Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Am J Physiol Lung Cell Mol Physiol ; 314(6): L936-L945, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29446321

RESUMO

Chorioamnionitis, mechanical ventilation, oxygen therapy, and postnatal infection promote inflammation in the newborn lung. The long-term consequences of pulmonary inflammation during infancy have not been well characterized. The aim of this study was to examine the impact of inflammation during the late saccular to alveolar stages of lung development on lung structure and function in adulthood. To induce IL-1ß expression in the pulmonary epithelium of mice with a tetracycline-inducible human IL-1ß transgene, doxycycline was administered via intraperitoneal injections to bitransgenic pups and their littermate controls on postnatal days (PN) 0, 0.5, and 1. Lung structure, inflammation, and airway reactivity were studied in adulthood. IL-1ß production in early life resulted in increased numbers of macrophages and neutrophils on PN21, but inflammation subsided by PN42. Permanent changes in alveolar structure, i.e., larger alveoli and thicker alveolar walls, were present from PN21 to PN84. Lack of alveolar septation thus persisted after IL-1ß production and inflammation had ceased. Early IL-1ß production caused goblet cell hyperplasia, enhanced calcium-activated chloride channel 3 (CLCA3) protein expression, and increased airway reactivity in response to methacholine on PN42. Lymphoid follicles were present adjacent to small airways in the lungs of adult bitransgenic mice, and levels of the B cell chemoattractant CXC-motif ligand (CXCL) 13 were elevated in the lungs of bitransgenic mice compared with controls. In conclusion, IL-1ß-induced pulmonary inflammation in early life causes a chronic lung disease in adulthood.


Assuntos
Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Neutrófilos/metabolismo , Alvéolos Pulmonares/crescimento & desenvolvimento , Mucosa Respiratória/crescimento & desenvolvimento , Animais , Quimiocina CXCL13/genética , Quimiocina CXCL13/metabolismo , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Interleucina-1beta/genética , Macrófagos/patologia , Camundongos , Camundongos Transgênicos , Mucoproteínas/genética , Mucoproteínas/metabolismo , Neutrófilos/patologia , Alvéolos Pulmonares/patologia , Mucosa Respiratória/patologia
2.
Exp Lung Res ; 43(2): 82-92, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28368678

RESUMO

BACKGROUND/AIM: Epithelial-mesenchymal communication plays a key role in tissue homeostasis and abnormal signaling contributes to chronic airways disease such as COPD. Most in vitro models are limited in complexity and poorly represent this epithelial-mesenchymal trophic unit. We postulated that cellular outgrowth from bronchial tissue would enable development of a mucosal structure that recapitulates better in vivo tissue architecture. MATERIALS AND METHODS: Bronchial tissue was embedded in Matrigel and outgrowth cultures monitored using time-lapse microscopy, electrical resistance, light and electron microscopy. Cultures were challenged repetitively with cigarette smoke extract (CSE). RESULTS: The outgrowths formed as a multicellular sheet with motile cilia becoming evident as the Matrigel was remodeled to provide an air interface; cultures were viable for more than one year. Immunofluorescence and electron microscopy (EM) identified an upper layer of mucociliary epithelium and a lower layer of highly organized extracellular matrix (ECM) interspersed with fibroblastic cells separated by a basement membrane. EM analysis of the mucosal construct after repetitive exposure to CSE revealed epithelial damage, loss of cilia, and ECM remodeling, as occurs in vivo. CONCLUSIONS: We have developed a robust bronchial mucosal model. The structural changes observed following CSE exposure suggest the model should have utility for drug discovery and preclinical testing, especially those targeting airway remodeling.


Assuntos
Modelos Biológicos , Fumaça/efeitos adversos , Brônquios/citologia , Brônquios/crescimento & desenvolvimento , Células Cultivadas , Colágeno , Combinação de Medicamentos , Células Epiteliais/citologia , Humanos , Laminina , Células-Tronco Mesenquimais/citologia , Microscopia , Proteoglicanas , Mucosa Respiratória/citologia , Mucosa Respiratória/crescimento & desenvolvimento
3.
Proc Natl Acad Sci U S A ; 111(22): E2291-300, 2014 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-24843139

RESUMO

Alveolar formation is coupled to the spatiotemporally regulated differentiation of alveolar myofibroblasts (AMYFs), which contribute to the morphological changes of interalveolar walls. Although the Ras-ERK signaling pathway is one of the key regulators for alveolar formation in developing lungs, the intrinsic molecular and cellular mechanisms underlying its role remain largely unknown. By analyzing the Ras-ERK signaling pathway during postnatal development of lungs, we have identified a critical role of DA-Raf1 (DA-Raf)-a dominant-negative antagonist for the Ras-ERK signaling pathway-in alveolar formation. DA-Raf-deficient mice displayed alveolar dysgenesis as a result of the blockade of AMYF differentiation. DA-Raf is predominantly expressed in type 2 alveolar epithelial cells (AEC2s) in developing lungs, and DA-Raf-dependent MEK1/2 inhibition in AEC2s suppresses expression of tissue inhibitor of matalloprotienase 4 (TIMP4), which prevents a subsequent proteolytic cascade matrix metalloproteinase (MMP)14-MMP2. Furthermore, MMP14-MMP2 proteolytic cascade regulates AMYF differentiation and alveolar formation. Therefore, DA-Raf-dependent inhibition of the Ras-ERK signaling pathway in AEC2s is required for alveolar formation via triggering MMP2 activation followed by AMYF differentiation. These findings reveal a pivotal role of the Ras-ERK signaling pathway in the dynamic regulation of alveolar development.


Assuntos
Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Proto-Oncogênicas A-raf/metabolismo , Alvéolos Pulmonares/crescimento & desenvolvimento , Alvéolos Pulmonares/metabolismo , Mucosa Respiratória/crescimento & desenvolvimento , Mucosa Respiratória/metabolismo , Animais , Diferenciação Celular/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Introdução de Genes , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/metabolismo , Masculino , Metaloproteinase 14 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Proto-Oncogênicas A-raf/genética , Proteínas Proto-Oncogênicas c-raf/metabolismo , Alvéolos Pulmonares/citologia , Mucosa Respiratória/citologia , Inibidores Teciduais de Metaloproteinases/genética , Inibidores Teciduais de Metaloproteinases/metabolismo , Inibidor Tecidual 4 de Metaloproteinase
4.
Artigo em Inglês | MEDLINE | ID: mdl-27768904

RESUMO

Gill morphometric and gill plasticity of the air-breathing striped catfish (Pangasianodon hypophthalmus) exposed to different temperatures (present day 27°C and future 33°C) and different air saturation levels (92% and 35%) during 6weeks were investigated using vertical sections to estimate the respiratory lamellae surface areas, harmonic mean barrier thicknesses, and gill component volumes. Gill respiratory surface area (SA) and harmonic mean water - blood barrier thicknesses (HM) of the fish were strongly affected by both environmental temperature and oxygen level. Thus initial values for 27°C normoxic fish (12.4±0.8g) were 211.8±21.6mm2g-1 and 1.67±0.12µm for SA and HM respectively. After 5weeks in same conditions or in the combinations of 33°C and/or PO2 of 55mmHg, this initial surface area scaled allometrically with size for the 33°C hypoxic group, whereas branchial SA was almost eliminated in the 27°C normoxic group, with other groups intermediate. In addition, elevated temperature had an astounding effect on growth with the 33°C group growing nearly 8-fold faster than the 27°C fish.


Assuntos
Peixes-Gato/fisiologia , Brânquias/fisiologia , Estresse Fisiológico , Termotolerância , Animais , Aquicultura , Peixes-Gato/crescimento & desenvolvimento , Peixes-Gato/parasitologia , Hipóxia Celular , Ingestão de Energia , Brânquias/crescimento & desenvolvimento , Brânquias/parasitologia , Aquecimento Global , Processamento de Imagem Assistida por Computador , Microscopia/veterinária , Carga Parasitária , Mucosa Respiratória/crescimento & desenvolvimento , Mucosa Respiratória/parasitologia , Mucosa Respiratória/fisiologia , Rios , Especificidade da Espécie , Tailândia , Fatores de Tempo , Aumento de Peso
5.
Ultrastruct Pathol ; 39(4): 245-54, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25867690

RESUMO

The ultrastructure of the developing bronchiolar cell was studied in six age groups: prenatal (60 d post-conception); postnatal (1-, 7-, 14- and 21-day-old); and adult. Following intratracheal fixation, the lung tissue was processed for scanning and transmission electron microscopy. The lining of terminal bronchioles consists of cuboidal to columnar nonciliated bronchiolar cells (NBCs) and ciliated with or without microvilli. NBCs were recognized by indented centrally located nucleus. The apical surface extended beyond the surface of neighboring cells and was covered by minute microvilli, except in prenatal kittens. The NBCs of the adult were characterized by abundant mitochondria and glycogen inclusions. In prenatal kittens, the cytoplasm was filled with patches of alpha and beta form of glycogen. Postnatally, glycogen was reduced in quantity, became scattered throughout the cytoplasm and was predominantly of the beta form. Islands of cytoplasm, separated from the apical cytoplasm were observed in the lumen of adult bronchioles. This suggests an apocrine mode of secretion. The NBCs attain maturity by three weeks of age.


Assuntos
Bronquíolos/crescimento & desenvolvimento , Bronquíolos/ultraestrutura , Gatos/crescimento & desenvolvimento , Células Epiteliais/ultraestrutura , Mucosa Respiratória/crescimento & desenvolvimento , Mucosa Respiratória/ultraestrutura , Animais , Gatos/anatomia & histologia , Feminino , Masculino , Microscopia Eletrônica
6.
Respir Res ; 15: 70, 2014 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-24970044

RESUMO

BACKGROUND: Claudins are transmembrane proteins expressed in tight junctions that prevent paracellular transport of extracellular fluid and a variety of other substances. However, the expression profile of Claudin-6 (Cldn6) in the developing lung has not been characterized. METHODS AND RESULTS: Cldn6 expression was determined during important periods of lung organogenesis by microarray analysis, qPCR and immunofluorescence. Expression patterns were confirmed to peak at E12.5 and diminish as lung development progressed. Immunofluorescence revealed that Cldn6 was detected in cells that also express TTF-1 and FoxA2, two critical transcriptional regulators of pulmonary branching morphogenesis. Cldn6 was also observed in cells that express Sox2 and Sox9, factors that influence cell differentiation in the proximal and distal lung, respectively. In order to assess transcriptional control of Cldn6, 0.5, 1.0, and 2.0-kb of the proximal murine Cldn6 promoter was ligated into a luciferase reporter and co-transfected with expression vectors for TTF-1 or two of its important transcriptional co-regulators, FoxA2 and Gata-6. In almost every instance, TTF-1, FoxA2, and Gata-6 activated gene transcription in cell lines characteristic of proximal airway epithelium (Beas2B) and distal alveolar epithelium (A-549). CONCLUSIONS: These data revealed for the first time that Cldn6 might be an important tight junctional component expressed by pulmonary epithelium during lung organogenesis. Furthermore, Cldn6-mediated aspects of cell differentiation may describe mechanisms of lung perturbation coincident with impaired cell junctions and abnormal membrane permeability.


Assuntos
Claudinas/fisiologia , Proteínas de Ligação a DNA/fisiologia , Fator de Transcrição GATA6/fisiologia , Fator 3-beta Nuclear de Hepatócito/fisiologia , Pulmão/crescimento & desenvolvimento , Transcrição Gênica/fisiologia , Animais , Animais Recém-Nascidos , Regulação da Expressão Gênica no Desenvolvimento , Pulmão/embriologia , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mucosa Respiratória/crescimento & desenvolvimento , Mucosa Respiratória/metabolismo , Fatores de Transcrição
7.
Am J Respir Cell Mol Biol ; 49(2): 190-6, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23526223

RESUMO

Tracheal brush cells (BCs) are specialized epithelial chemosensors that use the canonical taste transduction cascade to detect irritants. To test whether BCs are replaced at the same rate as other cells in the surrounding epithelium of adult mice, we used 5-bromo-2'-deoxyuridine (BrdU) to label dividing cells. Although scattered BrdU-labeled epithelial cells are present 5-20 days after BrdU, no BCs are labeled. These data indicate that BCs comprise a relatively static population. To determine how BCs are generated during development, we injected 5-day-old mice with BrdU and found labeled BCs and non-BC epithelial cells 5 days after BrdU. During the next 60 days, the percentage of labeled BCs increased, whereas the percentage of other labeled cell types decreased. These data suggest that BCs are generated from non-BC progenitor cells during postnatal tracheal growth. To test whether the adult epithelium retains the capacity to generate BCs, tracheal epithelial cells were recovered from adult mice and grown in an air-liquid interface (ALI) culture. After transition to differentiation conditions, BCs are detected, and comprise 1% of the total cell population by Day 14. BrdU added to cultures before the differentiation of BCs was chased into BCs, indicating that the increase in BC density is attributable to the proliferation of a non-BC progenitor. We conclude that: (1) BCs are normally a static population in adult mice; (2) BC progenitors proliferate and differentiate during neonatal development; and (3) BCs can be regenerated from a proliferative population resident in adult epithelium.


Assuntos
Células Quimiorreceptoras/citologia , Mucosa Respiratória/citologia , Mucosa Respiratória/crescimento & desenvolvimento , Células-Tronco/citologia , Traqueia/citologia , Traqueia/crescimento & desenvolvimento , Envelhecimento/fisiologia , Animais , Diferenciação Celular/fisiologia , Proliferação de Células , Células Cultivadas , Células Quimiorreceptoras/metabolismo , Camundongos , Camundongos Transgênicos , Mucosa Respiratória/metabolismo , Células-Tronco/metabolismo , Traqueia/metabolismo
8.
Exp Lung Res ; 39(2): 59-69, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23302116

RESUMO

Intranasally delivered human cord blood-derived CD34+ hematopoietic progenitor cells have the capacity to engraft and undergo transdifferentiation to surfactant-containing alveolar epithelial type II cell-like cells in lungs of newborn mice. The aim of this study was to determine the long-term fate of such transplanted cells as well as their effects on alveolar development in neonatally injured lungs. Double transgenic CCSP+/FasL+ mice with inducible lung-specific FasL expression, targeted to induce respiratory epithelial apoptosis in the perinatal period, served as model of neonatal lung injury. Non-injured single transgenic CCSP+/FasL- littermates served as controls. Freshly isolated umbilical cord blood CD34+ cells (0.5 to 1.0×10(6)) were administered at postnatal day 5 by intranasal inoculation; sham controls received equal volume PBS. Engraftment, alveolar epithelial differentiation, lung growth, and alveolarization were evaluated one year after transplantation. Engrafted cord blood-derived cells, detected by human-specific FISH (fluorescent in situ hybridization) analysis, and cord blood-derived alveolar type II-like cells, detected by double immunofluorescence analysis, while sparse, were seen in all conditions and more frequent in double than single transgenic recipients. The total lung volume and volume of air-exchanging parenchyma, assessed by stereological volumetry, were significantly greater in CD34-treated double transgenic animals than in PBS-treated double transgenic controls. Alveolarization, assessed by histomorphometry, was equivalent in these groups. These results suggest that transdifferentiated alveolar epithelial cells, derived from cord blood CD34+ cells, can persist up to one year after intrapulmonary delivery. Cord blood-CD34+ cell administration appears to have growth-promoting effects in injured newborn lungs, without affecting alveolar development in this model.


Assuntos
Displasia Broncopulmonar/terapia , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Sobrevivência de Enxerto , Células-Tronco Hematopoéticas/citologia , Alvéolos Pulmonares/citologia , Animais , Antígenos CD34/metabolismo , Diferenciação Celular , Sobrevivência Celular , Feminino , Células-Tronco Hematopoéticas/metabolismo , Humanos , Hibridização in Situ Fluorescente , Recém-Nascido , Masculino , Camundongos , Camundongos Transgênicos , Alvéolos Pulmonares/crescimento & desenvolvimento , Mucosa Respiratória/citologia , Mucosa Respiratória/crescimento & desenvolvimento , Fatores de Tempo , Transplante Heterólogo
9.
BMC Vet Res ; 9: 224, 2013 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-24206786

RESUMO

BACKGROUND: Airways progenitors may be involved in embryogenesis and lung repair. The characterization of these important populations may enable development of new therapeutics to treat acute or chronic lung disease. In this study, we aimed to establish the presence of bronchioloalveolar progenitors in ovine lungs and to characterize their potential to differentiate into specialized cells. RESULTS: Lung cells were studied using immunohistochemistry on frozen sections of the lung. Immunocytochemistry and flow cytometry were conducted on ex-vivo derived pulmonary cells. The bronchioloalveolar progenitors were identified by their co-expression of CCSP, SP-C and CD34. A minor population of CD34(pos)/SP-C(pos)/CCSP(pos) cells (0.33% ± 0.31) was present ex vivo in cell suspensions from dissociated lungs. Using CD34 magnetic positive-cell sorting, undifferentiated SP-C(pos)/CCSP(pos) cells were purified (>80%) and maintained in culture. Using synthetic media and various extracellular matrices, SP-C(pos)/CCSP(pos) cells differentiated into either club cells (formerly named Clara cells) or alveolar epithelial type-II cells. Furthermore, these ex vivo and in vitro derived bronchioloalveolar progenitors expressed NANOG, OCT4 and BMI1, specifically described in progenitors or stem cells, and during lung development. CONCLUSIONS: We report for the first time in a large animal the existence of bronchioloalveolar progenitors with dual differentiation potential and the expression of specialized genes. These newly described cell population in sheep could be implicated in regeneration of the lung following lesions or in development of diseases such as cancers.


Assuntos
Brônquios/citologia , Diferenciação Celular/fisiologia , Pulmão/citologia , Alvéolos Pulmonares/citologia , Células-Tronco/fisiologia , Animais , Brônquios/crescimento & desenvolvimento , Citometria de Fluxo/veterinária , Expressão Gênica/fisiologia , Imuno-Histoquímica/veterinária , Pulmão/crescimento & desenvolvimento , Alvéolos Pulmonares/crescimento & desenvolvimento , Proteína C Associada a Surfactante Pulmonar/biossíntese , Mucosa Respiratória/citologia , Mucosa Respiratória/crescimento & desenvolvimento , Ovinos
10.
Dev Biol ; 353(2): 242-58, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21385574

RESUMO

Six1 is a member of the six-homeodomain family of transcription factors. Six1 is expressed in multiple embryonic cell types and plays important roles in proliferation, differentiation and survival of precursor cells of different organs, yet its function during lung development was hitherto unknown. Herein we show that Six1(-/-) lungs are severely hypoplastic with greatly reduced epithelial branching and increased mesenchymal cellularity. Six1 is expressed at the distal epithelial tips of branching tubules as well as in the surrounding distal mesenchyme. Six1(-/-) lung epithelial cells show increased expression of differentiation markers, but loss of progenitor cell markers. Six1 overexpression in MLE15 lung epithelial cells in vitro inhibited cell differentiation, but increases the expression of progenitor cell markers. In addition, Six1(-/-) embryos and newborn mice exhibit mesenchymal overproliferation, decreased Fgf10 expression and severe defects in the smooth muscle component of the bronchi and major pulmonary vessels. These defects lead to rupture of major vessels in mutant lungs after birth. Treatment of Six1(-/-) epithelial explants in culture with recombinant Fgf10 protein restores epithelial branching. As Shh expression is abnormally increased in Six1(-/-) lungs, we also treated mutant mesenchymal explants with recombinant Shh protein and found that these explants were competent to respond to Shh and continued to grow in culture. Furthermore, inhibition of Shh signaling with cyclopamine stimulated Six1(-/-) lungs to grow and branch in culture. This study provides the first evidence for the requirement of Six1 in coordinating Shh-Fgf10 signaling in embryonic lung to ensure proper levels of proliferation and differentiation along the proximodistal axis of epithelial, mesenchymal and endothelial cells. These findings uncover novel and essential functions for Six1 as a critical coordinator of Shh-Fgf10 signaling during embryonic lung development. We propose that Six1 is hence critical for coordination of proper lung epithelial, mesenchymal and vascular development.


Assuntos
Proteínas de Homeodomínio/fisiologia , Pulmão/embriologia , Actinas/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Células-Tronco Embrionárias/citologia , Fator 10 de Crescimento de Fibroblastos/genética , Fator 10 de Crescimento de Fibroblastos/farmacologia , Fator 10 de Crescimento de Fibroblastos/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/antagonistas & inibidores , Proteínas Hedgehog/farmacologia , Proteínas Hedgehog/fisiologia , Proteínas de Homeodomínio/genética , Pulmão/anormalidades , Pulmão/irrigação sanguínea , Pulmão/crescimento & desenvolvimento , Mesoderma/embriologia , Mesoderma/crescimento & desenvolvimento , Mesoderma/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Camundongos Transgênicos , Morfogênese/efeitos dos fármacos , Morfogênese/genética , Morfogênese/fisiologia , Miócitos de Músculo Liso/citologia , Mucosa Respiratória/embriologia , Mucosa Respiratória/crescimento & desenvolvimento , Mucosa Respiratória/metabolismo , Transdução de Sinais/efeitos dos fármacos , Alcaloides de Veratrum/farmacologia
11.
Gene Ther ; 19(2): 201-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21654824

RESUMO

Viral vector-mediated gene transfer to the postnatal respiratory epithelium has, in general, been of low efficiency due to physical and immunological barriers, non-apical location of cellular receptors critical for viral uptake and limited transduction of resident stem/progenitor cells. These obstacles may be overcome using a prenatal strategy. In this study, HIV-1-based lentiviral vectors (LVs) pseudotyped with the envelope glycoproteins of Jaagsiekte sheep retrovirus (JSRV-LV), baculovirus GP64 (GP64-LV), Ebola Zaire-LV or vesicular stomatitis virus (VSVg-LV) and the adeno-associated virus-2/6.2 (AAV2/6.2) were compared for in utero transfer of a green fluorescent protein (GFP) reporter gene to ovine lung epithelium between days 65 and 78 of gestation. GFP expression was examined on day 85 or 136 of gestation (term is ∼145 days). The percentage of the respiratory epithelial cells expressing GFP in fetal sheep that received the JSRV-LV (3.18 × 10(8)-6.85 × 10(9) viral particles per fetus) was 24.6±0.9% at 3 weeks postinjection (day 85) and 29.9±4.8% at 10 weeks postinjection (day 136). Expression was limited to the surface epithelium lining fetal airways <100 µm internal diameter. Fetal airways were amenable to VSVg-LV transduction, although the percentage of epithelial expression was low (6.6±0.6%) at 1 week postinjection. GP64-LV, Ebola Zaire-LV and AAV2/6.2 failed to transduce the fetal ovine lung under these conditions. These data demonstrate that prenatal lung gene transfer with LV engineered to target apical surface receptors can provide sustained and high levels of transgene expression and support the therapeutic potential of prenatal gene transfer for the treatment of congenital lung diseases.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Proteínas de Fluorescência Verde/metabolismo , Retrovirus Jaagsiekte de Ovinos/genética , Pulmão/embriologia , Ovinos/genética , Animais , Baculoviridae/genética , Dependovirus/genética , Ebolavirus/genética , Feto , Células HEK293 , Humanos , Hialuronoglucosaminidase/genética , Hialuronoglucosaminidase/metabolismo , Lentivirus/genética , Pulmão/crescimento & desenvolvimento , Mucosa Respiratória/crescimento & desenvolvimento , Mucosa Respiratória/metabolismo , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
12.
Am J Respir Crit Care Med ; 184(4): 401-6, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21642246

RESUMO

Recent advances in cellular, molecular, and developmental biology have revolutionized our concepts regarding the process of organogenesis that have important implications for our understanding of both lung formation and pulmonary disease pathogenesis. Pulmonary investigators have long debated whether developmental processes are recapitulated during normal repair of the lung or in the setting of chronic pulmonary diseases. Although the cellular events involved in lung morphogenesis and those causing pulmonary disease are likely to include processes that are distinct, there is increasing evidence that the pathogenesis of many lung disorders involves the same genetic machinery that regulates cell growth,specification, and differentiation during normal lung development.


Assuntos
Pneumopatias/fisiopatologia , Pulmão/embriologia , Pulmão/crescimento & desenvolvimento , Animais , Diferenciação Celular , Linhagem da Célula , Doença Crônica , Proteínas de Ligação a DNA/metabolismo , Desenvolvimento Embrionário , Células-Tronco Embrionárias/citologia , Desenvolvimento Fetal , Fator 3-beta Nuclear de Hepatócito/metabolismo , Humanos , Inflamação/metabolismo , Pulmão/citologia , Pneumopatias/genética , Metaplasia/metabolismo , Morfogênese/genética , Células-Tronco Pluripotentes/citologia , Proteínas Proto-Oncogênicas c-ets/metabolismo , Receptores Notch/metabolismo , Mucosa Respiratória/citologia , Mucosa Respiratória/embriologia , Mucosa Respiratória/crescimento & desenvolvimento , Fatores de Transcrição SOXB1/metabolismo , Transdução de Sinais/fisiologia , Técnicas de Cultura de Tecidos , Fatores de Transcrição , Transcrição Gênica , Cicatrização/genética
13.
Ann Otol Rhinol Laryngol ; 121(10): 645-9, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23130538

RESUMO

OBJECTIVES: We performed a prospective observation study in an outpatient surgical and office setting to compare human post-tonsillectomy healing to human cutaneous wound healing and to established animal models of oral healing. METHODS: Fourteen teenaged patients underwent planned tonsillectomy. Intraoral digital photographs were collected at the time of tonsillectomy, during the management of complications, and at postoperative office visits. Serial intraoral photographs of one patient were taken at 48-hour intervals from the time of surgery until postoperative day 17. RESULTS: Intraoral photographs from the days after tonsillectomy revealed a pattern of inflammation and healing that closely paralleled that in human skin and in canine and porcine oral wound models. CONCLUSIONS: Edema and pain are greatest immediately after surgery, probably as a result of thermal effects and expression of inflammatory mediators that stimulate pharyngeal nociceptors. Pain gradually decreases over time, with an increase in analog pain measures on postoperative days 3 to 5 corresponding to the maximal wound inflammation documented in experimental models. Epithelial ingrowth beneath a fibrin clot begins shortly after wounding. Separation of the fibrin clot about 7 days after surgery exposes vascular stroma. Involution of the vascular stroma and completion of epithelial coverage correlate with decreased pain levels and a lessened risk of bleeding.


Assuntos
Mucosa Respiratória/fisiologia , Tonsilectomia , Cicatrização/fisiologia , Adolescente , Edema/patologia , Fibrina/metabolismo , Humanos , Inflamação/patologia , Neovascularização Fisiológica/fisiologia , Medição da Dor , Fotografação , Estudos Prospectivos , Mucosa Respiratória/crescimento & desenvolvimento , Fatores de Tempo
14.
Dev Biol ; 347(2): 301-14, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20816795

RESUMO

Foxm1 is a member of the Forkhead Box (Fox) family of transcription factors. Foxm1 (previously called Foxm1b, HFH-11B, Trident, Win, or MPP2) is expressed in multiple cell types and plays important roles in cellular proliferation, differentiation and tumorigenesis. Genetic deletion of Foxm1 from mouse respiratory epithelium during initial stages of lung development inhibits lung maturation and causes respiratory failure after birth. However, the role of Foxm1 during postnatal lung morphogenesis remains unknown. In the present study, Foxm1 expression was detected in epithelial cells of conducting and peripheral airways and changing dynamically with lung maturation. To discern the biological role of Foxm1 in the prenatal and postnatal lung, a novel transgenic mouse line that expresses a constitutively active form of FoxM1 (FoxM1 N-terminal deletion mutant or FoxM1-ΔN) under the control of lung epithelial-specific SPC promoter was produced. Expression of the FoxM1-ΔN transgene during embryogenesis caused epithelial hyperplasia, inhibited lung sacculation and expression of the type II epithelial marker, pro-SPC. Expression of FoxM1-ΔN mutant during the postnatal period did not influence alveologenesis but caused focal airway hyperplasia and increased proliferation of Clara cells. Likewise, expression of FoxM1-ΔN mutant in conducting airways with Scgb1a1 promoter was sufficient to induce Clara cell hyperplasia. Furthermore, FoxM1-ΔN cooperated with activated K-Ras to induce lung tumor growth in vivo. Increased activity of Foxm1 altered lung sacculation, induced proliferation in the respiratory epithelium and accelerated lung tumor growth, indicating that precise regulation of Foxm1 is critical for normal lung morphogenesis and development of lung cancer.


Assuntos
Fatores de Transcrição Forkhead/genética , Pulmão/embriologia , Pulmão/crescimento & desenvolvimento , Mucosa Respiratória/embriologia , Mucosa Respiratória/crescimento & desenvolvimento , Animais , Crescimento Celular , Feminino , Proteína Forkhead Box M1 , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Genes ras , Humanos , Hiperplasia , Pulmão/citologia , Pulmão/metabolismo , Neoplasias Pulmonares/genética , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Mutantes/genética , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/genética , Mucosa Respiratória/citologia , Mucosa Respiratória/metabolismo
15.
Thorax ; 66(4): 280-5, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21233480

RESUMO

BACKGROUND: The bronchial epithelium and underlying reticular basement membrane (RBM) have a close spatial and functional inter-relationship and are considered an epithelial-mesenchymal trophic unit (EMTU). An understanding of RBM development is critical to understanding the extent and time of appearance of its abnormal thickening that is characteristic of asthma. METHODS: RBM thickness and epithelial height were determined in histological sections of cartilaginous bronchi obtained postmortem from 47 preterm babies and infants (median age 40 weeks gestation (22 weeks gestation-8 months)), 40 children (2 years (1 month-17 years)) and 23 adults (44 (17-90) years) who had died from non-respiratory causes, and had no history of asthma. RESULTS: The RBM was visible by light microscopy at 30 weeks gestation. RBM thickness increased in successive age groups in childhood; in infants (r=0.63, p<0.001) and in children between 1 month and 17 years (r=0.82, p<0.001). After 18 years, RBM thickness decreased with increasing age (r=-0.42, p<0.05). Epithelial height showed a similar relationship with age, a positive relationship from preterm to 17 years (r=0.50, p<0.001) and a negative relationship in adulthood (r=-0.84, p<0.0001). There was a direct relationship between epithelial height and RBM thickness (r=0.6, p<0.001). CONCLUSIONS: The RBM in these subjects was microscopically identifiable by 30 weeks gestation. It thickened during childhood and adolescence. In adults, there was either no relationship with age, or a slow reduction in thickness in older age. Developmental changes of RBM thickness were accompanied by similar changes in epithelial height, supporting the close relationship between RBM and epithelium within the EMTU.


Assuntos
Brônquios/crescimento & desenvolvimento , Mucosa Respiratória/crescimento & desenvolvimento , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/patologia , Membrana Basal/anatomia & histologia , Membrana Basal/crescimento & desenvolvimento , Estatura/fisiologia , Peso Corporal/fisiologia , Brônquios/anatomia & histologia , Criança , Pré-Escolar , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro , Pessoa de Meia-Idade , Mucosa Respiratória/anatomia & histologia , Caracteres Sexuais , Adulto Jovem
16.
Respir Res ; 12: 134, 2011 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-21985298

RESUMO

BACKGROUND: Alveolar septation marks the beginning of the transition from the saccular to alveolar stage of lung development. Inflammation can disrupt this process and permanently impair alveolar formation resulting in alveolar hypoplasia as seen in bronchopulmonary dysplasia in preterm newborns. NF-κB is a transcription factor central to multiple inflammatory and developmental pathways including dorsal-ventral patterning in fruit flies; limb, mammary and submandibular gland development in mice; and branching morphogenesis in chick lungs. We have previously shown that epithelial overexpression of NF-κB accelerates lung maturity using transgenic mice. The purpose of this study was to test our hypothesis that targeted deletion of NF-κB signaling in lung epithelium would impair alveolar formation. METHODS: We generated double transgenic mice with lung epithelium-specific deletion of IKKß, a known activating kinase upstream of NF-κB, using a cre-loxP transgenic recombination strategy. Lungs of resulting progeny were analyzed at embryonic and early postnatal stages to determine specific effects on lung histology, and mRNA and protein expression of relevant lung morphoreulatory genes. Lastly, results measuring expression of the angiogenic factor, VEGF, were confirmed in vitro using a siRNA-knockdown strategy in cultured mouse lung epithelial cells. RESULTS: Our results showed that IKKß deletion in the lung epithelium transiently decreased alveolar type I and type II cells and myofibroblasts and delayed alveolar formation. These effects were mediated through increased alveolar type II cell apoptosis and decreased epithelial VEGF expression. CONCLUSIONS: These results suggest that epithelial NF-κB plays a critical role in early alveolar development possibly through regulation of VEGF.


Assuntos
Proteínas Reguladoras de Apoptose/deficiência , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Quinase I-kappa B/deficiência , Alvéolos Pulmonares/crescimento & desenvolvimento , Mucosa Respiratória/metabolismo , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Proteínas Reguladoras de Apoptose/genética , Quinase I-kappa B/genética , Pulmão/embriologia , Pulmão/crescimento & desenvolvimento , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Alvéolos Pulmonares/embriologia , Alvéolos Pulmonares/patologia , Mucosa Respiratória/embriologia , Mucosa Respiratória/crescimento & desenvolvimento , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/fisiologia
17.
J Immunol ; 182(7): 4357-67, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19299736

RESUMO

Overexpression of the epithelial Na(+) channel beta subunit (Scnn1b gene, betaENaC protein) in transgenic (Tg) mouse airways dehydrates mucosal surfaces, producing mucus obstruction, inflammation, and neonatal mortality. Airway inflammation includes macrophage activation, neutrophil and eosinophil recruitment, and elevated KC, TNF-alpha, and chitinase levels. These changes recapitulate aspects of complex human obstructive airway diseases, but their molecular mechanisms are poorly understood. We used genetic and pharmacologic approaches to identify pathways relevant to the development of Scnn1b-Tg mouse lung pathology. Genetic deletion of TNF-alpha or its receptor, TNFR1, had no measurable effect on the phenotype. Deletion of IL-4Ralpha abolished transient mucous secretory cell (MuSC) abundance and eosinophilia normally observed in neonatal wild-type mice. Similarly, IL-4Ralpha deficiency decreased MuSC and eosinophils in neonatal Scnn1b-Tg mice, which correlated with improved neonatal survival. However, chronic lung pathology in adult Scnn1b-Tg mice was not affected by IL-4Ralpha status. Prednisolone treatment ablated eosinophilia and MuSC in adult Scnn1b-Tg mice, but did not decrease mucus plugging or neutrophilia. These studies demonstrate that: 1) normal neonatal mouse airway development entails an IL-4Ralpha-dependent, transient abundance of MuSC and eosinophils; 2) absence of IL-4Ralpha improved neonatal survival of Scnn1b-Tg mice, likely reflecting decreased formation of asphyxiating mucus plugs; and 3) in Scnn1b-Tg mice, neutrophilia, mucus obstruction, and airspace enlargement are IL-4Ralpha- and TNF-alpha-independent, and only MuSC and eosinophilia are sensitive to glucocorticoids. Thus, manipulation of multiple pathways will likely be required to treat the complex pathogenesis caused by airway surface dehydration.


Assuntos
Imunidade nas Mucosas/fisiologia , Receptores de Superfície Celular/metabolismo , Mucosa Respiratória/fisiopatologia , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Líquido da Lavagem Broncoalveolar/química , Quimiocinas/análise , Desidratação/complicações , Modelos Animais de Doenças , Eosinofilia/tratamento farmacológico , Eosinofilia/etiologia , Canais Epiteliais de Sódio/genética , Glucocorticoides/uso terapêutico , Imunidade nas Mucosas/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Neutrófilos/imunologia , Prednisolona/uso terapêutico , Receptores de Superfície Celular/genética , Mucosa Respiratória/crescimento & desenvolvimento , Mucosa Respiratória/metabolismo , Sistema Respiratório/crescimento & desenvolvimento , Sistema Respiratório/imunologia , Sistema Respiratório/metabolismo
18.
Aging (Albany NY) ; 13(13): 16922-16937, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34238764

RESUMO

Idiopathic pulmonary fibrosis (IPF) is an age-related disorder that carries a universally poor prognosis and is thought to arise from repetitive micro injuries to the alveolar epithelium. To date, a major factor limiting our understanding of IPF is a deficiency of disease models, particularly in vitro models that can recapitulate the full complement of molecular attributes in the human condition. In this study, we aimed to develop a model that more closely resembles the aberrant IPF lung epithelium. By exposing mouse alveolar epithelial cells to repeated, low doses of bleomycin, instead of usual one-time exposures, we uncovered changes strikingly similar to those in the IPF lung epithelium. This included the acquisition of multiple phenotypic and functional characteristics of senescent cells and the adoption of previously described changes in mitochondrial homeostasis, including alterations in redox balance, energy production and activity of the mitochondrial unfolded protein response. We also uncovered dramatic changes in cellular metabolism and detected a profound loss of proteostasis, as characterized by the accumulation of cytoplasmic protein aggregates, dysregulated expression of chaperone proteins and decreased activity of the ubiquitin proteasome system. In summary, we describe an in vitro model that closely resembles the aberrant lung epithelium in IPF. We propose that this simple yet powerful tool could help uncover new biological mechanisms and assist in developing new pharmacological tools to treat the disease.


Assuntos
Fibrose Pulmonar Idiopática/patologia , Pulmão/crescimento & desenvolvimento , Pulmão/patologia , Mucosa Respiratória/crescimento & desenvolvimento , Mucosa Respiratória/patologia , Animais , Antibióticos Antineoplásicos/toxicidade , Bleomicina/toxicidade , Linhagem Celular , Senescência Celular , Modelos Animais de Doenças , Metabolismo Energético , Homeostase , Humanos , Camundongos , Mitocôndrias/metabolismo , Oxirredução , Complexo de Endopeptidases do Proteassoma , Proteínas/metabolismo , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/patologia , Resposta a Proteínas não Dobradas
19.
Cells ; 10(12)2021 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-34943975

RESUMO

Mesenchymal cells are an essential cell type because of their role in tissue support, their multilineage differentiation capacities and their potential clinical applications. They play a crucial role during lung development by interacting with airway epithelium, and also during lung regeneration and remodeling after injury. However, much less is known about their function in lung disease. In this review, we discuss the origins of mesenchymal cells during lung development, their crosstalk with the epithelium, and their role in lung diseases, particularly in chronic obstructive pulmonary disease.


Assuntos
Pulmão/crescimento & desenvolvimento , Células-Tronco Mesenquimais/metabolismo , Organogênese/genética , Doença Pulmonar Obstrutiva Crônica/genética , Remodelação das Vias Aéreas/genética , Diferenciação Celular/genética , Transição Epitelial-Mesenquimal/genética , Epitélio/crescimento & desenvolvimento , Epitélio/metabolismo , Epitélio/patologia , Humanos , Pulmão/metabolismo , Pulmão/patologia , Células-Tronco Mesenquimais/citologia , Doença Pulmonar Obstrutiva Crônica/patologia , Mucosa Respiratória/crescimento & desenvolvimento , Mucosa Respiratória/metabolismo
20.
J Clin Invest ; 117(10): 2929-40, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17909629

RESUMO

PTEN is a tumor suppressor gene mutated in many human cancers. We generated a bronchioalveolar epithelium-specific null mutation of Pten in mice [SP-C-rtTA/(tetO)(7)-Cre/Pten(flox/flox) (SOPten(flox/flox)) mice] that was under the control of doxycycline. Ninety percent of SOPten(flox/flox) mice that received doxycycline in utero [SOPten(flox/flox)(E10-16) mice] died of hypoxia soon after birth. Surviving SOPten(flox/flox)(E10-16) mice and mice that received doxycycline postnatally [SOPten(flox/flox)(P21-27) mice] developed spontaneous lung adenocarcinomas. Urethane treatment accelerated number and size of lung tumors developing in SOPten(flox/flox) mice of both ages. Histological and biochemical examinations of the lungs of SOPten(flox/flox)(E10-16) mice revealed hyperplasia of bronchioalveolar epithelial cells and myofibroblast precursors, enlarged alveolar epithelial cells, and impaired production of surfactant proteins. Numbers of bronchioalveolar stem cells (BASCs), putative initiators of lung adenocarcinomas, were increased. Lungs of SOPten(flox/flox)(E10-16) mice showed increased expression of Spry2, which inhibits the maturation of alveolar epithelial cells. Levels of Akt, c-Myc, Bcl-2, and Shh were also elevated in SOPten(flox/flox)(E10-16) and SOPten(flox/flox)(P21-27) lungs. Furthermore, K-ras was frequently mutated in adenocarcinomas observed in SOPten(flox/flox)(P21-27) lungs. These results indicate that Pten is essential for both normal lung morphogenesis and the prevention of lung carcinogenesis, possibly because this tumor suppressor is required for BASC homeostasis.


Assuntos
Adenocarcinoma/genética , Transformação Celular Neoplásica/genética , Neoplasias Pulmonares/genética , Pulmão/crescimento & desenvolvimento , Morfogênese/genética , Células-Tronco Neoplásicas/enzimologia , PTEN Fosfo-Hidrolase/fisiologia , Adenocarcinoma/induzido quimicamente , Adenocarcinoma/patologia , Animais , Brônquios/anormalidades , Brônquios/crescimento & desenvolvimento , Brônquios/patologia , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/patologia , Expressão Gênica , Pulmão/anormalidades , Pulmão/patologia , Neoplasias Pulmonares/induzido quimicamente , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Knockout , Mutação , PTEN Fosfo-Hidrolase/genética , Alvéolos Pulmonares/anormalidades , Alvéolos Pulmonares/crescimento & desenvolvimento , Alvéolos Pulmonares/patologia , Mucosa Respiratória/anormalidades , Mucosa Respiratória/crescimento & desenvolvimento , Mucosa Respiratória/patologia , Uretana/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA