción Forkhead/genética , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Noqueados , Células Madre Multipotentes/citología , Mutación , Cresta Neural/citología , Células-Madre Neurales/citología , Proteínas Represoras/deficiencia , Proteínas Represoras/genética
9.
Dev Biol ; 363(2): 373-87, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22266424

RESUMEN

The enteric nervous system (ENS) arises from the coordinated migration, expansion and differentiation of vagal and sacral neural crest progenitor cells. During development, vagal neural crest cells enter the foregut and migrate in a rostro-to-caudal direction, colonizing the entire gastrointestinal tract and generating the majority of the ENS. Sacral neural crest contributes to a subset of enteric ganglia in the hindgut, colonizing the colon in a caudal-to-rostral wave. During this process, enteric neural crest-derived progenitors (ENPs) self-renew and begin expressing markers of neural and glial lineages as they populate the intestine. Our earlier work demonstrated that the transcription factor Foxd3 is required early in neural crest-derived progenitors for self-renewal, multipotency and establishment of multiple neural crest-derived cells and structures including the ENS. Here, we describe Foxd3 expression within the fetal and postnatal intestine: Foxd3 was strongly expressed in ENPs as they colonize the gastrointestinal tract and was progressively restricted to enteric glial cells. Using a novel Ednrb-iCre transgene to delete Foxd3 after vagal neural crest cells migrate into the midgut, we demonstrated a late temporal requirement for Foxd3 during ENS development. Lineage labeling of Ednrb-iCre expressing cells in Foxd3 mutant embryos revealed a reduction of ENPs throughout the gut and loss of Ednrb-iCre lineage cells in the distal colon. Although mutant mice were viable, defects in patterning and distribution of ENPs were associated with reduced proliferation and severe reduction of glial cells derived from the Ednrb-iCre lineage. Analyses of ENS-lineage and differentiation in mutant embryos suggested activation of a compensatory population of Foxd3-positive ENPs that did not express the Ednrb-iCre transgene. Our findings highlight the crucial roles played by Foxd3 during ENS development including progenitor proliferation, neural patterning, and glial differentiation and may help delineate distinct molecular programs controlling vagal versus sacral neural crest development.


Asunto(s)
Sistema Nervioso Entérico/crecimiento & desarrollo , Factores de Transcripción Forkhead/metabolismo , Eliminación de Gen , Intestinos/inervación , Neurogénesis , Neuroglía/metabolismo , Proteínas Represoras/metabolismo , Células Madre/metabolismo , Animales , Movimiento Celular , Sistema Nervioso Entérico/embriología , Sistema Nervioso Entérico/metabolismo , Femenino , Factores de Transcripción Forkhead/genética , Intestinos/embriología , Intestinos/crecimiento & desarrollo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Cresta Neural/embriología , Proteínas Represoras/genética
10.
Pain ; 164(9): 1912-1926, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37326643

RESUMEN

ABSTRACT: Chronic pain affects more than 50 million Americans. Treatments remain inadequate, in large part, because the pathophysiological mechanisms underlying the development of chronic pain remain poorly understood. Pain biomarkers could potentially identify and measure biological pathways and phenotypical expressions that are altered by pain, provide insight into biological treatment targets, and help identify at-risk patients who might benefit from early intervention. Biomarkers are used to diagnose, track, and treat other diseases, but no validated clinical biomarkers exist yet for chronic pain. To address this problem, the National Institutes of Health Common Fund launched the Acute to Chronic Pain Signatures (A2CPS) program to evaluate candidate biomarkers, develop them into biosignatures, and discover novel biomarkers for chronification of pain after surgery. This article discusses candidate biomarkers identified by A2CPS for evaluation, including genomic, proteomic, metabolomic, lipidomic, neuroimaging, psychophysical, psychological, and behavioral measures. Acute to Chronic Pain Signatures will provide the most comprehensive investigation of biomarkers for the transition to chronic postsurgical pain undertaken to date. Data and analytic resources generatedby A2CPS will be shared with the scientific community in hopes that other investigators will extract valuable insights beyond A2CPS's initial findings. This article will review the identified biomarkers and rationale for including them, the current state of the science on biomarkers of the transition from acute to chronic pain, gaps in the literature, and how A2CPS will address these gaps.


Asunto(s)
Dolor Agudo , Dolor Crónico , Humanos , Proteómica , Dolor Postoperatorio/etiología , Dolor Agudo/complicaciones , Biomarcadores
11.
Dev Biol ; 349(2): 321-30, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21081123

RESUMEN

Interactions between cells from the ectoderm and mesoderm influence development of the endodermally-derived pancreas. While much is known about how mesoderm regulates pancreatic development, relatively little is understood about how and when the ectodermally-derived neural crest regulates pancreatic development and specifically, beta cell maturation. A previous study demonstrated that signals from the neural crest regulate beta cell proliferation and ultimately, beta cell mass. Here, we expand on that work to describe timing of neural crest arrival at the developing pancreatic bud and extend our knowledge of the non-cell autonomous role for neural crest derivatives in the process of beta cell maturation. We demonstrated that murine neural crest entered the pancreatic mesenchyme between the 26 and 27 somite stages (approximately 10.0 dpc) and became intermingled with pancreatic progenitors as the epithelium branched into the surrounding mesenchyme. Using a neural crest-specific deletion of the Forkhead transcription factor Foxd3, we ablated neural crest cells that migrate to the pancreatic primordium. Consistent with previous data, in the absence of Foxd3, and therefore the absence of neural crest cells, proliferation of insulin-expressing cells and insulin-positive area are increased. Analysis of endocrine cell gene expression in the absence of neural crest demonstrated that, although the number of insulin-expressing cells was increased, beta cell maturation was significantly impaired. Decreased MafA and Pdx1 expression illustrated the defect in beta cell maturation; we discovered that without neural crest, there was a reduction in the percentage of insulin-positive cells that co-expressed Glut2 and Pdx1 compared to controls. In addition, transmission electron microscopy analyses revealed decreased numbers of characteristic insulin granules and the presence of abnormal granules in insulin-expressing cells from mutant embryos. Together, these data demonstrate that the neural crest is a critical regulator of beta cell development on two levels: by negatively regulating beta cell proliferation and by promoting beta cell maturation.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Células Secretoras de Insulina/citología , Cresta Neural/embriología , Páncreas/embriología , Transducción de Señal/fisiología , Técnicas de Ablación , Factores de Edad , Animales , Cartilla de ADN/genética , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Eliminación de Gen , Técnicas Histológicas , Inmunohistoquímica , Células Secretoras de Insulina/fisiología , Células Secretoras de Insulina/ultraestructura , Ratones , Microscopía Electrónica de Transmisión , Páncreas/citología , Reacción en Cadena de la Polimerasa
12.
Genesis ; 49(1): 10-23, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21254333

RESUMEN

The transcription factors Foxd3 and Pax3 are important early regulators of neural crest (NC) progenitor cell properties. Homozygous mutations of Pax3 or a homozygous NC-specific deletion of Foxd3 cause marked defects in most NC derivatives, but neither loss of both Foxd3 alleles nor loss of one Pax3 allele alone greatly affects overall development of cardiac NC derivatives. In contrast, compound mutant embryos homozygous for a NC-specific Foxd3 mutation and heterozygous for Pax3 have fully penetrant persistent truncus arteriosus, severe thymus hypoplasia, and midgestation lethality. Foxd3; Pax3 compound mutant embryos have increased cell death in the neural folds and a drastic early reduction of NC cells, with an almost complete absence of NC caudal to the first pharyngeal arch. The genetic interaction between these genes implicates gene dosage-sensitive roles for Foxd3 and Pax3 in cardiac NC progenitors. Foxd3 and Pax3 act together to affect survival and maintenance of cardiac NC progenitors, and loss of these progenitors catastrophically affects key aspects of later cardiovascular development.


Asunto(s)
Factores de Transcripción Forkhead/genética , Cresta Neural/crecimiento & desarrollo , Factores de Transcripción Paired Box/genética , Proteínas Represoras/genética , Tronco Arterial Persistente/genética , Animales , Anomalías Craneofaciales/genética , Pérdida del Embrión/genética , Ratones , Ratones Mutantes , Cresta Neural/citología , Factor de Transcripción PAX3 , Eliminación de Secuencia , Células Madre/metabolismo , Células Madre/fisiología , Timo/anomalías
13.
Nat Cell Biol ; 6(11): 1082-93, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15517002

RESUMEN

A fundamental question in stem cell research is whether cultured multipotent adult stem cells represent endogenous multipotent precursor cells. Here we address this question, focusing on SKPs, a cultured adult stem cell from the dermis that generates both neural and mesodermal progeny. We show that SKPs derive from endogenous adult dermal precursors that exhibit properties similar to embryonic neural-crest stem cells. We demonstrate that these endogenous SKPs can first be isolated from skin during embryogenesis and that they persist into adulthood, with a niche in the papillae of hair and whisker follicles. Furthermore, lineage analysis indicates that both hair and whisker follicle dermal papillae contain neural-crest-derived cells, and that SKPs from the whisker pad are of neural-crest origin. We propose that SKPs represent an endogenous embryonic precursor cell that arises in peripheral tissues such as skin during development and maintains multipotency into adulthood.


Asunto(s)
Piel/citología , Células Madre/citología , Adulto , Animales , Western Blotting , Células Cultivadas , Embrión de Pollo , Embrión de Mamíferos/citología , Desarrollo Embrionario , Cabello/citología , Humanos , Inmunohistoquímica , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Dev Biol ; 333(1): 78-89, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19576203

RESUMEN

Identification of multipotent cardiac progenitors has provided important insights into the mechanisms of myocardial lineage specification, yet has done little to clarify the origin of the endocardium. Despite its essential role in heart development, characterization of the endocardial lineage has been limited by the lack of specific markers of this early vascular subpopulation. To distinguish endocardium from other vasculature, we generated an NFATc1-nuc-LacZ BAC transgenic mouse line capable of labeling this specific endothelial subpopulation at the earliest stages of cardiac development. To further characterize endocardiogenesis, embryonic stem cells (ESCs) derived from NFATc1-nuc-LacZ blastocysts were utilized to demonstrate that endocardial differentiation in vitro recapitulates the close temporal-spatial relationship observed between myocardium and endocardium seen in vivo. Endocardium is specified as a cardiac cell lineage, independent from other vascular populations, responding to BMP and Wnt signals that enhance cardiomyocyte differentiation. Furthermore, a population of Flk1+ cardiovascular progenitors, distinct from hemangioblast precursors, represents a mesodermal precursor of the endocardial endothelium, as well as other cardiovascular lineages. Taken together, these studies emphasize that the endocardium is a unique cardiac lineage and provides further evidence that endocardium and myocardium are derived from a common precursor.


Asunto(s)
Linaje de la Célula/fisiología , Células Madre Embrionarias/fisiología , Endocardio/embriología , Células Endoteliales/fisiología , Células Madre Multipotentes/fisiología , Miocitos Cardíacos/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Antígenos de Diferenciación/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Endocardio/citología , Endocardio/fisiología , Células Endoteliales/citología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Mesodermo/citología , Mesodermo/embriología , Ratones , Ratones Transgénicos , Células Madre Multipotentes/citología , Músculo Liso Vascular/citología , Músculo Liso Vascular/embriología , Miocitos Cardíacos/citología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/fisiología , Factores de Transcripción NFATC/genética
15.
Hum Mol Genet ; 17(23): 3740-60, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18799476

RESUMEN

Mutations in the aristaless-related homeobox (ARX) gene are associated with multiple neurologic disorders in humans. Studies in mice indicate Arx plays a role in neuronal progenitor proliferation and development of the cerebral cortex, thalamus, hippocampus, striatum, and olfactory bulbs. Specific defects associated with Arx loss of function include abnormal interneuron migration and subtype differentiation. How disruptions in ARX result in human disease and how loss of Arx in mice results in these phenotypes remains poorly understood. To gain insight into the biological functions of Arx, we performed a genome-wide expression screen to identify transcriptional changes within the subpallium in the absence of Arx. We have identified 84 genes whose expression was dysregulated in the absence of Arx. This population was enriched in genes involved in cell migration, axonal guidance, neurogenesis, and regulation of transcription and includes genes implicated in autism, epilepsy, and mental retardation; all features recognized in patients with ARX mutations. Additionally, we found Arx directly repressed three of the identified transcription factors: Lmo1, Ebf3 and Shox2. To further understand how the identified genes are involved in neural development, we used gene set enrichment algorithms to compare the Arx gene regulatory network (GRN) to the Dlx1/2 GRN and interneuron transcriptome. These analyses identified a subset of genes in the Arx GRN that are shared with that of the Dlx1/2 GRN and that are enriched in the interneuron transcriptome. These data indicate Arx plays multiple roles in forebrain development, both dependent and independent of Dlx1/2, and thus provides further insights into the understanding of the mechanisms underlying the pathology of mental retardation and epilepsy phenotypes resulting from ARX mutations.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Prosencéfalo/crecimiento & desarrollo , Factores de Transcripción/metabolismo , Transcripción Genética , Animales , Redes Reguladoras de Genes , Proteínas de Homeodominio/genética , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Prosencéfalo/metabolismo , Factores de Transcripción/genética
16.
Stem Cells ; 26(10): 2475-84, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18653770

