Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Mol Biol Evol ; 35(2): 417-430, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29177513

RESUMO

The origin of feathers is an important question in Evo-Devo studies, with the eventual evolution of vaned feathers which are aerodynamic, allowing feathered dinosaurs and early birds to fly and venture into new ecological niches. Studying how feathers and scales are developmentally specified provides insight into how a new organ may evolve. We identified feather-associated genes using genomic analyses. The candidate genes were tested by expressing them in chicken and alligator scale forming regions. Ectopic expression of these genes induced intermediate morphotypes between scales and feathers which revealed several major morphogenetic events along this path: Localized growth zone formation, follicle invagination, epithelial branching, feather keratin differentiation, and dermal papilla formation. In addition to molecules known to induce feathers on scales (retinoic acid, ß-catenin), we identified novel scale-feather converters (Sox2, Zic1, Grem1, Spry2, Sox18) which induce one or more regulatory modules guiding these morphogenetic events. Some morphotypes resemble filamentous appendages found in feathered dinosaur fossils, whereas others exhibit characteristics of modern avian feathers. We propose these morpho-regulatory modules were used to diversify archosaur scales and to initiate feather evolution. The regulatory combination and hierarchical integration may have led to the formation of extant feather forms. Our study highlights the importance of integrating discoveries between developmental biology and paleontology.


Assuntos
Escamas de Animais , Evolução Biológica , Plumas , Morfogênese/genética , Jacarés e Crocodilos , Animais , Embrião de Galinha , Perfilação da Expressão Gênica , Genômica , Fenótipo , Fatores de Transcrição , beta-Queratinas/genética , beta-Queratinas/metabolismo
2.
Hum Mol Genet ; 25(16): 3539-3554, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27418670

RESUMO

CHD7 mutations are implicated in a majority of cases of the congenital disorder, CHARGE syndrome. CHARGE, an autosomal dominant syndrome, is known to affect multiple tissues including eye, heart, ear, craniofacial nerves and skeleton and genital organs. Using a morpholino-antisense-oligonucleotide-based zebrafish model for CHARGE syndrome, we uncover a complex spectrum of abnormalities in the neural crest and the crest-derived cell types. We report for the first time, defects in myelinating Schwann cells, enteric neurons and pigment cells in a CHARGE model. We also observe defects in the specification of peripheral neurons and the craniofacial skeleton as previously reported. Chd7 morphants have impaired migration of neural crest cells and deregulation of sox10 expression from the early stages. Knocking down Sox10 in the zebrafish CHARGE model rescued the defects in Schwann cells and craniofacial cartilage. Our zebrafish CHARGE model thus reveals important regulatory roles for Chd7 at multiple points of neural crest development viz., migration, fate choice and differentiation and we suggest that sox10 deregulation is an important driver of the neural crest-derived aspects of Chd7 dependent CHARGE syndrome.


Assuntos
Síndrome CHARGE/genética , DNA Helicases/genética , Proteínas de Ligação a DNA/genética , Fatores de Transcrição SOXE/genética , Proteínas de Peixe-Zebra/genética , Animais , Síndrome CHARGE/patologia , Diferenciação Celular/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Morfolinos/genética , Crista Neural/crescimento & desenvolvimento , Crista Neural/patologia , Fenótipo , Células de Schwann/metabolismo , Células de Schwann/patologia , Peixe-Zebra/genética
3.
Am J Med Genet C Semin Med Genet ; 175(4): 478-486, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29082625

RESUMO

Neural crest cells are highly migratory pluripotent cells that give rise to diverse derivatives including cartilage, bone, smooth muscle, pigment, and endocrine cells as well as neurons and glia. Abnormalities in neural crest-derived tissues contribute to the etiology of CHARGE syndrome, a complex malformation disorder that encompasses clinical symptoms like coloboma, heart defects, atresia of the choanae, retarded growth and development, genital hypoplasia, ear anomalies, and deafness. Mutations in the chromodomain helicase DNA-binding protein 7 (CHD7) gene are causative of CHARGE syndrome and loss-of-function data in different model systems have firmly established a role of CHD7 in neural crest development. Here, we will summarize our current understanding of the function of CHD7 in neural crest development and discuss possible links of CHARGE syndrome to other developmental disorders.


Assuntos
Síndrome CHARGE/diagnóstico , Síndrome CHARGE/etiologia , Crista Neural/anormalidades , Fenótipo , Trifosfato de Adenosina/metabolismo , Animais , Síndrome CHARGE/metabolismo , Montagem e Desmontagem da Cromatina , DNA Helicases/genética , DNA Helicases/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Humanos , Complexos Multiproteicos/metabolismo , Mutação , Ligação Proteica
4.
Nature ; 470(7333): 279-83, 2011 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-21160473

RESUMO

Cell-fate transitions involve the integration of genomic information encoded by regulatory elements, such as enhancers, with the cellular environment. However, identification of genomic sequences that control human embryonic development represents a formidable challenge. Here we show that in human embryonic stem cells (hESCs), unique chromatin signatures identify two distinct classes of genomic elements, both of which are marked by the presence of chromatin regulators p300 and BRG1, monomethylation of histone H3 at lysine 4 (H3K4me1), and low nucleosomal density. In addition, elements of the first class are distinguished by the acetylation of histone H3 at lysine 27 (H3K27ac), overlap with previously characterized hESC enhancers, and are located proximally to genes expressed in hESCs and the epiblast. In contrast, elements of the second class, which we term 'poised enhancers', are distinguished by the absence of H3K27ac, enrichment of histone H3 lysine 27 trimethylation (H3K27me3), and are linked to genes inactive in hESCs and instead are involved in orchestrating early steps in embryogenesis, such as gastrulation, mesoderm formation and neurulation. Consistent with the poised identity, during differentiation of hESCs to neuroepithelium, a neuroectoderm-specific subset of poised enhancers acquires a chromatin signature associated with active enhancers. When assayed in zebrafish embryos, poised enhancers are able to direct cell-type and stage-specific expression characteristic of their proximal developmental gene, even in the absence of sequence conservation in the fish genome. Our data demonstrate that early developmental enhancers are epigenetically pre-marked in hESCs and indicate an unappreciated role of H3K27me3 at distal regulatory elements. Moreover, the wealth of new regulatory sequences identified here provides an invaluable resource for studies and isolation of transient, rare cell populations representing early stages of human embryogenesis.


Assuntos
Cromatina/genética , Desenvolvimento Embrionário/genética , Células-Tronco Embrionárias/metabolismo , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Acetilação , Animais , Diferenciação Celular , Linhagem Celular , Cromatina/metabolismo , Imunoprecipitação da Cromatina , DNA Helicases/metabolismo , Células-Tronco Embrionárias/citologia , Epigênese Genética/genética , Gastrulação/genética , Camadas Germinativas/embriologia , Camadas Germinativas/metabolismo , Histonas/química , Histonas/metabolismo , Humanos , Lisina/metabolismo , Mesoderma/citologia , Mesoderma/embriologia , Metilação , Placa Neural/citologia , Neurulação/genética , Proteínas Nucleares/metabolismo , RNA/análise , RNA/genética , Fatores de Transcrição/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Fatores de Transcrição de p300-CBP/metabolismo
5.
Nature ; 463(7283): 958-62, 2010 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-20130577

RESUMO

Heterozygous mutations in the gene encoding the CHD (chromodomain helicase DNA-binding domain) member CHD7, an ATP-dependent chromatin remodeller homologous to the Drosophila trithorax-group protein Kismet, result in a complex constellation of congenital anomalies called CHARGE syndrome, which is a sporadic, autosomal dominant disorder characterized by malformations of the craniofacial structures, peripheral nervous system, ears, eyes and heart. Although it was postulated 25 years ago that CHARGE syndrome results from the abnormal development of the neural crest, this hypothesis remained untested. Here we show that, in both humans and Xenopus, CHD7 is essential for the formation of multipotent migratory neural crest (NC), a transient cell population that is ectodermal in origin but undergoes a major transcriptional reprogramming event to acquire a remarkably broad differentiation potential and ability to migrate throughout the body, giving rise to craniofacial bones and cartilages, the peripheral nervous system, pigmentation and cardiac structures. We demonstrate that CHD7 is essential for activation of the NC transcriptional circuitry, including Sox9, Twist and Slug. In Xenopus embryos, knockdown of Chd7 or overexpression of its catalytically inactive form recapitulates all major features of CHARGE syndrome. In human NC cells CHD7 associates with PBAF (polybromo- and BRG1-associated factor-containing complex) and both remodellers occupy a NC-specific distal SOX9 enhancer and a conserved genomic element located upstream of the TWIST1 gene. Consistently, during embryogenesis CHD7 and PBAF cooperate to promote NC gene expression and cell migration. Our work identifies an evolutionarily conserved role for CHD7 in orchestrating NC gene expression programs, provides insights into the synergistic control of distal elements by chromatin remodellers, illuminates the patho-embryology of CHARGE syndrome, and suggests a broader function for CHD7 in the regulation of cell motility.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , DNA Helicases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Crista Neural/citologia , Crista Neural/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Xenopus/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Movimento Celular , Proteínas Cromossômicas não Histona/genética , DNA Helicases/química , DNA Helicases/deficiência , DNA Helicases/genética , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Embrião não Mamífero/citologia , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Crista Neural/embriologia , Ligação Proteica , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição da Família Snail , Síndrome , Fatores de Transcrição/genética , Transcrição Gênica , Proteína 1 Relacionada a Twist/genética , Proteína 1 Relacionada a Twist/metabolismo , Proteínas de Xenopus/química , Proteínas de Xenopus/deficiência , Proteínas de Xenopus/genética , Xenopus laevis/embriologia , Xenopus laevis/genética , Xenopus laevis/metabolismo
6.
Front Cell Dev Biol ; 10: 1030587, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36568983

RESUMO

Congenital heart defects occur in almost 80% of patients with CHARGE syndrome, a sporadically occurring disease causing craniofacial and other abnormalities due to mutations in the CHD7 gene. Animal models have been generated to mimic CHARGE syndrome; however, heart defects are not extensively described in zebrafish disease models of CHARGE using morpholino injections or genetic mutants. Here, we describe the co-occurrence of craniofacial abnormalities and heart defects in zebrafish chd7 mutants. These mutant phenotypes are enhanced in the maternal zygotic mutant background. In the chd7 mutant fish, we found shortened craniofacial cartilages and extra cartilage formation. Furthermore, the length of the ventral aorta is altered in chd7 mutants. Many CHARGE patients have aortic arch anomalies. It should be noted that the aberrant branching of the first branchial arch artery is observed for the first time in chd7 fish mutants. To understand the cellular mechanism of CHARGE syndrome, neural crest cells (NCCs), that contribute to craniofacial and cardiovascular tissues, are examined using sox10:Cre lineage tracing. In contrast to its function in cranial NCCs, we found that the cardiac NCC-derived mural cells along the ventral aorta and aortic arch arteries are not affected in chd7 mutant fish. The chd7 fish mutants we generated recapitulate some of the craniofacial and cardiovascular phenotypes found in CHARGE patients and can be used to further determine the roles of CHD7.

7.
Am J Med Genet A ; 155A(2): 270-9, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21271641

RESUMO

The craniofacial region is assembled through the active migration of cells and the rearrangement and sculpting of facial prominences and pharyngeal arches, which consequently make it particularly susceptible to a large number of birth defects. Genetic, molecular, and cellular processes must be temporally and spatially regulated to culminate in the three-dimension structures of the face. The starting constituent for the majority of skeletal and connective tissues in the face is a pluripotent population of cells, the cranial neural crest cells (NCCs). In this review we discuss the newest scientific findings in the development of the craniofacial complex as related to NCCs. Furthermore, we present recent findings on NCC diseases called neurocristopathies and, in doing so, provide clinicians with new tools for understanding a growing number of craniofacial genetic disorders.


Assuntos
Padronização Corporal , Movimento Celular/fisiologia , Anormalidades Craniofaciais/genética , Crista Neural/embriologia , Crista Neural/fisiopatologia , Transdução de Sinais/fisiologia , Crânio/embriologia , Diferenciação Celular/fisiologia , Proliferação de Células , Anormalidades Craniofaciais/patologia , Humanos
8.
J Cell Biol ; 170(3): 413-27, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16061694

RESUMO

Maternal embryonic leucine zipper kinase (MELK) was previously identified in a screen for genes enriched in neural progenitors. Here, we demonstrate expression of MELK by progenitors in developing and adult brain and that MELK serves as a marker for self-renewing multipotent neural progenitors (MNPs) in cultures derived from the developing forebrain and in transgenic mice. Overexpression of MELK enhances (whereas knockdown diminishes) the ability to generate neurospheres from MNPs, indicating a function in self-renewal. MELK down-regulation disrupts the production of neurogenic MNP from glial fibrillary acidic protein (GFAP)-positive progenitors in vitro. MELK expression in MNP is cell cycle regulated and inhibition of MELK expression down-regulates the expression of B-myb, which is shown to also mediate MNP proliferation. These findings indicate that MELK is necessary for proliferation of embryonic and postnatal MNP and suggest that it regulates the transition from GFAP-expressing progenitors to rapid amplifying progenitors in the postnatal brain.


Assuntos
Proliferação de Células , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Células-Tronco Multipotentes/fisiologia , Neurônios/fisiologia , Proteínas Serina-Treonina Quinases/biossíntese , Animais , Astrócitos/metabolismo , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Proteína Glial Fibrilar Ácida/biossíntese , Camundongos , Camundongos Transgênicos , Células-Tronco Multipotentes/metabolismo , Neurônios/metabolismo , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/biossíntese , Transativadores/metabolismo
9.
Mol Reprod Dev ; 75(5): 818-27, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18157870

RESUMO

Human embryonic stem cells (hESCs) hold great promise for cell-based therapies and drug screening applications. However, growing and processing large quantities of undifferentiated hESCs is a challenging task. Conventionally, hESCs are passaged as clusters, which can limit their growth efficiency and use in downstream applications. This study demonstrates that hESCs can be passaged as single cells using Accutase, a formulated mixture of digestive enzymes. In contrast to trypsin treatment, Accutase treatment does not significantly affect the viability and proliferation rate of hESC dissociation into single cells. Accutase-dissociated single cells can be separated by FACS and proliferate as fully pluripotent hESCs. An Oct4-eGFP reporter construct engineered into hESCs was used to monitor the pluripotency of hESCs passaged with Accutase. Compared to collagenase-passaged hESCs, Accutase-treated cultures contained a larger proportion of undifferentiated (Oct4-positive) cells. Additionally, Accutase-passaged undifferentiated hESCs could be grown as monolayers without the need for monitoring and/or selection for quality hESC colonies.


Assuntos
Técnicas de Cultura de Células/métodos , Proliferação de Células/efeitos dos fármacos , Células-Tronco Embrionárias/citologia , Enzimas/farmacologia , Células-Tronco Pluripotentes/citologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Células-Tronco Embrionárias/fisiologia , Humanos , Células-Tronco Pluripotentes/fisiologia
10.
Stem Cell Reports ; 5(4): 499-507, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26441305

RESUMO

Premigratory neural crest cells comprise a transient, embryonic population that arises within the CNS, but subsequently migrates away and differentiates into many derivatives. Previously, premigratory neural crest could not be maintained in a multipotent, adhesive state without spontaneous differentiation. Here, we report conditions that enable maintenance of neuroepithelial "crestospheres" that self-renew and retain multipotency for weeks. Moreover, under differentiation conditions, these cells can form multiple derivatives in vitro and in vivo after transplantation into chick embryos. Similarly, human embryonic stem cells directed to a neural crest fate can be maintained as crestospheres and subsequently differentiated into several derivatives. By devising conditions that maintain the premigratory state in vitro, these results demonstrate that neuroepithelial neural crest precursors are capable of long-term self-renewal. This approach will help uncover mechanisms underlying their developmental potential, differentiation and, together with the induced pluripotent stem cell techniques, the pathology of human neurocristopathies.


Assuntos
Diferenciação Celular , Células-Tronco Multipotentes/citologia , Crista Neural/citologia , Animais , Movimento Celular , Embrião de Galinha , Galinhas , Humanos , Células-Tronco Pluripotentes Induzidas/citologia
11.
Gene Expr Patterns ; 5(1): 113-21, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15533826

RESUMO

Growth and patterning during Drosophila wing development are mediated by signaling from its dorso-ventral (D/V) organizer. Wingless is expressed in the D/V boundary and functions as a morphogen to activate target genes at a distance. Wingless pathway and thereby D/V signaling is negatively regulated by the homeotic gene Ultrabithorax (Ubx) to mediate haltere development. In an enhancer-trap screen to identify genes that show differential expression between wing and haltere discs, we identified CG32062, which codes for a RNA-binding protein. In wing discs, CG32062 is expressed only in non-D/V cells. CG32062 expression in non-D/V cells is dependent on Notch-mediated signaling from the D/V boundary. However, CG32062 expression is independent of Wingless function, thus providing evidence for a second long-range signaling mechanism of the D/V organizer. In haltere discs, CG32062 is negatively regulated by Ubx. The non-cell autonomous nature of Ubx-mediated repression of CG32062 expression suggests that the novel component of D/V signaling is also negatively regulated during haltere specification.


Assuntos
Padronização Corporal/fisiologia , Proteínas de Drosophila/genética , Drosophila/crescimento & desenvolvimento , Proteínas Proto-Oncogênicas/genética , Proteínas de Ligação a RNA/genética , Asas de Animais/crescimento & desenvolvimento , Animais , Biomarcadores , Proteínas de Drosophila/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Organizadores Embrionários/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Asas de Animais/metabolismo , Proteína Wnt1
12.
Front Physiol ; 3: 188, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22737127

RESUMO

Interest regarding stem cell based therapies for the treatment of congenital or acquired craniofacial deformities is rapidly growing. Craniofacial problems such as periodontal disease, cleft lip and palate, ear microtia, craniofacial microsomia, and head and neck cancers are not only common but also some of the most burdensome surgical problems worldwide. Treatments often require a multi-staged multidisciplinary team approach. Current surgical therapies attempt to reduce the morbidity and social/emotional impact, yet outcomes can still be unpredictable and unsatisfactory. The concept of harvesting stem cells followed by expansion, differentiation, seeding onto a scaffold and re-transplanting them is likely to become a clinical reality. In this review, we will summarize the translational applications of stem cell therapy in tissue regeneration for craniofacial defects.

13.
Cell Stem Cell ; 11(5): 633-48, 2012 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-22981823

RESUMO

Neural crest cells (NCC) are a transient, embryonic cell population characterized by unusual migratory ability and developmental plasticity. To annotate and characterize cis-regulatory elements utilized by the human NCC, we coupled a hESC differentiation model with genome-wide profiling of histone modifications and of coactivator and transcription factor (TF) occupancy. Sequence analysis predicted major TFs binding at epigenomically annotated hNCC enhancers, including a master NC regulator, TFAP2A, and nuclear receptors NR2F1 and NR2F2. Although many TF binding events occur outside enhancers, sites coinciding with enhancer chromatin signatures show significantly higher sequence constraint, nucleosomal depletion, correlation with gene expression, and functional conservation in NCC isolated from chicken embryos. Simultaneous co-occupancy by TFAP2A and NR2F1/F2 is associated with permissive enhancer chromatin states, characterized by high levels of p300 and H3K27ac. Our results provide global insights into human NC chromatin landscapes and a rich resource for studies of craniofacial development and disease.


Assuntos
Elementos Facilitadores Genéticos , Epigênese Genética , Crista Neural/metabolismo , Animais , Fator I de Transcrição COUP/genética , Fator I de Transcrição COUP/metabolismo , Fator II de Transcrição COUP/genética , Fator II de Transcrição COUP/metabolismo , Diferenciação Celular , Embrião de Galinha/metabolismo , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica , Humanos , Crista Neural/citologia , Fator de Transcrição AP-2/genética , Fator de Transcrição AP-2/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
14.
Methods Mol Biol ; 767: 315-31, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21822885

RESUMO

Relatively safe, HIV-1-based lentiviral vectors have served as an efficient means of transducing human embryonic stem cells (hESCs). Here we describe the variety of lentiviral vector systems available with the basic strategy for designing viral vectors and methods for generating viruses for efficiently infecting and selecting transduced hESCs.


Assuntos
Lentivirus/metabolismo , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/virologia , Transdução Genética/métodos , Células Cultivadas , Células Clonais , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/virologia , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Lentivirus/efeitos dos fármacos , Lentivirus/fisiologia , Neomicina/farmacologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Transfecção , Transgenes/genética , Montagem de Vírus/efeitos dos fármacos
15.
PLoS One ; 4(4): e5046, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19352491

RESUMO

BACKGROUND: Developmental, physiological and tissue engineering studies critical to the development of successful myocardial regeneration therapies require new ways to effectively visualize and isolate large numbers of fluorescently labeled, functional cardiomyocytes. METHODOLOGY/PRINCIPAL FINDINGS: Here we describe methods for the clonal expansion of engineered hESCs and make available a suite of lentiviral vectors for that combine Blasticidin, Neomycin and Puromycin resistance based drug selection of pure populations of stem cells and cardiomyocytes with ubiquitous or lineage-specific promoters that direct expression of fluorescent proteins to visualize and track cardiomyocytes and their progenitors. The phospho-glycerate kinase (PGK) promoter was used to ubiquitously direct expression of histone-2B fused eGFP and mCherry proteins to the nucleus to monitor DNA content and enable tracking of cell migration and lineage. Vectors with T/Brachyury and alpha-myosin heavy chain (alphaMHC) promoters targeted fluorescent or drug-resistance proteins to early mesoderm and cardiomyocytes. The drug selection protocol yielded 96% pure cardiomyocytes that could be cultured for over 4 months. Puromycin-selected cardiomyocytes exhibited a gene expression profile similar to that of adult human cardiomyocytes and generated force and action potentials consistent with normal fetal cardiomyocytes, documenting these parameters in hESC-derived cardiomyocytes and validating that the selected cells retained normal differentiation and function. CONCLUSION/SIGNIFICANCE: The protocols, vectors and gene expression data comprise tools to enhance cardiomyocyte production for large-scale applications.


Assuntos
Células-Tronco Embrionárias/citologia , Vetores Genéticos , Lentivirus/genética , Miocárdio/citologia , Adulto , Sequência de Bases , Diferenciação Celular , Primers do DNA , Resistência a Medicamentos , Células-Tronco Embrionárias/metabolismo , Proteínas Fetais/genética , Citometria de Fluxo , Perfilação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Humanos , Imuno-Histoquímica , Miocárdio/metabolismo , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas com Domínio T/genética
16.
Cancer Res ; 68(19): 7882-6, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18829544

RESUMO

CD133 (Prominin1) is a pentaspan transmembrane glycoprotein expressed in several stem cell populations and cancers. Reactivity with an antibody (AC133) to a glycoslyated form of CD133 has been widely used for the enrichment of cells with tumor-initiating activity in xenograph transplantation assays. We have found by fluorescence-activated cell sorting that increased AC133 reactivity in human embryonic stem cells, colon cancer, and melanoma cells is correlated with increased DNA content and, reciprocally, that the least reactive cells are in the G(1)-G(0) portion of the cell cycle. Continued cultivation of cells sorted on the basis of high and low AC133 reactivity results in a normalization of the cell reactivity profiles, indicating that cells with low AC133 reactivity can generate highly reactive cells as they resume proliferation. The association of AC133 with actively cycling cells may contribute to the basis for enrichment for tumor-initiating activity.


Assuntos
Antígenos CD/metabolismo , Ciclo Celular/imunologia , Neoplasias do Colo/metabolismo , Células-Tronco Embrionárias/metabolismo , Glicoproteínas/metabolismo , Melanoma/metabolismo , Peptídeos/metabolismo , Antígeno AC133 , Animais , Antígenos CD/imunologia , Células CACO-2 , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células , Separação Celular/métodos , Técnicas de Cocultura , Neoplasias do Colo/patologia , DNA/análise , Epitopos/metabolismo , Perfilação da Expressão Gênica , Glicoproteínas/imunologia , Humanos , Imuno-Histoquímica , Melanoma/patologia , Camundongos , Peptídeos/imunologia
17.
PLoS One ; 3(10): e3451, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18941637

RESUMO

RNA expression data reveals that human embryonic stem (hES) cells differ from mouse ES (mES) cells in the expression of RNAs for keratin intermediate filament proteins. These differences were confirmed at the cellular and protein level and may reflect a fundamental difference in the epithelial nature of embryonic stem cells derived from mouse and human blastocysts. Mouse ES cells express very low levels of the simple epithelial keratins K8, K18 and K19. By contrast hES cells express moderate levels of the RNAs for these intermediate filament proteins as do mouse stem cells derived from the mouse epiblast. Expression of K8 and K18 RNAs are correlated with increased c-Jun RNA expression in both mouse and human ES cell cultures. However, decreasing K8 and K18 expression associated with differentiation to neuronal progenitor cells is correlated with increasing expression of the Snai2 (Slug) transcriptional repression and not decreased Jun expression. Increasing K7 expression is correlated with increased CDX2 and decreased Oct4 RNA expression associated with the formation of trophoblast derivatives by hES cells. Our study supports the view that hES cells are more similar to mouse epiblast cells than mouse ES cells and is consistent with the epithelial nature of hES cells. Keratin intermediate filament expression in hES cells may modulate sensitivity to death receptor mediated apoptosis and stress.


Assuntos
Células-Tronco Embrionárias/metabolismo , Queratinas/genética , RNA Mensageiro/análise , Animais , Diferenciação Celular , Células Epiteliais/citologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Queratina-18/genética , Queratina-19/genética , Queratina-8/genética , Camundongos , Neurônios/citologia , Especificidade da Espécie
18.
J Neurosci Res ; 86(1): 48-60, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17722061

RESUMO

Emerging evidence suggests that neural stem cells and brain tumors regulate their proliferation via similar pathways. In a previous study, we demonstrated that maternal embryonic leucine zipper kinase (Melk) is highly expressed in murine neural stem cells and regulates their proliferation. Here we describe how MELK expression is correlated with pathologic grade of brain tumors, and its expression levels are significantly correlated with shorter survival, particularly in younger glioblastoma patients. In normal human astrocytes, MELK is only faintly expressed, and MELK knockdown does not significantly influence their growth, whereas Ras and Akt overexpressing astrocytes have up-regulated MELK expression, and the effect of MELK knockdown is more prominent in these transformed astrocytes. In primary cultures from human glioblastoma and medulloblastoma, MELK knockdown by siRNA results in inhibition of the proliferation and survival of these tumors. Furthermore, we show that MELK siRNA dramatically inhibits proliferation and, to some extent, survival of stem cells isolated from glioblastoma in vitro. These results demonstrate a critical role for MELK in the proliferation of brain tumors, including their stem cells, and suggest that MELK may be a compelling molecular target for treatment of high-grade brain tumors.


Assuntos
Neoplasias Encefálicas/patologia , Proliferação de Células , Glioblastoma/patologia , Células-Tronco Neoplásicas/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Adulto , Idoso , Animais , Células Cultivadas , Feminino , Citometria de Fluxo/métodos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Espectrometria de Massas/métodos , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Receptores Patched , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/deficiência , RNA Interferente Pequeno/farmacologia , Receptores de Superfície Celular/deficiência , Transfecção/métodos
19.
Cryobiology ; 53(2): 194-205, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16839540

RESUMO

We tested a "standard" cryopreservation protocol (slow cooling with 10% DMSO) on the human embryonic stem cell (hESC) line H9 containing an Oct-4 (POU5F1) promoter-driven, enhanced green fluorescent protein (EGFP) reporter to monitor maintenance of pluripotency. Cells were cooled to -80 degrees C in cryovials and then transferred to a -80 degrees C freezer. Cells were held at -80 degrees C for 3 days ("short-term storage") or 3 months ("long-term storage"). Vials were thawed in a +36 degrees C water bath and cells were cultured for 3, 7, or 14 days. Propidium iodide (PI) was used to assess cell viability by flow cytometry. Control cells were passaged on the same day that the frozen cells were thawed. The majority of cells in control hESC cultures were Oct-4 positive and almost 99% of EGFP+ cells were alive as determined by exclusion of PI. In contrast, the frozen cells, even after 3 days of culture, contained only 50% live cells, and only 10% were EGFP-positive. After 7 days in culture, the proportion of dead cells decreased and there was an increase in the Oct-4-positive population but microscopic examination revealed large patches of EGFP-negative cells within clusters of colonies even after 14 days of culturing. After 3 months of storage at -80 degrees C the deleterious effect of freezing was even more pronounced: the samples regained a quantifiable number of EGFP-positive cells only after 7 days of culturing following thawing. It is concluded that new protocols and media are required for freezing hESC and safe storage at -80 degrees C as well as studies of the mechanisms of stress-related events associated with cell cryopreservation.


Assuntos
Criopreservação/métodos , Crioprotetores/farmacologia , Dimetil Sulfóxido/farmacologia , Embrião de Mamíferos/citologia , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco/citologia , Animais , Diferenciação Celular , Sobrevivência Celular , Criopreservação/instrumentação , Sequestradores de Radicais Livres/farmacologia , Proteínas de Fluorescência Verde/metabolismo , Humanos , Cinética , Camundongos , Microscopia de Fluorescência
20.
Development ; 130(8): 1537-47, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12620980

RESUMO

In the third thoracic segment of Drosophila, wing development is suppressed by the homeotic selector gene Ultrabithorax (Ubx) in order to mediate haltere development. Previously, we have shown that Ubx represses dorsoventral (DV) signaling to specify haltere fate. Here we examine the mechanism of Ubx-mediated downregulation of DV signaling. We show that Wingless (Wg) and Vestigial (Vg) are differentially regulated in wing and haltere discs. In wing discs, although Vg expression in non-DV cells is dependent on DV boundary function of Wg, it maintains its expression by autoregulation. Thus, overexpression of Vg in non-DV cells can bypass the requirement for Wg signaling from the DV boundary. Ubx functions, at least, at two levels to repress Vestigial expression in non-DV cells of haltere discs. At the DV boundary, it functions downstream of Shaggy/GSK3 beta to enhance the degradation of Armadillo (Arm), which causes downregulation of Wg signaling. In non-DV cells, Ubx inhibits event(s) downstream of Arm, but upstream of Vg autoregulation. Repression of Vg at multiple levels appears to be crucial for Ubx-mediated specification of the haltere fate. Overexpression of Vg in haltere discs is enough to override Ubx function and cause haltere-to-wing homeotic transformations.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , Asas de Animais/crescimento & desenvolvimento , Animais , Proteínas do Domínio Armadillo , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/anatomia & histologia , Drosophila melanogaster/metabolismo , Genes Reporter , Quinase 3 da Glicogênio Sintase/metabolismo , Morfogênese/fisiologia , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/fisiologia , Transativadores/metabolismo , Fatores de Transcrição/genética , Asas de Animais/anatomia & histologia , Asas de Animais/metabolismo , Proteína Wnt1
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa