当前位置: 首页 > 医学版 > 期刊论文 > 医药卫生总论 > 美国呼吸和危急护理医学 > 2005年 > 第6期 > 正文
编号:11259661
Rescue of the Hypoplastic Lung by Prenatal Cyclical Strain
     Division of Maternal and Child Health Sciences, University of Dundee, Dundee

    Department of Paediatric Surgery, Royal Hospital for Sick Children and University of Glasgow

    Department of Obstetrics and Fetal Medicine, Queen Mother's Hospital

    Department of Veterinary Anatomy, University of Glasgow, Glasgow, Scotland, United Kingdom.

    ABSTRACT

    We determined the effects of sustained and cyclical prenatal mechanical strain on the hypoplastic lung of the ovine model of congenital diaphragmatic hernia. Over a period of 4 weeks in late gestation, repeated cyclical tracheal occlusion for 23 hours with 1-hour release stimulated minimal growth, but promoted maturation with the development of a saccular lung. In contrast, a cycle consisting of 47 hours with 1-hour release induced optimal lung growth and morphologic maturation of the hypoplastic lung parenchyma. Sustained occlusion resulted in exaggerated lung growth, exceeding that of unaffected controls, and abnormal alveolar development. The extent of induction of lung growth by mechanical strain was inversely proportional to the number of alveolar type II cells remaining in the lung epithelium. These studies show that, although mechanical strain is capable of inducing lung growth and differentiation, cyclical strain is a prerequisite for normal development and that mechanically induced growth occurs at the expense of the alveolar type II cell. We conclude that cyclical strain may allow optimal alveolar development while maintaining a population of alveolar type II cells and may thus facilitate an improvement in postnatal lung function in infants with congenital diaphragmatic hernia.

    Key Words: diaphragmatic hernia fetoscopy fetus lung

    Despite advances in neonatal care, 50% of infants affected by isolated severe congenital diaphragmatic hernia (CDH) die with respiratory insufficiency secondary to pulmonary hypoplasia (1eC3). Survival is associated with significant long-term morbidity, primarily from pulmonary, neurologic, and gastrointestinal disease (4eC8).

    It is well established that lung growth can be modified by manipulating lung liquid volume. Thus drainage of fetal lung fluid retards growth, resulting in pulmonary hypoplasia (9, 10), whereas tracheal occlusion results in pulmonary hyperplasia (9eC16). Tracheal occlusion has been evaluated as an in utero treatment for surgically created CDH in fetal lambs. Although this procedure expands the lung, increases mass, and temporarily improves gas exchange (12), the lungs differentiate abnormally (11), are deplete of type II alveolar cells (17) and surfactant (18), and develop respiratory failure and pulmonary hypertension after birth (19). Further studies have examined the reversal of tracheal occlusion before birth with variable results (20, 21). Prenatal tracheal occlusion has also been applied to human fetuses (22); the initial results were poor (23), but even more recent studies show survival similar to standard postnatal care (2).

    Sustained strain causes injury to organotypic lung cultures (24), whereas in vivo sustained strain after tracheal occlusion increases DNA synthesis, which peaks 2 days postocclusion and is largely complete by 4 days (25); subsequently, there is a resumption of growth at control rates (26). The lack of cyclical variation in the degree of mechanical strain, a prerequisite for optimal growth in a number of tissues (27, 28), including lung (24), may be responsible for this plateauing of lung growth (27, 28). In organotypic lung culture, rapid cyclical elongation is essential for proliferation of epithelial cells and fibroblasts (27eC29) and causes a selective increase in steady state mRNA levels of surfactant protein type C (30).

    In the latter part of gestation, fetal breathing movements (FBM) occur approximately 30% of the time (31) and, although individually they are virtually isovolumetric (32, 33), continuous periods of FBM are associated with diminished outflow resistance and thus increased egress of fluid ("deflation") (31). Furthermore, the abolition of a cyclical strain to the fetal lung arrests normal differentiation (34). We hypothesized that the periodic loss of volume (and consequent decrease in mechanical strain) during periods of FBM is necessary for normal differentiation (17), and we considered it possible that the fetal lung growth and development achieved by static occlusion could be further improved by applying a cyclical variation in strain. Because DNA synthesis is at its highest during the first 2 days of tracheal occlusion (25), we chose to evaluate cycles of 23 and 47 hours' occlusion, each with 1-hour release, to define which best achieved lung growth with preservation of the alveolar type II cell population.

    METHODS

    Experimental Design

    All animal work was overseen by the Animal Ethics Committee of Glasgow University Veterinary School, and the United Kingdom Home Office approved all procedures under the terms of current United Kingdom animal legislation. Five groups of lamb fetuses from Texel cross ewes (full term 145 ± 1 days) were studied: (1) the CDH group (n = 6) was obtained by creating CDH in utero at 80 days' gestation; (2) the CDH/TO23h group (n = 3) underwent CDH creation at 80 days and tracheal occlusion with a endoluminal silicone balloon between 110 and 138 days—tracheal occlusion was for 23 hours with 1-hour release, and this cyclical regimen was repeated continuously for 28 days; (3) the CDH/TO47h group (n = 4) underwent CDH creation at 80 days and cyclical tracheal occlusion for 47 hours with 1-hour release repeatedly from 110 to 138 days; (4) the CDH/TOstatic group (n = 3) underwent CDH creation at 80 days and sustained tracheal occlusion from 110 to 138 days; and (5) the control group (n = 6), consisted of unoperated fetuses derived from twins of the intervention groups. A total of 58 CDH fetuses were created at 80 days; 33 ewes underwent a second intervention at 110 days for placement of fetal endoluminal balloons. Because of a combination of fetal deaths, abortions, balloon failures, and healed diaphragms, 42 fetuses were excluded from analysis and were not included in the study groups. These losses reduced the numbers of animals in each of the three treatment groups, but further studies were not permitted by the United Kingdom Home Office on the grounds that, in spite of the small numbers, the 47-hour cycle had been shown to be statistically superior for the outcomes studied compared with the CDH, 23-hour cyclical, and static occlusion regimes.

    Fetal Surgical Procedures

    At 80 days' gestation, maternal and fetal anesthesia was induced and the left-sided diaphragmatic defect created via a laparotomy and hysterotomy as previously described (19, 35). At 110 days' gestation, the ewes underwent a second laparotomy and hysterotomy and a Syntel Latex-Free 4F Arterial Embolectomy Catheter (Applied Medical Resources Corp., Medlink Europe, Eindhoven, The Netherlands) was placed under direct vision into the fetal trachea, secured to the fetal mandible, and tunnelled subcutaneously onto the maternal flank. Externally, the catheter was connected to a three-way tap facilitating balloon inflation and deflation.

    At 138 days' gestation, the ewes were killed by an intravenous bolus of sodium pentobarbital and the fetuses retrieved. Each lamb was weighed, the chest wall removed, and the lungs excised and weighed. The isolated left lung was fixed by concurrent immersion and bronchial instillation of 10% buffered formalin at constant pressure of 25 cm H2O.

    Histologic and Immunohistochemical Studies

    Design-based sampling methods were combined with blinded morphometric assessment of the left lung (36, 37). The data sets and hierarchical equations studied were as previously described (38). Morphometric analysis of the alveolar type II cell population was performed as described previously (39). Additional details on the histologic and stereologic techniques are provided in the online supplement.

    Statistical Analyses

    Continuous variables were summarized by the mean and SEM. Comparison of three or more means was made using analysis of variance (ANOVA). Multiple comparisons after ANOVA were performed using Tukey's method. Correlation was determined by Pearson correlation coefficient and Spearman's rho. Statistical significance was assumed at the 5% level and analysis was performed using SPSS version 11.0 (SPSS Inc., Chicago, IL).

    RESULTS

    Body and Lung Weights

    In the CDH group, the total (p < 0.0001) and individual right (p < 0.0001) and left (p < 0.0001) lung weights were reduced compared with the control group (Table 1). In the CDH/TO23h group, lung weights and lung weight to body weight ratios were similar to the CDH group. In contrast, the CDH/TO47h group had increased total and individual lung weights (p < 0.0001) compared with the CDH group and were similar to the control group (Table 1). Lung weights were much increased in the CDH/TOstatic group (p < 0.0001) compared with all groups, including the controls (Table 1). The dry:wet ratios did not differ between groups (p = 0.09) (Table 1), although the CDH lung had an increased ratio consistent with a more cellular lung. That progressive occlusions decreased this ratio is suggestive of an increased water content with longer occlusions.

    Lung Volumes

    The creation of a diaphragmatic defect resulted in a reduction in lung volume (p < 0.001) and parenchyma (p < 0.001) compared with the control group (Table 2). An alteration in lung composition is evident with 58% parenchymal tissue in the CDH group compared with 90% in control fetuses (p < 0.001). The CDH/TO23h group had a small, nonsignificant increase in fixed lung volume and parenchymal tissue, but the CDH/TO47h group had significantly increased lung (p = 0.014) and parenchymal (p < 0.01) volumes compared with control values. The CDH/TOstatic group showed an excessive increase in volume that exceeded control values (p < 0.0001). Pleural (p < 0.05) volumes were significantly increased only in the CDH/TOstatic group. There were no differences in interlobular septal volumes for any group.

    Alveolar Morphometry

    Alveolar tissue volume fractions, wall thicknesses, and total surface areas differed between the groups (Figure 1). The CDH group had a significantly smaller alveolar surface area compared with the control group, consistent with the decrease in lung volume. This reduction in alveolar total surface area was reversed by the three tracheal occlusion treatments, and longer periods of occlusion were associated with greater increments in alveolar surface area (Figure 1). As the alveolar total surface area increased, the alveolar tissue volume fractions were significantly reduced in the CDH/TO47h and CDH/TOstatic groups compared with the CDH group. Alveolar wall thickness was reduced in the CDH/TO47h group, but not the CDH/TO23h group, compared with the CDH group (p = 0.026) (Figure 1).

    Total alveolar number in the CDH group was significantly smaller than the control group (p < 0.001), but the alveolar numerical density did not differ (Figure 2). The total alveolar number was significantly reduced in all three occlusion groups compared with the control group; alveolar numerical density was also decreased but not significantly. Average alveolar volume was only increased in the CDH/TOStatic group compared with the control group (p < 0.001) (Figure 2).

    Lung Morphology

    The pulmonary parenchyma of the CDH group had a very immature structure resembling the architecture of the normal late canalicular phase with significant heterogeneity resulting from the coexistence of hypoplastic and developmentally advanced parenchyma (Figure 3). Lungs from the CDH/TO23h group were quite markedly different from the CDH group and resembled the saccular phase of lung development (Figure 3). The parenchymal component was characterized by thick-walled saccular airspaces of variable size with little evidence of alveolarization and secondary crest development. Lungs from the CDH/TO47h group were further advanced than either the CDH group or the CDH/TO23h group and were very similar to the control group of 138 days in appearance (Figure 3). There was a dramatic increase in the parenchymal component, with the conducting airways being larger, smoother, and rounder than untreated CDH. The terminal and respiratory bronchioles were well defined, with alveolar ducts frequently originating from them. The alveolar ducts were interrupted by alveolar crests forming a large complex number of thin-walled alveoli. As determined by high-powered light microscopy, the aireCblood barrier was well developed with single capillary complexes, little connective tissue, and attenuated alveolar epithelial type I cells. Similar to the control group, the large bronchi were lined with simple cuboidal cells and contained discrete glands within the underlying wall. Cartilage plates, blood vessels, and ganglia were also enmeshed in a thick fibromuscular coat. The lungs from the CDH/TOstatic group appeared more mature than those of the CDH group, although overall the alveoli were considerably larger than the control group and were abnormal in structure and appearance, with some similarities to an emphysematous lung (Figure 3).

    Alveolar Type II Cells

    The CDH group alveolar type II cells were more frequent in hypoplastic areas, but in developmentally advanced areas were of similar frequency and distribution to those of the control group. Alveolar type II cells were reduced in both the CDH/TO23h and CDH/TO47h groups when compared with both the control and CDH groups. The distended alveoli of the CDH/TOstatic group were virtually devoid of alveolar type II cells.

    The total number of alveolar type II cells was greatest in the control group, with significantly fewer (p = 0.02) and smaller (p = 0.006) cells present in the CDH group (Figure 4). Reductions in alveolar type II cell number, but not size, were observed in the three occlusion groups compared with CDH and control groups (Figure 4). Alveolar type II cell numerical density was negatively correlated with fixed left lung volume (Figure 5); Pearson's correlation coefficient eC0.63, (p = 0.004).

    DISCUSSION

    The aim of this study was to analyze the growth and maturation of fetal lungs after cyclical or sustained tracheal occlusion in a surgical model of diaphragmatic hernia. CDH results in a reduction in lung weight, volume, airway generation, total number of acini and alveoli, abnormal development of bronchiolar cartilage, abnormal muscularization of the arteriolar wall (40, 41), and decreased cross-sectional area of the pulmonary vascular bed (42). Our surgical model of CDH, with its associated lung hypoplasia that is present by 96 days' gestation and increases in severity toward term (43), is consistent with the known effects of CDH in both animals (35, 44, 45) and humans (40, 46, 47).

    During the last third of gestation in humans and sheep, lung development is characterized by expansion of the primitive airways, interstitial thinning with reduction of aireCblood barrier thickness, and the subdivision of terminal saccules to form alveoli, thus increasing the surface area for gas exchange (48). These developmental changes are dependent on adequate lung liquid volumes generated by an active secretory process which gives rise to secretion rates of between 2.7 and 3.7 ml/kg/h over the last trimester (49). Reductions in luminal liquid volume are associated with pulmonary hypoplasia (9, 26); conversely, tracheal occlusion to prevent egress of lung liquid accelerates fetal lung growth (9, 50eC53). Lung liquid volume increases rapidly at the onset of occlusion, doubling within the first 24 hours, and then increasing more slowly to plateau after 7 days (25). Intraluminal tracheal pressure similarly increases after tracheal occlusion but, unlike lung volume, does not correlate with lung growth (25).

    We compared the potential of tracheal occlusion for 23 hours with 1-hour deflation (CDH/TO23h) and tracheal occlusion for 47 hours with 1-hour deflation (CDH/TO47h) with static occlusion (CDH/TOstatic) applied over 4 weeks to rescue the hypoplastic lung phenotype. Because of the small numbers in the three treatment groups, we cannot exclude the possibility that Type II errors are responsible for the apparent lack effect of CDH/TO23h, compared with CDH, on any of the outcomes measured. Nevertheless, despite the small numbers, we are confident in our conclusion that CDH/TO47h provides significantly better lung development than CDH/TO23h. The outcomes we found to be significantly different by ANOVA and post hoc analysis remain significant if subjected to nonparametric statistical analysis and, for these outcomes, the CDH/TO47h 95% confidence interval lower boundaries lie well above the CDH/TO23h 95% confidence interval upper boundaries. Our observation that static occlusion (CDH/TOstatic) results in lung hyperplasia and poor differentiation is consistent with previous reports (11, 12).

    Our finding that, of the two cyclical protocols, only CDH/TO47h resulted in lung growth, indicates that the cellular mechanisms responsible for transducing the increase of lung volume into lung growth only become effective during the second 23-hour period of obstruction, even though it has been observed that neither fetal lung liquid volume nor tracheal pressure increase during the second half of the 47-hour occlusion period (25). We would expect the growth transduction signal to the lung to be similar after 23 hours or 47 hours of occlusion if lung lumen volume was the only factor underlying the induction of lung growth, but the duration and frequency of exposure to this increased luminal volume may be a key factor in determining the fetal lung response.

    After release of tracheal occlusion, we anticipated an immediate egress of fetal lung fluid as a result of the increased pressure gradient between tracheal lumen and amniotic liquid; this was confirmed by postmortem observations. Had there been no volume loss, we would have expected growth to be equivalent to that of the CDH/TOstatic group (i.e., hyperplastic with abnormal differentiation). That this was not the outcome suggests that the periodic loss of volume in our cyclical CDH/TO47h group is essential for lung differentiation.

    The periods of increased intraluminal volume and mechanical strain in the cyclical occlusion groups have demonstrable effects on lung maturation that are consistent with in vitro observations (54). The mechanism by which increased intraluminal volume and, consequently, pressure is translated into acceleration of fetal lung growth is not clear. The differences in the maturation of the CDH/TO23h and CDH/TO47h groups suggest that time exposed to mechanical stress is a key determinant of the fetal lung response. The tensegrity model for mechanoregulation based on adhesion molecules and the cytoskeleton (55) would be consistent with exposure time as a key factor (56). In this system, prolonged physical distortion of the lung from tracheal occlusion would change force distributions across adhesion receptors, resulting in both restructuring of the cytoskeleton network and activation of diverse cell signaling activities. Local changes in the cellular force balance may contribute to pattern formation and differentiation during tissue morphogenesis. In support of this, inhibition of Rho-kinase, a GTPase, which promotes myosin light chain kinase phosphorylation and stimulates cytoskeleton contraction, decreases morphogenetic branching of E12 mouse lung rudiments (57). Conversely, treatment to increase cytoskeleton tension with a low dose of a specific activator of Rho, cytotoxic necrotizing factor-1, increases branching (57).

    The increase in the alveolar type II cell population in the CDH lung as measured by immunohistochemical computer-assisted stereology is in agreement with previous studies using electron microscopy (58) and in situ hybridization for surfactant-B mRNA (59). The application of a cyclical stretch in the CDH/TO23h and CDH/47h groups caused reductions in the alveolar type II cell population compared with controls, but in each case the reductions were less than in the CDH/TOstatic group. Alveolar type II cell depletion by tracheal occlusion may be attributable to increased transdifferentiation to alveolar type I cells rather than decreased cell proliferation or cell death. Thus distension of alveolar type II cells increases RTI40 mRNA expression, a type I phenotype marker, and decreases mRNA expression of type II markers, surfactant proteins B and C (60). These in vitro observations are supported by in vivo studies that show that sustained tracheal occlusion over a 10-day period decreases alveolar type II cell and increases type I cell numbers, with the transient appearance of an intermediary cell type displaying a mixed phenotype without any evidence of increased proliferation or apoptosis (61). Additionally, it has been shown that tracheal occlusion is associated with reductions in disaturated phosphatidylcholine (62) and surfactant protein expression (62eC64). Deflation of the fetal lung induces differentiation in the opposite direction with an increase in alveolar type II cells and surfactant protein mRNA levels (65). Similar changes are also described in cultured fetal explants (66).

    The correlation of increased fetal lung volume with a decreased number of alveolar type II cells is consistent with the concept of differentiation of alveolar type II into type I cells to facilitate the increase in alveolar surface area. The application of a cyclical strain to the alveolar type II cell with its periodic break from growth stimuli may be responsible for the increased number of alveolar type II cells maintained in the CDH/TO23h and CDH/TO47h groups compared with the CDH/TOstatic group. The level of lung transforming growth factor (TGF)-2, one of a family of inhibitory growth factors known to inhibit alveolar type II cell development (67), has been observed to increase in response to tracheal occlusion (68); therefore, the periodic escape from TGF-2 inhibition could explain the link between periodic lung deflation and the retention of a population of alveolar type II cells in the cyclical strain groups. The observed morphologic differences between the cyclical strain groups and the untreated CDH and CDH/TOstatic lung groups suggest that the maintenance of a population of alveolar type II cells with improved postnatal lung function may be achieved in the CDH/TO47h group.

    We have demonstrated that mechanical distension by prenatal tracheal occlusion accelerates lung growth and that cyclical strain induces optimal growth and differentiation. We speculate that these findings may also explain the postnatal induction of lung growth by liquid mediated cyclical distension of airspaces. Extracorporeal membrane oxygenation (ECMO) has been extensively used in the management of CDH, principally in the rescue of infants with severe hypoxemia after surgical repair (69eC71). Although pulmonary growth and vascular remodeling do take place in CDH infants after birth, ECMO does not offer enough time for these changes to have a significant impact on outcome (72). Active controlled acceleration of postnatal lung growth would be necessary for the pulmonary hypoplasia to be meaningfully reversed. Lung growth has been achieved in postnatal animal models with continuous intrapulmonary distension by perfluorocarbon for 3 weeks (73). This approach has now been piloted in human patients on ECMO, with survival of unsalvageable infants marooned on ECMO (74, 75). In both of these human studies, a cyclical strain was applied to the lungs as the perfluorocarbons were exchanged daily. However, in contrast to the CDH/TO23h group, which was dependent on the lung liquid secretory mechanism to generate a positive intrapulmonary distending pressure, a continuous positive airway pressure of 4eC7 mm Hg was used in the ECMO patients, a value only achieved after 15eC24 hours of prenatal tracheal occlusion in nonhypoplastic fetal lungs (76). Therefore, these postnatal results achieved with artificial liquid distension of the developing lung bear some resemblance to the CDH/47h prenatal group, in which the lungs were distended by endogenous lung liquid at these pressures for at least 24 hours.

    The optimal strategy for rescuing the hypoplastic lung, as measured by morphologic criteria, would appear to be a 47/1-hour cyclical occlusion, performed between 0.76 and 0.95 gestation, given the 145-day gestation of sheep. In contrast, the human fetus has a gestation of 283 days (77), and this intervention period would correspond to 30.7eC38 weeks, much later than previous fetal interventions have been performed (2). It is not clear whether this would provide sufficient time for fetal lung growth to occur. It is similarly unclear whether 47-hour occlusion with 1-hour release would be the optimal regimen in human fetuses because lung liquid production rates in human fetuses have not been quantified, although, as suggested from the postnatal perfluorodecalin studies, this occlusion frequency may be sufficient (75). Moreover, before the application of these experiments to human fetuses, further animal studies are required to confirm that the improved growth, differentiation, and preservation of the small population of alveolar type II cells observed in the CDH/TO47h group correlates with an improved postnatal lung function. These functional studies could be combined usefully with further studies of differentiation by electron microscopy to determine the maturity of the aireCblood barrier. Last, the use of exteriorized fetal catheters is not applicable to humans and significant technologic development is required to create a system comprising a detachable balloon (78) and a miniaturized remotely activated implantable pump (79), which could be used in human gestations.

    These caveats notwithstanding, prenatal cyclical tracheal occlusion is a novel potential treatment for the hypoplastic lung of congenital diaphragmatic hernia. It remains to be seen whether additional strategies for increasing the population of alveolar type II cells, such as a period of continuous deflation after a period of cyclical strain or corticosteroids, are effective.

    Acknowledgments

    The authors thank Christine Stirton and her staff for their excellent technical assistance in the care of the animals.

    This article has an online supplement, which is accessible from this issue's table of contents at www.atsjournals.org

    REFERENCES

    Langham MR Jr, Kays DW, Ledbetter DJ, Frentzen B, Sanford LL, Richards DS. Congenital diaphragmatic hernia: epidemiology and outcome. Clin Perinatol 1996;23:671eC688.

    Harrison MR, Keller RL, Hawgood SB, Kitterman JA, Sandberg PL, Farmer DL, Lee H, Filly RA, Farrell JA, Albanese CT. A randomized trial of fetal endoscopic tracheal occlusi for severe fetal congenital diaphragmatic hernia. N Engl J Med 2003;349:1916eC1924.

    Beresford MW, Shaw NJ. Outcome of congenital diaphragmatic hernia. Pediatr Pulmonol 2000;30:249eC256.

    Muratore CS, Kharasch V, Lund DP, Sheils C, Friedman S, Brown C, Utter S, Jaksic T, Wilson JM. Pulmonary morbidity in 100 survivors of congenital diaphragmatic hernia monitored in a multidisciplinary clinic. J Pediatr Surg 2001;36:133eC140.

    Schwartz IP, Bernbaum JC, Rychik J, Grunstein M, D'Agostino J, Polin RA. Pulmonary hypertension in children following extracorporeal membrane oxygenation therapy and repair of congenital diaphragmatic hernia. J Perinatol 1999;19:220eC226.

    Lund DP, Mitchell J, Kharasch V, Quigley S, Kuehn M, Wilson JM. Congenital diaphragmatic hernia: the hidden morbidity. J Pediatr Surg 1994;29:258eC262.

    Reid IS, Hutcherson RJ. Long-term follow-up of patients with congenital diaphragmatic hernia. J Pediatr Surg 1976;11:939eC942.

    Muratore CS, Utter S, Jaksic T, Lund DP, Wilson JM. Nutritional morbidity in survivors of congenital diaphragmatic hernia. J Pediatr Surg 2001;36:1171eC1176.

    Alcorn D, Adamson TM, Lambert TF, Maloney JE, Ritchie BC, Robinson PM. Morphological effects of chronic tracheal ligation and drainage in the fetal lamb lung. J Anat 1977;123:649eC660.

    Carmel J, Friedman F, Adams F. Fetal tracheal ligation and tracheal development. Am J Dis Child 1965;109:452.

    Lipsett J, Cool JC, Runciman SI, Ford WD, Kennedy JD, Martin AJ. Effect of antenatal tracheal occlusion on lung development in the sheep model of congenital diaphragmatic hernia: a morphometric analysis of pulmonary structure and maturity. Pediatr Pulmonol 1998;25:257eC269.

    DiFiore JW, Fauza DO, Slavin R, Peters CA, Fackler JC, Wilson JM. Experimental fetal tracheal ligation reverses the structural and physiological effects of pulmonary hypoplasia in congenital diaphragmatic hernia. J Pediatr Surg 1994;29:248eC256.

    Bullard KM, Sonne J, Hawgood S, Harrison MR, Adzick NS. Tracheal ligation increases cell proliferation but decreases surfactant protein in fetal murine lungs in vitro. J Pediatr Surg 1997;32:207eC211. [Discussion J Pediatr Surg 1997;32:211eC213).

    Zgleszewski SE, Cilley RE, Krummel TM, Chinoy MR. Effects of dexamethasone, growth factors, and tracheal ligation on the development of nitrofen-exposed hypoplastic murine fetal lungs in organ culture. J Pediatr Surg 1999;34:1187eC1195.

    Kitano Y, Yang EY, von Allmen D, Quinn TM, Adzick NS, Flake AW. Tracheal occlusion in the fetal rat: a new experimental model for the study of accelerated lung growth. J Pediatr Surg 1998;33:1741eC1744.

    De Paepe ME, Johnson BD, Papadakis K, Sueishi K, Luks FI. Temporal pattern of accelerated lung growth after tracheal occlusion in the fetal rabbit. Am J Pathol 1998;152:179eC190.

    De Paepe ME, Papadakis K, Johnson BD, Luks FI. Fate of the type II pneumocyte following tracheal occlusion in utero: a time-course study in fetal sheep. Virchows Arch 1998;432:7eC16.

    O'Toole SJ, Sharma A, Karamanoukian HL, Holm B, Azizkhan RG, Glick PL. Tracheal ligation does not correct the surfactant deficiency associated with congenital diaphragmatic hernia. J Pediatr Surg 1996;31:546eC550.

    O'Toole SJ. The pulmonary surfactant system in congenital diaphragmatic hernia and the influence of fetal surgery on its development. MD thesis 1999; University of Newcastle upon Tyne, England.

    Wild YK, Piasecki GJ, De Paepe ME, Luks FI. Short-term tracheal occlusion in fetal lambs with diaphragmatic hernia improves lung function, even in the absence of lung growth. J Pediatr Surg 2000;35:775eC779.

    Davey MG, Hedrick HL, Bouchard S, Mendoza JM, Schwarz U, Adzick NS, Flake AW. Temporary tracheal occlusion in fetal sheep with lung hypoplasia does not improve postnatal lung function. J Appl Physiol 2003;94:1054eC1062.

    VanderWall KJ, Skarsgard ED, Filly RA, Eckert J, Harrison MR. Fetendo-clip: a fetal endoscopic tracheal clip procedure in a human fetus. J Pediatr Surg 1997;32:970eC972.

    Flake AW, Crombleholme TM, Johnson MP, Howell LJ, Adzick NS. Treatment of severe congenital diaphragmatic hernia by fetal tracheal occlusion: clinical experience with fifteen cases. Am J Obstet Gynecol 2000;183:1059eC1066.

    Liu M, Skinner SJ, Xu J, Han RN, Tanswell AK, Post M. Stimulation of fetal rat lung cell proliferation in vitro by mechanical stretch. Am J Physiol 1992;263:L376eCL383.

    Nardo L, Hooper SB, Harding R. Stimulation of lung growth by tracheal obstruction in fetal sheep: relation to luminal pressure and lung liquid volume. Pediatr Res 1998;43:184eC190.

    Hooper SB, Han VK, Harding R. Changes in lung expansion alter pulmonary DNA synthesis and IGF-II gene expression in fetal sheep. Am J Physiol 1993;265:L403eCL409.

    Liu M, Xu J, Liu J, Kraw ME, Tanswell AK, Post M. Mechanical strain-enhanced fetal lung cell proliferation is mediated by phospholipase C and D and protein kinase C. Am J Physiol 1995;268:L729eCL738.

    Stelnicki EJ, Komuves LG, Holmes D, Clavin W, Harrison MR, Adzick NS, Largman C. The human homeobox genes MSX-1, MSX-2, and MOX-1 are differentially expressed in the dermis and epidermis in fetal and adult skin. Differentiation 1997;62:33eC41.

    Liu M, Xu J, Souza P, Tanswell B, Tanswell AK, Post M. The effect of mechanical strain on fetal rat lung cell proliferation: comparison of two- and three-dimensional culture systems. In Vitro Cell Dev Biol Anim 1995;31:858eC866.

    Nakamura T, Liu M, Slutsky AS, Post M. Glucocorticoids differentially affect mechanical strain induced SP-C and tropoelastin mRNA expression in fetal rat lung cells. Am J Respir Crit Care Med 1997;155:A273.

    Harding R, Bocking AD, Sigger JN. Influence of upper respiratory tract on liquid flow to and from fetal lungs. J Appl Physiol 1986;61:68eC74.

    Dawes GS, Fox HE, Leduc BM, Liggins GC, Richards RT. Respiratory movements and rapid eye movement sleep in the foetal lamb. J Physiol 1972;220:119eC143.

    Harding R, Hooper SB. Regulation of lung expansion and lung growth before birth. J Appl Physiol 1996;81:209eC224.

    Wigglesworth JS, Desai R. Effect on lung growth of cervical cord section in the rabbit fetus. Early Hum Dev 1979;3:51eC65.

    de Lorimier AA, Tierney DF, Parker HR. Hypoplastic lungs in fetal lambs with surgically produced congenital diaphragmatic hernia. Surgery 1967;62:12eC17.

    Bolender RP, Hyde DM, Dehoff RT. Lung morphometry: a new generation of tools and experiments for organ, tissue, cell, and molecular biology. Am J Physiol 1993;265:L521eCL548.

    Scherle W. A simple method for volumetry of organs in quantitative stereology. Mikroskopie 1970;26:57.

    Willet KE, Jobe AH, Ikegami M, Newnham J, Brennan S, Sly PD. Antenatal endotoxin and glucocorticoid effects on lung morphometry in preterm lambs. Pediatr Res 2000;48:782eC788.

    De Paepe ME, Papadakis K, Johnson BD, Luks FI. Fate of the type II pneumocyte following tracheal occlusion in utero: a time-course study in fetal sheep. Virchows Arch 1998;432:7eC16.

    Nakamura Y, Yamamoto I, Fukuda S, Hashimoto T. Pulmonary acinar development in diaphragmatic hernia. Arch Pathol Lab Med 1991;115:372eC376.

    Kitagawa M, Hislop A, Boyden EA, Reid L. Lung hypoplasia in congenital diaphragmatic hernia: a quantitative study of airway, artery, and alveolar development. Br J Surg 1971;58:342eC346.

    Adzick NS, Outwater KM, Harrison MR, Davies P, Glick PL, deLorimier AA, Reid LM. Correction of congenital diaphragmatic hernia in utero: IV: an early gestational fetal lamb model for pulmonary vascular morphometric analysis. J Pediatr Surg 1985;20:673eC680.

    Evrard V. Towards an endoscopic intra-uterine treatment for congenital diaphragmatic hernia: preclinical experimental studies in the foetal lamb. PhD thesis 2002; Katholieke Universiteit Leuven, The Netherlands.

    Lipsett J, Cool JC, Runciman SC, Kennedy JD, Martin AJ, Byard RW, Ford WD. Morphometric analysis of pulmonary development in the sheep following creation of fetal diaphragmatic hernia. Pediatr Pathol Lab Med 1997;17:789eC807.

    Ohi R, Suzuki H, Kato T, Kasai M. Development of the lung in fetal rabbits with experimental diaphragmatic hernia. J Pediatr Surg 1976;11:955eC959.

    Areechon W, Reid L. Hypoplasia of the lung with congenital diaphragmatic hernia. BMJ 1963;1:230eC233.

    George DK, Cooney TP, Chiu BK, Thurlbeck WM. Hypoplasia and immaturity of the terminal lung unit (acinus) in congenital diaphragmatic hernia. Am Rev Respir Dis 1987;136:947eC950.

    Alcorn DG, Alexander IG, Maloney JE, Ritchie BC, Walker AM. Morphological development of the lung: a review. Aust Paediatr J 1974;10:189eC198.

    Olver RE, Schneeberger EE, Walters DV. Epithelial solute permeability, ion transport and tight junction morphology in the developing lung of the fetal lamb. J Physiol 1981;315:395eC412.

    Wilson JM, DiFiore JW, Peters CA. Experimental fetal tracheal ligation prevents the pulmonary hypoplasia associated with fetal nephrectomy: possible application for congenital diaphragmatic hernia. J Pediatr Surg 1993;28:1433eC1439.

    DiFiore JW, Fauza DO, Slavin R, Peters CA, Fackler JC, Wilson JM. Experimental fetal tracheal ligation reverses the structural and physiological effects of pulmonary hypoplasia in congenital diaphragmatic hernia. J Pediatr Surg 1994;29:248eC256.

    Bullard KM, Sonne J, Hawgood S, Harrison MR, Adzick NS. Tracheal ligation increases cell proliferation but decreases surfactant protein in fetal murine lungs in vitro. J Pediatr Surg 1997;32:207eC211.

    Kitano Y, Davies P, von Allmen D, Adzick NS, Flake AW. Fetal tracheal occlusion in the rat model of nitrofen-induced congenital diaphragmatic hernia. J Appl Physiol 1999;87:769eC775.

    Liu M, Tanswell AK, Post M. Mechanical force-induced signal transduction in lung cells. Am J Physiol 1999;277:L667eCL683.

    Ingber D. Integrins as mechanochemical transducers. Curr Opin Cell Biol 1991;3:841eC848.

    Ingber DE. Tensegrity: the architectural basis of cellular mechanotransduction. Annu Rev Physiol 1997;59:575eC599.

    Moore KA, Huang S, Kong Y, Sunday ME, Ingber DE. Control of embryonic lung branching morphogenesis by the Rho activator, cytotoxic necrotizing factor 1. J Surg Res 2002;104:95eC100.

    Hashimoto EG, Pringle KC, Soper RT, Brown CK. The creation and repair of diaphragmatic hernia in fetal lambs: morphology of the type II alveolar cell. J Pediatr Surg 1985;20:354eC356.

    Benachi A, Chailley-Heu B, Delezoide AL, Dommergues M, Brunelle F, Dumez Y, Bourbon JR. Lung growth and maturation after tracheal occlusion in diaphragmatic hernia. Am J Respir Crit Care Med 1998;157:921eC927.

    Gutierrez JA, Gonzalez RF, Dobbs LG. Mechanical distension modulates pulmonary alveolar epithelial phenotypic expression in vitro. Am J Physiol 1998;274:L196eCL202.

    Flecknoe S, Harding R, Maritz G, Hooper SB. Increased lung expansion alters the proportions of type I and type II alveolar epithelial cells in fetal sheep. Am J Physiol Lung Cell Mol Physiol 2000;278:L1180eCL1185.

    Joe P, Wallen LD, Chapin CJ, Lee CH, Allen L, Han VK, Dobbs LG, Hawgood S, Kitterman JA. Effects of mechanical factors on growth and maturation of the lung in fetal sheep. Am J Physiol 1997;272:L95eC105.

    Piedboeuf B, LaBerge JM, Ghitulescu G, Gamache M, Petrov P, Belanger S, Chen MF, Hashim E, Possmayer F. Deleterious effect of tracheal obstruction on type II pneumocytes in fetal sheep. Pediatr Res 1997;41:473eC479.

    Flageole H, Evrard VA, Piedboeuf B, LaBerge JM, Lerut TE, Deprest JA. The plug-unplug sequence: an important step to achieve type II pneumocyte maturation in the fetal lamb model. J Pediatr Surg 1998;33:299eC303.

    Flecknoe SJ, Wallace MJ, Harding R, Hooper SB. Determination of alveolar epithelial cell phenotypes in fetal sheep: evidence for the involvement of basal lung expansion. J Physiol 2002;542:245eC253.

    Gutierrez JA, Ertsey R, Scavo LM, Collins E, Dobbs LG. Mechanical distention modulates alveolar epithelial cell phenotypic expression by transcriptional regulation. Am J Respir Cell Mol Biol 1999;21:223eC229.

    Fraslon C, Lacaze-Masmonteil T, Zupan V, Chailley-Heu B, Bourbon JR. Fetal rat lung type II cell differentiation in serum-free isolated cell culture: modulation and inhibition. Am J Physiol 1993;264:L504eCL516.

    Quinn TM, Sylvester KG, Kitano Y, Kitano Y, Liechty KW, Jarrett BP, Adzick NS, Flake AW. TGF-beta2 is increased after fetal tracheal occlusion. J Pediatr Surg 1999;34:701eC704.

    Frenckner B, Ehren H, Granholm T, Linden V, Palmer K. Improved results in patients who have congenital diaphragmatic hernia using preoperative stabilization, extracorporeal membrane oxygenation, and delayed surgery. J Pediatr Surg 1997;32:1185eC1189.

    Reickert CA, Hirschl RB, Atkinson JB, Dudell G, Georgeson K, Glick P, Greenspan J, Kays D, Klein M, Lally KP, et al. Congenital diaphragmatic hernia survival and use of extracorporeal life support at selected level III nurseries with multimodality support. Surgery 1998;123:305eC310.

    Ssemakula N, Stewart DL, Goldsmith LJ, Cook LN, Bond SJ. Survival of patients with congenital diaphragmatic hernia during the ECMO era: an 11-year experience. J Pediatr Surg 1997;32:1683eC1689.

    Beals DA, Schloo BL, Vacanti JP, Reid LM, Wilson JM. Pulmonary growth and remodeling in infants with high-risk congenital diaphragmatic hernia. J Pediatr Surg 1992;27:997eC1001.

    Nobuhara KK, Fauza DO, DiFiore JW, Hines MH, Fackler JC, Slavin R, Hirschl R, Wilson JM. Continuous intrapulmonary distension with perfluorocarbon accelerates neonatal (but not adult) lung growth. J Pediatr Surg 1998;33:292eC298.

    Fauza DO, Hirschl RB, Wilson JM. Continuous intrapulmonary distension with perfluorocarbon accelerates lung growth in infants with congenital diaphragmatic hernia: initial experience. J Pediatr Surg 2001;36:1237eC1240.

    Walker GM, Kasem KF, O'Toole SJ, Watt A, Skeoch CH, Davis CF. Early perfluorodecalin lung distension in infants with congenital diaphragmatic hernia. J Pediatr Surg 2003;38:17eC20.

    Kitano Y, Flake AW, Quinn TM, Kanai M, Davies P, Sablich TJ, Schneider C, Adzick NS, von Allmen D. Lung growth induced by tracheal occlusion in the sheep is augmented by airway pressurization. J Pediatr Surg 2000;35:216eC221.

    Smith GC. Use of time to event analysis to estimate the normal duration of human pregnancy. Hum Reprod 2001;16:1497eC1500.

    Deprest JA, Evrard VA, Van Ballaer PP, Verbeken E, Vandenberghe K, Lerut TE, Flageole H. Tracheoscopic endoluminal plugging using an inflatable device in the fetal lamb model. Eur J Obstet Gynecol Reprod Biol 1998;81:165eC169.

    Wang Q, Yambe T, Shiraishi Y, Esashi M, Haga Y, Yoshizawa M, Sato F, Matsuki H, Imachi K, Abe Y, et al. Component engineering for an implantable system. Artif Organs 2004;28:869eC873.(Scott M. Nelson, Constant)