RESUMEN

The Foxd3 forkhead transcription factor is required for maintaining pluripotent cells in the early mouse embryo and for the establishment of murine embryonic stem cell (ESC) lines. To begin to understand the role of Foxd3 in ESC maintenance, we derived ESC lines from blastocysts that carried two conditional Foxd3 alleles and a tamoxifen-inducible Cre transgene. Tamoxifen treatment produced a rapid and near complete loss of Foxd3 mRNA and protein. Foxd3-deficient ESCs maintained a normal proliferation rate but displayed increased apoptosis, and clonally dispersed ESCs showed a decreased ability to self-renew. Under either self-renewal or differentiation-promoting culture conditions we observed a strong, precocious differentiation of Foxd3 mutant ESCs along multiple lineages, including trophectoderm, endoderm, and mesendoderm. This profound alteration in biological behavior occurred in the face of continued expression of factors known to induce pluripotency, including Oct4, Sox2, and Nanog. We present a model for the role of Foxd3 in repressing differentiation, promoting self-renewal, and maintaining survival of mouse ESCs. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Células Madre Embrionarias/citología , Factores de Transcripción Forkhead/metabolismo , Células Madre Pluripotentes/citología , Proteínas Represoras/metabolismo , Animales , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , División Celular , Línea Celular , Linaje de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Factores de Transcripción Forkhead/genética , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Ratones , Modelos Biológicos , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Proteínas Represoras/genética , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología
17.
Stem Cells ; 25(12): 3101-10, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17823241

RESUMEN

A novel population of tissue-resident endothelial precursors (TEPs) was isolated from small blood vessels in dermal, adipose, and skeletal muscle of mouse based on their ability to be grown as spheres. Cellular and molecular analyses of these cells revealed that they were highly related regardless of the tissue of origin and distinct from embryonic neural stem cells. Notably, TEPs did not express hematopoietic markers, but they expressed numerous characteristics of angiogenic precursors and their differentiated progeny, such as CD34, Flk-1, Tie-1, CD31, and vascular endothelial cadherin (VE-cadherin). TEPs readily differentiated into endothelial cells in newly formed vascular networks following transplantation into regenerating skeletal muscle. Taken together, these experiments suggest that TEPs represent a novel class of endothelial precursors that are closely associated with small blood vessels in muscle, adipose, and dermal tissue. This finding is of particular interest since it could bring new insight in cancer angiogenesis and collateral blood vessels developed following ischemia. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Tejido Adiposo/irrigación sanguínea , Tejido Adiposo/crecimiento & desarrollo , Dermis/crecimiento & desarrollo , Endotelio/irrigación sanguínea , Endotelio/crecimiento & desarrollo , Músculos/fisiología , Neovascularización Fisiológica/fisiología , Células Madre/fisiología , Tejido Adiposo/citología , Animales , Animales Recién Nacidos , Diferenciación Celular/fisiología , Células Cultivadas , Dermis/citología , Endotelio/citología , Ratones , Ratones Endogámicos BALB C , Morfogénesis/fisiología , Músculos/citología , Esferoides Celulares/citología , Esferoides Celulares/fisiología , Células Madre/citología
18.
Gene Expr Patterns ; 6(8): 971-7, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16750430

RESUMEN

Multipotent progenitor cells self renew throughout an animal's lifetime and can differentiate to give rise to different cell types. Before we can fully understand the developmental potential of progenitor cells and control their differentiation both in vivo and in vitro as stem cells, identification and characterization of the genes that control stem cell fate must first be obtained. Foxd3, a member of the forkhead family of transcriptional regulators, is required for the maintenance of embryonic stem cells and trophoblast stem cells of the early mouse embryo. We describe here the expression of this protein in the developing pancreas. Foxd3 is expressed in most beta cells and infrequently in alpha and PP cells but is not expressed in somatostatin cells. The subcellular localization of Foxd3 varies with fat content in the diet; with a high fat diet the protein is found primarily in the cytoplasm while a low fat diet results in nuclear localization. Foxd3 is differentially localized in a rat model of diabetes: it is nuclear in ZDF rats but cytoplasmic in their lean counterparts. Foxd3 is nuclear in Lep(Ob/Ob) mice.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Páncreas/embriología , Páncreas/crecimiento & desarrollo , Páncreas/metabolismo , Proteínas Represoras/metabolismo , Animales , Linaje de la Célula , Núcleo Celular/metabolismo , Células Cultivadas , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/fisiología , Femenino , Humanos , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Ratones Transgénicos , Especificidad de Órganos , Ratas , Ratas Zucker , Distribución Tisular
19.
Adv Exp Med Biol ; 589: 206-12, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17076284

RESUMEN

Stem cells are defined by their ability to both self-renew and give rise to multiple lineages in vivo and/or in vitro. As discussed in other chapters in this volume, the embryonic neural crest is a multipotent tissue that gives rise to a plethora of differentiated cell types in the adult organism and is unique to vertebrate embryos. From the point of view of stem cell biology, the neural crest is an ideal source for multipotent adult stem cells. Significant advances have been made in the past few years isolating neural crest stem cell lines that can be maintained in vitro and can give rise to many neural crest derivatives either in vitro or when placed back into the context of an embryo. The initial work identifying these stem cells was carried out with premigratory neural crest from the embryonic neural tube. Later, neural crest stem cells were isolated from postmigratory neural crest, presumably more restricted in developmental potential. More recently it has been demonstrated that neural crest stem cell progenitors persist in the adult in at least two differentiated tissues, the enteric nervous system of the gut and the whisker follicles of the facial skin. In all cases, the properties of the stem cells derived reflect their tissue of origin and the potential of the progenitors becomes more restricted with age. In this chapter we will review this work and speculate on future possibilities with respect to combining our knowledge of neural crest gene function in the embryo and the manipulation of adult neural crest stem cells in vitro and eventually in vivo.


Asunto(s)
Cresta Neural/citología , Células Madre/citología , Animales , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Proliferación Celular , Biología Evolutiva/métodos , Sistema Nervioso Entérico/citología , Humanos , Neuronas/metabolismo
20.
Cell Stem Cell ; 18(1): 104-17, 2016 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-26748757

RESUMEN

Early development is governed by the ability of pluripotent cells to retain the full range of developmental potential and respond accurately to developmental cues. This property is achieved in large part by the temporal and contextual regulation of gene expression by enhancers. Here, we evaluated regulation of enhancer activity during differentiation of embryonic stem to epiblast cells and uncovered the forkhead transcription factor FOXD3 as a major regulator of the developmental potential of both pluripotent states. FOXD3 bound to distinct sites in the two cell types priming enhancers through a dual-functional mechanism. It recruited the SWI/SNF chromatin remodeling complex ATPase BRG1 to promote nucleosome removal while concurrently inhibiting maximal activation of the same enhancers by recruiting histone deacetylases1/2. Thus, FOXD3 prepares cognate genes for future maximal expression by establishing and simultaneously repressing enhancer activity. Through switching of target sites, FOXD3 modulates the developmental potential of pluripotent cells as they differentiate.


Asunto(s)
ADN Helicasas/metabolismo , Elementos de Facilitación Genéticos , Factores de Transcripción Forkhead/fisiología , Regulación de la Expresión Génica , Proteínas Nucleares/metabolismo , Células Madre Pluripotentes/citología , Proteínas Represoras/fisiología , Factores de Transcripción/metabolismo , Acetilación , Secuencias de Aminoácidos , Animales , Sitios de Unión , Linaje de la Célula , Factores de Transcripción Forkhead/genética , Perfilación de la Expresión Génica , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Ratones , Ratones Noqueados , Nucleosomas/metabolismo , Proteínas Represoras/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA