Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 8(1): e54957, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23383012

RESUMO

Epigenetic modifications are heritable changes in gene expression without changes in DNA sequence. DNA methylation has been implicated in the control of several cellular processes including differentiation, gene regulation, development, genomic imprinting and X-chromosome inactivation. Methylated cytosine residues at CpG dinucleotides are commonly associated with gene repression; conversely, strategic loss of methylation during development could lead to activation of lineage-specific genes. Evidence is emerging that bone development and growth are programmed; although, interestingly, bone is constantly remodelled throughout life. Using human embryonic stem cells, human fetal bone cells (HFBCs), adult chondrocytes and STRO-1(+) marrow stromal cells from human bone marrow, we have examined a spectrum of developmental stages of femur development and the role of DNA methylation therein. Using pyrosequencing methodology we analysed the status of methylation of genes implicated in bone biology; furthermore, we correlated these methylation levels with gene expression levels using qRT-PCR and protein distribution during fetal development evaluated using immunohistochemistry. We found that during fetal femur development DNA methylation inversely correlates with expression of genes including iNOS (NOS2) and COL9A1, but not catabolic genes including MMP13 and IL1B. Furthermore, significant demethylation was evident in the osteocalcin promoter between the fetal and adult developmental stages. Increased TET1 expression and decreased expression of DNA (cytosine-5-)-methyltransferase 1 (DNMT1) in adult chondrocytes compared to HFBCs could contribute to the loss of methylation observed during fetal development. HFBC multipotency confirms these cells to be an ideal developmental system for investigation of DNA methylation regulation. In conclusion, these findings demonstrate the role of epigenetic regulation, specifically DNA methylation, in bone development, informing and opening new possibilities in development of strategies for bone repair/tissue engineering.


Assuntos
Metilação de DNA/genética , Epigênese Genética , Fêmur/embriologia , Feto/embriologia , Adulto , Células da Medula Óssea/metabolismo , Condrócitos/metabolismo , Colágeno Tipo IX/genética , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , Proteínas de Ligação a DNA/genética , Células-Tronco Embrionárias/metabolismo , Fêmur/metabolismo , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Interleucina-1beta/genética , Metaloproteinase 13 da Matriz/genética , Óxido Nítrico Sintase Tipo II/genética , Osteocalcina/genética , Regiões Promotoras Genéticas/genética
2.
ACS Nano ; 7(3): 1867-81, 2013 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-23414366

RESUMO

Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have the capacity to differentiate into any specialized cell type of the human body, and therefore, ESC/iPSC-derived cell types offer great potential for regenerative medicine. However, key to realizing this potential requires a strong understanding of stem cell biology, techniques to maintain stem cells, and strategies to manipulate cells to efficiently direct cell differentiation toward a desired cell type. As nanoscale science and engineering continues to produce novel nanotechnology platforms, which inform, infiltrate, and impinge on many aspects of everyday life, it is no surprise that stem cell research is turning toward developments in nanotechnology to answer research questions and to overcome obstacles in regenerative medicine. Here we discuss recent advances in ESC and iPSC manipulation using nanomaterials and highlight future challenges within this area of research.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Animais , Diferenciação Celular , Células-Tronco Embrionárias/transplante , Humanos , Células-Tronco Pluripotentes Induzidas/transplante , Nanomedicina , Nanoestruturas , Nanotecnologia , Medicina Regenerativa , Nicho de Células-Tronco , Transplante de Células-Tronco
3.
Small ; 9(12): 2140-51, 2013 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-23362187

RESUMO

The production of bone-forming osteogenic cells for research purposes or transplantation therapies remains a significant challenge. Using planar polycarbonate substrates lacking in topographical cues and substrates displaying a nanotopographical pattern, mesenchymal differentiation of human embryonic stem cells is directed in the absence of chemical factors and without induction of differentiation by embryoid body formation. Cells incubated on nanotopographical substrates show enhanced expression of mesenchymal or stromal markers and expression of early osteogenic progenitors at levels above those detected in cells on planar substrates in the same basal media. Evidence of epithelial-to-mesenchymal transition during substrate differentiation and DNA methylation changes akin to chemical induction are also observed. These studies provide a suitable approach to overcome regenerative medical challenges and describe a defined, reproducible platform for human embryonic stem cell differentiation.


Assuntos
Células-Tronco Embrionárias/citologia , Diferenciação Celular/efeitos dos fármacos , Metilação de DNA , Transição Epitelial-Mesenquimal , Humanos , Células-Tronco Mesenquimais/citologia , Nanotecnologia , Osteogênese/fisiologia
4.
ACS Nano ; 6(11): 10239-49, 2012 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-23072705

RESUMO

It is emerging that mesenchymal stem cell (MSC) metabolic activity may be a key regulator of multipotency. The metabolome represents a "snapshot" of the stem cell phenotype, and therefore metabolic profiling could, through a systems biology approach, offer and highlight critical biochemical pathways for investigation. To date, however, it has remained difficult to undertake unbiased experiments to study MSC multipotency in the absence of strategies to retain multipotency without recourse to soluble factors that can add artifact to experiments. Here we apply a nanotopographical systems approach linked to metabolomics to regulate plasticity and demonstrate rapid metabolite reorganization, allowing rational selection of key biochemical targets of self-renewal (ERK1/2, LDL, and Jnk). We then show that these signaling effectors regulate functional multipotency.


Assuntos
Metaboloma/fisiologia , Nanoestruturas/química , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteogênese/fisiologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Células Cultivadas , Humanos , Nanoestruturas/ultraestrutura , Proteoma , Propriedades de Superfície
5.
Nat Mater ; 10(8): 637-44, 2011 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-21765399

RESUMO

There is currently an unmet need for the supply of autologous, patient-specific stem cells for regenerative therapies in the clinic. Mesenchymal stem cell differentiation can be driven by the material/cell interface suggesting a unique strategy to manipulate stem cells in the absence of complex soluble chemistries or cellular reprogramming. However, so far the derivation and identification of surfaces that allow retention of multipotency of this key regenerative cell type have remained elusive. Adult stem cells spontaneously differentiate in culture, resulting in a rapid diminution of the multipotent cell population and their regenerative capacity. Here we identify a nanostructured surface that retains stem-cell phenotype and maintains stem-cell growth over eight weeks. Furthermore, the study implicates a role for small RNAs in repressing key cell signalling and metabolomic pathways, demonstrating the potential of surfaces as non-invasive tools with which to address the stem cell niche.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais/citologia , Células-Tronco Multipotentes/citologia , Fenótipo , Linhagem da Célula , Proliferação de Células , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Multipotentes/metabolismo , Propriedades de Superfície
6.
Biochem Soc Trans ; 39(2): 674-8, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21428960

RESUMO

ESCs (embryonic stem cells) are derived from the inner cell mass of pre-implantation embryos and are pluripotent, meaning they can differentiate into all of the cells that make up the adult organism. This property of pluripotency makes ESCs attractive as a model system for studying early development and for the generation of specific cell types for use in regenerative medicine and drug screening. In order to harness their potential, the molecular mechanisms regulating ESC pluripotency, proliferation and differentiation (i.e. cell fate) need to be understood so that pluripotency can be maintained during expansion, while differentiation to specific lineages can be induced accurately when required. The present review focuses on the potential roles that PI3K (phosphoinositide 3-kinase) and GSK-3 (glycogen synthase kinase 3)-dependent signalling play in the co-ordination and integration of mouse ESC pluripotency and proliferation and contrast this with our understanding of their functions in human ESCs.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/fisiologia , Quinase 3 da Glicogênio Sintase/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Células-Tronco Pluripotentes/fisiologia , Adulto , Animais , Células-Tronco Embrionárias/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Camundongos , Modelos Biológicos , Fosfatidilinositol 3-Quinases/metabolismo , Células-Tronco Pluripotentes/metabolismo
7.
J Cell Sci ; 122(Pt 13): 2311-21, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19509054

RESUMO

Self-renewal of embryonic stem cells (ESCs) is essential for maintenance of pluripotency, which is defined as the ability to differentiate into any specialised cell type comprising the adult organism. Understanding the mechanisms that regulate ESC self-renewal and proliferation is required before ESCs can fulfil their potential in regenerative therapies, and murine ESCs (mESCs) have been widely used as a model. Members of the class-IA phosphoinositide 3-kinase (PI3K) family of lipid kinases regulate a variety of physiological responses, including cell migration, proliferation and survival. PI3Ks have been reported to regulate both proliferation and self-renewal of mESCs. Here we investigate the contribution of specific class-IA PI3K isoforms to the regulation of mESC fate using small-molecule inhibitors with selectivity for particular class-IA PI3K catalytic isoforms, and siRNA-mediated knockdown. Pharmacological inhibition or knockdown of p110beta promoted mESC differentiation, accompanied by a decrease in expression of Nanog. By comparison, pharmacological inhibition or siRNA-mediated knockdown of p110alpha had no effect on mESC self-renewal per se, but instead appeared to reduce proliferation, which was accompanied by inhibition of leukaemia inhibitory factor (LIF) and insulin-induced PI3K signalling. Our results suggest that PI3Ks contribute to the regulation of both mESC pluripotency and proliferation by differential coupling to selected p110 catalytic isoforms.


Assuntos
Células-Tronco Embrionárias/fisiologia , Isoenzimas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Subunidades Proteicas/metabolismo , Animais , Proliferação de Células , Classe I de Fosfatidilinositol 3-Quinases , Células-Tronco Embrionárias/citologia , Inibidores Enzimáticos/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Isoenzimas/genética , Camundongos , Proteína Homeobox Nanog , Fosfatidilinositol 3-Quinases/genética , Subunidades Proteicas/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...