Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Respir Res ; 25(1): 237, 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38849894

RESUMO

Pulmonary hypertension (PH) refers to a group of deadly lung diseases characterized by vascular lesions in the microvasculature and a progressive increase in pulmonary vascular resistance. The prevalence of PH has increased over time. Currently, the treatment options available for PH patients have limited efficacy, and none of them can fundamentally reverse pulmonary vascular remodeling. Stem cells represent an ideal seed with proven efficacy in clinical studies focusing on liver, cardiovascular, and nerve diseases. Since the potential therapeutic effect of mesenchymal stem cells (MSCs) on PH was first reported in 2006, many studies have demonstrated the efficacy of stem cells in PH animal models and suggested that stem cells can help slow the deterioration of lung tissue. Existing PH treatment studies basically focus on the paracrine action of stem cells, including protein regulation, exosome pathway, and cell signaling; however, the specific mechanisms have not yet been clarified. Apoptotic and afunctional pulmonary microvascular endothelial cells (PMVECs) and alveolar epithelial cells (AECs) are two fundamental promoters of PH although they have not been extensively studied by researchers. This review mainly focuses on the supportive communication and interaction between PMVECs and AECs as well as the potential restorative effect of stem cells on their injury. In the future, more studies are needed to prove these effects and explore more radical cures for PH.


Assuntos
Hipertensão Pulmonar , Humanos , Hipertensão Pulmonar/terapia , Hipertensão Pulmonar/metabolismo , Animais , Transplante de Células-Tronco/métodos , Transplante de Células-Tronco Mesenquimais/métodos
2.
Stem Cells ; 32(2): 402-13, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24123810

RESUMO

Human induced pluripotent stem cells (hiPSCs) have great therapeutic potential in repairing defective lung alveoli. However, genetic abnormalities caused by vector integrations and low efficiency in generating hiPSCs, as well as difficulty in obtaining transplantable hiPSC-derived cell types are still major obstacles. Here we report a novel strategy using a single nonviral site-specific targeting vector with a combination of Tet-On inducible gene expression system, Cre/lox P switching gene expression system, and alveolar epithelial type II cell (ATIIC)-specific Neomycin(R) transgene expression system. With this strategy, a single copy of all of the required transgenes can be specifically knocked into a site immediately downstream of ß-2-microglobulin (B2M) gene locus at a high frequency, without causing B2M dysfunction. Thus, the expression of reprogramming factors, Oct4, Sox2, cMyc, and Klf4, can be precisely regulated for efficient reprogramming of somatic cells into random integration-free or genetic mutation-free hiPSCs. The exogenous reprogramming factor transgenes can be subsequently removed after reprogramming by transient expression of Cre recombinase, and the resulting random integration-free and exogenous reprogramming factor-free hiPSCs can be selectively differentiated into a homogenous population of ATIICs. In addition, we show that these hiPSC-derived ATIICs exhibit ultrastructural characteristics and biological functions of normal ATIICs. When transplanted into bleomycin-challenged mice lungs, hiPSC-derived ATIICs efficiently remain and re-epithelialize injured alveoli to restore pulmonary function, preventing lung fibrosis and increasing survival without tumorigenic side effect. This strategy allows for the first time efficient generation of patient-specific ATIICs for possible future clinical applications.


Assuntos
Diferenciação Celular/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Alvéolos Pulmonares/metabolismo , Animais , Linhagem Celular , Reprogramação Celular/genética , Células Epiteliais/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Integrases/genética , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/biossíntese , Fatores de Transcrição Kruppel-Like/genética , Pulmão , Camundongos , Fator 3 de Transcrição de Octâmero/genética , Fatores de Transcrição SOXB1/biossíntese , Fatores de Transcrição SOXB1/genética
3.
Annu Rev Med ; 62: 95-105, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21226612

RESUMO

Embryonic stem (ES) cells derived from preimplantation blastocysts and induced pluripotent stem (iPS) cells generated from somatic cell sources are pluripotent and capable of indefinite expansion in vitro. They provide a possible unlimited source of cells that could be differentiated into lung progenitor cells for potential clinical use in pulmonary regenerative medicine. Because of inherent difficulties in deriving endodermal cells from undifferentiated cell cultures, applications using lung epithelial cells derived from ES and iPS cells have lagged behind similar efforts devoted to other tissues, such as the heart and spinal cord. However, during the past several years, significant advances in culture, differentiation, and purification protocols, as well as in bioengineering methodologies, have fueled enthusiasm for the development of stem cell-based lung therapeutics. This article provides an overview of recent research achievements and discusses future technical challenges that must be met before the promise of stem cell applications for lung disease can be realized.


Assuntos
Células-Tronco Embrionárias/fisiologia , Células Epiteliais/citologia , Células Epiteliais/transplante , Células-Tronco Pluripotentes Induzidas/fisiologia , Pneumopatias/terapia , Animais , Técnicas de Cultura de Células/métodos , Células-Tronco Embrionárias/citologia , Células Epiteliais/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Pulmão/anatomia & histologia , Pulmão/fisiologia , Camundongos , Ratos , Medicina Regenerativa/métodos , Engenharia Tecidual/métodos
4.
Stem Cell Rev Rep ; 18(8): 2629-2645, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35871209

RESUMO

Several attempts have been made to reconstruct the whole lung using pluripotent stem cells (PSCs) to treat terminal stage diseases, such as chronic obstructive pulmonary disease [COPD] and idiopathic pulmonary fibrosis [IPF], for which whole-organ transplantation is currently the only treatment option. The development of induced differentiation technologies has made it possible to regenerate lungs from the 'bottom-up' via stepwise protocols. Nonetheless, the earliest lung multipotent progenitors, namely lung primordial stem cells, have not been identified to date. Considering the intricate crosstalk network that regulates lung development, stepwise protocols to differentiate PSCs into lung progenitors have raised some key questions: (1) the heterogeneity of these induced progenitors, and (2) obtaining a high-purity population. One important strategy to overcome these hurdles is to identify relevant markers or factors that regulate the complex network in lung morphogenesis according to those erected in vivo and ex vivo experiments. For screening lung primordial stem cells, several markers are 'on the shelf', and this review explores the most common or substantiated candidates. We artificially divided these markers into positive selecting and negative limiting proximal or distal markers as well as early progenitor markers that can be used to identify lung primordial stem cell, which represents the earliest progenitor during lung morphogenesis.


Assuntos
Fibrose Pulmonar Idiopática , Células-Tronco Pluripotentes , Humanos , Diferenciação Celular , Pulmão , Biomarcadores
5.
Stem Cell Res Ther ; 13(1): 492, 2022 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-36195893

RESUMO

Pulmonary fibrosis (PF) is a chronic and relentlessly progressive interstitial lung disease in which the accumulation of fibroblasts and extracellular matrix (ECM) induces the destruction of normal alveolar structures, ultimately leading to respiratory failure. Patients with advanced PF are unable to perform physical labor and often have concomitant cough and dyspnea, which markedly impair their quality of life. However, there is a paucity of available pharmacological therapies, and to date, lung transplantation remains the only possible treatment for patients suffering from end-stage PF; moreover, the complexity of transplantation surgery and the paucity of donors greatly restrict the application of this treatment. Therefore, there is a pressing need for alternative therapeutic strategies for this complex disease. Due to their capacity for pluripotency and paracrine actions, stem cells are promising therapeutic agents for the treatment of interstitial lung disease, and an extensive body of literature supports the therapeutic efficacy of stem cells in lung fibrosis. Although stem cell transplantation may play an important role in the treatment of PF, some key issues, such as safety and therapeutic efficacy, remain to be resolved. In this review, we summarize recent preclinical and clinical studies on the stem cell-mediated regeneration of fibrotic lungs and present an analysis of concerning issues related to stem cell therapy to guide therapeutic development for this complex disease.


Assuntos
Transplante de Células-Tronco Mesenquimais , Fibrose Pulmonar , Fibroblastos , Humanos , Fibrose Pulmonar/terapia , Qualidade de Vida
6.
Front Oncol ; 12: 988528, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36353537

RESUMO

Although arsenic trioxide (ATO) shows a strong anti-tumor effect in the treatment of acute promyelocytic leukemia, it does not benefit patients with hepatocellular carcinoma (HCC). Thus, combination therapy is proposed to enhance the efficacy of ATO. Parthenolide (PTL), a natural compound, selectively eradicates cancer cells and cancer stem cells with no toxicity to normal cells. In this study, we chose PTL and ATO in combination and found that nontoxic dosage of PTL and ATO co-treatment can synergistically inhibit the in vitro and in vivo proliferation activity of HCC cells through suppressing stemness and self-renewal ability and inducing mitochondria-dependent apoptosis. More importantly, USP7-HUWE1-p53 pathway is involved in PTL enhancing ATO-induced apoptosis of HCC cell lines. Meanwhile, accompanied by induction of apoptosis, PTL and ATO evoke autophagic activity via inhibiting PI3K/Akt/mTOR pathway, and consciously controlling autophagy can improve the anti-HCC efficacy of a combination of PTL and ATO. In short, our conclusion represents a novel promising approach to the treatment of HCC.

7.
J Immunol ; 182(10): 6533-9, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19414808

RESUMO

Carboxypeptidase N (CPN) is a plasma zinc metalloprotease, which consists of two enzymatically active small subunits (CPN1) and two large subunits (CPN2) that protect the protein from degradation. Historically, CPN has been implicated as a major regulator of inflammation by its enzymatic cleavage of functionally important arginine and lysine amino acids from potent phlogistic molecules, such as the complement anaphylatoxins C3a and C5a. Because of no known complete CPN deficiencies, the biological impact of CPN in vivo has been difficult to evaluate. Here, we report the generation of a mouse with complete CPN deficiency by targeted disruption of the CPN1 gene. CPN1(-/-) mice were hypersensitive to lethal anaphylactic shock due to acute complement activation by cobra venom factor. This hypersensitivity was completely resolved in CPN1(-/-)/C5aR(-/-) but not in CPN1(-/-)/C3aR(-/-) mice. Moreover, CPN1(-/-) mice given C5a i.v., but not C3a, experienced 100% mortality. This C5a-induced mortality was reduced to 20% when CPN1(-/-) mice were treated with an antihistamine before C5a challenge. These studies describe for the first time a complete deficiency of CPN and demonstrate 1) that CPN plays a requisite role in regulating the lethal effects of anaphylatoxin-mediated shock, 2) that these lethal effects are mediated predominantly by C5a-induced histamine release, and 3) that C3a does not contribute significantly to shock following acute complement activation.


Assuntos
Complemento C5a/metabolismo , Lisina Carboxipeptidase/genética , Choque/genética , Animais , Southern Blotting , Complemento C3a/imunologia , Complemento C3a/metabolismo , Complemento C5a/imunologia , Inativadores do Complemento/toxicidade , Suscetibilidade a Doenças/imunologia , Venenos Elapídicos/toxicidade , Feminino , Histamina/imunologia , Histamina/metabolismo , Humanos , Lisina Carboxipeptidase/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Choque/imunologia
8.
Mol Ther ; 18(3): 625-34, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20087316

RESUMO

Respiratory diseases are a major cause of mortality and morbidity worldwide. Current treatments offer no prospect of cure or disease reversal. Transplantation of pulmonary progenitor cells derived from human embryonic stem cells (hESCs) may provide a novel approach to regenerate endogenous lung cells destroyed by injury and disease. Here, we examine the therapeutic potential of alveolar type II epithelial cells derived from hESCs (hES-ATIICs) in a mouse model of acute lung injury. When transplanted into lungs of mice subjected to bleomycin (BLM)-induced acute lung injury, hES-ATIICs behaved as normal primary ATIICs, differentiating into cells expressing phenotypic markers of alveolar type I epithelial cells. Without experiencing tumorigenic side effects, lung injury was abrogated in mice transplanted with hES-ATIICs, demonstrated by recovery of body weight and arterial blood oxygen saturation, decreased collagen deposition, and increased survival. Therefore, transplantation of hES-ATIICs shows promise as an effective therapeutic to treat acute lung injury.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Células-Tronco Embrionárias/citologia , Células Epiteliais/citologia , Alvéolos Pulmonares/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Humanos , Camundongos , Oxigênio/metabolismo , Fenótipo , Regiões Promotoras Genéticas , Transplante de Células-Tronco/métodos , Transfecção , Transgenes
9.
Mol Cell Biol ; 24(13): 6021-8, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15199155

RESUMO

SUMO (also called Sentrin) is a ubiquitin-like protein that plays an important role in regulating protein function and localization. It is known that several nuclear receptors are modified by SUMO; however, the effect of desumoylation in regulating nuclear receptor function has not been elucidated. Here we show that androgen receptor (AR)-mediated transcription is markedly enhanced by SENP1, a member of SUMO-specific protease family. SENP1's ability to enhance AR-dependent transcription is not mediated through desumoylation of AR, but rather through its ability to deconjugate histone deacetylase 1 (HDAC1), thereby reducing its deacetylase activity. HDAC1's repressive effect on AR-dependent transcription could be reversed by SENP1 and by deletion of its sumoylation sites. RNA interference depletion of endogenous HDAC1 also reduced SENP1's effect. Thus, SENP1 could regulate AR-dependent transcription through desumoylation of HDAC1. These studies provide insights on the potential role of desumoylation in the regulation of nuclear receptor activity.


Assuntos
Endopeptidases/fisiologia , Histona Desacetilases/metabolismo , Receptores Androgênicos/fisiologia , Transcrição Gênica , Linhagem Celular Tumoral , Cisteína Endopeptidases , Regulação da Expressão Gênica , Histona Desacetilase 1 , Histona Desacetilases/genética , Humanos , RNA Interferente Pequeno/farmacologia , Receptores Citoplasmáticos e Nucleares , Proteína SUMO-1/metabolismo , Ubiquitinas/metabolismo
10.
Stem Cell Res Ther ; 8(1): 138, 2017 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-28595637

RESUMO

BACKGROUND: Pathways directing endogenous stem/progenitor cells to restore normal architecture and function of damaged/diseased lungs remain underexplored. Published data have revealed that alveolar progenitor type II cell (ATIIC)-derived signaling promotes re-epithelialization of injured alveoli, yet the underlying mechanism is unknown. Here we aim to define the role of ATIIC-derived exosome miRNA signaling in controlling ATIIC-specific proliferation or differentiation in response to injury. METHODS: Pluripotent stem cell-derived cultures, which contain early lung stem/progenitor populations that can subsequently differentiate into ATIICs, were used as a model for unbiased screening and identification of ATIIC phenotype-specific exosome miRNA signaling, and human induced pluripotent stem cell-derived ATIICs (hiPSC-ATIICs) were employed to examine the molecular basis of key exosome miRNA signaling in promoting ATIIC-specific proliferation. QRT-PCR was performed to examine expression pattern of ATIIC-derived key exosome miRNA in an alveolar injury model and in injured human lungs. RESULTS: We show that human ATIIC line (A549)-derived exosome miR-371b-5p promotes ATIIC-specific proliferation, but not differentiation, in differentiating cultures of pluripotent stem cells. Using 3'UTR-driven luciferase reporters, we identified PTEN as a direct target of miR-371b-5p. Transfection of miR-371b-5p mimic into hiPSC-ATIICs leads to significantly decreased expression of endogenous PTEN, which stimulates phosphorylation of Akt and its downstream substrates, GSK3ß and FOXOs, promoting cell proliferation. While not expressed in normal ATIIC phenotypes, the exosome miR-371b-5p expression is significantly induced after hiPSC-ATIICs or hATIICs (human primary ATIICs) are subjected to bleomycin-induced injury. To rule out that the ATIIC-derived exosome-miRNAs are merely a cell culture phenomenon, we transplanted hiPSC-ATIICs into bleomycin-challenged lungs of mice, and found that the transplanted hiPSC-ATIICs engraft and express exosome miR-371b-5p, along with additional survival of numerous mouse ATIICs in bleomycin-injured lungs. Consistent with these findings, significant levels of exosome miR-371b-5p were also detected in lavage samples of patients with acute pneumonia, but not in those from patients without pulmonary disorders. CONCLUSIONS: Collectively, our data strongly suggest that ATIIC-derived exosome miR-371b-5p may serve as a niche signaling to augment ATIIC survival/proliferation, promoting re-epithelialization of injured alveoli, and thus provide a promising novel target to develop treatment for currently incurable lung diseases.


Assuntos
Diferenciação Celular/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Pneumopatias/genética , MicroRNAs/genética , Regiões 3' não Traduzidas , Animais , Proliferação de Células/genética , Exossomos/genética , Exossomos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Pulmão/citologia , Pulmão/metabolismo , Pneumopatias/patologia , Camundongos , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/metabolismo , Transdução de Sinais/genética , Células-Tronco/metabolismo
11.
Circulation ; 110(25): 3803-7, 2004 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-15596566

RESUMO

BACKGROUND: Adult human peripheral blood CD34-positive (CD34+) cells appear to transform into cardiomyocytes in the injured hearts of severe combined immunodeficient mice. It remains unclear, however, whether the apparent transformation is the result of transdifferentiation of the donor stem cells or of fusion of the donor cell with the cardiomyocyte of the recipients. METHODS AND RESULTS: We performed flow cytometry analyses of cells isolated from the hearts of mice that received human CD34+ cells. Human HLA-ABC antigen and cardiac troponin T or Nkx2.5 were used as markers for cardiomyocytes derived from human CD34+ cells, and HLA-ABC and VE-cadherin were used to identify the transformed endothelial cells. The double-positive cells were collected and interphase fluorescence in situ hybridization was used to detect the expression of human and mouse X chromosomes in these cells. We found that 73.3% of nuclei derived from HLA+ and troponin T+ or Nkx2.5+ cardiomyocytes contain both human and mouse X chromosomes and 23.7% contain only human X chromosome. In contrast, the nuclei of HLA-, troponin T+ cells contain only mouse X chromosomes. Furthermore, 97.3% of endothelial cells derived from CD34+ cells contained human X chromosome only. CONCLUSIONS: Thus, both cell fusion and transdifferentiation may account for the transformation of peripheral blood CD34+ cells into cardiomyocytes in vivo.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Infarto do Miocárdio/terapia , Miócitos Cardíacos/citologia , Animais , Antígenos CD , Antígenos CD34/análise , Biomarcadores , Caderinas/análise , Diferenciação Celular/fisiologia , Fusão Celular , Células Cultivadas/citologia , Cromossomos Humanos/química , Células Endoteliais/química , Células Endoteliais/ultraestrutura , Feminino , Sobrevivência de Enxerto , Antígenos HLA/análise , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/análise , Humanos , Células Híbridas/química , Células Híbridas/ultraestrutura , Hibridização in Situ Fluorescente , Interfase , Camundongos , Camundongos Endogâmicos C3H , Camundongos SCID , Infarto do Miocárdio/patologia , Miocárdio/patologia , Especificidade da Espécie , Fatores de Transcrição/análise , Transplante Heterólogo , Troponina T/análise , Cromossomo X
12.
Stem Cells Transl Med ; 4(10): 1234-45, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26285657

RESUMO

UNLABELLED: Human embryonic stem cells (hESCs) are a promising source of cells for tissue regeneration, yet histoincompatibility remains a major challenge to their clinical application. Because the human leukocyte antigen class I (HLA-I) molecules are the primary mediators of immune rejection, we hypothesized that cells derived from a hESC line lacking HLA-I expression could be transplanted without evoking a robust immune response from allogeneic recipients. In the present study, we used the replacement targeting strategy to delete exons 2 and 3 of ß2-microglobulin on both gene alleles in hESCs. Because ß2-microglobulin serves as the HLA-I light chain, disruption of the ß2-microglobulin gene led to complete HLA-I deficiency on the cell surface of hESCs and their derivatives. Therefore, these cells were resistant to CD8+ T-cell-mediated destruction. Although interferon-γ (IFN-γ) treatment significantly induced ß2-microglobulin expression, promoting CD8+ T cell-mediated killing of control hESCs and their derivatives, CD8+ T-cell-mediated cytotoxicity was barely observed with ß2-microglobulin-null hESCs and their derivatives treated with IFN-γ. This genetic manipulation to disrupt HLA-I expression did not affect the self-renewal capacity, genomic stability, or pluripotency of hESCs. Despite being relatively sensitive to natural killer (NK) cell-mediated killing due to the lack of HLA-I expression, when transplanted into NK cell-depleted immunocompetent mice, ß2-microglobulin-null hESCs developed into tumors resembling those derived from control hESCs in severe combined immunodeficiency mice. These results demonstrate that ß2-microglobulin-null hESCs significantly reduce immunogenicity to CD8+ T cells and might provide a renewable source of cells for tissue regeneration without the need for HLA matching in the future. SIGNIFICANCE: This study reports the generation of a novel ß2-microglobulin (B2M)-/- human embryonic stem cell (hESC) line. Differentiated mature cells from this line do not express cell surface human leukocyte antigen molecules even after interferon-γ stimulation and are resistant to alloreactive CD8+ T cells. Moreover, this B2M-/- hESC line contains no off-target integration or cleavage events, is devoid of stable B2M mRNA, exhibits a normal karyotype, and retains its self-renewal capacity, genomic stability, and pluripotency. Although B2M-/- hESC-derived cells are more susceptible to natural killer (NK) cells, murine transplantation studies have indicated that they are, overall, much less immunogenic than normal hESCs. Thus, these data show for the first time that, in vivo, the advantages provided by B2M-/- hESC-derived cells in avoiding CD8+ T-cell killing appear significantly greater than any disadvantage caused by increased susceptibility to NK cells.


Assuntos
Técnicas de Silenciamento de Genes , Células-Tronco Embrionárias Humanas/imunologia , Microglobulina beta-2/genética , Alelos , Animais , Linfócitos T CD8-Positivos/imunologia , Autorrenovação Celular , Sobrevivência Celular , Transplante de Células/efeitos adversos , Citotoxicidade Imunológica , Éxons/genética , Perfilação da Expressão Gênica , Vetores Genéticos , Instabilidade Genômica , Rejeição de Enxerto/prevenção & controle , Antígenos HLA , Xenoenxertos , Histocompatibilidade , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/transplante , Humanos , Interferon gama/farmacologia , Células Matadoras Naturais/imunologia , Camundongos , Camundongos SCID , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/imunologia , Células-Tronco Pluripotentes/transplante , Deleção de Sequência , Teratoma/etiologia , Teratoma/imunologia , Microglobulina beta-2/fisiologia
13.
Clin Transl Med ; 3: 15, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24995122

RESUMO

Lung possesses very limited regenerative capacity. Failure to maintain homeostasis of lung epithelial cell populations has been implicated in the development of many life-threatening pulmonary diseases leading to substantial morbidity and mortality worldwide, and currently there is no known cure for these end-stage pulmonary diseases. Embryonic stem cells (ESCs) and somatic cell-derived induced pluripotent stem cells (iPSCs) possess unlimited self-renewal capacity and great potential to differentiate to various cell types of three embryonic germ layers (ectodermal, mesodermal, and endodermal). Therapeutic use of human ESC/iPSC-derived lung progenitor cells for regeneration of injured or diseased lungs will have an enormous clinical impact. This article provides an overview of recent advances in research on pluripotent stem cells in lung tissue regeneration and discusses technical challenges that must be overcome for their clinical applications in the future.

14.
Tissue Eng Part C Methods ; 20(6): 464-72, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24102479

RESUMO

The use of embryonic stem cells (ESCs) to regenerate distal lung epithelia damaged by injuries or diseases requires development of safe and efficient methodologies that direct ESC differentiation into transplantable distal lung epithelial progenitors. Time-consuming culture procedure and low differentiation efficiency are major problems that are associated with conventional differentiation approaches via embryoid body formation. The use of a growth factor cocktail or a lung-specific cell-conditioned medium to enrich definitive endoderm for efficient differentiation of mouse ESCs (mESC) into alveolar epithelial progenitor type II cells (ATIICs) has been reported, but not yet successful for generating a homogenous population of ATIICs for tissue regeneration purpose, and it remains unclear whether or not those mESC-derived ATIICs possess normal biological functions. Here, we report a novel method using a genetically modified mESC line harboring an ATIIC-specific neomycin(R) transgene in Rosa 26 locus. We showed that ATIICs can be efficiently differentiated from mESCs as early as day 7 by culturing them directly on Matrigel-coated plates in DMEM containing 15% knockout serum replacement. With this culture condition, the genetically modified mESCs can be selectively differentiated into a homogenous population (>99%) of ATIICs. Importantly, the mESC-derived ATIICs (mESC-ATIICs) exhibited typical lamellar bodies and expressed surfactant protein A, B, and C as normal control ATIICs. When cultured with an air-liquid-interface culture system in Small Airway Epithelial Cell Growth Medium, the mESC-ATIICs can be induced to secrete surfactant proteins after being treated with dibutyryl cAMP+dexamethasone. These mESC-ATIICs can synthesize and secrete surfactant lipid in response to secretagogue, demonstrating active surfactant metabolism in mESC-ATIICs as that seen in normal control ATIICs. In addition, we demonstrated that the selected mESC-ATIICs can be maintained on Matrigel-coated plates for at least 4 days with robust proliferative capacity. When cultured in DMEM medium containing 10% FBS, mESC-ATIICs spontaneously differentiated into alveolar epithelial type I cells. Collectively, these data demonstrate that the genetically modified mESCs can be selectively differentiated into a homogenous population of functional ATIICs, providing a reliable cell source to explore their therapeutic potential in lung tissue regeneration.


Assuntos
Técnicas de Cultura Celular por Lotes/métodos , Células-Tronco Embrionárias/citologia , Alvéolos Pulmonares/citologia , Mucosa Respiratória/citologia , Engenharia Tecidual/métodos , Animais , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Separação Celular/métodos , Sobrevivência Celular/fisiologia , Células Cultivadas , Células-Tronco Embrionárias/fisiologia , Melhoramento Genético/métodos , Camundongos , Camundongos Endogâmicos C57BL , Alvéolos Pulmonares/fisiologia , Mucosa Respiratória/fisiologia
15.
Proc Natl Acad Sci U S A ; 104(11): 4449-54, 2007 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-17360544

RESUMO

Alveolar epithelial type II (ATII) cells are small, cuboidal cells that constitute approximately 60% of the pulmonary alveolar epithelium. These cells are crucial for repair of the injured alveolus by differentiating into alveolar epithelial type I cells. ATII cells derived from human ES (hES) cells are a promising source of cells that could be used therapeutically to treat distal lung diseases. We have developed a reliable transfection and culture procedure, which facilitates, via genetic selection, the differentiation of hES cells into an essentially pure (>99%) population of ATII cells (hES-ATII). Purity, as well as biological features and morphological characteristics of normal ATII cells, was demonstrated for the hES-ATII cells, including lamellar body formation, expression of surfactant proteins A, B, and C, alpha-1-antitrypsin, and the cystic fibrosis transmembrane conductance receptor, as well as the synthesis and secretion of complement proteins C3 and C5. Collectively, these data document the successful generation of a pure population of ATII cells derived from hES cells, providing a practical source of ATII cells to explore in disease models their potential in the regeneration and repair of the injured alveolus and in the therapeutic treatment of genetic diseases affecting the lung.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Embrionárias/citologia , Alvéolos Pulmonares/citologia , Diferenciação Celular , Linhagem Celular , Complemento C3/metabolismo , Complemento C5/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Humanos , Pulmão/metabolismo , Microscopia Eletrônica de Transmissão , Modelos Genéticos , Regiões Promotoras Genéticas , Tensoativos/química , alfa 1-Antitripsina/metabolismo
16.
Mol Microbiol ; 47(5): 1407-17, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12603744

RESUMO

The vls locus of Borrelia burgdorferi B31 consists of 15 silent cassettes and one expression site (vlsE), and the presence of the encoding plasmid lp28-1 correlates with high infectivity. Recombination between the expression cassette and silent cassettes occurs in vivo, and this process may enable B. burgdorferi to evade the immune response. To determine the characteristics of the vls loci in other Borrelia strains, we have cloned and characterized the vls silent cassette loci of Borrelia garinii Ip90 and Borrelia afzelii ACAI, consisting of 11 vls silent cassettes and 14 vls silent cassettes respectively. The silent cassettes share 90-97% nucleotide sequence identity with one another within the Ip90 vls locus and 84-91% within the ACAI vls locus. In both organisms, the silent cassettes resemble the B31 Vls sequences in overall amino acid similarity (50-65%) and in the presence of six variable regions interspersed between six relatively invariant regions. The vlsE expression sites of these two strains have not been isolated, but transcripts of vlsE were detected by reverse transcriptase-polymerase chain reaction for both Ip90 and ACAI. In addition, the occurrence of sequence variation within the vlsE cassette region of these transcripts was verified. This study indicates that the vls loci present in B. garinii Ip90 and B. afzelii ACAI have characteristics similar to those found in B. burgdorferi B31.


Assuntos
Variação Antigênica/genética , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Grupo Borrelia Burgdorferi/genética , Genes Bacterianos , Lipoproteínas/genética , Doença de Lyme/microbiologia , Acrodermatite/microbiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Borrelia burgdorferi/genética , Borrelia burgdorferi/imunologia , Grupo Borrelia Burgdorferi/imunologia , Regulação Bacteriana da Expressão Gênica , Humanos , Dados de Sequência Molecular , Recombinação Genética , Sequências Repetitivas de Ácido Nucleico , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico , Especificidade da Espécie , Telômero/genética , Carrapatos/microbiologia , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA