Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Exp Lung Res ; 36(5): 307-20, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20497026

RESUMO

The fibroblast growth factors (FGFs) play an important role in the development of embryonic lung. In this study, we investigated the effects of mainly FGF 1, 2, and 10 at concentrations selected on the basis of data obtained from previous in vitro culture on the derivation of the pulmonary progenitors from murine embryonic stem cells cultured on gelatin or Matrigel-coated plates. For cells cultured on a gelatin-coated plate, high concentrations of FGF1 were found to enhance the expression of mRNAs for SPC and CC10, markers of distal airway epithelium, while high levels of FGF2 decreased the expression of RNAs for not only SPC, CC10 but also for the additional markers SPD and aquaporin 5. FGF10 at all tested concentrations was found to have no effect on the differentiation of pneumocytes when ESCs were grown on gelatin-coated plates. However, when differentiation was performed on Matrigel-coated plates, the addition of 60 ng/ml FGF10 enhanced the expression of pneumocyte markers, suggesting a synergic effect of FGF10 and extracellular matrix. In conclusion, growth factors were proven to be effective in the differentiation of pulmonary progenitors from mESCs. The need of signals from extracellular matrix proteins depends on the growth factors supplemented.


Assuntos
Células Epiteliais Alveolares/metabolismo , Diferenciação Celular , Células-Tronco Embrionárias/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Pulmão/metabolismo , Animais , Diferenciação Celular/genética , Linhagem Celular , Fator 1 de Crescimento de Fibroblastos/metabolismo , Fator 10 de Crescimento de Fibroblastos/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intercelular , Pulmão/citologia , Camundongos , Proteínas Nucleares/metabolismo , Peptídeos/genética , Peptídeos/metabolismo , Reação em Cadeia da Polimerase , Proteína C Associada a Surfactante Pulmonar , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/metabolismo , Fator Nuclear 1 de Tireoide , Fatores de Tempo , Fatores de Transcrição/metabolismo , Transfecção
2.
Biomaterials ; 29(29): 3946-52, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18639332

RESUMO

The majority of methodologies for maintaining human embryonic stem cell (hESC) pluripotency require the use of human or animal feeder cell layers, the most common being murine embryonic fibroblasts. In this study, we applied a protocol aimed at maintaining hESCs in culture without exposure to animal cells or proteins. hESCs were encapsulated in 1.1% (w/v) calcium alginate hydrogels and grown in basic maintenance medium for a period of up to 260 days. Investigation of the cell aggregates formed within the hydrogels yielded no evidence of the formation of any of the three germ layers, although the hESCs retained their pluripotency and could differentiate when they were subsequently cultured in a conditioned environment. Immunohistochemistry and RT-PCR showed that the hESC aggregates expressed protein and gene markers characteristic of pluripotency including Oct-4, Nanog, SSEA-4, TRA-1-60 and TRA-1-81. At the ultrastructural level, the cells were arranged in closely packed clusters and showed no cytoplasmic organelles, suggesting an undifferentiated state. These data show that it is possible to maintain hESCs in an undifferentiated state, without passaging or embryoid body formation, and without animal contamination.


Assuntos
Técnicas de Cultura de Células/métodos , Técnicas de Cocultura , Células-Tronco Embrionárias/fisiologia , Células-Tronco Pluripotentes/fisiologia , Animais , Biomarcadores/metabolismo , Forma Celular , Células-Tronco Embrionárias/citologia , Expressão Gênica , Humanos , Hidrogéis/química , Hidrogéis/metabolismo , Teste de Materiais , Camundongos , Células-Tronco Pluripotentes/citologia
3.
Cloning Stem Cells ; 9(3): 407-16, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17907951

RESUMO

We have previously shown that embryonic stem cells (ESC) can be directed to differentiate into alveolar type II cells by provision of a serum-free medium designed for in vitro maintenance of mature alveolar epithelial cells (small airway growth medium: SAGM), although the target cell yield was low. SAGM comprises a basal serum-free medium (SABM) plus a series of defined supplements. In order to try increase the proportion of pneumocytes in differentiated cultures, we aimed in this study to determine the effects on murine ESC of each of the individual growth factors in SAGM. In accordance with our previous reports, expression of surfactant protein C (SPC) and its mRNA was used to monitor differentiation of type II pneumocytes. Surprisingly, we found that addition of each factor separately to SABM decreased the expression of SPC mRNA when compared with the effect of SABM alone. Thus, it seems that the observed enhancement by SAGM of pneumocyte differentiation from murine ESC can, in fact, be attributed to the provision of a serum-free environment.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Alvéolos Pulmonares/citologia , Animais , Diferenciação Celular/efeitos dos fármacos , Meios de Cultura , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Camundongos , Peptídeos/genética , Peptídeos/metabolismo , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/metabolismo , Proteína C Associada a Surfactante Pulmonar , RNA Mensageiro/metabolismo
4.
Methods Enzymol ; 418: 333-49, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17141045

RESUMO

Repair or regeneration of defective lung epithelium would be of great therapeutic potential. Cellular sources for such repair have long been searched for within the lung, but the identification and characterization of stem or progenitor cells have been hampered by the complexity and cellular heterogeneity of the organ. In recent years, various pulmonary cells have been identified that meet the criteria for stem cells but it remains to be seen how far manipulation of these tissue-specific cell pools can upregulate epithelial repair. The initial excitement that greeted the results of animal experiments showing cells of bone marrow origin in murine lung has been tempered by more recent data suggesting that the cells do not repair pulmonary epithelium. However, there are reports of engraftment of bone marrow-derived cells in human lung, albeit at a low level, so the administration of cell therapy via the circulation, for repair and/or gene delivery, needs further investigation. The potential of human embryonic stem cells to generate any cell, tissue, or organ on demand for tissue repair or replacement is promising to revolutionize the treatment of human disease. Although some headway has been made into making pulmonary epithelium from these stem cells, human embryonic stem cell technology is still in its infancy and many technical, safety, and ethical hurdles must be cleared before clinical trials can begin. This chapter focuses on the potential role of stem cells in future approaches to lung repair and regeneration.


Assuntos
Pulmão/citologia , Pulmão/fisiologia , Mucosa Respiratória/citologia , Mucosa Respiratória/fisiologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Técnicas de Cocultura/métodos , Meios de Cultura , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Humanos , Mesoderma/citologia , Regeneração , Mucosa Respiratória/transplante , Células-Tronco/citologia , Células-Tronco/fisiologia
5.
Respir Res ; 7: 1, 2006 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-16390543

RESUMO

BACKGROUND: To elucidate further the pathogenesis of sporadic, idiopathic pulmonary arterial hypertension (IPAH) and identify potential therapeutic avenues, differential gene expression in IPAH was examined by suppression subtractive hybridisation (SSH). METHODS: Peripheral lung samples were obtained immediately after removal from patients undergoing lung transplant for IPAH without familial disease, and control tissues consisted of similarly sampled pieces of donor lungs not utilised during transplantation. Pools of lung mRNA from IPAH cases containing plexiform lesions and normal donor lungs were used to generate the tester and driver cDNA libraries, respectively. A subtracted IPAH cDNA library was made by SSH. Clones isolated from this subtracted library were examined for up regulated expression in IPAH using dot blot arrays of positive colony PCR products using both pooled cDNA libraries as probes. Clones verified as being upregulated were sequenced. For two genes the increase in expression was verified by northern blotting and data analysed using Student's unpaired two-tailed t-test. RESULTS: We present preliminary findings concerning candidate genes upregulated in IPAH. Twenty-seven upregulated genes were identified out of 192 clones examined. Upregulation in individual cases of IPAH was shown by northern blot for tissue inhibitor of metalloproteinase-3 and decorin (P < 0.01) compared with the housekeeping gene glyceraldehydes-3-phosphate dehydrogenase. CONCLUSION: Four of the up regulated genes, magic roundabout, hevin, thrombomodulin and sucrose non-fermenting protein-related kinase-1 are expressed specifically by endothelial cells and one, muscleblind-1, by muscle cells, suggesting that they may be associated with plexiform lesions and hypertrophic arterial wall remodelling, respectively.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Hipertensão Pulmonar/genética , Pulmão/fisiologia , Adulto , Citoplasma/fisiologia , Enzimas/genética , Espaço Extracelular/fisiologia , Feminino , Humanos , Hibridização Genética , Hipertensão Pulmonar/fisiopatologia , Masculino , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Proteínas Nucleares/genética , Artéria Pulmonar/fisiopatologia , Regulação para Cima/genética
6.
Tissue Eng ; 12(7): 2025-30, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16889530

RESUMO

Embryonic stem cells (ESCs) are being investigated increasingly for their potential as a cell source for tissue engineering. Antibiotics are regularly used in ESC culture media to control contamination, although they can be cytotoxic and interfere with protein synthesis. Our aim was to examine the effects of the frequently used antibiotics gentamicin and combined penicillin and streptomycin on ESC culture using differentiation of murine ESC into type II pneumocytes as a model. Antibiotics reduced the expression of the specific marker for type II pneumocytes, SPC mRNA, by up to 60%. We also identified an adverse effect on the growth rate of differentiating embryoid bodies, causing a significant ( p < 0.05) reduction of up to 40%, and an increase in population doubling time of up to 48%. No contamination was seen in any of the cultures. Our findings suggest that the routine use of antibiotics in ESC culture should be avoided as it may reduce the efficiency of the culture system.


Assuntos
Antibacterianos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Embrião de Mamíferos/metabolismo , Células-Tronco/metabolismo , Animais , Antígenos de Diferenciação/biossíntese , Linhagem Celular , Embrião de Mamíferos/citologia , Camundongos , Células-Tronco/citologia
7.
Tissue Eng ; 12(6): 1687-97, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16846363

RESUMO

We have previously induced differentiation of embryonic stem cells (ESC) to specific phenotypes by manipulating the culture conditions, including the use of indirect co-culture. In this study, we hypothesized that co-culture with primary chondrocytes can induce human embryonic stem cells (hESC) to differentiate towards the chondrocyte lineage. Co-cultures of hESC and chondrocytes were established using well inserts, with control comprising hESC grown alone or with fibroblasts. After 28 days, after removal of the chondrocyte inserts, hESC differentiation was assessed, by morphology, immunocytochemistry, and reverse transcription polymerase chain reaction. hESC, co-cultured or grown alone, were also implanted into SCID mice on a poly-D, L-lactide scaffold, harvested 35 days later and assessed in the same way. hESC co-cultured with chondrocytes formed colonies and secreted extracellular matrix containing glycosaminoglycans (GAG). Quantitative assay showed increased synthesis of sulfated GAG in co-culture as compared with control hESC grown alone for the same period (p < 0.0001). In addition, co-cultured hESC expressed Sox 9 and collagen type II, unlike control hESC. Co-culture with fibroblasts did not induce chondrogenic differentiation. The implanted constructs with co-cultured hESC contained significantly more type II collagen (p < 0.01), type I collagen (p < 0.05), total collagen (p < 0.01), and GAG (p < 0.01) than those with hESC grown alone. Thus, we show for the first time differentiation of hESC to chondrocytes. Our results confirm the potential of the culture micro-environment to influence ESC differentiation and could provide the basis for future generation of chondrogenic cells for use in tissue repair and increase our understanding of the mechanisms that direct differentiation.


Assuntos
Diferenciação Celular/fisiologia , Condrócitos/citologia , Condrogênese/fisiologia , Células-Tronco/citologia , Linhagem Celular , Células Cultivadas , Técnicas de Cocultura , Humanos
8.
Tissue Eng ; 12(4): 867-75, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16674299

RESUMO

The pluripotency of embryonic stem cells (ESC) is offering new opportunities in tissue engineering and cell therapy. We have shown previously that alveolar epithelial cells, specifically type II pneumocytes, can be derived from murine ESC and hypothesized that a similar protocol could be used successfully on human ESC. Undifferentiated human ESC were induced to form embryoid bodies that were transferred into adherent culture conditions and grown in a medium designed for the maintenance of mature small airway epithelium. On inverted microscopy, the generated cells showed the cobblestone-like morphology of epithelium. The presence of surfactant protein C, a specific marker of type II pneumocytes, and its corresponding RNA were demonstrated by immunostaining and reverse transcription polymerase chain reaction, respectively. Electron microscopy revealed frequent cells with the typical ultrastructure of type II pneumocytes. This study provides evidence for in vitro induction of the differentiation from human ESC of alveolar type II cells, which have the potential for therapeutic use or construction of an in vitro model of human lung.


Assuntos
Embrião de Mamíferos/citologia , Células Epiteliais/citologia , Alvéolos Pulmonares/citologia , Células-Tronco/citologia , Engenharia Tecidual/métodos , Animais , Biomarcadores/metabolismo , Adesão Celular , Técnicas de Cultura de Células , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Colagenases/farmacologia , Meios de Cultura/química , Meios de Cultura/farmacologia , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fibroblastos/citologia , Humanos , Camundongos , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/ultraestrutura , Surfactantes Pulmonares/metabolismo , Proteínas Recombinantes/farmacologia , Fatores de Tempo
9.
Ann N Y Acad Sci ; 1068: 352-66, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16831937

RESUMO

Tissue engineering is an interdisciplinary field that brings together the principles of the life sciences and medicine with those of engineering. The increase in its development over the past decade has resulted from a variety of factors; advances in genomics and proteomics, the advent of new biomaterials as potential templates for tissue growth, improvements in bioreactor design, and increased understanding of healing processes. Possibly the greatest contribution has come from our increased knowledge and understanding of stem cell biology, which is paving the way for the generation of unlimited cells of specific phenotypes for incorporation into engineered tissue constructs. Thus, tissue engineering approaches for expanding and engrafting the differentiated progeny of embryonic, fetal, or adult stem cells have major potential for tissue repair and will make a major contribution to medicine in the 21st century.


Assuntos
Células-Tronco/citologia , Células-Tronco/fisiologia , Engenharia Tecidual/métodos , Animais , Materiais Biocompatíveis , Diferenciação Celular , Sangue Fetal/citologia , Humanos , Recém-Nascido , Engenharia Tecidual/tendências
10.
Methods Mol Biol ; 330: 233-48, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16846028

RESUMO

We present a protocol that has been developed for induction of the differentiation of murine embryonic stem (ES) cells to alveolar type II cells. With this protocol, undifferentiated murine ES cells are induced to form embryoid bodies (EBs). The 10-d-old EBs are transferred to adherent culture conditions and are fed with high-glucose Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% v/v fetal bovine serum, 2 mM L-glutamine, and 0.1 mM 2-mercaptoethanol for 20 d without splitting. Then, the cells are fed with a medium designed for the maintenance and growth of mature distal airway epithelial cells (small airway growth medium, SAGM) for 3 d. Characterization of the alveolar type II cells was done using real-time reverse transcriptase polymerase chain reaction detection of surfactant protein C mRNA and immunocytochemical detection of prosurfactant protein C. Real-time reverse transcriptase polymerase chain reaction revealed that SAGM increases the mRNA expression level of SPC by a factor of 8 when compared to that of cells grown in supplemented high-glucose DMEM (p < 0.05, Student t-test). Immunocytochemistry revealed that proSPC-expressing cells comprised 2.8 +/- 0.23% of the total cell population in SAGM-treated samples and 0.5 +/- 0.1% in samples treated with supplemented high-glucose DMEM (p < 0.05, chi2 test).


Assuntos
Técnicas de Cultura de Células , Diferenciação Celular/fisiologia , Células Epiteliais/fisiologia , Alvéolos Pulmonares/citologia , Células-Tronco/fisiologia , Animais , Células Cultivadas , Embrião de Mamíferos/anatomia & histologia , Células Epiteliais/citologia , Camundongos , Precursores de Proteínas/metabolismo , Proteína C Associada a Surfactante Pulmonar/metabolismo , Células-Tronco/citologia
11.
Transplantation ; 73(11): 1724-9, 2002 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12084993

RESUMO

BACKGROUND: Inflammation, epithelial cell injury, and development of fibrosis and airway obliteration are the major histological features of posttransplant obliterative bronchiolitis (OB). The expression of inducible nitric oxide synthase (iNOS) in the damaged epithelium, accompanied by peroxynitrite, suggests that endogenous nitric oxide (NO) mediates the epithelial destruction preceding obliteration. To elucidate the role of NO in this cascade, heterotopic bronchial allografts were studied in pigs. METHODS: Allografts or autografts were harvested serially 3-90 days after transplantation and processed for histology and immunocytochemistry for iNOS, nitrotyrosine, a marker of peroxynitrite formation, and superoxide dismutase (SOD). RESULTS: During initial ischemic damage to the epithelium, iNOS, nitrotyrosine, and SOD were found to be strongly expressed in the epithelium of all implants as well as later, after partial recovery, parallel to onset of epithelial destruction and subsequent airway obliteration in allografts. The levels of expression of iNOS in fibroblasts during the early phase of obliteration paralleled the onset of fibrosis. Constant expression of iNOS and SOD, but not nitrotyrosine, occurred in autografts and allografts with blocked alloimmune response. CONCLUSIONS: These findings suggest that an excessive amount of NO promotes posttransplant obliterative bronchiolitis by destroying airway epithelium and stimulating fibroblast activity. SOD may provide protection by binding reactive molecules and preventing peroxynitrite formation.


Assuntos
Bronquiolite Obliterante/etiologia , Bronquiolite Obliterante/imunologia , Transplante de Pulmão/efeitos adversos , Óxido Nítrico/metabolismo , Tirosina/análogos & derivados , Animais , Brônquios/metabolismo , Brônquios/patologia , Bronquiolite Obliterante/patologia , Divisão Celular , Modelos Animais de Doenças , Fibroblastos/patologia , Transplante de Pulmão/imunologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Ácido Peroxinitroso/metabolismo , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia , Superóxido Dismutase/metabolismo , Suínos , Tirosina/metabolismo
12.
Tissue Eng ; 10(9-10): 1456-66, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15588405

RESUMO

Osterix is a transcription factor crucial for the normal development of the osteoblast. Here we have investigated whether the osteogenic differentiation of murine embryonic stem (ES) cells can be induced by overexpression of osterix. Differentiation was initiated by formation of embryoid bodies (EB) which were then dispersed and cultured in alpha-minimum essential medium supplemented with L-ascorbate phosphate and alpha-glycerophosphate for up to 21 days. osterix was found to induce expression of several osteoblast-specific markers, as confirmed by immunostaining and real-time RT-PCR. The expression of genes encoding osteocalcin and Cbfa1 was upregulated and the formation of mineralized bone nodules was significantly increased by osterix transfection. In combination with dexamethasone, bone nodule formation was further increased in osterix-transfected cells. Expression of both Sox-9 and PPAR-gamma, genes that are associated with chondrocyte and adipocyte differentiation, was initially increased in the osterix-transfected cells but was downregulated after day 7. This suggests that the process of osterix-induced differentiation of ES cells involves transition through an intermediate bi- or tripotential progenitor cell population. In conclusion, this cell differentiation strategy is useful not only for generating osteoblastic cells from ES cells, but also for investigating factors that influence this process and potentially delineating the ontogeny of the osteoblast.


Assuntos
Osteoblastos/citologia , Osteoblastos/fisiologia , Osteogênese/fisiologia , Células-Tronco/citologia , Células-Tronco/fisiologia , Engenharia Tecidual/métodos , Fatores de Transcrição/metabolismo , Animais , Calcificação Fisiológica/fisiologia , Diferenciação Celular/fisiologia , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Melhoramento Genético/métodos , Camundongos , Camundongos Endogâmicos C3H , Engenharia de Proteínas/métodos , Proteínas Recombinantes/metabolismo , Fator de Transcrição Sp7 , Fatores de Transcrição/genética , Transfecção/métodos
13.
Tissue Eng ; 8(4): 541-50, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12201994

RESUMO

Embryonic stem (ES) cell pluripotency is being investigated increasingly to obtain specific cell lineages for tissue engineering. However, the possibility that ES cells can give rise to lung tissue has not been tested. We hypothesized that lung epithelial cells (type II pneumocytes) can be derived in vitro from murine ES cells. After withdrawal of leukemia inhibitory factor (LIF) and formation of embryoid bodies in maintenance medium for 10, 20, and 30 days, differentiating ES cells were kept in the same medium or transferred to serum-free small airway growth medium (SAGM) for a further 3 or 14 days of culture. The presence of type II pneumocytes in the resulting mixed cultures was demonstrated by reverse transcriptase-polymerase chain reaction (RT-PCR) of surfactant protein C (SPC) mRNA, immunostaining of SPC, and electron microscopy of osmiophilic lamellar bodies only at 30 days sampling time. SAGM appeared to be more favorable for type II cell formation than ES medium. No SPC transcripts were found in differentiating cells grown under the same conditions without formation of embryoid bodies. These findings could form the basis for the enrichment of ES cell-derived cultures with type II pneumocytes, and provide an in vitro system for investigating mechanisms of lung repair and regeneration.


Assuntos
Células Epiteliais/citologia , Alvéolos Pulmonares/citologia , Células-Tronco/citologia , Animais , Biomarcadores , Diferenciação Celular , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Imuno-Histoquímica , Camundongos , Microscopia de Contraste de Fase , Células-Tronco Pluripotentes/citologia , Surfactantes Pulmonares/metabolismo , RNA Mensageiro , Células-Tronco/metabolismo , Células-Tronco/ultraestrutura
14.
Tissue Eng Part C Methods ; 18(4): 263-72, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22047052

RESUMO

Application of stem cells for cell therapy of respiratory diseases is a developing field. We have previously established several protocols for the differentiation of embryonic stem cells (ESC) into alveolar epithelial cells, which require a high degree of operator interference and result in a low yield of target cells. Herein, we have shown that, by provision of a medium conditioned using A549 cells and by integration of classic steps of ESC differentiation into a single step through encapsulation in hydrogels (three-dimensional) and culture in a rotary bioreactor, murine ESC (mESC) could be directed to differentiate into distal respiratory epithelial cells. Type I and II pneumocytes (with a yield of 50% for type II) and Clara cells were demonstrated by the expression of aquaporin 5, surfactant protein C, and Clara cell secretory protein, respectively. We identified target cells as early as day 5 of culture and stably maintained our differentiated cells in vitro for 100 days. Electron microscopy demonstrated microvilli and intracellular lamellar bodies (LB), and fluorescent staining confirmed the active process of exocytosis of these LB in differentiated type II cells. When these cells were decapsulated and cultured in static conditions in flask cultures (two-dimensional), they retained their characteristic type II phenotype and morphology. In conclusion, our protocol offers integrated bioprocessing, shorter time of differentiation, lower cost, no use of growth factors, high reproducibility, and high phenotypic and functional stability, as well as being amenable to automation and being scalable, which would move this field closer to future clinical applications.


Assuntos
Reatores Biológicos , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Alvéolos Pulmonares/citologia , Rotação , Animais , Anticorpos/imunologia , Automação , Agregação Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Células Imobilizadas/citologia , Células Imobilizadas/efeitos dos fármacos , Células Imobilizadas/metabolismo , Meios de Cultivo Condicionados/farmacologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/ultraestrutura , Humanos , Camundongos , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/ultraestrutura , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Tissue Eng Part A ; 16(5): 1515-26, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20001250

RESUMO

We have previously differentiated lung epithelium from human and murine embryonic stem cells (mESCs) and are now exploring the potential applications of these cells, including in the engineering of lung tissue constructs. In this study, we hypothesized that the differentiation and maintenance of lung epithelium derived from mESCs can be enhanced by extracellular matrix (ECM) proteins. Our established differentiation protocol was applied to mESCs grown on a range of ECMs: collagen I, laminin 332, fibronectin, Matrigel, and, as an experimental control, gelatin. The ECMs were coated onto tissue culture plastic (TCP) and poly-DL-lactic acid (PDLLA), a biodegradable polymer we have previously shown to support the growth of mature pneumocytes. Matrigel or Laminin-332 coating of either TCP or PDLLA film resulted in enhanced surfactant protein C gene expression in differentiating mESCs, a direct indication of the upregulation of lung epithelial differentiation. For each combination, changes in the contact angle and zeta potential of protein-coated TCP and PDLLA film confirmed protein adsorption. We conclude that the choice of the coating protein can greatly affect the differentiation of ESCs, and laminin-332-coated PDLLA provided an ECM-degradable scaffold combination that is suitable for engineering of lung tissue constructs.


Assuntos
Células Epiteliais Alveolares/citologia , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Matriz Extracelular/metabolismo , Pulmão/fisiologia , Engenharia Tecidual/métodos , Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/metabolismo , Animais , Bioensaio , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Matriz Extracelular/efeitos dos fármacos , Citometria de Fluxo , Proteínas de Fluorescência Verde/metabolismo , Pulmão/citologia , Pulmão/efeitos dos fármacos , Camundongos , Microscopia de Fluorescência , Poliésteres/farmacologia , Proteína C Associada a Surfactante Pulmonar/genética , Proteína C Associada a Surfactante Pulmonar/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Técnicas de Cultura de Tecidos
16.
Proc Am Thorac Soc ; 5(6): 717-22, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18684724

RESUMO

Embryonic stem cells (ESCs) derived from the preimplantation blastocyst are pluripotent and capable of indefinite expansion in vitro. As such, they present a cell source to derive a potentially inexhaustible supply of pulmonary cells and tissue. ESC-derived pulmonary epithelium could be used for in vitro cell or tissue models or, in the future, implanted into the damaged or diseased lung to effect repair. Efforts to date have largely focused on obtaining distal lung epithelial phenotypes from ESCs, notably alveolar epithelium. Several disparate methods have been developed to enhance differentiation of ESCs into pulmonary epithelial lineages; these are broadly based on recapitulating developmental signaling events, mimicking the physical environment, or forcibly reprogramming the ESC nucleus. Early findings of our preclinical experiments implanting differentiated ESCs into the injured lung are also described here. Future efforts will focus on maximizing ESC differentiation efficiency and yield of the target phenotype, as well as characterizing the function of derived cells in vivo and in vitro.


Assuntos
Células-Tronco Embrionárias/fisiologia , Células-Tronco Embrionárias/transplante , Células Epiteliais/citologia , Regeneração/fisiologia , Mucosa Respiratória/citologia , Mucosa Respiratória/fisiologia , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Humanos , Camundongos
17.
Regen Med ; 2(4): 407-15, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17635048

RESUMO

Repair or regeneration of defective lung tissue would be of great clinical use. Potential cellular sources for the regeneration of lung tissue in vivo or lung tissue engineering in vitro include endogenous pulmonary stem cells, extrapulmonary circulating stem cells and embryonic stem cells. This review summarizes the recent research on each of these stem cell types and their potential for use in the treatment of lung injury and disease.


Assuntos
Pulmão/citologia , Regeneração , Células-Tronco/citologia , Humanos , Pulmão/fisiologia
18.
Curr Protoc Stem Cell Biol ; Chapter 1: Unit 1G.1, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18785171

RESUMO

The potential for embryonic stem (ES) cells to differentiate into cells with a distal lung epithelial phenotype has been demonstrated using different in vitro culture methods. Three separate protocols are described here that utilize both murine and human ES cells. The distal lung epithelial phenotype is induced through the use of embryonic distal lung mesenchyme in coculture systems with differentiating embryoid bodies or the use of soluble factors in defined media to maximize definitive endoderm formation and select and maintain the desired phenotype. Phenotypic analysis is demonstrated using immunocytochemistry and SP-C promoter-eGFP reporter gene expression in transgenic ES cells. These methods provide an increased efficiency of distal lung epithelial derivation from ES cells and, therefore, they provide the foundation for the development of a cell replacement product to treat chronic lung disease or a useful in vitro model for the study of lung disease and development.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Embrionárias/citologia , Pulmão/citologia , Animais , Diferenciação Celular , Técnicas de Cocultura , Primers do DNA/genética , Células-Tronco Embrionárias/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Expressão Gênica , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos , Fenótipo , Proteína C Associada a Surfactante Pulmonar/genética , Proteína C Associada a Surfactante Pulmonar/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Expert Opin Biol Ther ; 6(8): 751-8, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16856797

RESUMO

Repair or regeneration of defective lung epithelium would be of great therapeutic potential. It is estimated by the British Lung Foundation that 1 in 7 people in the UK is affected by a lung disease and that 1 in 4 admissions to children's wards are as a result of respiratory problems. Potential cellular sources for the regeneration of lung tissue in vivo or lung tissue engineering in vitro include endogenous pulmonary epithelial stem cells, extrapulmonary circulating stem cells and embryonic stem cells. This article discusses the potential role of each of these stem cell types in future approaches to the treatment of lung injury and disease.


Assuntos
Embrião de Mamíferos/citologia , Transplante de Pulmão/métodos , Pulmão/patologia , Regeneração , Células-Tronco/citologia , Animais , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Humanos , Camundongos , Engenharia Tecidual/métodos , Obtenção de Tecidos e Órgãos/métodos
20.
Stem Cells ; 24(5): 1389-98, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16456134

RESUMO

Embryonic stem cells (ESCs) are a potential source for the cell-based therapy of a wide variety of lung diseases for which the only current treatment is transplantation. However, distal lung epithelium, like many other endodermally derived somatic cell lineages, is proving difficult to obtain from both murine and human ESCs. We have previously obtained alveolar epithelium from ESCs, although final cell yield remained extremely low. Here, we present an optimized three-step protocol for the derivation of distal lung epithelial cells from murine ESCs. This protocol incorporates (a) treatment of early differentiating embryoid bodies with activin A to enhance the specification of the endodermal germ layer, followed by (b) adherent culture in serum-free medium and (c) the final application of a commercial, lung-specific medium. As well as enhancing the specification of distal lung epithelium, this protocol was found to yield cells with a phenotype most closely resembling that of lung-committed progenitor cells present in the foregut endoderm and the early lung buds during embryonic development. This is in contrast to our previous differentiation method, which drives differentiation through to mature type II alveolar epithelial cells. The derivation of a committed lung progenitor cell type from ESCs is particularly significant for regenerative medicine because the therapeutic implantation of progenitor cells has several clear advantages over the transplantation of mature, terminally differentiated somatic cells.


Assuntos
Diferenciação Celular , Embrião de Mamíferos/citologia , Células Epiteliais/citologia , Pulmão/citologia , Células-Tronco/citologia , Ativinas/farmacologia , Animais , Biomarcadores/análise , Técnicas de Cultura de Células/métodos , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA