Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Redox Biol ; 22: 101156, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30851670

RESUMO

Adult progenitor cells reside in specialized microenvironments which maintain their undifferentiated cell state and trigger regenerative responses following injury. Although these environments are well described in several tissues, the cellular components that comprise the cardiac environment where progenitor cells are located remain unknown. Here we use Bmi1CreERT and Bmi1GFP mice as genetic tools to trace cardiac damage-responsive cells throughout the mouse lifespan. In adolescent mice, Bmi1+ damage-responsive cells are broadly distributed throughout the myocardium. In adult mice, however, Bmi1+ cells are confined predominately in perivascular areas with low levels of reactive oxygen species (ROS) and their number decline in an age-dependent manner. In vitro co-culture experiments with endothelial cells supported a regulatory role of the endothelium in damage-responsive cell behavior. Accordingly, in vivo genetic decrease of ROS levels in adult heart disengaged Bmi1+ cells from the cardiovascular network, recapitulating an adolescent-like Bmi1 expression profile. Thus, we identify cardiac perivascular regions as low-stress microenvironments that favor the maintenance of adult damage-responsive cells.


Assuntos
Miocárdio/metabolismo , Estresse Oxidativo , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fatores Etários , Animais , Permeabilidade Capilar , Proliferação de Células , Células Endoteliais/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Estresse Oxidativo/genética , Complexo Repressor Polycomb 1/genética , Proteínas Proto-Oncogênicas/genética , Espécies Reativas de Oxigênio/metabolismo
2.
PLoS One ; 13(11): e0206534, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30395586

RESUMO

BACKGROUND: Human adult adipose-derived stem cells (hADSCs) have become the most promising cell source for regenerative medicine. However the prolonged ex vivo expansion periods required to obtain the necessary therapeutic dose promotes progressive senescence, with the concomitant reduction of their therapeutic potential. AIM AND SCOPE: A better understanding of the determinants of hADSC senescence is needed to improve biosafety while preserving therapeutic efficiency. Here, we investigated the association between deregulation of the imprinted DLK1-DIO3 region and replicative senescence in hADSC cultures. METHODS: We compared hADSC cultures at short (PS) and prolonged (PL) passages, both in standard and low [O2] (21 and 3%, respectively), in relation to replicative senescence. hADSCs were evaluated for expression alterations in the DLK1-DIO3 region on chromosome 14q32, and particularly in its main miRNA cluster. RESULTS: Comparison of hADSCs cultured at PL or PS surprisingly showed a quite significant fraction (69%) of upregulated miRNAs in PL cultures mapping to the imprinted 14q32 locus, the largest miRNA cluster described in the genome. In agreement, expression of the lncRNA MEG3 (Maternally Expressed 3; Meg3/Gtl2), cultured at 21 and 3% [O2], was also significantly higher in PL than in PS passages. During hADSC replicative senescence the AcK16H4 activating mark was found to be significantly associated with the deregulation of the entire DLK1-DIO3 locus, with a secondary regulatory role for the methylation of DMR regions. CONCLUSION: A direct relationship between DLK1-DIO3 deregulation and replicative senescence of hADSCs is reported, involving upregulation of a very significant fraction of its largest miRNA cluster (14q32.31), paralleled by the progressive overexpression of the lncRNA MEG3, which plays a central role in the regulation of Dlk1/Dio3 activation status in mice.


Assuntos
Impressão Genômica , Peptídeos e Proteínas de Sinalização Intercelular/genética , Iodeto Peroxidase/genética , Proteínas de Membrana/genética , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Animais , Proteínas de Ligação ao Cálcio , Proliferação de Células/genética , Células Cultivadas , Senescência Celular/genética , Cromossomos Humanos Par 14/genética , Epigênese Genética , Regulação da Expressão Gênica , Humanos , Camundongos , MicroRNAs/genética , Regulação para Cima
3.
Arterioscler Thromb Vasc Biol ; 38(9): 2160-2173, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29930004

RESUMO

Objective- Cardiac progenitor cells reside in the heart in adulthood, although their physiological relevance remains unknown. Here, we demonstrate that after myocardial infarction, adult Bmi1+ (B lymphoma Mo-MLV insertion region 1 homolog [PCGF4]) cardiac cells are a key progenitor-like population in cardiac neovascularization during ventricular remodeling. Approach and Results- These cells, which have a strong in vivo differentiation bias, are a mixture of endothelial- and mesenchymal-related cells with in vitro spontaneous endothelial cell differentiation capacity. Genetic lineage tracing analysis showed that heart-resident Bmi1+ progenitor cells proliferate after acute myocardial infarction and differentiate to generate de novo cardiac vasculature. In a mouse model of induced myocardial infarction, genetic ablation of these cells substantially deteriorated both heart angiogenesis and the ejection fraction, resulting in an ischemic-dilated cardiac phenotype. Conclusions- These findings imply that endothelial-related Bmi1+ progenitor cells are necessary for injury-induced neovascularization in adult mouse heart and highlight these cells as a suitable therapeutic target for preventing dysfunctional left ventricular remodeling after injury.


Assuntos
Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia , Neovascularização Patológica , Complexo Repressor Polycomb 1/fisiologia , Células-Tronco/patologia , Células-Tronco/fisiologia , Remodelação Ventricular , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-kit/metabolismo , Fatores de Transcrição/metabolismo
4.
Cell Death Differ ; 25(4): 809-822, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29323265

RESUMO

Accumulation of reactive oxygen species (ROS) is associated with several cardiovascular pathologies and with cell cycle exit by neonanatal cardiomyocytes, a key limiting factor in the regenerative capacity of the adult mammalian heart. The polycomb complex component BMI1 is linked to adult progenitors and is an important partner in DNA repair and redox regulation. We found that high BMI1 expression is associated with an adult Sca1+ cardiac progenitor sub-population with low ROS levels. In homeostasis, BMI1 repressed cell fate genes, including a cardiogenic differentiation program. Oxidative damage nonetheless modified BMI1 activity in vivo by derepressing canonical target genes in favor of their antioxidant and anticlastogenic functions. This redox-mediated mechanism is not restricted to damage situations, however, and we report ROS-associated differentiation of cardiac progenitors in steady state. These findings demonstrate how redox status influences the cardiac progenitor response, and identify redox-mediated BMI1 regulation with implications in maintenance of cellular identity in vivo.


Assuntos
Células-Tronco Adultas/metabolismo , Diferenciação Celular , Miocárdio/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Células-Tronco Adultas/citologia , Animais , Camundongos , Camundongos Transgênicos , Miocárdio/citologia , Oxirredução , Complexo Repressor Polycomb 1/genética , Proteínas Proto-Oncogênicas/genética
5.
DNA Repair (Amst) ; 54: 40-45, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28460268

RESUMO

Non-homologous end joining (NHEJ) is the main mechanism for double strand break (DSB) DNA repair. The error-prone DNA polymerase mu (Polµ) is involved in immunoglobulin variable region rearrangement and in general, NHEJ in non-lymphoid cells. Deletion of NHEJ factors in P53-/- mice, which are highly prone to development of T cell lymphoma, generally increases cancer incidence and shifts the tumor spectrum towards aggressive pro-B lymphoma. In contrast, Polµ deletion increased sarcoma incidence, proportionally reducing pro-B lymphoma development on the P53-deficient background. Array comparative genomic hybridization (aCGH) analyses showed DNA copy number alterations in both P53-/- and Polµ-/-P53-/- lymphomas. Our results also indicate that the increase in sarcoma incidence in Polµ-/-P53-/- mice could be associated with Cdk4 and Kub3 amplification and overexpression. These results identify a role for Polµ in the prevention of sarcomagenesis on a murine P53-deficient background, in contrast to most other NHEJ factors.


Assuntos
Carcinogênese , Reparo do DNA por Junção de Extremidades , DNA Polimerase Dirigida por DNA/genética , Sarcoma/metabolismo , Proteína Supressora de Tumor p53/genética , Animais , Proteínas de Transporte/genética , Quinase 4 Dependente de Ciclina/genética , DNA/metabolismo , Variações do Número de Cópias de DNA , Amplificação de Genes , Deleção de Genes , Instabilidade Genômica , Linfoma/genética , Linfoma/metabolismo , Linfoma/patologia , Camundongos , Camundongos Knockout , Sarcoma/genética , Sarcoma/patologia , Regulação para Cima
6.
J Tissue Eng Regen Med ; 11(3): 787-799, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-25492026

RESUMO

miRNA-1 (miR-1) and miRNA-133a (miR-133a) are muscle-specific miRNAs that play an important role in heart development and physiopathology. Although both miRNAs have been broadly studied during cardiogenesis, the mechanisms by which miR-1 and miR-133a could influence linage commitment in pluripotent stem cells remain poorly characterized. In this study we analysed the regulation of miR-1 and miR-133a expression during pluripotent stem cell differentiation [P19.CL6 cells; embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs)] and investigated their role in DMSO and embryoid body (EB)-mediated mesodermal and cardiac differentiation by gain- and loss-of-function studies, as well as in vivo, by the induction of teratomas. Gene expression analysis revealed that miR-1 and miR-133a are upregulated during cardiac differentiation of P19.CL6 cells, and also during ESC and iPSC EB differentiation. Forced overexpression of both miRNAs promoted mesodermal commitment and a concomitant decrease in the expression of neural differentiation markers. Moreover, overexpression of miR-1 enhanced the cardiac differentiation of P19.CL6, while miR-133a reduced it with respect to control cells. Teratoma formation experiments with P19.CL6 cells confirmed the influence of miR-1 and miR-133a during in vivo differentiation. Finally, inhibition of both miRNAs during P19.CL6 cardiac differentiation had opposite results to their overexpression. In conclusion, gene regulation involving miR-1 and miR-133a controls the mesodermal and cardiac fate of pluripotent stem cells. Copyright © 2014 John Wiley & Sons, Ltd.


Assuntos
Diferenciação Celular/genética , Linhagem da Célula/genética , MicroRNAs/metabolismo , Miocárdio/citologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Animais , Regulação da Expressão Gênica , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Mesoderma/citologia , Camundongos SCID , MicroRNAs/genética , Modelos Biológicos , Neurônios/citologia , Neurônios/metabolismo
7.
J Mol Cell Cardiol ; 99: 162-173, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27545043

RESUMO

MicroRNAs (miR) have considerable potential as therapeutic tools in cardiac diseases. Alterations in atrial miR are involved in the development of atrial fibrillation (AF), but the molecular mechanism underlying their contribution to atrial remodeling in chronic atrial fibrillation (CAF) is only partially understood. Here we used miR array to analyze the miR profile of atrial biopsies from sinus rhythm (SR) and CAF patients. qRT-PCR identified a distinctive CAF-miR signature and described conserved miR-208b upregulation in human and ovine AF atrial tissue. We used bioinformatics analysis to predict genes and signaling pathways as putative miR-208b targets, which highlighted genes from the cardiac muscle gene program and from canonical WNT, gap-junction and Ca2+ signaling networks. Results from analysis of miR-208b-overexpressing HL-1 atrial myocytes and from myocytes isolated from CAF patients showed that aberrant miR-208b levels reduced the expression and function of L-type Ca2+ channel subunits (CACNA1C and CACNB2) as well as the sarcoplasmic reticulum-Ca2+ pump SERCA2. These findings clearly pointed to CAF-specific upregulated miR-208b as an important mediator in Ca2+ handling impairment during atrial remodeling.


Assuntos
Fibrilação Atrial/genética , Fibrilação Atrial/metabolismo , Cálcio/metabolismo , Átrios do Coração/citologia , Átrios do Coração/metabolismo , MicroRNAs/genética , Miócitos Cardíacos/metabolismo , Regiões 3' não Traduzidas , Animais , Fibrilação Atrial/fisiopatologia , Remodelamento Atrial , Sequência de Bases , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Linhagem Celular , Doença Crônica , Conexina 43/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Átrios do Coração/fisiopatologia , Humanos , Miosinas/genética , Ligação Proteica , Interferência de RNA , RNA Mensageiro/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Ovinos , Proteínas Wnt/metabolismo
8.
Stem Cell Reports ; 3(6): 1029-42, 2014 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-25465869

RESUMO

miR-133a and miR-1 are known as muscle-specific microRNAs that are involved in cardiac development and pathophysiology. We have shown that both miR-1 and miR-133a are early and progressively upregulated during in vitro cardiac differentiation of adult cardiac progenitor cells (CPCs), but only miR-133a expression was enhanced under in vitro oxidative stress. miR-1 was demonstrated to favor differentiation of CPCs, whereas miR-133a overexpression protected CPCs against cell death, targeting, among others, the proapoptotic genes Bim and Bmf. miR-133a-CPCs clearly improved cardiac function in a rat myocardial infarction model by reducing fibrosis and hypertrophy and increasing vascularization and cardiomyocyte proliferation. The beneficial effects of miR-133a-CPCs seem to correlate with the upregulated expression of several relevant paracrine factors and the plausible cooperative secretion of miR-133a via exosomal transport. Finally, an in vitro heart muscle model confirmed the antiapoptotic effects of miR-133a-CPCs, favoring the structuration and contractile functionality of the artificial tissue.


Assuntos
MicroRNAs/genética , Mioblastos Cardíacos/metabolismo , Infarto do Miocárdio/genética , Animais , Cardiomegalia/genética , Cardiomegalia/patologia , Biologia Computacional , Expressão Gênica , Perfilação da Expressão Gênica , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Interferência de RNA , RNA Mensageiro/genética , Ratos
9.
Dev Cell ; 30(4): 410-22, 2014 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-25127056

RESUMO

The first lineage choice in mammalian embryogenesis is that between the trophectoderm, which gives rise to the trophoblast of the placenta, and the inner cell mass, from which is derived the embryo proper and the yolk sac. The establishment of these lineages is preceded by the inside-versus-outside positioning of cells in the early embryo and stochastic expression of key transcription factors, which is then resolved into lineage-restricted expression. The regulatory inputs that drive this restriction and how they relate to cell position are largely unknown. Here, we show an unsuspected role of Notch signaling in regulating trophectoderm-specific expression of Cdx2 in cooperation with TEAD4. Notch activity is restricted to outer cells and is able to influence positional allocation of blastomeres, mediating preferential localization to the trophectoderm. Our results show that multiple signaling inputs at preimplantation stages specify the first embryonic lineages.


Assuntos
Blastocisto/metabolismo , Linhagem da Célula , Ectoderma/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptor Notch1/metabolismo , Fatores de Transcrição/metabolismo , Animais , Blastocisto/citologia , Fator de Transcrição CDX2 , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Ectoderma/citologia , Ectoderma/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Via de Sinalização Hippo , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Proteínas Serina-Treonina Quinases/genética , Receptor Notch1/genética , Fatores de Transcrição de Domínio TEA , Fatores de Transcrição/genética , Transcrição Gênica
10.
Nat Struct Mol Biol ; 18(6): 708-14, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21602820

RESUMO

Many genomic alterations associated with human diseases localize in noncoding regulatory elements located far from the promoters they regulate, making it challenging to link noncoding mutations or risk-associated variants with target genes. The range of action of a given set of enhancers is thought to be defined by insulator elements bound by the 11 zinc-finger nuclear factor CCCTC-binding protein (CTCF). Here we analyzed the genomic distribution of CTCF in various human, mouse and chicken cell types, demonstrating the existence of evolutionarily conserved CTCF-bound sites beyond mammals. These sites preferentially flank transcription factor-encoding genes, often associated with human diseases, and function as enhancer blockers in vivo, suggesting that they act as evolutionarily invariant gene boundaries. We then applied this concept to predict and functionally demonstrate that the polymorphic variants associated with multiple sclerosis located within the EVI5 gene impinge on the adjacent gene GFI1.


Assuntos
DNA/metabolismo , Genoma , Proteínas Repressoras/metabolismo , Animais , Fator de Ligação a CCCTC , Proteínas de Ciclo Celular , Linhagem Celular , Galinhas , Sequência Conservada , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas Ativadoras de GTPase , Humanos , Camundongos , Esclerose Múltipla/patologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Polimorfismo Genético , Ligação Proteica , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
11.
Cell Cycle ; 10(11): 1731-8, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21527827

RESUMO

Early mammalian development is characterized by a highly specific stage, the blastocyst, by which embryonic and extraembryonic lineages have been determined, but pattern formation has not yet begun. The blastocyst is also of interest because cell precursors of the embryo proper retain for a certain time the capability to generate all the cell types of the adult animal. This embryonic pluripotency is established and maintained by a regulatory network under the control of a small set of transcription factors, comprising Oct4, Sox2 and Nanog. This network is largely conserved in eutherian mammals, but there is scarce information about how it arose in vertebrates. We have analysed the conservation of gene regulatory networks controlling blastocyst lineages and pluripotency in the mouse by comparison with the chick. We found that few of elements of the network are novel to mammals; rather, most of them were present before the separation of the mammalian lineage from other amniotes, but acquired novel expression domains during early mammalian development. Our results strongly support the hypothesis that mammalian blastocyst regulatory networks evolved through rewiring of pre-existing components, involving the co-option and duplication of existing genes and the establishment of new regulatory interactions among them.


Assuntos
Evolução Biológica , Blastocisto/citologia , Linhagem da Célula/genética , Células-Tronco Pluripotentes/citologia , Animais , Redes Reguladoras de Genes/genética , Humanos , Mamíferos , Fatores de Transcrição/genética
12.
Proc Natl Acad Sci U S A ; 107(46): 19955-60, 2010 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-21048080

RESUMO

Embryonic pluripotency in the mouse is established and maintained by a gene-regulatory network under the control of a core set of transcription factors that include octamer-binding protein 4 (Oct4; official name POU domain, class 5, transcription factor 1, Pou5f1), sex-determining region Y (SRY)-box containing gene 2 (Sox2), and homeobox protein Nanog. Although this network is largely conserved in eutherian mammals, very little information is available regarding its evolutionary conservation in other vertebrates. We have compared the embryonic pluripotency networks in mouse and chick by means of expression analysis in the pregastrulation chicken embryo, genomic comparisons, and functional assays of pluripotency-related regulatory elements in ES cells and blastocysts. We find that multiple components of the network are either novel to mammals or have acquired novel expression domains in early developmental stages of the mouse. We also find that the downstream action of the mouse core pluripotency factors is mediated largely by genomic sequence elements nonconserved with chick. In the case of Sox2 and Fgf4, we find that elements driving expression in embryonic pluripotent cells have evolved by a small number of nucleotide changes that create novel binding sites for core factors. Our results show that the network in charge of embryonic pluripotency is an evolutionary novelty of mammals that is related to the comparatively extended period during which mammalian embryonic cells need to be maintained in an undetermined state before engaging in early differentiation events.


Assuntos
Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Evolução Molecular , Redes Reguladoras de Genes , Mamíferos/embriologia , Mamíferos/genética , Células-Tronco Pluripotentes/metabolismo , Animais , Sequência de Bases , Embrião de Galinha , Sequência Conservada/genética , Elementos Facilitadores Genéticos/genética , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/metabolismo , Gastrulação/genética , Regulação da Expressão Gênica no Desenvolvimento , Genoma/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Dados de Sequência Molecular , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Especificidade da Espécie
13.
Dev Dyn ; 239(2): 620-9, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20014105

RESUMO

In the mouse blastocyst, Eomes and Cdx2 are critical for establishing the trophoectoderm, the precursor of the placenta. To better understand how the trophoectoderm lineage arose in mammals during evolution, we examined the expression of their orthologues in the pregastrulation chick embryo and found that, while both genes are expressed in extraembryonic tissues, their temporal pattern of expression differs from what occurs in mouse. Moreover, we failed to detect expression of other genes specific from the mouse trophoectoderm in extraembryonic regions of the chick. Also unlike the mouse, chick Eomes is expressed in primordial germ cells. Finally, conserved noncoding elements in the Eomes genomic region are unable to drive trophoectoderm restricted expression in the mouse blastocyst, but do so in conserved sites of expression such as the forebrain. These results suggest that critical changes in the gene regulatory networks controlling extraembryonic development accompanied the appearance of the trophoectoderm in mammals.


Assuntos
Evolução Biológica , Linhagem da Célula , Embrião de Galinha/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/metabolismo , Animais , Blastocisto/metabolismo , Fator de Transcrição CDX2 , Sequência Conservada , Elementos Facilitadores Genéticos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Placenta/embriologia , Gravidez
14.
Dev Dyn ; 235(10): 2889-94, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16921504

RESUMO

Nanog is a pluripotency-associated factor expressed in embryonic stem cells and in the epiblast and primordial germ cells of the mouse embryo. We have identified the chick orthologue of Nanog and found that its expression is limited to primordial germ cells in the early embryo and is not found throughout the epiblast. Genomic analysis has shown that Nanog is an amniote-specific gene and is absent from anamniotes and invertebrates. Furthermore, other pluripotency associated genes that are located in close proximity to Nanog in human and mouse are absent from the chick genome. Such observations lead to a scenario of sequential addition of novel genes to a genomic region associated to pluripotency. These results have profound implications for the study of the evolution of pluripotent lineages in the embryo and of vertebrate stem cells.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Células Germinativas/metabolismo , Proteínas de Homeodomínio/genética , Fatores de Transcrição/genética , Sequência de Aminoácidos , Animais , Embrião de Galinha , Proteínas de Ligação a DNA/genética , Etiquetas de Sequências Expressas , Humanos , Camundongos , Modelos Genéticos , Dados de Sequência Molecular , Proteína Homeobox Nanog , Gambás/embriologia , Gambás/genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
15.
Hear Res ; 196(1-2): 19-25, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15464297

RESUMO

Insulin-like growth factors (IGFs) have a pivotal role during nervous system development and in its functional maintenance. IGF-I and its high affinity receptor (IGF1R) are expressed in the developing inner ear and in the postnatal cochlear and vestibular ganglia. We recently showed that trophic support by IGF-I is essential for the early neurogenesis of the chick cochleovestibular ganglion (CVG). In the chicken embryo otic vesicle, IGF-I regulates developmental death dynamics by regulating the activity and/or levels of key intracellular molecules, including lipid and protein kinases such as ceramide kinase, Akt and Jun N-terminal kinase (JNK). Mice lacking IGF-I lose many auditory neurons and present increased auditory thresholds at early postnatal ages. Neuronal loss associated to IGF-I deficiency is caused by apoptosis of the auditory neurons, which presented abnormally increased levels of activated caspase-3. It is worth noting that in man, homozygous deletion of the IGF-1 gene causes sensory-neural deafness. IGF-I is thus necessary for normal development and maintenance of the inner ear. The trophic actions of IGF-I in the inner ear suggest that this factor may have therapeutic potential for the treatment of hearing loss.


Assuntos
Orelha Interna/embriologia , Fator de Crescimento Insulin-Like I/fisiologia , Animais , Animais Recém-Nascidos/crescimento & desenvolvimento , Senescência Celular/fisiologia , Cóclea/citologia , Cóclea/crescimento & desenvolvimento , Desenvolvimento Embrionário/fisiologia
16.
J Cell Sci ; 116(Pt 3): 475-86, 2003 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-12508109

RESUMO

Nerve growth factor induces cell death in organotypic cultures of otic vesicle explants. This cell death has a restricted pattern that reproduces the in vivo pattern of apoptosis occurring during inner ear development. In this study, we show that binding of nerve growth factor to its low affinity p75 neurotrophin receptor is essential to achieve the apoptotic response. Blockage of binding to p75 receptor neutralized nerve-growth-factor-induced cell death, as measured by immunoassays detecting the presence of cytosolic oligonucleosomes and by TUNEL assay to visualize DNA fragmentation. Nerve growth factor also induced a number of cell-death-related intracellular events including ceramide generation, caspase activation and poly-(ADP ribose) polymerase cleavage. Again, p75 receptor blockade completely abolished all of these effects. Concerning the intracellular pathway, ceramide increase depended on initiator caspases, whereas its actions depended on both initiator and effector caspases, as shown by using site-specific caspase inhibitors. Conversely, insulin-like growth factor I, which promotes cell growth and survival in the inner ear, abolished apoptosis induced by nerve growth factor. Insulin-like growth factor cytoprotective actions were accomplished, at least in part, by decreasing endogenous ceramide levels and activating Akt. Taken together, these results strongly suggest that regulation of nerve-growth-factor-induced apoptosis in the otocysts occurs via p75 receptor binding and is strictly controlled by the interaction with survival signalling pathways.


Assuntos
Apoptose/fisiologia , Orelha Interna/embriologia , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Neural/metabolismo , Proteínas Serina-Treonina Quinases , Receptor de Fator de Crescimento Neural/metabolismo , Animais , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/fisiologia , Caspases/efeitos dos fármacos , Caspases/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Ceramidas/metabolismo , Embrião de Galinha , Fragmentação do DNA/efeitos dos fármacos , Fragmentação do DNA/fisiologia , Interações Medicamentosas/fisiologia , Orelha Interna/efeitos dos fármacos , Orelha Interna/metabolismo , Inibidores Enzimáticos/farmacologia , Fator de Crescimento Insulin-Like I/farmacologia , Fator de Crescimento Neural/farmacologia , Técnicas de Cultura de Órgãos , Poli(ADP-Ribose) Polimerases/efeitos dos fármacos , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Receptor de Fator de Crescimento Neural/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA