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.
Angiogenesis ; 25(1): 113-128, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34478025

RESUMO

Embryonic stem cells (ES) are a valuable source of endothelial cells. By co-culturing ES cells with the stromal PA6 cells, the endothelial commitment can be achieved by adding exogenous FGF2 or BMP4. In this work, the molecular pathways that direct the differentiation of ES cells toward endothelium in response to FGF2 are evaluated and compared to those activated by BMP4. To this purpose the genes expression profiles of both ES/PA6 co-cultures and of pure cultures of PA6 cells were obtained by microarray technique at different time points. The bioinformatics processing of the data indicated TGFß1 as the most represented upstream regulator in FGF2-induced endothelial commitment while WNT pathway as the most represented in BMP4-activated endothelial differentiation. Loss of function experiments were performed to validate the importance of TGFß1 and WNT6 respectively in FGF2 and BMP4-induced endothelial differentiation. The loss of TGFß1 expression significantly impaired the accomplishment of the endothelial commitment unless exogenous recombinant TGFß1 was added to the culture medium. Similarly, silencing WNT6 expression partially affected the endothelial differentiation of the ES cells upon BMP4 stimulation. Such dysfunction was recovered by the addition of recombinant WNT6 to the culture medium. The ES/PA6 co-culture system recreates an in vitro complete microenvironment in which endothelial commitment is accomplished in response to alternative signals through different mechanisms. Given the importance of WNT and TGFß1 in mediating the crosstalk between tumor and stromal cells this work adds new insights in the mechanism of tumor angiogenesis and of its possible inhibition.


Assuntos
Células Endoteliais , Fator 2 de Crescimento de Fibroblastos , Fator de Crescimento Transformador beta1/fisiologia , Animais , Proteína Morfogenética Óssea 4 , Diferenciação Celular , Células-Tronco Embrionárias , Fator 2 de Crescimento de Fibroblastos/farmacologia , Camundongos , Proteínas Proto-Oncogênicas , Células Estromais , Fator de Crescimento Transformador beta1/genética , Proteínas Wnt
2.
Cytotherapy ; 13(9): 1140-52, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21846293

RESUMO

BACKGROUND AIMS: Bone marrow (BM)-derived cells appear to be a promising therapeutic source for the treatment of acute myocardial infarction (AMI). However, the quantity and quality of the cells to be used, along with the appropriate time of administration, still need to be defined. We thus investigated the use of BM CD34(+)-derived cells as cells suitable for a cell therapy protocol (CTP) in the treatment of experimental AMI. METHODS: The need for a large number of cells was satisfied by the use of a previously established protocol allowing the expansion of human CD34(+) cells isolated from neonatal and adult hematopoietic tissues. We evaluated gene expression, endothelial differentiation potential and cytokine release by BM-derived cells during in vitro culture. Basal and expanded CD34(+) cells were used as a delivery product in a murine AMI model consisting of a coronary artery ligation (CAL). Cardiac function recovery was evaluated after injecting basal or expanded cells. RESULTS: Gene expression analysis of in vitro-expanded cells revealed that endothelial markers were up-regulated during culture. Moreover, expanded cells generated a CD14(+) subpopulation able to differentiate efficiently into VE-cadherin-expressing cells. In vivo, we observed a cardiac function recovery in mice sequentially treated with basal and expanded cells injected 4 h and 7 days after CAL, respectively. CONCLUSIONS: Our data suggest that combining basal and expanded BM-derived CD34(+) cells in a specific temporal pattern of administration might represent a promising strategy for a successful cell-based therapy.


Assuntos
Vasos Coronários/cirurgia , Ligadura , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Infarto do Miocárdio/terapia , Animais , Antígenos CD/metabolismo , Antígenos CD34/biossíntese , Medula Óssea/patologia , Caderinas/metabolismo , Linhagem Celular , Proliferação de Células , Modelos Animais de Doenças , Endotélio/metabolismo , Perfilação da Expressão Gênica , Humanos , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Endogâmicos NOD , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Recuperação de Função Fisiológica
3.
J Cell Mol Med ; 15(10): 2200-15, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21070596

RESUMO

The connections existing between vessels and nerves go beyond the structural architecture of vascular and nervous systems to comprise cell fate determination. The analysis of functional/molecular links that interconnect endothelial and neural commitments requires a model in which the two differentiation programs take place at the same time in an artificial controllable environment. To this regard, this work presents an in vitro model to differentiate embryonic stem (ES) cells simultaneously into mature neurons and endothelial cells. Murine ES cells are differentiated within an artificial environment composed of PA6 stromal cells and a serum-free medium. Upon these basal culture conditions ES cells preferentially differentiate into neurons. The addition of basic fibroblast growth factor (FGF2) to the medium allows the simultaneous maturation of neurons and endothelial cells, whereas bone morphogenetic protein (BMP)4 drives endothelial differentiation to the disadvantage of neural commitment. The responsiveness of the system to exogenous cytokines was confirmed by genes expression analysis that revealed a significant up-regulation of endothelial genes in presence of FGF2 and a massive down-regulation of the neural markers in response to BMP4. Furthermore, the role played by single genes in determining endothelial and neural fate can be easily explored by knocking down the expression of the target gene with lentiviruses carrying the corresponding shRNA sequence. The possibility to address the neural and the endothelial fate separately or simultaneously by exogenous stimuli combined with an efficient gene silencing strategy make this model an optimal tool to identify environmental signals and genes pathways involved in both endothelial and neural specification.


Assuntos
Células-Tronco Embrionárias/citologia , Endotélio/citologia , Endotélio/crescimento & desenvolvimento , Neurônios/citologia , Animais , Biomarcadores/metabolismo , Proteína Morfogenética Óssea 4/farmacologia , Técnicas de Cultura de Células , Diferenciação Celular , Fator 2 de Crescimento de Fibroblastos/farmacologia , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Camundongos , Células Estromais/citologia
4.
Microvasc Res ; 79(3): 178-83, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20053368

RESUMO

Embryonic stem (ES) cells are pluripotent cells capable of differentiating in all the cell types present in a living organism. They derive from the inner cell mass of blastocysts of different species including humans. Given their unlimited potential, ES cells represent an invaluable resource of different cell types for transplantation and tissue engineering applications. However, in order to accomplish these therapeutic purposes, efficient and controlled in vitro systems of directing ES cell differentiation are mandatory. ES cell differentiation is strongly influenced by physical, chemical and cellular signals provided by the local microenvironment. Understanding the relationships occurring between differentiating cells and surrounding environment is pivotal for a successful ES cells-based therapy. This review describes three different methods of in vitro differentiation of ES cells by outlining the environmental elements required for endothelial fate specification. For each system, the efficiency of endothelial differentiation, the accessibility and the advantages are discussed. The main conclusion that arises from this analysis is that the knowledge of the role played by microenvironment in cell fate determination is essential to control and take advantage of ES cells potential.


Assuntos
Técnicas de Cultura de Células , Diferenciação Celular , Células-Tronco Embrionárias/fisiologia , Células Endoteliais/fisiologia , Nicho de Células-Tronco , Animais , Comunicação Celular , Linhagem da Célula , Meios de Cultura/metabolismo , Células-Tronco Embrionárias/metabolismo , Células Endoteliais/metabolismo , Matriz Extracelular/metabolismo , Humanos , Camundongos , Neovascularização Fisiológica
5.
FASEB J ; 23(1): 68-78, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18757501

RESUMO

The observation that the architecture of the cardiovascular and nervous systems is drawn by common guidance cues and the closeness between neural progenitors and endothelial cells in the vascular niche strongly suggests the existence of links between endothelial and neural cell fates. We identified an embryonic stem cell-derived discrete, nonclonal cell population expressing the two vascular endothelial growth factor receptors neuropilin-1 (Nrp1) and Flk1 that differentiates in vitro toward endothelial or neural phenotypes depending on microenvironmental cues. When microinjected in the chick embryo, Nrp1(+) cells integrate within the host, developing vessels and brain, and acquire endothelial and neural markers, respectively. These results show that precursors of endothelial cells and precursors of neural cells arise from the same pool of differentiating embryonic stem cells and share the expression of Nrp1 and Flk1. These data reinforce the parallelism between vascular and nervous system at the level of cell fate and commitment and open new perspective in regenerative medicine of neurovascular diseases.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Neuropilina-1/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Diferenciação Celular/fisiologia , Linhagem Celular , Embrião de Galinha , Fibroblastos/metabolismo , Camundongos , Neuropilina-1/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
6.
Gene ; 308: 31-41, 2003 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-12711388

RESUMO

Latent transforming growth factor binding proteins (Ltbp-1, -2, -3 and -4) and fibrillins (Fbn-1 and -2) are structurally related cysteine-rich extracellular matrix proteins that localize to the 10 nm microfibrils. Ltbp-1 is thought to promote the secretion and proper folding of the small latent transforming growth factor beta (TGF-beta) complex (TGF-beta plus its propeptide) and is implicated in sequestering it in the extracellular matrix. Here we report the isolation of the mouse Ltbp-1 complementary DNA (cDNA) and gene. The longer form of the Ltbp-1 cDNA encodes a predicted 1713 amino acid protein containing 18 epidermal growth factor-like repeats, four 8-cysteine domains and several motifs that suggest interactions with alpha(IV)beta(1) and alpha(9)beta(1) integrins. Northern blotting analyses indicate that long and short Ltbp-1 transcripts are widely expressed in adult mouse tissues and most abundantly expressed in heart. Ltbp-1 is a single copy gene that maps to chromosome 17, band E (1-3) and encompasses more than 212 kb. The Ltbp-1 gene contains 34 exons and shows a similar organization to the LTBP-2 gene, suggesting that these genes originated from a common ancestral gene.


Assuntos
Processamento Alternativo , Proteínas de Transporte/genética , Peptídeos e Proteínas de Sinalização Intracelular , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting , Mapeamento Cromossômico , Clonagem Molecular , DNA/química , DNA/genética , DNA Complementar/genética , Éxons , Expressão Gênica , Genes/genética , Hibridização in Situ Fluorescente , Íntrons , Proteínas de Ligação a TGF-beta Latente , Camundongos , Camundongos Endogâmicos , Dados de Sequência Molecular , Isoformas de Proteínas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Análise de Sequência de DNA
7.
Angiogenesis ; 1(1): 102-116, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-14517397

RESUMO

Basic fibroblast growth factor (FGF-2) is expressed in vascular endothelium during tumor neovascularization and angioproliferative diseases, including vascular tumors and Kaposi's sarcoma (KS). We have investigated the in vivo biological consequences of endothelial cell activation by endogenous FGF-2 in a mouse aortic endothelial cell line transfected with a retroviral expression vector harboring a human FGF-2 cDNA and the neomycin resistance gene. FGF-2 transfectants, named pZipbFGF2-MAE cells, caused the rapid growth of highly vascularized, non-infiltrating tumors when injected in nude mice. In contrast, lesions grew poorly when cells were injected in immunocompetent syngeneic animals. Histologically, the tumors had the appearance of hemangioendothelioma with spindled areas resembling KS and with numerous CD31+ blood vessels and lacunae. Southern blot analysis of tumor DNA, as well as disaggregation of the lesion followed by in vitro cell culture, revealed that less than 10% of the cells in the tumor mass retain FGF-2 overexpression and neomycin resistance at 6-8 weeks post-injection. Nevertheless, in vitro G418 selection allowed the isolation from the tumor of a FGF-2-overexpressing cell population showing biochemical and biological characteristics similar to those of pZipbFGF2-MAE cells, including the capacity to originate vascular lesions when re-injected in nude mice. To evaluate the effect of angiostatic compounds on the growth and vascularization of pZipbFGF2-MAE cell-induced lesions, nude mice were treated weekly (100mg/kg, i.p.) with the angiostatic sulfonated distamycin A derivative 2,2'-(carbonyl-bis-[imino-N-methyl-4,2-pyrrole carbonyl-imino-{N-methyl-4,2-pyrrole}carbonylimino])-bis-(1,5-naphthalene) disulfonic acid (PNU 153429). The results demonstrate that PNU 153429 inhibits the growth of the lesions and causes a approximately 50% decrease in CD31+ microvessel density. In conclusion, the data indicate that FGF-2-overexpressing endothelial cells cause vascular lesions in immunodeficient mice which may represent a novel model for opportunistic vascular tumors suitable for the evaluation of angiostatic compounds.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...