Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Hum Gene Ther ; 19(1): 39-52, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18052721

RESUMO

Type II alveolar epithelial (AT2) cell-specific reporter expression has been highly useful in the study of embryology and alveolar regeneration in transgenic mice. Technologies enabling efficient gene transfer and cell type-restricted transgene expression in AT2 cells would allow for correction of AT2 cell-based diseases such as genetic surfactant deficiencies. Moreover, such approaches are urgently required to investigate differentiation of AT2 cells from adult and embryonic stem cells of other species than mouse. Using a human surfactant protein C (SP-C) promoter fragment, we have constructed lentiviral vectors enabling AT2-restricted transgene expression and identification of stem cell-derived AT2 cells. Lung epithelial cell lines M3E3/C3, H441, RLE-6TN, A549, MLE-12, and MLE-15 were characterized at the molecular and ultrastructural levels to identify cell lines useful to assess the cell type specificity of our vector constructs. After transduction, no green fluorescent protein (GFP) expression was observed in nontarget cells including bronchial H441 cells, pulmonary A549 cells, fibroblasts, smooth muscle cells, and endothelial cells. In contrast, and in correlation with endogenous SP-C expression, lentiviral transduction resulted in stable GFP expression in MLE-12 and MLE-15 AT2 cells. In conclusion, we have constructed a lentiviral vector mediating SP-C promoter-dependent GFP expression. Transgene expression strictly corresponds with an AT2 phenotype of the transduced cells. In particular, the generated vector should facilitate local alveolar gene therapy and investigation of alveolar regeneration and stem cell differentiation.


Assuntos
Vetores Genéticos , Proteínas de Fluorescência Verde/metabolismo , Lentivirus/genética , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/metabolismo , Proteína C Associada a Surfactante Pulmonar/genética , Animais , Células Cultivadas , Células Epiteliais/metabolismo , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Fenótipo , Regiões Promotoras Genéticas , Ratos , Transdução Genética , Transgenes
2.
Cloning Stem Cells ; 10(1): 49-64, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18241124

RESUMO

Alveolar type II (AT2) epithelial cells have important functions including the production of surfactant and regeneration of lost alveolar type I epithelial cells. The ability of in vitro production of AT2 cells would offer new therapeutic options in treating pulmonary injuries and disorders including genetically based surfactant deficiencies. Aiming at the generation of AT2-like cells, the differentiation of murine embryonic stem cells (mESCs) toward mesendodermal progenitors (MEPs) was optimized using a "Brachyury-eGFP-knock in" mESC line. eGFP expression demonstrated generation of up to 65% MEPs at day 4 after formation of embryoid bodies (EBs) under serum-free conditions. Plated EBs were further differentiated into AT2-like cells for a total of 25 days in serum-free media resulting in the expression of endodermal marker genes (FoxA2, Sox17, TTR, TTF-1) and of markers for distal lung epithelium (surfactant proteins (SP-) A, B, C, and D, CCSP, aquaporin 5). Notably, expression of SP-C as the only known AT2 cell specific marker could be detected after serum-induction as well as under serum-free conditions. Cytoplasmic localization of SP-C was demonstrated by confocal microscopy. The presence of AT2-like cells was confirmed by electron microscopy providing evidence for polarized cells with apical microvilli and lamellar body-like structures. Our results demonstrate the differentiation of AT2-like cells from mESCs after serum-induction and under serum-free conditions. The established serum-free differentiation protocol will facilitate the identification of key differentiation factors leading to a more specific and effective generation of AT2-like cells from ESCs.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Alvéolos Pulmonares/efeitos dos fármacos , Algoritmos , Animais , Técnicas de Cultura de Células , Diferenciação Celular/genética , Células Cultivadas , Meios de Cultura Livres de Soro/farmacologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/fisiologia , Células-Tronco Embrionárias/ultraestrutura , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Células Epiteliais/ultraestrutura , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/fisiologia , Alvéolos Pulmonares/ultraestrutura , Proteína B Associada a Surfactante Pulmonar/metabolismo , Proteína C Associada a Surfactante Pulmonar/metabolismo , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/fisiologia , Soro/fisiologia , Fatores de Tempo
3.
Stem Cells ; 24(6): 1423-32, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16543489

RESUMO

Embryonic stem cells (ESCs) from mice and humans (hESCs) have been shown to be able to efficiently differentiate toward cardiomyocytes (CMs). Because murine ESCs and hESCs do not allow for establishment of pre-clinical allogeneic transplantation models, the aim of our study was to generate functional CMs from rhesus monkey ESCs (rESCs). Although formation of ectodermal and neuronal/glial cells appears to be the default pathway of the rESC line R366.4, we were able to change this commitment and to direct generation of endodermal/mesodermal cells and further differentiation toward CMs. Differentiation of rESCs resulted in an average of 18% of spontaneously contracting embryoid bodies (EBs) from rESCs. Semiquantitative reverse transcription-polymerase chain reaction analyses demonstrated expression of marker genes typical for endoderm, mesoderm, cardiac mesoderm, and CMs, including brachyury, goosecoid, Tbx-5, Tbx-20, Mesp1, Nkx2.5, GATA-4, FOG-2, Mlc2a, MLC2v, ANF, and alpha-MHC in rESC-derived CMs. Immunohistological and ultrastructural studies showed expression of CM-typical proteins, including sarcomeric actinin, troponin T, titin, connexin 43, and cross-striated muscle fibrils. Electrophysiological studies by means of multielectrode arrays revealed evidence of functionality, electrical coupling, and beta-adrenergic signaling of the generated CMs. This is the first study demonstrating generation of functional CMs derived from rESCs. In contrast to hESCs, rESCs allow for establishment of pre-clinical allogeneic transplantation models. Moreover, rESC-derived CMs represent a cell source for the development of high-throughput assays for cardiac safety pharmacology.


Assuntos
Miócitos Cardíacos/citologia , Células-Tronco/citologia , Animais , Diferenciação Celular , Células Cultivadas , Eletrofisiologia , Embrião de Mamíferos/citologia , Expressão Gênica , Humanos , Macaca mulatta , Camundongos , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Miofibrilas/ultraestrutura , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/metabolismo
4.
Stem Cells ; 23(6): 761-71, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15917472

RESUMO

Until recently, culture and expansion of nondifferentiated human embryonic stem cells (hESCs) depended on coculture with murine embryonic fibroblasts. Because mice are known to harbor a variety of pathogens, such culture conditions implicate the risk of xenozoonoses. Among these pathogens, endogenous retroviruses, including murine leukemia viruses (MuLVs), are of special importance. It is well known that some strains cause pathogenic (e.g., leukemic) effects and that xenotropic, polytropic, and amphotropic MuLVs are able to infect human cells. In view of potential clinical applications of hESC lines, it is therefore imperative to investigate potential infection of hESCs by mouse feeder cell-derived viruses. As a first step towards a comprehensive infection risk assessment, we have analyzed embryonic fibroblasts derived from different mouse strains for expression and release of xenotropic, polytropic, and amphotropic MuLVs. Moreover, several hESC lines have been investigated for expression of specific receptors for xenotropic/polytropic MuLVs, as well as for MuLV infection and expression. Evidence for expression of humantropic MuLVs was found in cultures of mouse embryonic fibroblasts (MEFs). Moreover, expression of specific receptors for xenotropic/ polytropic MuLV on human HEK293 and hESC lines and infection after coculture with an MuLV-producing mink cell line could be demonstrated. In contrast, no evidence of MuLV transmission from MEFs to human HEK293 cells or to the hESC lines I-3, I-6, I-8, and H-9 has been obtained. Our results suggest that recently established hESC lines are free of MuLV infections despite long-term close contact with MEFs.


Assuntos
Embrião de Mamíferos/citologia , Vírus da Leucemia Murina/metabolismo , Células-Tronco/citologia , Animais , Diferenciação Celular , Linhagem Celular , Técnicas de Cocultura , Meios de Cultura/farmacologia , DNA Complementar/metabolismo , Embrião de Mamíferos/virologia , Retrovirus Endógenos/genética , Fibroblastos/metabolismo , Humanos , Infecções , Camundongos , Vison , Reação em Cadeia da Polimerase , Retroviridae/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Especificidade da Espécie , Células-Tronco/virologia
5.
Transpl Int ; 17(12): 848-58, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15864489

RESUMO

Clinical pig-to-human xenotransplantation might be associated with the risk of transmission of xenozoonoses, especially porcine endogenous retroviruses (PERVs). We have established a pig-to-humanised-cynomolgus monkey xenotransplantation model allowing the analysis of potential PERV-transmission from normal or transgenic porcine organs to human vascular tissue. Pig-to-human kidney xenotransplantation was performed in cynomolgus monkeys. An interposition graft constructed from a human saphena vein replaced the porcine kidney vein. After graft rejection and/or death of the recipient (survival 2, 4, 6, 13, 16, 19 days), the human interposition grafts were removed. Human endothelial cells (huECs) were isolated from the interposition grafts and cultivated in vitro. Explanted human vascular tissue, isolated huECs, plasma and serum samples of the graft recipients were characterised by flow cytometry and immunohistochemistry and screened for indications of PERV transmission by quantitative polymerase chain reaction (PCR), reverse transcriptase-polymerase chain reaction (RT-PCR) and RT assay. PERV-specific immune response of recipients was analysed by Western blot. No evidence of PERV infection or PERV-specific immune response was detected.


Assuntos
Retrovirus Endógenos , Transplante de Rim/efeitos adversos , Transplante Heterólogo/efeitos adversos , Zoonoses/transmissão , Zoonoses/virologia , Animais , Sobrevivência Celular , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/virologia , Citometria de Fluxo , Rejeição de Enxerto/virologia , Sobrevivência de Enxerto , Humanos , Macaca fascicularis , Complicações Pós-Operatórias/virologia , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA