Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Thorax ; 72(5): 451-459, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28070015

RESUMEN

BACKGROUND: Lung ageing, a significant risk factor for chronic human lung diseases such as COPD and emphysema, is characterised by airspace enlargement and decreasing lung function. Likewise, in prematurely ageing telomerase null (terc-/-) mice, p53 stabilisation within diminishing numbers of alveolar epithelial type 2 cells (AEC2) accompanies reduced lung function. Resveratrol (RSL) is a plant phytoalexin that has previously showed efficacy in enhancing invertebrate longevity and supporting mammalian muscle metabolism when delivered orally. Here, we tested whether inhaled RSL could protect young, terc-/- mice from accelerated ageing of the lung. METHODS: terc-/- mice aged 2 months inhaled 1 mg/kg RSL that was instilled intratracheally once per month for 3 months. One month after the last inhalation, whole lung function, structure and cellular DNA damage were evaluated and AEC2 survival was assessed by western blotting for survival pathway gene expression. RESULTS: RSL treatments delayed the loss of lung compliance (p<0.05), maintained lung structure (p<0.001) and blocked parenchymal cell DNA damage as measured by TdT Nick-End Labeling (TUNEL). RSL, a known agonist of deacetylase SIRT1, supported AEC2 survival by stimulating SIRT1 expression, promoting p53 destabilisation and decreasing Bax expression and by maintaining expression levels of Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), activated p-Akt and p-Mdm2 and inactivated Phospho-Phosphatase and tensin homolog (p-PTEN). CONCLUSIONS: RSL prophylaxis by inhalation is a potential approach for slowing ageing-related deterioration of lung function and structure by maintaining AEC2 integrity.


Asunto(s)
Envejecimiento/efectos de los fármacos , Antioxidantes/administración & dosificación , Antioxidantes/farmacología , Pulmón/efectos de los fármacos , Estilbenos/administración & dosificación , Estilbenos/farmacología , Administración por Inhalación , Animales , Western Blotting , Daño del ADN/efectos de los fármacos , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos C57BL , Pruebas de Función Respiratoria , Resveratrol
2.
Gerontology ; 63(3): 270-280, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27825158

RESUMEN

Natural lung aging is marked by molecular changes that occur during development, maturation, and late-life decline. At the cellular and whole organ level, degenerative changes that are a hallmark of natural aging (shorter telomeres, increased expression of cellular senescence markers, increased DNA damage, oxidative stress, and apoptosis, accompanied by diminished elasticity) reach pathological levels in aging humans in the form of chronic respiratory disease. Aging strongly correlates with the development and incidence of chronic respiratory diseases, including cancer and idiopathic pulmonary fibrosis, but is most strongly linked with development of chronic obstructive pulmonary disease. Lung failure due to aging can be traced to loss of lung stem cell regenerative capacity within the distinctive stem cell niches found within each compartment of the lung. Current knowledge about the identity and function of these stem cell compartments has been largely drawn from a variety of transgenic and spontaneously mutated mouse models that are characterized by rapid rates of aging or have been used to examine regeneration from injury in the context of natural or accelerated aging. While much work has focused on the failure of epithelial cell populations as a key component of the aging process, additional studies have shown that aging, as a global phenomenon in the lung, also impacts resident endothelial, mesenchymal, and immune cell populations. In this review, we examine aging as a process dependent on specific changes in molecular pathways within multiple lung cell populations.


Asunto(s)
Envejecimiento/fisiología , Pulmón/fisiología , Regeneración/fisiología , Envejecimiento/patología , Animales , Senescencia Celular/fisiología , Humanos , Pulmón/patología , Enfermedades Pulmonares/etiología , Enfermedades Pulmonares/patología , Enfermedades Pulmonares/fisiopatología , Ratones , Modelos Animales , Nicho de Células Madre/fisiología
3.
Am J Respir Cell Mol Biol ; 54(3): 319-30, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26203800

RESUMEN

Type 2 alveolar epithelial cells (AEC2) are regarded as the progenitor population of the alveolus responsible for injury repair and homeostatic maintenance. Depletion of this population is hypothesized to underlie various lung pathologies. Current models of lung injury rely on either uncontrolled, nonspecific destruction of alveolar epithelia or on targeted, nontitratable levels of fixed AEC2 ablation. We hypothesized that discrete levels of AEC2 ablation would trigger stereotypical and informative patterns of repair. To this end, we created a transgenic mouse model in which the surfactant protein-C promoter drives expression of a mutant SR39TK herpes simplex virus-1 thymidine kinase specifically in AEC2. Because of the sensitivity of SR39TK, low doses of ganciclovir can be administered to these animals to induce dose-dependent AEC2 depletion ranging from mild (50%) to lethal (82%) levels. We demonstrate that specific levels of AEC2 depletion cause altered expression patterns of apoptosis and repair proteins in surviving AEC2 as well as distinct changes in distal lung morphology, pulmonary function, collagen deposition, and expression of remodeling proteins in whole lung that persist for up to 60 days. We believe SPCTK mice demonstrate the utility of cell-specific expression of the SR39TK transgene for exerting fine control of target cell depletion. Our data demonstrate, for the first time, that specific levels of type 2 alveolar epithelial cell depletion produce characteristic injury repair outcomes. Most importantly, use of these mice will contribute to a better understanding of the role of AEC2 in the initiation of, and response to, lung injury.


Asunto(s)
Células Epiteliales Alveolares/patología , Lesión Pulmonar/patología , Fibrosis Pulmonar/patología , Regeneración , Células Epiteliales Alveolares/enzimología , Animales , Apoptosis , Proliferación Celular , Forma de la Célula , Células Cultivadas , Colágeno/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ganciclovir/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Predisposición Genética a la Enfermedad , Humanos , Hiperoxia/complicaciones , Lesión Pulmonar/genética , Lesión Pulmonar/metabolismo , Lesión Pulmonar/fisiopatología , Ratones Transgénicos , Fenotipo , Regiones Promotoras Genéticas , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/fisiopatología , Proteína C Asociada a Surfactante Pulmonar/genética , Simplexvirus/enzimología , Simplexvirus/genética , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Factores de Tiempo , Proteínas Virales/genética , Proteínas Virales/metabolismo
4.
Br Med Bull ; 101: 147-61, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22279079

RESUMEN

INTRODUCTION OR BACKGROUND: The adult lung is a complex organ whose large surface area interfaces extensively with both the environment and circulatory system. Yet, in spite of the high potential for exposure to environmental or systemic harm, epithelial cell turnover in adult lung is comparatively slow. Moreover, loss of lung function with advancing age is becoming an increasingly costly healthcare problem. Cell-based therapies stimulating endogenous stem/progenitor cells or supplying exogenous ones have therefore become a prime translational goal. Alternatively when lung repair becomes impossible, replacement with tissue-engineered lung is an attractive emerging alternative using a decellularized matrix or bioengineered scaffold. SOURCES OF DATA: Endogenous and exogenous stem cells for lung therapy are being characterized by defining developmental lineages, surface marker expression, functions within the lung and responses to injury and disease. Seeding decellularized lung tissue or bioengineered matrices with various stem and progenitor cells is an approach that has already been used to replace bronchus and trachea in human patients and awaits further development for whole lung tissue. AREAS OF AGREEMENT: Cellular therapies have clear potential for respiratory disease. However, given the surface size and complexity of lung structure, the probability of a single cellular population sufficing to regenerate the entire organ, as in the bone marrow, remains low. Hence, lung regenerative medicine is currently focused around three aims: (i) to identify and stimulate resident cell populations that respond to injury or disease, (ii) to transplant exogenous cells which can ameliorate disease and (iii) to repopulate decellularized or bioengineered lung matrix creating a new implantable organ. AREAS OF CONTROVERSY: Lack of consensus on specific lineage markers for lung stem and progenitor cells in development and disease constrains transferability of research between laboratories and sources of cellular therapy. Furthermore, effectiveness of individual cellular therapies to correct gas exchange and provide other critical lung functions remains unproven. Finally, feasibility of autologous whole organ replacement has not been confirmed as a durable therapy. Growing points Cellular therapies for lung regeneration would be enhanced by better lineage tracing within the lung, the ability to direct differentiation of exogenous stem or progenitor cells, and the development of functional assays for cellular viability and regenerative properties. Whether endogenous or exogeneous cells will ultimately play a greater therapeutic role remains to be seen. Reducing the need for lung replacement via endogenous cell-mediated repair is a key goal. Thereafter, improving the potential of donor lungs in transplant recipients is a further area where cell-based therapies may be beneficial. Ultimately, lung replacement with autologous tissue-engineered lungs is another goal for cell-based therapy. Areas timely for developing research Defining 'lung stem or progenitor cell' populations in both animal models and human tissue may help. Additionally, standardizing assays for assessing the potential of endogenous or exogenous cells within the lung is important. Understanding cell-matrix interactions in real time and with biomechanical insight will be central for lung engineering. Cautionary note Communicating the real potential for cell-based lung therapy needs to remain realistic, given the keen expectations of patients with end-stage lung disease.


Asunto(s)
Enfermedades Pulmonares/terapia , Trasplante de Células Madre/métodos , Animales , Humanos , Pulmón/fisiología , Trasplante de Pulmón/métodos , Ratones , Regeneración , Trasplante de Células Madre/tendencias , Ingeniería de Tejidos/métodos
5.
Am J Respir Cell Mol Biol ; 45(6): 1212-21, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21700959

RESUMEN

Alveolar epithelial integrity is dependent upon the alveolar milieu, yet the milieu of the damaged alveolar epithelial cell type 2 (AEC2) has been little studied. Characterization of its components may offer the potential for ex vivo manipulation of stem cells to optimize their therapeutic potential. We examined the cytokine profile of AEC2 damage milieu, hypothesizing that it would promote endogenous epithelial repair while recruiting cells from other locations and instructing their engraftment and differentiation. Bronchoalveolar lavage and lung extract from hyperoxic rats represented AEC2 in vivo damage milieu, and medium from a scratch-damaged AEC2 monolayer represented in vitro damage. CINC-2 and ICAM, the major cytokines detected by proteomic cytokine array in AEC2 damage milieu, were chemoattractive to normoxic AECs and expedited in vitro wound healing, which was blocked by their respective neutralizing antibodies. The AEC2 damage milieu was also chemotactic for exogenous uncommitted human amniotic fluid stem cells (hAFSCs), increasing migration greater than 20-fold. hAFSCs attached within an in vitro AEC2 wound and expedited wound repair by contributing cytokines migration inhibitory factor and plasminogen activator inhibitor 1 to the AEC2 damage milieu, which promoted wound healing. The AEC2 damage milieu also promoted differentiation of a subpopulation of hAFSCs to express SPC, TTF-1, and ABCA3, phenotypic markers of distal alveolar epithelium. Thus, the microenvironment created by AEC2 damage not only promotes autocrine repair but also can attract uncommitted stem cells, which further augment healing through cytokine secretion and differentiation.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Comunicación Autocrina , Diferenciación Celular , Citocinas/metabolismo , Regeneración , Células Madre/metabolismo , Transportadoras de Casetes de Unión a ATP/biosíntesis , Células Epiteliales Alveolares/patología , Animales , Células Cultivadas , Proteínas de Unión al ADN/biosíntesis , Regulación de la Expresión Génica , Humanos , Hiperoxia/metabolismo , Hiperoxia/patología , Masculino , Proteínas Nucleares/biosíntesis , Ratas , Ratas Sprague-Dawley , Células Madre/patología , Factor Nuclear Tiroideo 1 , Factores de Transcripción/biosíntesis
6.
Am J Physiol Lung Cell Mol Physiol ; 300(6): L898-909, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21460122

RESUMEN

Telomerase mutations and significantly shortened chromosomal telomeres have recently been implicated in human lung pathologies. Natural telomere shortening is an inevitable consequence of aging, which is also a risk factor for development of lung disease. However, the impact of shortened telomeres and telomerase dysfunction on the ability of lung cells to respond to significant challenge is still largely unknown. We have previously shown that lungs of late generation, telomerase null B6.Cg-Terc(tm1Rdp) mice feature alveolar simplification and chronic stress signaling at baseline, a phenocopy of aged lung. To determine the role telomerase plays when the lung is challenged, B6.Cg-Terc(tm1Rdp) mice carrying shortened telomeres and wild-type controls were subjected to partial pneumonectomy. We found that telomerase activity was strongly induced in alveolar epithelial type 2 cells (AEC2) of the remaining lung immediately following surgery. Eighty-six percent of wild-type animals survived the procedure and exhibited a burst of early compensatory growth marked by upregulation of proliferation, stress response, and DNA repair pathways in AEC2. In B6.Cg-Terc(tm1Rdp) mice carrying shortened telomeres, response to pneumonectomy was characterized by decreased survival, diminished compensatory lung growth, attenuated distal lung progenitor cell response, persistent DNA damage, and cell growth arrest. Overall, survival correlated strongly with telomere length. We conclude that functional telomerase and properly maintained telomeres play key roles in both long-term survival and the early phase of compensatory lung growth following partial pneumonectomy.


Asunto(s)
Envejecimiento , Proliferación Celular , Neumonectomía , Alveolos Pulmonares/citología , Telomerasa/fisiología , Telómero/fisiología , Animales , Peso Corporal , Daño del ADN , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos , Alveolos Pulmonares/metabolismo , Transducción de Señal , Células Madre
7.
Am J Respir Crit Care Med ; 180(5): 424-36, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19498056

RESUMEN

RATIONALE: Fibroblast growth factor-10 (FGF10) controls survival, proliferation, and differentiation of distal-alveolar epithelial progenitor cells during lung development. OBJECTIVES: To test for the protective and regenerative effect of Fgf10 overexpression in a bleomycin-induced mouse model of pulmonary inflammation and fibrosis. METHODS: In SP-C-rtTA; tet(O)Fgf10 double-transgenic mice, lung fibrosis was induced in 2-month-old transgenic mice by subcutaneous delivery of bleomycin (BLM), using an osmotic minipump for 1 week. Exogenous Fgf10 expression in the alveolar epithelium was induced for 7 days with doxycycline during the first, second, and third weeks after bleomycin pump implantation, and lungs were examined at 28 days. MEASUREMENTS AND MAIN RESULTS: Fgf10 overexpression during Week 1 (inflammatory phase) resulted in increased survival and attenuated lung fibrosis score and collagen deposition. In these Fgf10-overexpressing mice, an increase in regulatory T cells and a reduction in both transforming growth factor-beta(1) and matrix metalloproteinase-2 activity were observed in bronchoalveolar lavage fluids whereas the number of surfactant protein C (SP-C)-positive, alveolar epithelial type II cells (AEC2) was markedly elevated. Analysis of SP-C and TUNEL (terminal deoxynucleotidyltransferase dUTP nick end labeling) double-positive cells and isolation of AEC2 from lungs overexpressing Fgf10 demonstrated increased AEC2 survival. Expression of Fgf10 during Weeks 2 and 3 (fibrotic phase) showed significant attenuation of the lung fibrosis score and collagen deposition. CONCLUSIONS: In the bleomycin model of lung inflammation and fibrosis, Fgf10 overexpression during both the inflammatory and fibrotic phases results in a greatly reduced extent of lung fibrosis, suggesting that FGF10 may be useful as a novel approach to the treatment of pulmonary fibrosis.


Asunto(s)
Factor 10 de Crecimiento de Fibroblastos/metabolismo , Neumonía/metabolismo , Fibrosis Pulmonar/metabolismo , Animales , Antibióticos Antineoplásicos , Bleomicina , Modelos Animales de Enfermedad , Doxiciclina , Pulmón/patología , Ratones , Ratones Transgénicos , Neumonía/inducido químicamente , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/metabolismo , Fibrosis Pulmonar/inducido químicamente , Valores de Referencia , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Factor de Crecimiento Transformador beta1/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo
8.
Stem Cells ; 26(11): 2902-11, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18719226

RESUMEN

A new source of stem cells has recently been isolated from amniotic fluid; these amniotic fluid stem cells have significant potential for regenerative medicine. These cells are multipotent, showing the ability to differentiate into cell types from each embryonic germ layer. We investigated the ability of human amniotic fluid stem cells (hAFSC) to integrate into murine lung and to differentiate into pulmonary lineages after injury. Using microinjection into cultured mouse embryonic lungs, hAFSC can integrate into the epithelium and express the early human differentiation marker thyroid transcription factor 1 (TTF1). In adult nude mice, following hyperoxia injury, tail vein-injected hAFSC localized in the distal lung and expressed both TTF1 and the type II pneumocyte marker surfactant protein C. Specific damage of Clara cells through naphthalene injury produced integration and differentiation of hAFSC at the bronchioalveolar and bronchial positions with expression of the specific Clara cell 10-kDa protein. These results illustrate the plasticity of hAFSC to respond in different ways to different types of lung damage by expressing specific alveolar versus bronchiolar epithelial cell lineage markers, depending on the type of injury to recipient lung. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Líquido Amniótico/citología , Células Epiteliales/citología , Pulmón/citología , Mucosa Respiratoria/citología , Células Madre/citología , Animales , Diferenciación Celular , Linaje de la Célula , Quimiocina CXCL12/metabolismo , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos , Femenino , Humanos , Pulmón/metabolismo , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/patología , Lesión Pulmonar/terapia , Masculino , Mesodermo/citología , Ratones , Ratones Desnudos , Microinyecciones , Naftalenos , Surfactantes Pulmonares/metabolismo , Receptores CXCR4/metabolismo , Trasplante de Células Madre , Factores de Transcripción
9.
Front Pediatr ; 4: 28, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27066462

RESUMEN

Clinical and basic experimental approaches to pediatric acute lung injury (ALI), including acute respiratory distress syndrome (ARDS), have historically focused on acute care and management of the patient. Additional efforts have focused on the etiology of pediatric ALI and ARDS, clinically defined as diffuse, bilateral diseases of the lung that compromise function leading to severe hypoxemia within 7 days of defined insult. Insults can include ancillary events related to prematurity, can follow trauma and/or transfusion, or can present as sequelae of pulmonary infections and cardiovascular disease and/or injury. Pediatric ALI/ARDS remains one of the leading causes of infant and childhood morbidity and mortality, particularly in the developing world. Though incidence is relatively low, ranging from 2.9 to 9.5 cases/100,000 patients/year, mortality remains high, approaching 35% in some studies. However, this is a significant decrease from the historical mortality rate of over 50%. Several decades of advances in acute management and treatment, as well as better understanding of approaches to ventilation, oxygenation, and surfactant regulation have contributed to improvements in patient recovery. As such, there is a burgeoning interest in the long-term impact of pediatric ALI/ARDS. Chronic pulmonary deficiencies in survivors appear to be caused by inappropriate injury repair, with fibrosis and predisposition to emphysema arising as irreversible secondary events that can severely compromise pulmonary development and function, as well as the overall health of the patient. In this chapter, the long-term effectiveness of current treatments will be examined, as will the potential efficacy of novel, acute, and long-term therapies that support repair and delay or even impede the onset of secondary events, including fibrosis.

10.
Tissue Eng Part C Methods ; 22(11): 1049-1057, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27796199

RESUMEN

The cellular and molecular mechanisms that underpin regeneration of the human lung are unknown, and the study of lung repair has been impeded by the necessity for reductionist models that may exclude key components. We hypothesized that multicellular epithelial and mesenchymal cell clusters or lung organoid units (LuOU) could be transplanted to recapitulate proximal and distal cellular structures of the native lung and airways. Transplantation of LuOU resulted in the growth of tissue-engineered lung (TELu) that contained the necessary cell types consistent with native adult lung tissue and demonstrated proliferative cells at 2 and 4 weeks. This technique recapitulated important elements of both mouse and human lungs featuring key components of both the proximal and distal lung regions. When LuOU were generated from whole lung, TELu contained key epithelial and mesenchymal cell types, and the origin of the cells was traced from both ActinGFP and SPCGFP donors to indicate that the cells in TELu were derived from the transplanted LuOU. Alveolar epithelial type 2 cells (AEC2s), club cells, ciliated cells marked by beta-tubulin IV, alveolar epithelial type I cells, Sox-2-positive proximal airway progenitors, p63-positive basal cells, and CGRP-positive pulmonary neuroendocrine cells were identified in the TELu. The mesenchymal components of peribronchial smooth muscle and nerve were identified with a CD31-positive donor endothelial cell contribution to TELu vasculature. TELu successfully grew from postnatal tissues from whole murine and human lung, distal murine lung, as well as murine and human trachea. These data support a model of postnatal lung regeneration containing the diverse cell types present in the entirety of the respiratory tract.


Asunto(s)
Linaje de la Célula , Proliferación Celular , Pulmón/citología , Ingeniería de Tejidos/métodos , Tráquea/metabolismo , Cicatrización de Heridas , Animales , Células Cultivadas , Humanos , Pulmón/fisiología , Trasplante de Pulmón , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Tráquea/citología
11.
PLoS One ; 11(2): e0148323, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26863115

RESUMEN

RATIONALE: Neonatal respiratory distress syndrome is a restrictive lung disease characterized by surfactant deficiency. Decreased vascular endothelial growth factor (VEGF), which demonstrates important roles in angiogenesis and vasculogenesis, has been implicated in the pathogenesis of restrictive lung diseases. Current animal models investigating VEGF in the etiology and outcomes of RDS require premature delivery, hypoxia, anatomically or temporally limited inhibition, or other supplemental interventions. Consequently, little is known about the isolated effects of chronic VEGF inhibition, started at birth, on subsequent developing lung structure and function. OBJECTIVES: To determine whether inducible, mesenchyme-specific VEGF inhibition in the neonatal mouse lung results in long-term modulation of AECII and whole lung function. METHODS: Triple transgenic mice expressing the soluble VEGF receptor sFlt-1 specifically in the mesenchyme (Dermo-1/rtTA/sFlt-1) were generated and compared to littermate controls at 3 months to determine the impact of neonatal downregulation of mesenchymal VEGF expression on lung structure, cell composition and function. Reduced tissue VEGF bioavailability has previously been demonstrated with this model. MEASUREMENTS AND MAIN RESULTS: Triple transgenic mice demonstrated restrictive lung pathology. No differences in gross vascular development or protein levels of vascular endothelial markers was noted, but there was a significant decrease in perivascular smooth muscle and type I collagen. Mutants had decreased expression levels of surfactant protein C and hypoxia inducible factor 1-alpha without a difference in number of type II pneumocytes. CONCLUSIONS: These data show that mesenchyme-specific inhibition of VEGF in neonatal mice results in late restrictive disease, making this transgenic mouse a novel model for future investigations on the consequences of neonatal RDS and potential interventions.


Asunto(s)
Enfermedades Pulmonares/metabolismo , Pulmón/crecimiento & desarrollo , Pulmón/metabolismo , Mesodermo/metabolismo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Apoptosis , Peso Corporal , Colágeno/química , Femenino , Regulación de la Expresión Génica , Hidroxiprolina/química , Modelos Lineales , Masculino , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa , Pruebas de Función Respiratoria , Transducción de Señal , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética
12.
Cancer Lett ; 338(1): 89-93, 2013 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-22906416

RESUMEN

Epithelial stem cells are critical for tissue generation during development and for repair following injury. In both gestational and postnatal stages, the highly branched and compartmentalized organization of the lung is maintained by multiple, resident stem/progenitor cell populations that are responsible for the homeostatic maintenance and injury repair of pulmonary epithelium. Though lung epithelial injury in the absence of oncogenic mutation is more commonly expressed as chronic lung disease, lung cancer is the most common form of death worldwide and poses a highly significant risk to human health. Cancer is defined by the cell of origin, responsible for initiating the disease. The Cancer Stem Cell Hypothesis proposes that cancer stem cells, identified by stem-like properties of self-renewal and generation of differentiated progeny, are responsible for propagating growth and spread of the disease. In lung cancer, it is hypothesized that cancer stem cells derive from several possible cell sources. The stem cell-like resistance to injury and proliferative potentials of bronchioalveolar stem cells (BASCs) and alveolar epithelial type II cells (AEC2), as well as cells that express the cancer stem cell marker glycoprotein prominin-1 (CD133) or markers for side populations make them potential reservoirs of lung cancer stem cells. The abnormal activation of pathways that normally regulate embryonic lung development, as well as adult tissue maintenance and injury repair, including the Wnt, Hedgehog (Hh) and Notch pathways, has also been identified in lung tumor cells. It is postulated that therapies for lung cancer that specifically target stem cell signaling pathways utilized by lung cancer stem cells could be beneficial in combating this disease.


Asunto(s)
Neoplasias Pulmonares/patología , Células Madre Neoplásicas/patología , Alveolos Pulmonares/patología , Nicho de Células Madre , Transformación Celular Neoplásica , Daño del ADN , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Células Madre Neoplásicas/metabolismo , Alveolos Pulmonares/metabolismo , Especies Reactivas de Oxígeno/metabolismo
13.
PLoS One ; 8(8): e71679, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23967234

RESUMEN

The potential for amniotic fluid stem cell (AFSC) treatment to inhibit the progression of fibrotic lung injury has not been described. We have previously demonstrated that AFSC can attenuate both acute and chronic-fibrotic kidney injury through modification of the cytokine environment. Fibrotic lung injury, such as in Idiopathic Pulmonary Fibrosis (IPF), is mediated through pro-fibrotic and pro-inflammatory cytokine activity. Thus, we hypothesized that AFSC treatment might inhibit the progression of bleomycin-induced pulmonary fibrosis through cytokine modulation. In particular, we aimed to investigate the effect of AFSC treatment on the modulation of the pro-fibrotic cytokine CCL2, which is increased in human IPF patients and is correlated with poor prognoses, advanced disease states and worse fibrotic outcomes. The impacts of intravenous murine AFSC given at acute (day 0) or chronic (day 14) intervention time-points after bleomycin injury were analyzed at either day 3 or day 28 post-injury. Murine AFSC treatment at either day 0 or day 14 post-bleomycin injury significantly inhibited collagen deposition and preserved pulmonary function. CCL2 expression increased in bleomycin-injured bronchoalveolar lavage (BAL), but significantly decreased following AFSC treatment at either day 0 or at day 14. AFSC were observed to localize within fibrotic lesions in the lung, showing preferential targeting of AFSC to the area of fibrosis. We also observed that MMP-2 was transiently increased in BAL following AFSC treatment. Increased MMP-2 activity was further associated with cleavage of CCL2, rendering it a putative antagonist for CCL2/CCR2 signaling, which we surmise is a potential mechanism for CCL2 reduction in BAL following AFSC treatment. Based on this data, we concluded that AFSC have the potential to inhibit the development or progression of fibrosis in a bleomycin injury model during both acute and chronic remodeling events.


Asunto(s)
Líquido Amniótico/citología , Lavado Broncoalveolar , Quimiocina CCL2/metabolismo , Fibrosis Pulmonar/metabolismo , Células Madre/metabolismo , Células Epiteliales Alveolares/metabolismo , Animales , Bleomicina/efectos adversos , Quimiotaxis/inmunología , Técnicas de Cocultivo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Feto , Humanos , Mediadores de Inflamación/metabolismo , Pulmón/metabolismo , Pulmón/patología , Pulmón/fisiopatología , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Modelos Biológicos , Embarazo , Proteolisis , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/terapia , Trasplante de Células Madre , Células Madre/inmunología , Factores de Tiempo
14.
Proc Am Thorac Soc ; 9(3): 117-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22802284

RESUMEN

Fibrotic lung injury is often attributed to a myriad of factors, including environmental exposure, age, genetic predisposition, epigenetics, coexisting conditions, acute lung injury, and viral infection. No effective therapies, other than lung transplantation, have proven effective against lung fibrosis. Loss of cellular homeostasis mechanisms in alveolar epithelial type I cells and any inability of type II progenitor cells to resist and repair epithelial injury are indicators that impaired response to injury and regeneration is a critical component of this disorder. The alveolar epithelium has a limited repertoire of responses to injury, which are dictated by the alveolar milieu, a repository of cytokines and growth factors that affect recruitment of other cells to the site of injury, or the proliferation of resident cells at the site of injury. The identification and characterization of the cytokines, growth factors, and other biomarkers that dictate the response to disease is key to understanding, diagnosing, treating, and determining the trajectory of various lung disorders. Corrective therapy of the alveolar milieu may therefore prove to be beneficial in many presently serious and incurable lung diseases that likely begin and progress with injury to the alveolar epithelium.


Asunto(s)
Lesión Pulmonar/terapia , Alveolos Pulmonares/fisiología , Fibrosis Pulmonar/terapia , Trasplante de Células Madre/métodos , Líquido Amniótico/citología , Humanos , Lesión Pulmonar/diagnóstico , Fibrosis Pulmonar/diagnóstico , Cicatrización de Heridas/fisiología
15.
Methods Mol Biol ; 879: 109-22, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22610556

RESUMEN

The majority of epithelial cells in the distal lung of rodents and humans are quiescent in vivo, yet certain cell populations retain an intrinsic capacity to proliferate and differentiate in response to lung injury or in appropriate culture settings, thus giving them properties of stem/progenitor cells. Here, we describe the isolation of two such populations from adult mouse lung: alveolar epithelial type 2 cells (AEC2), which can generate alveolar epithelial type 1 cells, and bronchioalveolar stem cells (BASCs), which in culture can reproduce themselves, as well as generate a small number of other distal lung epithelial cell types. These primary epithelial cells are typically isolated using enzyme digestion, mechanical disruption, and serial filtration. AEC2 and BASCs are distinguished from other distal lung cells by expression of specific markers as detected by fluorescence-activated cell sorting, immunohistochemistry, or a combination of both of these techniques.


Asunto(s)
Separación Celular/métodos , Pulmón/citología , Células Madre/citología , Animales , Citometría de Flujo , Inmunohistoquímica , Ratones , Ratones Endogámicos
16.
J Invest Surg ; 24(2): 81-6, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21345008

RESUMEN

BACKGROUND: Partial pneumonectomy (PNX) in mice results in compensatory growth in the remaining lung and is a useful model for lung repair. However, common pitfalls to the technique present a challenge for researchers. A complete description of murine PNX is thus provided, with a modification that, in our hands, enhanced animal survival. MATERIALS AND METHODS: 10 ± 2 weeks old mice were anesthetized using 5% inhalational isoflurane via tracheotomy. Mechanical ventilation was provided using a Harvard Model 687 ventilator. In a procedure optimized to be performed in ≤20 min, left lateral thoracotomy was used to access to the left lung, which was grasped with a blunt forceps just distal to the hilum and clipped using a single 5-mm neuro clip. The left lung was resected, leaving a small rim of lung tissue immediately adjacent to the clip. The thoracotomy was closed, and while anesthesia was titrated, sterile saline was injected subcutaneously to replace insensible fluid losses. Upon return of spontaneous breaths, the trachea was decannulated, and the tracheotomy was closed. RESULTS: Even when performed by a single operator, this modified technique produced a survival rate of >85% during the procedure and >90% up to seven days postoperatively in wild-type C57BL/6J mice. CONCLUSIONS: Minimizing the time required to perform left lobe pneumonectomy is critical for animal survival. Using a 5-mm neuro clip, rather than silk suture, to isolate the lobe streamlines the procedure, helps reduce cardiac arrythmia, and results in significantly increased rates of intraoperative and immediate postoperative survival.


Asunto(s)
Neumonectomía/métodos , Animales , Pulmón/fisiología , Ratones , Ratones Endogámicos C57BL , Neumonectomía/veterinaria , Regeneración , Toracotomía/veterinaria
17.
Am J Physiol Lung Cell Mol Physiol ; 296(1): L57-70, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18952756

RESUMEN

Shortened telomeres are a normal consequence of cell division. However, telomere shortening past a critical point results in cellular senescence and death. To determine the effect of telomere shortening on lung, four generations of B6.Cg-Terc(tm1Rdp) mice, null for the terc component of telomerase, the holoenzyme that maintains telomeres, were bred and analyzed. Generational inbreeding of terc-/- mice caused sequential shortening of telomeres. Lung histology from the generation with the shortest telomeres (terc-/- F4) showed alveolar wall thinning and increased alveolar size. Morphometric analysis confirmed a significant increase in mean linear intercept (MLI). terc-/- F4 lung showed normal elastin deposition but had significantly decreased collagen content. Both airway and alveolar epithelial type 1 cells (AEC1) appeared normal by immunohistochemistry, and the percentage of alveolar epithelial type 2 cells (AEC2) per total cell number was similar to wild type. However, because of a decrease in distal lung cellularity, the absolute number of AEC2 in terc-/- F4 lung was significantly reduced. In contrast to wild type, terc-/- F4 distal lung epithelium from normoxia-maintained mice exhibited DNA damage by terminal deoxynucleotidyltransferase (TdT)-mediated dUTP nick end labeling (TUNEL) and 8-oxoguanine immunohistochemistry. Western blotting of freshly isolated AEC2 lysates for stress signaling kinases confirmed that the stress-activated protein kinase (SAPK)/c-Jun NH(2)-terminal kinase (JNK) stress response pathway is stimulated in telomerase-null AEC2 even under normoxic conditions. Expression of downstream apoptotic/stress markers, including caspase-3, caspase-6, Bax, and HSP-25, was also observed in telomerase-null, but not wild-type, AEC2. TUNEL analysis of freshly isolated normoxic AEC2 showed that DNA strand breaks, essentially absent in wild-type cells, increased with each successive terc-/- generation and correlated strongly with telomere length (R(2) = 0.9631). Thus lung alveolar integrity, particularly in the distal epithelial compartment, depends on proper telomere maintenance.


Asunto(s)
Alveolos Pulmonares/patología , ARN/genética , Telomerasa/genética , Telómero/genética , Telómero/patología , Actinas/metabolismo , Animales , Biomarcadores/metabolismo , Colágeno/metabolismo , Daño del ADN , Elastina/metabolismo , Femenino , Etiquetado Corte-Fin in Situ , Péptidos y Proteínas de Señalización Intercelular , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Estrés Oxidativo/fisiología , Péptidos/metabolismo , Alveolos Pulmonares/metabolismo , Proteína C Asociada a Surfactante Pulmonar , ARN/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Transducción de Señal/fisiología , Telomerasa/metabolismo
18.
Proc Am Thorac Soc ; 5(6): 703-6, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18684721

RESUMEN

At least two populations of epithelial stem/progenitor cells give rise to the lung anlage, comprising the laryngo-tracheal complex versus the distal lung below the first bronchial bifurcation. Amplification of the distal population requires FGF9-FGF10-FGFR2b-Sprouty signaling. Residual pools of adult stem cells are hypothesized to be the source of lung regeneration and repair. These pools have been located within the basal layer of the upper airways, within or near pulmonary neuroendocrine cell rests, at the bronchoalveolar junction as well as within the alveolar epithelial surface. Rapid repair of the denuded alveolar surface after injury is clearly key to survival. Strategies to enhance endogenous alveolar epithelial repair could include protection of epithelial progenitors from injury and/or stimulation of endogenous progenitor cell function. Protection with inosine or FGF signaling are possible small molecule therapeutic options. Alternatively, exogenous stem/progenitor cells can be delivered into the lung either intravenously, intratracheally, or by direct injection. Sources of exogenous stem/progenitor cells that are currently under evaluation in the context of acute lung injury repair include embryonic stem cells, bone marrow- or fat-derived mesenchymal stem cells, circulating endothelial progenitors, and, recently, amniotic fluid stem/progenitor cells. Further work will be needed to translate stem/progenitor cell therapy for the lung.


Asunto(s)
Pulmón/embriología , Pulmón/fisiología , Regeneración/fisiología , Células Madre/fisiología , Diferenciación Celular , Células Epiteliales/citología , Humanos , Pulmón/citología , Mucosa Respiratoria/citología , Mucosa Respiratoria/embriología , Mucosa Respiratoria/fisiología , Trasplante de Células Madre/tendencias
19.
Am J Physiol Lung Cell Mol Physiol ; 290(4): L685-L694, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16299057

RESUMEN

In this study, C57BL/6J mice were exposed to hyperoxia and allowed to recover in room air. The sublethal dose of hyperoxia for C57BL/6J was 48 h. Distal lung cellular isolates from treated animals were characterized as 98% epithelial, with minor fibroblast and endothelial cell contaminants. Cells were then verified as 95% pure alveolar epithelial type II cells (AEC2) by surfactant protein C (SP-C) expression. After hyperoxia exposure in vivo, fresh, uncultured AEC2 were analyzed for proliferation by cell yield, cell cycle, PCNA expression, and telomerase activity. DNA damage was assessed by TdT-dUTP nick-end labeling, whereas induction of DNA repair was evaluated by GADD-153 expression. A baseline level for proliferation and damage was observed in cells from control animals that did not alter significantly during acute hyperoxia exposure. However, a rise in these markers was observed 24 h into recovery. Over 72 h of recovery, markers for proliferation remained elevated, whereas those for DNA damage and repair peaked at 48 h and then returned back to baseline. The expression of GADD-153 followed a distinct course, rising significantly during acute exposure and peaking at 48 h recovery. These data demonstrate that in healthy, adult male C57BL/6J mice, AEC2 proliferation, damage, and repair follow separate courses during hyperoxia recovery and that both proliferation and efficient repair may be required to ensure AEC2 survival.


Asunto(s)
Proliferación Celular , Reparación del ADN , Hiperoxia/patología , Hiperoxia/fisiopatología , Alveolos Pulmonares/patología , Alveolos Pulmonares/fisiopatología , Animales , Biomarcadores/metabolismo , Daño del ADN , Células Epiteliales/clasificación , Células Epiteliales/metabolismo , Hiperoxia/genética , Hiperoxia/metabolismo , Queratinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Antígeno Nuclear de Célula en Proliferación/metabolismo , Alveolos Pulmonares/metabolismo , Proteína C Asociada a Surfactante Pulmonar/metabolismo , Recuperación de la Función , Fase S , Telomerasa/metabolismo , Factores de Tiempo , Factor de Transcripción CHOP/metabolismo , Regulación hacia Arriba
20.
Am J Physiol Lung Cell Mol Physiol ; 286(4): L658-67, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12922980

RESUMEN

Alveolar epithelial type 2 cells (AEC2) isolated from hyperoxia-treated animals exhibit increases in both proliferation and DNA damage in response to culture. AEC2 express the zonula adherens proteins E-cadherin, -, - and -catenin, desmoglein, and pp120, as demonstrated by Western blotting. Immunohistochemical analysis of cultured AEC2 showed expression of E-cadherin on cytoplasmic membranes varying from strongly to weakly staining. When cultured AEC2 placed in suspension were labeled with fluorescent-tagged antibodies to E-cadherin, cells could be sorted into at least two subpopulations, either dim or brightly staining for this marker. With the use of antibody to E-cadherin bound to magnetic beads, cells were physically separated into E-cadherin-positive and -negative subpopulations, which were then analyzed for differences in proliferation and DNA damage. The E-cadherin-positive subpopulation contained the majority of damaged cells, was quiescent, and expressed low levels of telomerase activity, whereas the E-cadherin-negative subpopulation was undamaged, proliferative, and expressed high levels of telomerase activity.


Asunto(s)
Alveolos Pulmonares/citología , Mucosa Respiratoria/citología , Células Madre/citología , Uniones Adherentes/metabolismo , Animales , Cadherinas/metabolismo , División Celular , Separación Celular , Hiperoxia/patología , Etiquetado Corte-Fin in Situ , Masculino , Alveolos Pulmonares/fisiología , Ratas , Ratas Sprague-Dawley , Regeneración , Mucosa Respiratoria/fisiología , Células Madre/metabolismo , Telomerasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA