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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Angiogenesis ; 17(3): 541-51, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24150281

RESUMO

In most rodents and some other mammals, the removal of one lung results in compensatory growth associated with dramatic angiogenesis and complete restoration of lung capacity. One pivotal mechanism in neoalveolarization is neovascularization, because without angiogenesis new alveoli can not be formed. The aim of this study is to image and analyze three-dimensionally the different patterns of neovascularization seen following pneumonectomy in mice on a sub-micron-scale. C57/BL6 mice underwent a left-sided pneumonectomy. Lungs were harvested at various timepoints after pneumonectomy. Volume analysis by microCT revealed a striking increase of 143 percent in the cardiac lobe 14 days after pneumonectomy. Analysis of microvascular corrosion casting demonstrated spatially heterogenous vascular densitities which were in line with the perivascular and subpleural compensatory growth pattern observed in anti-PCNA-stained lung sections. Within these regions an expansion of the vascular plexus with increased pillar formations and sprouting angiogenesis, originating both from pre-existing bronchial and pulmonary vessels was observed. Also, type II pneumocytes and alveolar macrophages were seen to participate actively in alveolar neo-angiogenesis after pneumonectomy. 3D-visualizations obtained by high-resolution synchrotron radiation X-ray tomographic microscopy showed the appearance of double-layered vessels and bud-like alveolar baskets as have already been described in normal lung development. Scanning electron microscopy data of microvascular architecture also revealed a replication of perialveolar vessel networks through septum formation as already seen in developmental alveolarization. In addition, the appearance of pillar formations and duplications on alveolar entrance ring vessels in mature alveoli are indicative of vascular remodeling. These findings indicate that sprouting and intussusceptive angiogenesis are pivotal mechanisms in adult lung alveolarization after pneumonectomy. Various forms of developmental neoalveolarization may also be considered to contribute in compensatory lung regeneration.


Assuntos
Neovascularização Fisiológica , Pneumonectomia , Alvéolos Pulmonares/irrigação sanguínea , Alvéolos Pulmonares/crescimento & desenvolvimento , Animais , Molde por Corrosão , Camundongos Endogâmicos C57BL , Alvéolos Pulmonares/patologia , Alvéolos Pulmonares/ultraestrutura , Síncrotrons , Tomografia , Remodelação Vascular
2.
Angiogenesis ; 15(1): 23-32, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21969134

RESUMO

Growth of the remaining lung after pneumonectomy has been observed in many mammalian species; nonetheless, the pattern and morphology of alveolar angiogenesis during compensatory growth is unknown. Here, we investigated alveolar angiogenesis in a murine model of post-pneumonectomy lung growth. As expected, the volume and weight of the remaining lung returned to near-baseline levels within 21 days of pneumonectomy. The percentage increase in lobar weight was greatest in the cardiac lobe (P < 0.001). Cell cycle flow cytometry demonstrated a peak of lung cell proliferation (12.02 ± 1.48%) 6 days after pneumonectomy. Spatial autocorrelation analysis of the cardiac lobe demonstrated clustering of similar vascular densities (positive autocorrelation) that consistently mapped to subpleural regions of the cardiac lobe. Immunohistochemical staining demonstrated increased cell density and enhanced expression of angiogenesis-related factors VEGFA, and GLUT1 in these subpleural regions. Corrosion casting and scanning electron microscopy 3-6 days after pneumonectomy demonstrated subpleural vessels with angiogenic sprouts. The monopodial sprouts appeared to be randomly oriented along the vessel axis with interbranch distances of 11.4 ± 4.8 µm in the regions of active angiogenesis. Also present within the regions of increased vascular density were frequent "holes" or "pillars" consistent with active intussusceptive angiogenesis. The mean pillar diameter was 4.2 ± 3.8 µm, and the pillars were observed in all regions of active angiogenesis. These findings indicate that the process of alveolar construction involves discrete regions of regenerative growth, particularly in the subpleural regions of the cardiac lobe, characterized by both sprouting and intussusceptive angiogenesis.


Assuntos
Neovascularização Fisiológica , Pneumonectomia , Alvéolos Pulmonares/irrigação sanguínea , Alvéolos Pulmonares/crescimento & desenvolvimento , Animais , Ciclo Celular , Citometria de Fluxo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Tamanho do Órgão , Pleura/patologia , Alvéolos Pulmonares/patologia , Alvéolos Pulmonares/ultraestrutura , Fatores de Tempo
3.
Exp Lung Res ; 38(8): 396-405, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22905715

RESUMO

In many species, pneumonectomy results in compensatory growth in the remaining lung. Although the late mechanical consequences of murine pneumonectomy are known, little is known about the anatomic adaptations and respiratory mechanics during compensatory lung growth. To investigate the structural and mechanical changes during compensatory growth, mice were studied for 21 days after left pneumonectomy using microCT and respiratory system impedance (FlexiVent). Anatomic changes after left pneumonectomy included minimal mediastinal shift or chestwall remodeling, but significant displacement of the heart and cardiac lobe. Mean displacement of the cardiac lobe centroid was 5.2 ± 0.8 mm. Lung impedance measurements were used to investigate the associated changes in respiratory mechanics. Quasi-static pressure-volume loops demonstrated progressive increase in volumes with decreased distensibility. Measures of quasi-static compliance and elastance were increased at all time points postpneumonectomy (P < .01). Oscillatory mechanics demonstrated a significant change in tissue impedance on the third day after pneumonectomy. The input impedance on day 3 after pneumonectomy demonstrated a significant increase in tissue damping (5.8 versus 4.3 cm H(2)O/mL) and elastance (36.7 versus 26.6 cm H(2)O/mL) when compared to controls. At all points, hysteresivity was unchanged (0.17). We conclude that the timing and duration of the mechanical changes was consistent with a mechanical signal for compensatory growth.


Assuntos
Adaptação Fisiológica/fisiologia , Pulmão/patologia , Pneumonectomia , Regeneração/fisiologia , Animais , Modelos Animais de Doenças , Elasticidade , Condutividade Elétrica , Pulmão/crescimento & desenvolvimento , Pulmão/cirurgia , Complacência Pulmonar , Medidas de Volume Pulmonar , Camundongos , Camundongos Endogâmicos C57BL , Tamanho do Órgão , Período Pós-Operatório , Respiração , Parede Torácica/fisiopatologia , Microtomografia por Raio-X
4.
Exp Lung Res ; 37(5): 301-9, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21574875

RESUMO

Thoracic surgical procedures in mice have been applied to a wide range of investigations, but little is known about the murine physiologic response to pulmonary surgery. Using continuous arterial oximetry monitoring and the FlexiVent murine ventilator, the authors investigated the effect of anesthesia and pneumonectomy on mouse oxygen saturation and lung mechanics. Sedation resulted in a dose-dependent decline of oxygen saturation that ranged from 55% to 82%. Oxygen saturation was restored by mechanical ventilation with increased rate and tidal volumes. In the mouse strain studied, optimal ventilatory rates were a rate of 200/minute and a tidal volume of 10 mL/kg. Sustained inflation pressures, referred to as a "recruitment maneuver," improved lung volumes, lung compliance, and arterial oxygenation. In contrast, positive end-expiratory pressure (PEEP) had a detrimental effect on oxygenation; an effect that was ameliorated after pneumonectomy. These results confirm that lung volumes in the mouse are dynamically determined and suggest a threshold level of mechanical ventilation to maintain perioperative oxygen saturation.


Assuntos
Complacência Pulmonar/fisiologia , Pulmão/fisiologia , Oxigênio/metabolismo , Pneumonectomia/métodos , Anestesia/métodos , Animais , Pulmão/metabolismo , Medidas de Volume Pulmonar/métodos , Camundongos , Camundongos Endogâmicos C57BL , Respiração com Pressão Positiva/métodos , Respiração Artificial/métodos , Volume de Ventilação Pulmonar/fisiologia , Capacidade Pulmonar Total/fisiologia
5.
Int J Mol Med ; 33(4): 833-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24452195

RESUMO

In the present study, we investigated whether proangiogenic growth factors and endothelial progenitor cells (EPCs) induce favourable effects on cutaneous incisional wound healing in diabetic mice. The proangiogenic effects of human EPCs were initially analyzed using a HUVEC in vitro angiogenesis assay and an in vivo Matrigel assay in nude mice (n=12). For the diabetic wound model, 48 Balb/c mice with streptozotocin (STZ)-induced diabetes were divided randomly into 4 groups (12 mice in each group). Subsequently, 3, 5 and 7 days before a 15-mm full-thickness incisional skin wound was set, group 1 was pre-treated subcutaneously with a mixture of vascular endothelial growth factor (VEGF)/basic fibroblast growth factor (bFGF)/platelet-derived growth factor (PDGF) (3.5 µg of each), group 2 with 3.5 µg PDGF and group 3 with an aliquot of two million EPCs, whereas the control animals (group 4) were pre-treated with 0.2 ml saline solution. The wounds were assessed daily and the repaired tissues were harvested 7 days after complete wound closure. The angiogenesis assay demonstrated significantly increased sprout densities, areas and lengths in the EPC-treated group (all p<0.01). In the Matrigel assay, significantly increased microvessel densities, areas and sizes (all p<0.001) were also detected in the EPC-treated group. In the STZ-induced model of diabetes, the animals pre-treated with a combination of proangiogenic factors and EPCs showed in general, a more rapid wound closure. Vessel densities were >2-fold higher in the mice treated with a combination of proangiogenic factors and EPCs (p<0.05) and tensile strengths were higher in the groups treated with proangiogenic growth factors compared to the controls (p<0.05). These results suggest a beneficial effect of pre-treatment with proangiogenic growth factors and EPCs in incisional wound healing.


Assuntos
Diabetes Mellitus Experimental/patologia , Células Endoteliais/citologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Transplante de Células-Tronco , Células-Tronco/citologia , Cicatrização/efeitos dos fármacos , Animais , Bioensaio , Colágeno/metabolismo , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Experimental/cirurgia , Combinação de Medicamentos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Laminina/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Microvasos/efeitos dos fármacos , Microvasos/patologia , Proteoglicanas/metabolismo , Células-Tronco/efeitos dos fármacos , Resistência à Tração/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa