Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Pain ; 164(9): 1912-1926, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37326643

RESUMO

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.


Assuntos
Dor Aguda , Dor Crônica , Humanos , Proteômica , Dor Pós-Operatória/etiologia , Dor Aguda/complicações , Biomarcadores
2.
FASEB J ; 34(3): 3570-3582, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31960495

RESUMO

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.


Assuntos
National Institutes of Health (U.S.)/organização & administração , Pesquisa Biomédica/educação , Humanos , Pesquisadores/educação , Estados Unidos
4.
Cell Stem Cell ; 18(1): 104-17, 2016 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-26748757

RESUMO

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.


Assuntos
DNA Helicases/metabolismo , Elementos Facilitadores Genéticos , Fatores de Transcrição Forkhead/fisiologia , Regulação da Expressão Gênica , Proteínas Nucleares/metabolismo , Células-Tronco Pluripotentes/citologia , Proteínas Repressoras/fisiologia , Fatores de Transcrição/metabolismo , Acetilação , Motivos de Aminoácidos , Animais , Sítios de Ligação , Linhagem da Célula , Fatores de Transcrição Forkhead/genética , Perfilação da Expressão Gênica , Histona Desacetilases/metabolismo , Histonas/metabolismo , Camundongos , Camundongos Knockout , Nucleossomos/metabolismo , Proteínas Repressoras/genética
5.
FASEB J ; 30(2): 507-14, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26432783

RESUMO

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.


Assuntos
Disciplinas das Ciências Biológicas/educação , Educação de Pós-Graduação/economia , National Institutes of Health (U.S.) , Pesquisa/educação , Educação de Pós-Graduação/estatística & dados numéricos , Humanos , Estados Unidos
6.
J Extracell Vesicles ; 4: 27493, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26320938

RESUMO

The Extracellular RNA (exRNA) Communication Consortium, funded as an initiative of the NIH Common Fund, represents a consortium of investigators assembled to address the critical issues in the exRNA research arena. The overarching goal is to generate a multi-component community resource for sharing fundamental scientific discoveries, protocols, and innovative tools and technologies. The key initiatives include (a) generating a reference catalogue of exRNAs present in body fluids of normal healthy individuals that would facilitate disease diagnosis and therapies, (b) defining the fundamental principles of exRNA biogenesis, distribution, uptake, and function, as well as development of molecular tools, technologies, and imaging modalities to enable these studies,

7.
Development ; 141(7): 1480-91, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24574008

RESUMO

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.


Assuntos
Vasos Sanguíneos/fisiologia , Comunicação Celular/genética , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/inervação , Neovascularização Fisiológica/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Vasos Sanguíneos/embriologia , Células Cultivadas , Embrião de Mamíferos , Endotélio Vascular/embriologia , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiologia , Feminino , Ilhotas Pancreáticas/embriologia , Camundongos , Camundongos Transgênicos , Fator A de Crescimento do Endotélio Vascular/genética
8.
J Mol Cell Cardiol ; 69: 88-96, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24508561

RESUMO

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.


Assuntos
Aorta/embriologia , Diferenciação Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Variação Genética , Músculo Liso Vascular/embriologia , Animais , Aorta/metabolismo , Biomarcadores/metabolismo , Western Blotting , Células Cultivadas , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos , Músculo Liso Vascular/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Stem Cell Res ; 12(1): 233-40, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24270162

RESUMO

Understanding gene regulatory networks controlling properties of pluripotent stem cells will facilitate development of stem cell-based therapies. The transcription factor Foxd3 is critical for maintenance of self-renewal, survival, and pluripotency in murine embryonic stem cells (ESCs). Using a conditional deletion of Foxd3 followed by gene expression analyses, we demonstrate that genes required for several developmental processes including embryonic organ development, epithelium development, and epithelial differentiation were misregulated in the absence of Foxd3. Additionally, we identified 6 novel targets of Foxd3 (Sox4, Safb, Sox15, Fosb, Pmaip1 and Smarcd3). Finally, we present data suggesting that Foxd3 functions upstream of genes required for skeletal muscle development. Together, this work provides further evidence that Foxd3 is a critical regulator of murine development through the regulation of lineage specific differentiation.


Assuntos
Células-Tronco Embrionárias/citologia , Fatores de Transcrição Forkhead/metabolismo , Proteínas Repressoras/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Imunoprecipitação da Cromatina , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Análise por Conglomerados , Células-Tronco Embrionárias/metabolismo , Fatores de Transcrição Forkhead/deficiência , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Ligação à Região de Interação com a Matriz/genética , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Camundongos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Fatores de Transcrição SOX/genética , Fatores de Transcrição SOX/metabolismo , Fatores de Transcrição SOXC/genética , Fatores de Transcrição SOXC/metabolismo
10.
Methods Mol Biol ; 1032: 1-17, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23943440

RESUMO

The generation of transgenic mouse models has been a powerful technique for several decades and is still widely used. There have been many manuals and general reviews of this technology. This chapter is designed to be a "how-to" resource with detailed specifics.


Assuntos
Modelos Animais de Doenças , Camundongos Transgênicos , Animais , Humanos , Camundongos
11.
Proc Natl Acad Sci U S A ; 110(31): 12709-14, 2013 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-23858437

RESUMO

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.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Melanócitos/metabolismo , Crista Neural/embriologia , Células-Tronco Neurais/metabolismo , Proteínas Repressoras/metabolismo , Células de Schwann/metabolismo , Animais , Embrião de Galinha , Galinhas , Gânglios Sensitivos/citologia , Gânglios Sensitivos/embriologia , Melanócitos/citologia , Camundongos , Crista Neural/citologia , Células-Tronco Neurais/citologia , Neuroglia/citologia , Neuroglia/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Células de Schwann/citologia
12.
Development ; 140(11): 2269-79, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23615280

RESUMO

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.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Melanócitos/metabolismo , Tubo Neural/embriologia , Tubo Neural/fisiologia , Neurônios/metabolismo , Proteínas Repressoras/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula , Embrião de Galinha , Melaninas/metabolismo , Camundongos , Fator de Transcrição Associado à Microftalmia/metabolismo , Microscopia de Fluorescência , Receptor de Endotelina B/metabolismo , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição da Família Snail , Fatores de Tempo , Fatores de Transcrição/metabolismo
13.
Stem Cells Dev ; 22(12): 1779-88, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23316968

RESUMO

Skin-derived precursors (SKPs) are an attractive stem cell model for cell-based therapies. SKPs can be readily generated from embryonic and adult mice and adult humans, exhibit a high degree of multipotency, and have the potential to serve as a patient autologous stem cell. The advancement of these cells toward therapeutic use depends on the ability to control precisely the self-renewal and differentiation of SKPs. Here we show that two well-known stem cell factors, Foxd3 and Sox2, are critical regulators of the stem cell properties of SKPs. Deletion of Foxd3 completely abolishes the sphere-forming potential of these cells. In the absence of Sox2, SKP spheres can be formed, but with reduced size and frequency. Our results provide entry points into the gene regulatory networks dictating SKP behavior, and pave the way for future studies on a therapeutically relevant stem cell.


Assuntos
Células-Tronco Embrionárias/metabolismo , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Multipotentes/metabolismo , Proteínas Repressoras/genética , Fatores de Transcrição SOXB1/genética , Pele/metabolismo , Transcrição Gênica , Animais , Diferenciação Celular , Embrião de Mamíferos , Células-Tronco Embrionárias/citologia , Fatores de Transcrição Forkhead/deficiência , Camundongos , Camundongos Transgênicos , Células-Tronco Multipotentes/citologia , Cultura Primária de Células , Proteínas Repressoras/deficiência , Fatores de Transcrição SOXB1/deficiência , Transdução de Sinais , Pele/citologia , Pele/embriologia
14.
J Vis Exp ; (64): e4134, 2012 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-22688801

RESUMO

The embryonic neural crest (NC) is a multipotent progenitor population that originates at the dorsal aspect of the neural tube, undergoes an epithelial to mesenchymal transition (EMT) and migrates throughout the embryo, giving rise to diverse cell types. NC also has the unique ability to influence the differentiation and maturation of target organs. When explanted in vitro, NC progenitors undergo self-renewal, migrate and differentiate into a variety of tissue types including neurons, glia, smooth muscle cells, cartilage and bone. NC multipotency was first described from explants of the avian neural tube. In vitro isolation of NC cells facilitates the study of NC dynamics including proliferation, migration, and multipotency. Further work in the avian and rat systems demonstrated that explanted NC cells retain their NC potential when transplanted back into the embryo. Because these inherent cellular properties are preserved in explanted NC progenitors, the neural tube explant assay provides an attractive option for studying the NC in vitro. To attain a better understanding of the mammalian NC, many methods have been employed to isolate NC populations. NC-derived progenitors can be cultured from post-migratory locations in both the embryo and adult to study the dynamics of post-migratory NC progenitors, however isolation of NC progenitors as they emigrate from the neural tube provides optimal preservation of NC cell potential and migratory properties. Some protocols employ fluorescence activated cell sorting (FACS) to isolate a NC population enriched for particular progenitors. However, when starting with early stage embryos, cell numbers adequate for analyses are difficult to obtain with FACS, complicating the isolation of early NC populations from individual embryos. Here, we describe an approach that does not rely on FACS and results in an approximately 96% pure NC population based on a Wnt1-Cre activated lineage reporter. The method presented here is adapted from protocols optimized for the culture of rat NC. The advantages of this protocol compared to previous methods are that 1) the cells are not grown on a feeder layer, 2) FACS is not required to obtain a relatively pure NC population, 3) premigratory NC cells are isolated and 4) results are easily quantified. Furthermore, this protocol can be used for isolation of NC from any mutant mouse model, facilitating the study of NC characteristics with different genetic manipulations. The limitation of this approach is that the NC is removed from the context of the embryo, which is known to influence the survival, migration and differentiation of the NC.


Assuntos
Separação Celular/métodos , Crista Neural/citologia , Tubo Neural/citologia , Animais , Embrião de Mamíferos/citologia , Humanos , Camundongos , Proteína Wnt1/genética
15.
Dev Biol ; 363(2): 373-87, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22266424

RESUMO

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.


Assuntos
Sistema Nervoso Entérico/crescimento & desenvolvimento , Fatores de Transcrição Forkhead/metabolismo , Deleção de Genes , Intestinos/inervação , Neurogênese , Neuroglia/metabolismo , Proteínas Repressoras/metabolismo , Células-Tronco/metabolismo , Animais , Movimento Celular , Sistema Nervoso Entérico/embriologia , Sistema Nervoso Entérico/metabolismo , Feminino , Fatores de Transcrição Forkhead/genética , Intestinos/embriologia , Intestinos/crescimento & desenvolvimento , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Crista Neural/embriologia , Proteínas Repressoras/genética
16.
Development ; 139(1): 179-90, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22115757

RESUMO

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.


Assuntos
Diferenciação Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fator 6 Nuclear de Hepatócito/metabolismo , Proteínas de Homeodomínio/metabolismo , Interneurônios/citologia , Medula Espinal/citologia , Medula Espinal/embriologia , Fatores de Transcrição/metabolismo , Animais , Bromodesoxiuridina , Cruzamentos Genéticos , Eletrofisiologia , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Interneurônios/metabolismo , Interneurônios/fisiologia , Camundongos , Fatores de Tempo
17.
Endocrinology ; 152(12): 4589-600, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21952247

RESUMO

A complete molecular understanding of ß-cell mass expansion will be useful for the improvement of therapies to treat diabetic patients. During normal periods of metabolic challenges, such as pregnancy, ß-cells proliferate, or self-renew, to meet the new physiological demands. The transcription factor Forkhead box D3 (Foxd3) is required for maintenance and self-renewal of several diverse progenitor cell lineages, and Foxd3 is expressed in the pancreatic primordium beginning at 10.5 d postcoitum, becoming localized predominantly to ß-cells after birth. Here, we show that mice carrying a pancreas-specific deletion of Foxd3 have impaired glucose tolerance, decreased ß-cell mass, decreased ß-cell proliferation, and decreased ß-cell size during pregnancy. In addition, several genes known to regulate proliferation, Foxm1, Skp2, Ezh2, Akt2, and Cdkn1a, are misregulated in islets isolated from these Foxd3 mutant mice. Together, these data place Foxd3 upstream of several pathways critical for ß-cell mass expansion in vivo.


Assuntos
Proliferação de Células , Fatores de Transcrição Forkhead/deficiência , Intolerância à Glucose/etiologia , Células Secretoras de Insulina/patologia , Proteínas Repressoras/deficiência , Animais , Tamanho Celular , Feminino , Fatores de Transcrição Forkhead/fisiologia , Regulação da Expressão Gênica , Células Secretoras de Insulina/citologia , Camundongos , Pâncreas , Gravidez , Proteínas Repressoras/fisiologia , Deleção de Sequência
18.
Development ; 138(4): 641-52, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21228004

RESUMO

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.


Assuntos
Linhagem da Célula , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Células-Tronco Multipotentes/metabolismo , Crista Neural/metabolismo , Células-Tronco Neurais/metabolismo , Proteínas Repressoras/metabolismo , Animais , Diferenciação Celular , Embrião de Mamíferos/inervação , Fatores de Transcrição Forkhead/deficiência , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Knockout , Células-Tronco Multipotentes/citologia , Mutação , Crista Neural/citologia , Células-Tronco Neurais/citologia , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética
19.
Genesis ; 49(1): 10-23, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21254333

RESUMO

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.


Assuntos
Fatores de Transcrição Forkhead/genética , Crista Neural/crescimento & desenvolvimento , Fatores de Transcrição Box Pareados/genética , Proteínas Repressoras/genética , Persistência do Tronco Arterial/genética , Animais , Anormalidades Craniofaciais/genética , Perda do Embrião/genética , Camundongos , Camundongos Mutantes , Crista Neural/citologia , Fator de Transcrição PAX3 , Deleção de Sequência , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Timo/anormalidades
20.
Sci Rep ; 1: 106, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22355623

RESUMO

Formation of higher-order structure of nucleic acids (hairpins or loops, for example) may impact not only gene regulation, but also molecular biology techniques and approaches critical for design and production of vectors needed for genetic engineering approaches. In the course of designing vectors aimed to modify the murine Foxd3 locus through homologous recombination in embryonic stem cells, we discovered a 370 nucleotide segment of DNA resistant to polymerase read-through. In addition to sequencing and PCR disruptions, we were unable to use BAC recombineering strategies to exchange sequences within the Foxd3 locus. This segment corresponds to a putative DNA hairpin region just upstream of the 5' untranslated region of Foxd3. This region is also highly conserved across vertebrate species, suggesting possible functional significance. Our findings provide a cautionary note for researchers experiencing technical challenges with BAC recombineering or other molecular biology methods requiring recombination or polymerase activity.


Assuntos
Cromossomos Artificiais Bacterianos , Reação em Cadeia da Polimerase/métodos , Recombinação Genética , Análise de Sequência de DNA/métodos , Regiões 5' não Traduzidas , Animais , Sequência de Bases , Análise por Conglomerados , Primers do DNA , Fatores de Transcrição Forkhead/genética , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA