Comprehensive Physiology Wiley Online Library

Bioengineering the Blood‐gas Barrier

Full Article on Wiley Online Library



Abstract

The pulmonary blood‐gas barrier represents a remarkable feat of engineering. It achieves the exquisite thinness needed for gas exchange by diffusion, the strength to withstand the stresses and strains of repetitive and changing ventilation, and the ability to actively maintain itself under varied demands. Understanding the design principles of this barrier is essential to understanding a variety of lung diseases, and to successfully regenerating or artificially recapitulating the barrier ex vivo. Many classical studies helped to elucidate the unique structure and morphology of the mammalian blood‐gas barrier, and ongoing investigations have helped to refine these descriptions and to understand the biological aspects of blood‐gas barrier function and regulation. This article reviews the key features of the blood‐gas barrier that enable achievement of the necessary design criteria and describes the mechanical environment to which the barrier is exposed. It then focuses on the biological and mechanical components of the barrier that preserve integrity during homeostasis, but which may be compromised in certain pathophysiological states, leading to disease. Finally, this article summarizes recent key advances in efforts to engineer the blood‐gas barrier ex vivo, using the platforms of lung‐on‐a‐chip and tissue‐engineered whole lungs. © 2020 American Physiological Society. Compr Physiol 10:415‐452, 2020.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1. Timeline of early lung development and associated milestones in blood‐gas barrier maturation. Blue trapezoids illustrate the magnitudes of morphometric changes during alveolarization and microvascular maturation in the rat. E, embryonic day. P, postnatal day. Gest wks, gestational weeks.
Figure 2. Figure 2. Hematoxylin and eosin (H&E) staining of native rat lung fixed by formalin inflation at postnatal days (A) 1, (B) 4, (C) 7, (D) 14, (E) 21, and (F) 60. Scale bars, 50 μm.
Figure 3. Figure 3. Scales of blood‐gas barrier organization. Top of each panel, native lung image. Bottom of each panel, schematic. Tops of panels: (A) Scanning electron micrograph of distal lung parenchyma in mouse. (B) Light micrograph of alveolar septa in a saline‐filled rabbit lung. (C) Transmission electron micrograph of an alveolar capillary in monkey. (D) Enlarged detail by transmission electron micrograph of the gas exchange surface. Scale bars in lower panels representative of human lung. (A) Reused, with permission, from Bastacky J and Goerke J, 1992 18, copyright the American Physiological Society; (B) Reused, with permission, from Gil J, et al., 1979 80, copyright the American Physiological Society; (C,D) Reused, with permission, from Weibel ER, 1970 246, with permission from Elsevier.
Figure 4. Figure 4. Layers of the blood‐gas barrier.
Figure 5. Figure 5. Alveolar‐capillary morphology. (A) Arteriolar “islands” and venous “lakes” in cat lung. (B) Pulmonary capillary beds in frog lung (left) and in the alveolar walls of cat lung (right). AL, alveolar sac. l, line delineating interalveolar wall. P, Intercapillary connective tissue posts. (C) Schematic of capillary network between a pulmonary arteriole (left) and venule (right). (D) Schematic of the alveolar‐capillary “sheet” with associated dimensions in cat. (E) Comparison of pulmonary capillary sheet width to the height of the Empire State Building. (A) Reprinted with slight modification from Sobin SS, et al. 1980 204, with permission from Elsevier; (B, left) Reprinted from Maloney JE and Castle BL, 1969 138, with permission from Elsevier; (B, right) Reprinted from Sobin SS, et al. 1980 204, with permission from Elsevier.
Figure 6. Figure 6. Alveolar epithelial type I cell morphology. (A) Scanning electron micrograph of the alveolar surface of a human lung, with an alveolar epithelial type I cell (AEC1) highlighted in yellow. (B) Drawing of an AEC1 (in yellow, with inner and outer cell membranes in red and green, respectively) branching to either side of an alveolar septum. (C) Alternative view of the AEC1 in (B) illustrating the cell's structure of intercapillary stalks and multiple cytoplasmic plates on both sides of the septum. Ep1, type I epithelial cell, Ep2, type II epithelial cell. Reprinted from Weibel ER, 2015 250 with permission of the American Thoracic Society. Copyright © 2019 American Thoracic Society. The American Journal of Respiratory and Critical Care Medicine is an official journal of the American Thoracic Society.
Figure 7. Figure 7. Lung pressure‐volume curve.
Figure 8. Figure 8. Schematic of micromechanical forces acting upon the alveolar septum.
Figure 9. Figure 9. Alveolar septal pleats. An alveolar septum is shown by transmission electron micrograph and by line drawing. Arrowheads, alveolar luminal surface. Dotted line, pleated epithelial basement membrane. C, capillaries. SLL, surface lining layer. EPII, alveolar epithelial type II cell. M, alveolar macrophage. Modified from image originally printed in Bachofen H, et al., 1987 11, copyright the American Physiological Society.
Figure 10. Figure 10. Morphological differences of air‐ and saline‐filled lungs at different volumes. (A,B) Scanning electron micrographs of air‐filled lungs fixed by vascular perfusion at 40% and 80% TLC, respectively. (C,D) Saline‐filled lungs fixed by vascular perfusion at 40% and 80% TLC, respectively. Originally printed in Gil J, et al. 1979 80, copyright the American Physiological Society.
Figure 11. Figure 11. Zonation of pulmonary blood flow. (A) Location of zones, with Zone 1 near the apex of the lung and Zone 3 near the base. (B) Relative pressures, (C) capillary profiles, and (D) capillary cross‐sections in Zones 1 to 3. PA, alveolar pressure. Pa, arterial pressure. Pv, venous pressure.
Figure 12. Figure 12. Starling's law and alveolar protective mechanisms against edema. (A) Schematic of the alveolar septum in homeostasis. Block arrows at left depict typical relative magnitudes of the Starling forces Pc, Pis, πc, and πis across the alveolar‐capillary wall in homeostasis. Dashed arrow at right depicts small amount of lymph flow from the perivascular spaces. (B) Altered Starling forces in the setting of increased hydrostatic pressure Pc, and the structural and biological protective mechanisms that may limit edema.
Figure 13. Figure 13. Schematic of major extracellular matrix and interstitial components in the alveolar wall.
Figure 14. Figure 14. Lung stress and strain. Lung collagen and elastin have been described to behave as a “string‐spring” pair of mechanical elements. Collagen and elastin at low (A) and high (B) lung volumes. (C) Stress‐strain curve for the lung.
Figure 15. Figure 15. Evolution of lung‐on‐a‐chip designs. (A) A rudimentary model of the alveolar blood‐gas barrier can be made by plating the underside of a porous Transwell membrane with endothelium and the apical side with pulmonary epithelium. This allows intermittent TEER measurements and visualization of the cultured cells but not stretch or flow. (B) A pioneering design by Huh et al. applied cyclic stretch to an elastic porous silicone membrane, allowing modeling of alveolar ventilatory mechanics. This design also allows media flow, thereby mimicking capillary and airway shear dynamics. (C) A later chip‐based design with integrated gold‐plated electrodes allowing simultaneous real‐time microscopy and TEER underflow dynamics. (D) A recent microimpedance tomography‐based lung‐on‐a‐chip design, which segregates all electrodes on one side of the modeled barrier and opens the apical side of this chip for direct visualization and/or intervention. Chips with integrated barrier‐measuring electrodes are generally capable of producing more repeatable and cross‐laboratory translatable results.
Figure 16. Figure 16. Overview of decellularization‐recellularization paradigm for whole lung engineering. (A) Intact native lungs are decellularized via detergent rinsing, creating an extracellular matrix scaffold free of intact cells (B). (C) The scaffold is subsequently seeded with cells into the airway and/or vascular compartments. (D) The reseeded construct is transferred to a bioreactor for culture under physiologically relevant conditions. The resulting engineered organ undergoes (E) ex vivo characterization before final (F) orthotopic transplantation for in vivo functional assessment.
Figure 17. Figure 17. Endothelial cell seeding strategy for decellularized lung. The leaky nature of the decellularized scaffold leads to a “sieving effect” whereby fluid preferentially leaks across the capillary wall. (A) Cells clump in the early capillary segments due to fluid leakage. (B) Seeding a dilute cell suspension from both ends of the vasculature, under higher pressure and with pulsatile perfusion, leads to enhanced distribution of endothelial cells within the decellularized scaffold.


Figure 1. Timeline of early lung development and associated milestones in blood‐gas barrier maturation. Blue trapezoids illustrate the magnitudes of morphometric changes during alveolarization and microvascular maturation in the rat. E, embryonic day. P, postnatal day. Gest wks, gestational weeks.


Figure 2. Hematoxylin and eosin (H&E) staining of native rat lung fixed by formalin inflation at postnatal days (A) 1, (B) 4, (C) 7, (D) 14, (E) 21, and (F) 60. Scale bars, 50 μm.


Figure 3. Scales of blood‐gas barrier organization. Top of each panel, native lung image. Bottom of each panel, schematic. Tops of panels: (A) Scanning electron micrograph of distal lung parenchyma in mouse. (B) Light micrograph of alveolar septa in a saline‐filled rabbit lung. (C) Transmission electron micrograph of an alveolar capillary in monkey. (D) Enlarged detail by transmission electron micrograph of the gas exchange surface. Scale bars in lower panels representative of human lung. (A) Reused, with permission, from Bastacky J and Goerke J, 1992 18, copyright the American Physiological Society; (B) Reused, with permission, from Gil J, et al., 1979 80, copyright the American Physiological Society; (C,D) Reused, with permission, from Weibel ER, 1970 246, with permission from Elsevier.


Figure 4. Layers of the blood‐gas barrier.


Figure 5. Alveolar‐capillary morphology. (A) Arteriolar “islands” and venous “lakes” in cat lung. (B) Pulmonary capillary beds in frog lung (left) and in the alveolar walls of cat lung (right). AL, alveolar sac. l, line delineating interalveolar wall. P, Intercapillary connective tissue posts. (C) Schematic of capillary network between a pulmonary arteriole (left) and venule (right). (D) Schematic of the alveolar‐capillary “sheet” with associated dimensions in cat. (E) Comparison of pulmonary capillary sheet width to the height of the Empire State Building. (A) Reprinted with slight modification from Sobin SS, et al. 1980 204, with permission from Elsevier; (B, left) Reprinted from Maloney JE and Castle BL, 1969 138, with permission from Elsevier; (B, right) Reprinted from Sobin SS, et al. 1980 204, with permission from Elsevier.


Figure 6. Alveolar epithelial type I cell morphology. (A) Scanning electron micrograph of the alveolar surface of a human lung, with an alveolar epithelial type I cell (AEC1) highlighted in yellow. (B) Drawing of an AEC1 (in yellow, with inner and outer cell membranes in red and green, respectively) branching to either side of an alveolar septum. (C) Alternative view of the AEC1 in (B) illustrating the cell's structure of intercapillary stalks and multiple cytoplasmic plates on both sides of the septum. Ep1, type I epithelial cell, Ep2, type II epithelial cell. Reprinted from Weibel ER, 2015 250 with permission of the American Thoracic Society. Copyright © 2019 American Thoracic Society. The American Journal of Respiratory and Critical Care Medicine is an official journal of the American Thoracic Society.


Figure 7. Lung pressure‐volume curve.


Figure 8. Schematic of micromechanical forces acting upon the alveolar septum.


Figure 9. Alveolar septal pleats. An alveolar septum is shown by transmission electron micrograph and by line drawing. Arrowheads, alveolar luminal surface. Dotted line, pleated epithelial basement membrane. C, capillaries. SLL, surface lining layer. EPII, alveolar epithelial type II cell. M, alveolar macrophage. Modified from image originally printed in Bachofen H, et al., 1987 11, copyright the American Physiological Society.


Figure 10. Morphological differences of air‐ and saline‐filled lungs at different volumes. (A,B) Scanning electron micrographs of air‐filled lungs fixed by vascular perfusion at 40% and 80% TLC, respectively. (C,D) Saline‐filled lungs fixed by vascular perfusion at 40% and 80% TLC, respectively. Originally printed in Gil J, et al. 1979 80, copyright the American Physiological Society.


Figure 11. Zonation of pulmonary blood flow. (A) Location of zones, with Zone 1 near the apex of the lung and Zone 3 near the base. (B) Relative pressures, (C) capillary profiles, and (D) capillary cross‐sections in Zones 1 to 3. PA, alveolar pressure. Pa, arterial pressure. Pv, venous pressure.


Figure 12. Starling's law and alveolar protective mechanisms against edema. (A) Schematic of the alveolar septum in homeostasis. Block arrows at left depict typical relative magnitudes of the Starling forces Pc, Pis, πc, and πis across the alveolar‐capillary wall in homeostasis. Dashed arrow at right depicts small amount of lymph flow from the perivascular spaces. (B) Altered Starling forces in the setting of increased hydrostatic pressure Pc, and the structural and biological protective mechanisms that may limit edema.


Figure 13. Schematic of major extracellular matrix and interstitial components in the alveolar wall.


Figure 14. Lung stress and strain. Lung collagen and elastin have been described to behave as a “string‐spring” pair of mechanical elements. Collagen and elastin at low (A) and high (B) lung volumes. (C) Stress‐strain curve for the lung.


Figure 15. Evolution of lung‐on‐a‐chip designs. (A) A rudimentary model of the alveolar blood‐gas barrier can be made by plating the underside of a porous Transwell membrane with endothelium and the apical side with pulmonary epithelium. This allows intermittent TEER measurements and visualization of the cultured cells but not stretch or flow. (B) A pioneering design by Huh et al. applied cyclic stretch to an elastic porous silicone membrane, allowing modeling of alveolar ventilatory mechanics. This design also allows media flow, thereby mimicking capillary and airway shear dynamics. (C) A later chip‐based design with integrated gold‐plated electrodes allowing simultaneous real‐time microscopy and TEER underflow dynamics. (D) A recent microimpedance tomography‐based lung‐on‐a‐chip design, which segregates all electrodes on one side of the modeled barrier and opens the apical side of this chip for direct visualization and/or intervention. Chips with integrated barrier‐measuring electrodes are generally capable of producing more repeatable and cross‐laboratory translatable results.


Figure 16. Overview of decellularization‐recellularization paradigm for whole lung engineering. (A) Intact native lungs are decellularized via detergent rinsing, creating an extracellular matrix scaffold free of intact cells (B). (C) The scaffold is subsequently seeded with cells into the airway and/or vascular compartments. (D) The reseeded construct is transferred to a bioreactor for culture under physiologically relevant conditions. The resulting engineered organ undergoes (E) ex vivo characterization before final (F) orthotopic transplantation for in vivo functional assessment.


Figure 17. Endothelial cell seeding strategy for decellularized lung. The leaky nature of the decellularized scaffold leads to a “sieving effect” whereby fluid preferentially leaks across the capillary wall. (A) Cells clump in the early capillary segments due to fluid leakage. (B) Seeding a dilute cell suspension from both ends of the vasculature, under higher pressure and with pulsatile perfusion, leads to enhanced distribution of endothelial cells within the decellularized scaffold.
References
 1.Akei H, Whitsett JA, Buroker M, Ninomiya T, Tatsumi H, Weaver TE, Ikegami M. Surface tension influences cell shape and phagocytosis in alveolar macrophages. Am J Physiol Lung Cell Mol Physiol 291: L572‐L579, 2006.
 2.Al Jamal R, Roughley PJ, Ludwig MS. Effect of glycosaminoglycan degradation on lung tissue viscoelasticity. Am J Physiol Lung Cell Mol Physiol 280: L306‐L315, 2001.
 3.Albert RK, Lakshminarayan S, Hildebrandt J, Kirk W, Butler J. Increased surface tension favors pulmonary edema formation in anesthetized dogs' lungs. J Clin Invest 63: 1015‐1018, 1979.
 4.Angelidis I, Simon LM, Fernandez IE, Strunz M, Mayr CH, Greiffo FR, Tsitsiridis G, Graf E, Strom TM, Eickelberg O, Mann M, Theis FJ, Schiller HB. An atlas of the aging lung mapped by single cell transcriptomics and deep tissue proteomics. Nat Commun 10 (1): Article 963, 1–17, 2019.
 5.Arık YB, Van Der Helm MW, Odijk M, Segerink LI, Passier R, Van Den Berg A, Van Der Meer AD. Barriers‐on‐chips: Measurement of barrier function of tissues in organs‐on‐chips. Biomicrofluidics 12: 042218, 2018.
 6.Ashbaugh D, Boyd Bigelow D, Petty T, Levine B. Acute Respiratory distress in adults. Lancet 290: 319‐323, 1967.
 7.Ashino Y, Ying X, Dobbs LG, Bhattacharya J. [Ca(2+)](i) oscillations regulate type II cell exocytosis in the pulmonary alveolus. Am J Physiol Lung Cell Mol Physiol 279: L5‐L13, 2000.
 8.Avery ME, Mead J. Surface properties in relation to atelectasis and hyaline membrane disease. AMA J Dis Child 97: 517‐523, 1959.
 9.Bachofen H, Gehr P, Weibel ER. Alterations of mechanical properties and morphology in excised rabbit lungs rinsed with a detergent. J Appl Physiol Respir Environ Exerc Physiol 47: 1002‐1010, 1979.
 10.Bachofen H, Schürch S. Alveolar surface forces and lung architecture. Comp Biochem Physiol A Mol Integr Physiol 129: 183‐193, 2001.
 11.Bachofen H, Schürch S, Urbinelli M, Weibel ER. Relations among alveolar surface tension, surface area, volume, and recoil pressure. J Appl Physiol 62: 1878‐1887, 1987.
 12.Bachofen H, Weber J, Wangensteen D, Weibel ER. Morphometric estimates of diffusing capacity in lungs fixed under zone II and zone III conditions. Respir Physiol 52: 41‐52, 1983.
 13.Bagnato G, Harari S. Cellular interactions in the pathogenesis of interstitial lung diseases. Eur Respir Rev 24: 102‐114, 2015.
 14.Balestrini JL, Gard AL, Gerhold KA, Wilcox EC, Liu A, Schwan J, Le AV, Baevova P, Dimitrievska S, Zhao L, Sundaram S, Sun H, Rittié L, Dyal R, Broekelmann TJ, Mecham RP, Schwartz MA, Niklason LE, White ES. Comparative biology of decellularized lung matrix: Implications of species mismatch in regenerative medicine. Biomaterials 102: 220‐230, 2016.
 15.Balestrini JL, Gard AL, Liu A, Leiby KL, Schwan J, Kunkemoeller B, Calle EA, Sivarapatna A, Lin T, Dimitrievska S, Cambpell SG, Niklason LE. Production of decellularized porcine lung scaffolds for use in tissue engineering. Integr Biol 7 (12): 1‐13, 2015.
 16.Ball WC Jr, Stewart PB, Newsham LG, Bates DV. Regional pulmonary function studied with xenon 133. J Clin Invest 41: 519‐531, 1962.
 17.Barré SF, Haberthür D, Cremona TP, Stampanoni M, Schittny JC. The total number of acini remains constant throughout postnatal rat lung development. Am J Physiol Lung Cell Mol Physiol 311: L1082‐L1089, 2016.
 18.Bastacky J, Goerke J. Pores of Kohn are filled in normal lungs: Low‐temperature scanning electron microscopy. J Appl Physiol (1985) 73: 88‐95, 1992.
 19.Bastacky J, Lee C, Goerke J, Koushafar H, Yager D, Kenaga L, Speed TP, Chen Y, Clements JA. Alveolar lining layer is thin and continuous: Low‐temperature scanning electron microscopy of rat lung. J Appl Physiol 79: 1615‐1628, 1995.
 20.Bates JHT, Irvin CG, Farré R, Hantos Z. Oscillation mechanics of the respiratory system. Compr Physiol 1: 1233‐1272, 2011.
 21.Beers MF, Moodley Y. When is an alveolar type 2 cell an alveolar type 2 cell? A conundrum for lung stem cell biology and regenerative medicine. Am J Respir Cell Mol Biol 57: 18‐27, 2017.
 22.Bellani G, Laffey JG, Pham T, Fan E, Brochard L, Esteban A, Gattinoni L, van Haren F, Larsson A, McAuley DF, Ranieri M, Rubenfeld G, Thompson BT, Wrigge H, Slutsky AS, Pesenti A, Investigators LS, Group ET. Epidemiology, patterns of care, and mortality for patients with acute respiratory distress syndrome in intensive care units in 50 countries. JAMA 315: 788‐800, 2016.
 23.Benam KH, Villenave R, Lucchesi C, Varone A, Hubeau C, Lee H‐H, Alves SE, Salmon M, Ferrante TC, Weaver JC. Small airway‐on‐a‐chip enables analysis of human lung inflammation and drug responses in vitro. Nat Methods 13: 151, 2015.
 24.Bhattacharya J, Matthay MA. Regulation and repair of the alveolar‐capillary barrier in acute lung injury. Annu Rev Physiol 75: 593‐615, 2013.
 25.Birks EK, Mathieu‐Costello O, Fu Z, Tyler WS, West JB. Comparative aspects of the strength of pulmonary capillaries in rabbit, dog, and horse. Respir Physiol 97: 235‐246, 1994.
 26.Birukov KG, Zebda N, Birukova AA. Barrier enhancing signals in pulmonary edema. Compr Physiol 3: 429‐484, 2013.
 27.Bonvillain RW, Danchuk S, Sullivan DE, Betancourt AM, Semon JA, Eagle ME, Mayeux JP, Gregory AN, Wang G, Townley IK, Borg ZD, Weiss DJ, Bunnell BA. A nonhuman primate model of lung regeneration: Detergent‐mediated decellularization and initial in vitro recellularization with mesenchymal stem cells. Tissue Eng Part A 18: 2437‐2452, 2012.
 28.Booth AJ, Hadley R, Cornett AM, Dreffs AA, Matthes SA, Tsui JL, Weiss K, Horowitz JC, Fiore VF, Barker TH, Moore BB, Martinez FJ, Niklason LE, White ES. Acellular normal and fibrotic human lung matrices as a culture system for in vitro investigation. Am J Respir Crit Care Med 186: 866‐876, 2012.
 29.Bredenberg CE, Paskanik AM, Nieman GF. High surface tension pulmonary edema. J Surg Res 34: 515‐523, 1983.
 30.Brody JS, Vaccaro CA, Hill NS, Rounds S. Binding of charged ferritin to alveolar wall components and charge selectivity of macromolecular transport in permeability pulmonary edema in rats. Circ Res 55: 155‐167, 1984.
 31.Brown ES, Johnson RP, Clements JA. Pulmonary surface tension. J Appl Physiol (1985) 14: 717‐720, 1959.
 32.Burri P. Fetal and postnatal development of the lung. Annu Rev Physiol 46: 617‐628, 1984.
 33.Burri P, Weibel E. Morphometric estimation of pulmonary diffusion capacity: II. Effect of PO2 on the growing lung adaption of the growing rat lung to hypoxia and hyperoxia. Respir Physiol 11: 247‐264, 1971.
 34.Burri PH. The postnatal growth of the rat lung. 3. Morphology. Anat Rec 180: 77‐98, 1974.
 35.Burri PH. Morphology and respiratory function of the alveolar unit. Int Arch Allergy Appl Immunol 76 (Suppl 1): 2‐12, 1985.
 36.Burri PH, Dbaly J, Weibel ER. The postnatal growth of the rat lung. I. Morphometry. Anat Rec 178: 711‐730, 1974.
 37.Buschmann MD, Grodzinsky AJ. A molecular model of proteoglycan‐associated electrostatic forces in cartilage mechanics. J Biomech Eng 117: 179‐192, 1995.
 38.Butler C II, Kleinerman J. Capillary density: Alveolar diameter, a morphometric approach to ventilation and perfusion. Am Rev Respir Dis 102: 886‐894, 1970.
 39.Caduff JH, Fischer LC, Burri PH. Scanning electron microscope study of the developing microvasculature in the postnatal rat lung. Anat Rec 216: 154‐164, 1986.
 40.Calle EA. Alveolar barrier formation in engineered lung tissue. In: Biomedical Engineering. New Haven, CT: Yale University, 2014, p. 193‐252.
 41.Calle EA, Hill RC, Leiby KL, Le AV, Gard AL, Madri JA, Hansen KC, Niklason LE. Targeted proteomics effectively quantifies differences between native lung and detergent‐decellularized lung extracellular matrices. Acta Biomater 46: 91‐100, 2016.
 42.Cavalcante FSA, Ito S, Brewer K, Sakai H, Alencar AM, Almeida MP, Andrade JS Jr, Majumdar A, Ingenito EP, Suki B. Mechanical interactions between collagen and proteoglycans: Implications for the stability of lung tissue. J Appl Physiol 98: 672‐679, 2005.
 43.Charest JM, Okamoto T, Kitano K, Yasuda A, Gilpin SE, Mathisen DJ, Ott HC. Design and validation of a clinical‐scale bioreactor for long‐term isolated lung culture. Biomaterials 52: 79‐87, 2015.
 44.Chung M‐I, Bujnis M, Barkauskas CE, Kobayashi Y, Hogan BLM. Niche‐mediated BMP/SMAD signaling regulates lung alveolar stem cell proliferation and differentiation. Development 145, 2018. DOI: 10.1242/dev.163014.
 45.Clements JA. Pulmonary edema and permeability of alveolar membranes. Arch Environ Health Int J 2: 104‐107, 1961.
 46.Clements JA, Brown ES, Johnson RP. Pulmonary surface tension and the mucus lining of the lungs: Some theoretical considerations. J Appl Physiol 12: 262‐268, 1958.
 47.Conforti E, Fenoglio C, Bernocchi G, Bruschi O, Miserocchi GA. Morpho‐functional analysis of lung tissue in mild interstitial edema. Am J Physiol Lung Cell Mol Physiol 282: L766‐L774, 2002.
 48.Cook CD, Cochran WD. The respiratory‐distress syndrome of newborn infants. N Engl J Med 270: 673‐676, 1964.
 49.Cordingley JL. Pores of Kohn. Thorax 27: 433‐441, 1972.
 50.Cox G, Kable E, Jones A, Fraser I, Manconi F, Gorrell MD. 3‐Dimensional imaging of collagen using second harmonic generation. J Struct Biol 141: 53‐62, 2003.
 51.Crapo JD, Barry BE, Gehr P, Bachofen M, Weibel ER. Cell number and cell characteristics of the normal human lung 1–3. Am Rev Respir Dis 126: 332‐337, 1982.
 52.Crapo PM, Gilbert TW, Badylak SF. An overview of tissue and whole organ decellularization processes. Biomaterials 32: 3233‐3243, 2011.
 53.Daly AB, Wallis JM, Borg ZD, Bonvillain RW, Deng B, Ballif BA, Jaworski DM, Allen GB, Weiss DJ. Initial binding and recellularization of decellularized mouse lung scaffolds with bone marrow‐derived mesenchymal stromal cells. Tissue Eng Part A 18: 1‐16, 2012.
 54.Doi R, Tsuchiya T, Mitsutake N, Nishimura S, Matsuu‐Matsuyama M, Nakazawa Y, Ogi T, Akita S, Yukawa H, Baba Y, Yamasaki N, Matsumoto K, Miyazaki T, Kamohara R, Hatachi G, Sengyoku H, Watanabe H, Obata T, Niklason LE, Nagayasu T. Transplantation of bioengineered rat lungs recellularized with endothelial and adipose‐derived stromal cells. Sci Rep 7: 8447, 2017.
 55.Dorrello NV, Guenthart BA, O'Neill JD, Kim J, Cunningham K, Chen Y‐W, Biscotti M, Swayne T, Wobma HM, Huang SXL, Snoeck H‐W, Bacchetta M, Vunjak‐Novakovic G. Functional vascularized lung grafts for lung bioengineering. Sci Adv 3: e1700521, 2017.
 56.Dreyfuss D, Saumon G. Ventilator‐induced lung injury: Lessons from experimental studies. Am J Respir Crit Care Med 157: 294‐323, 1998.
 57.Dunnill MS. Postnatal growth of the lung. Thorax 17: 329‐333, 1962.
 58.Edington CD, Chen WLK, Geishecker E, Kassis T, Soenksen LR, Bhushan BM, Freake D, Kirschner J, Maass C, Tsamandouras N. Interconnected microphysiological systems for quantitative biology and pharmacology studies. Sci Rep 8: 4530, 2018.
 59.Effros RM, Mason GR, Silverman P, Reid E, Hukkanen J. Movement of ions and small solutes across endothelium and epithelium of perfused rabbit lungs. J Appl Physiol 60: 100‐107, 1986.
 60.Effros RM, Parker JC. Pulmonary vascular heterogeneity and the Starling hypothesis. Microvasc Res 78: 71‐77, 2009.
 61.Elbrecht DH, Long CJ, Hickman JJ. Transepithelial/endothelial electrical resistance (TEER) theory and applications for microfluidic body‐on‐a‐chip devices. J Rare Dis Res Treat 1 (3): 46‐52, 2016.
 62.Elenz E. Ueber das Lungenepithel. Naturwissenschaften 5: 1‐18, 1864.
 63.Elliott AR, Fu Z, Tsukimoto K, Prediletto R, Mathieu‐Costello O, West JB. Short‐term reversibility of ultrastructural changes in pulmonary capillaries caused by stress failure. J Appl Physiol 73: 1150‐1158, 1992.
 64.Enhorning G. Pulsating bubble technique for evaluating pulmonary surfactant. J Appl Physiol Respir Environ Exerc Physiol 43: 198‐203, 1977.
 65.Fan E, Brodie D, Slutsky AS. Acute respiratory distress syndrome: Advances in diagnosis and treatment. JAMA 319: 698‐710, 2018.
 66.Fishman AP. Pulmonary edema. The water‐exchanging function of the lung. Circulation 46: 390‐408, 1972.
 67.Frank JA, Briot R, Lee JW, Ishizaka A, Uchida T, Matthay MA. Physiological and biochemical markers of alveolar epithelial barrier dysfunction in perfused human lungs. Am J Physiol Lung Cell Mol Physiol 293: L52‐L59, 2007.
 68.Frank JA, Matthay MA. Science review: Mechanisms of ventilator‐induced injury. Crit Care 7: 233‐241, 2003.
 69.Fu Z, Costello ML, Tsukimoto K, Prediletto R, Elliott AR, Mathieu‐Costello O, West JB. High lung volume increases stress failure in pulmonary capillaries. J Appl Physiol 73: 123‐133, 1992.
 70.Fung Y. Blood flow in the lung. In: Biomechanics: Circulation. Springer, 1997, p. 333‐445.
 71.Fung Y, Sobin S. Theory of sheet flow in lung alveoli. J Appl Physiol 26: 472‐488, 1969.
 72.Fung Y, Sobin S. Pulmonary alveolar blood flow. Circ Res 30: 470‐490, 1972.
 73.Fung YC. Does the surface tension make the lung inherently unstable? Circ Res 37: 497‐502, 1975.
 74.Galili U. The alpha‐gal epitope and the anti‐Gal antibody in xenotransplantation and in cancer immunotherapy. Immunol Cell Biol 83: 674‐686, 2005.
 75.Gattinoni L, Pesenti A. The concept of “baby lung”. Intensive Care Med 31: 776‐784, 2005.
 76.Gattinoni L, Protti A, Caironi P, Carlesso E. Ventilator‐induced lung injury: The anatomical and physiological framework. Crit Care Med 38: S539‐S548, 2010.
 77.Gehr P, Bachofen M, Weibel ER. The normal human lung: Ultrastructure and morphometric estimation of diffusion capacity. Respir Physiol 32: 121‐140, 1978.
 78.Gehr P, Sehovic S, Burri PH, Claassen H, Weibel ER. The lung of shrews: Morphometric estimation of diffusion capacity. Respir Physiol 40: 33‐47, 1980.
 79.Ghaedi M, Le AV, Hatachi G, Beloiartsev A, Rocco K, Sivarapatna A, Mendez JJ, Baevova P, Dyal RN, Leiby KL, White ES, Niklason LE. Bioengineered lungs generated from human iPSCs‐derived epithelial cells on native extracellular matrix. J Tissue Eng Regen Med 12: e1623‐e1635, 2018.
 80.Gil J, Bachofen H, Gehr P, Weibel ER. Alveolar volume‐surface area relation in air‐ and saline‐filled lungs fixed by vascular perfusion. J Appl Physiol Respir Environ Exerc Physiol 47: 990‐1001, 1979.
 81.Gil J, Weibel ER. Improvements in demonstration of lining layer of lung alveoli by electron microscopy. Respir Physiol 8: 13‐36, 1969.
 82.Gilpin SE, Charest JM, Ren X, Tapias LF, Wu T, Evangelista‐Leite D, Mathisen DJ, Ott HC. Regenerative potential of human airway stem cells in lung epithelial engineering. Biomaterials 108: 111‐119, 2016.
 83.Gilpin SE, Guyette JP, Gonzalez G, Ren X, Asara JM, Mathisen DJ, Vacanti JP, Ott HC. Perfusion decellularization of human and porcine lungs: Bringing the matrix to clinical scale. J Heart Lung Transplant 33: 298‐308, 2014.
 84.Gilpin SE, Ren X, Okamoto T, Guyette JP, Mou H, Rajagopal J, Mathisen DJ, Vacanti JP, Ott HC. Enhanced lung epithelial specification of human induced pluripotent stem cells on decellularized lung matrix. Ann Thorac Surg 98: 1712‐1719, 2014.
 85.Giwa S, Lewis JK, Alvarez L, Langer R, Roth AE, Church GM, Markmann JF, Sachs DH, Chandraker A, Wertheim JA, Rothblatt M, Boyden ES, Eidbo E, Lee WPA, Pomahac B, Brandacher G, Weinstock DM, Elliott G, Nelson D, Acker JP, Uygun K, Schmalz B, Weegman BP, Tocchio A, Fahy GM, Storey KB, Rubinsky B, Bischof J, Elliott JAW, Woodruff TK, Morris GJ, Demirci U, Brockbank KGM, Woods EJ, Ben RN, Baust JG, Gao D, Fuller B, Rabin Y, Kravitz DC, Taylor MJ, Toner M. The promise of organ and tissue preservation to transform medicine. Nat Biotechnol 35: 530‐542, 2017.
 86.Glazier JB, Hughes JM, Maloney JE, West JB. Measurements of capillary dimensions and blood volume in rapidly frozen lungs. J Appl Physiol (1985) 26: 65‐76, 1969.
 87.Glenny RW, Lamm WJ, Albert RK, Robertson HT. Gravity is a minor determinant of pulmonary blood flow distribution. J Appl Physiol (1985) 71: 620‐629, 1991.
 88.Gordon S, Daneshian M, Bouwstra J, Caloni F, Constant S, Davies DE, Dandekar G, Hartung T, Leist M, Lehr C‐M. Non‐animal models of epithelial barriers (skin, intestine and lung) in research, industrial applications and regulatory toxicology. ALTEX 32: 327‐378, 2015.
 89.Gouveia L, Betsholtz C, Andrae J. PDGF‐A signaling is required for secondary alveolar septation and controls epithelial proliferation in the developing lung. Development 145. dev161976, 2018.
 90.Gregory TJ, Longmore WJ, Moxley MA, Whitsett JA, Reed CR, Fowler AA, Hudson LD, Maunder RJ, Crim C, Hyers TM. Surfactant chemical composition and biophysical activity in acute respiratory distress syndrome. J Clin Invest 88: 1976‐1981, 1991.
 91.Griese M. Pulmonary surfactant in health and human lung diseases: State of the art. Eur Respir J 13: 1455‐1476, 1999.
 92.Guenthart BA, O'Neill JD, Kim J, Fung K, Vunjak‐Novakovic G, Bacchetta M. Cell replacement in human lung bioengineering. J Heart Lung Transplant 38 (2): 215‐224, 2019.
 93.Günther A, Ruppert C, Schmidt R, Markart P, Grimminger F, Walmrath D, Seeger W. Surfactant alteration and replacement in acute respiratory distress syndrome. Respir Res 2: 353, 2001.
 94.Guo M, Du Y, Gokey JJ, Ray S, Bell SM, Adam M, Sudha P, Perl AK, Deshmukh H, Potter SS, Whitsett JA, Xu Y. Single cell RNA analysis identifies cellular heterogeneity and adaptive responses of the lung at birth. Nat Commun 10: 37, 2019.
 95.Guyton AC, Granger HJ, Taylor AE. Interstitial fluid pressure. Physiol Rev 51: 527‐563, 1971.
 96.Hallman M, Spragg R, Harrell JH, Moser KM, Gluck L. Evidence of lung surfactant abnormality in respiratory failure. Study of bronchoalveolar lavage phospholipids, surface activity, phospholipase activity, and plasma myoinositol. J Clin Invest 70: 673‐683, 1982.
 97.Henry OY, Villenave R, Cronce MJ, Leineweber WD, Benz MA, Ingber DE. Organs‐on‐chips with integrated electrodes for trans‐epithelial electrical resistance (TEER) measurements of human epithelial barrier function. Lab Chip 17: 2264‐2271, 2017.
 98.Hermanns MI, Unger RE, Kehe K, Peters K, Kirkpatrick CJ. Lung epithelial cell lines in coculture with human pulmonary microvascular endothelial cells: Development of an alveolo‐capillary barrier in vitro. Lab Invest 84: 736, 2004.
 99.Hill RC, Calle EA, Dzieciatkowska M, Niklason LE, Hansen KC. Quantification of extracellular matrix proteins from a rat lung scaffold to provide a molecular readout for tissue engineering. Mol Cell Proteomics 14: 961‐973, 2015.
 100.Hobi N, Ravasio A, Haller T. Interfacial stress affects rat alveolar type II cell signaling and gene expression. Am J Physiol Lung Cell Mol Physiol 303: L117‐L129, 2012.
 101.Hopkins SR, Schoene RB, Henderson WR, Spragg RG, Martin TR, West JB. Intense exercise impairs the integrity of the pulmonary blood‐gas barrier in elite athletes. Am J Respir Crit Care Med 155: 1090‐1094, 1997.
 102.Hopkins SR, Schoene RB, Henderson WR, Spragg RG, West JB. Sustained submaximal exercise does not alter the integrity of the lung blood‐gas barrier in elite athletes. J Appl Physiol 84: 1185‐1189, 1998.
 103.Horie T, Hildebrandt J. Dynamic compliance, limit cycles, and static equilibria of excised cat lung. J Appl Physiol (1985) 31: 423‐430, 1971.
 104.Horsfield K. Morphometry of the small pulmonary arteries in man. Circ Res 42: 593‐597, 1978.
 105.Hotchkiss JR, Simonson DA, Marek DJ, Marini JJ, Dries DJ. Pulmonary microvascular fracture in a patient with acute respiratory distress syndrome. Crit Care Med 30: 2368‐2370, 2002.
 106.Hsia CC, Hyde DM, Ochs M, Weibel ER, ATS/ERS Joint Task Force on Quantitative Assessment of Lung Structure. An official research policy statement of the American Thoracic Society/European Respiratory Society: Standards for quantitative assessment of lung structure. Am J Respir Crit Care Med 181: 394‐418, 2010.
 107.Huang W, Yen R, McLaurine M, Bledsoe G. Morphometry of the human pulmonary vasculature. J Appl Physiol 81: 2123‐2133, 1996.
 108.Huh D, Leslie DC, Matthews BD, Fraser JP, Jurek S, Hamilton GA, Thorneloe KS, McAlexander MA, Ingber DE. A human disease model of drug toxicity‐induced pulmonary edema in a lung‐on‐a‐chip microdevice. Sci Transl Med 4: 159ra147, 2012.
 109.Huh D, Matthews BD, Mammoto A, Montoya‐Zavala M, Hsin HY, Ingber DE. Reconstituting organ‐level lung functions on a chip. Science 328: 1662‐1668, 2010.
 110.Humayun M, Chow C‐W, Young EW. Microfluidic lung airway‐on‐a‐chip with arrayable suspended gels for studying epithelial and smooth muscle cell interactions. Lab Chip 18: 1298‐1309, 2018.
 111.Hussell T, Bell TJ. Alveolar macrophages: Plasticity in a tissue‐specific context. Nat Rev Immunol 14: 81‐93, 2014.
 112.Jacob A, Morley M, Hawkins F, McCauley KB, Jean JC, Heins H, Na CL, Weaver TE, Vedaie M, Hurley K, Hinds A, Russo SJ, Kook S, Zacharias W, Ochs M, Traber K, Quinton LJ, Crane A, Davis BR, White FV, Wambach J, Whitsett JA, Cole FS, Morrisey EE, Guttentag SH, Beers MF, Kotton DN. Differentiation of human pluripotent stem cells into functional lung alveolar epithelial cells. Cell Stem Cell 21: 472‐488.e10, 2017.
 113. Ranieri VM, Rubenfeld GD, Thompson BT, Ferguson ND, Caldwell E, Fan E, Camporota L, Slutsky AS. ARDS Definition Task Force. Acute respiratory distress syndrome. JAMA 307: 1‐8, 2012.
 114.Jones AT, Hansell DM, Evans TW. Pulmonary perfusion in supine and prone positions: An electron‐beam computed tomography study. J Appl Physiol (1985) 90: 1342‐1348, 2001.
 115.Kauffman SL, Burri PH, Weibel ER. The postnatal growth of the rat lung. II. Autoradiography. Anat Rec 180: 63‐76, 1974.
 116.Kikkawa Y. Morphology of alveolar lining layer. Anat Rec 167: 389‐400, 1970.
 117.Klaus MH, Clements JA, Havel RJ. Composition of surface‐active material isolated from beef lung. Proc Natl Acad Sci 47: 1858‐1859, 1961.
 118.Kolliker A. Zur Kenntnis des Baues der Lunge des Menschen. Verh Phys‐Med Ges Wurzburg 16: 1‐24, 1881.
 119.Kotton DN, Morrisey EE. Lung regeneration: Mechanisms, applications and emerging stem cell populations. Nat Med 20: 822‐832, 2014.
 120.Koyama S, Lamm WJ, Hildebrandt J, Albert RK. Flow characteristics of open vessels in zone 1 rabbit lungs. J Appl Physiol 66: 1817‐1823, 1989.
 121.Kuehn A, Kletting S, de Souza Carvalho‐Wodarz C, Repnik U, Griffiths G, Fischer U, Meese E, Huwer H, Wirth D, May T, Schneider‐Daum N, Lehr CM. Human alveolar epithelial cells expressing tight junctions to model the air‐blood barrier. ALTEX 33: 251‐260, 2016.
 122.Kuhn K. Basement membrane (type IV) collagen. Matrix Biol 14: 439‐445, 1995.
 123.Lafemina MJ, Rokkam D, Chandrasena A, Pan J, Bajaj A, Johnson M, Frank JA. Keratinocyte growth factor enhances barrier function without altering claudin expression in primary alveolar epithelial cells. Am J Physiol Lung Cell Mol Physiol 299: L724‐L734, 2010.
 124.Lamm WJ, Kirk KR, Hanson WL, Wagner WW, Albert RK. Flow through zone 1 lungs utilizes alveolar corner vessels. J Appl Physiol 70: 1518‐1523, 1991.
 125.Le AV, Hatachi G, Beloiartsev A, Ghaedi M, Engler AJ, Baevova P, Niklason LE, Calle EA. Efficient and functional endothelial repopulation of whole lung organ scaffolds. ACS Biomater Sci Eng 3: 2000‐2010, 2017.
 126.Leach JP, Morrisey EE. Repairing the lungs one breath at a time: How dedicated or facultative are you? Genes Dev 32: 1461‐1471, 2018.
 127.Lecuona E, Saldías F, Comellas A, Ridge K, Guerrero C, Sznajder JI. Ventilator‐associated lung injury decreases lung ability to clear edema in rats. Am J Respir Crit Care Med 159: 603‐609, 1999.
 128.Lee JW, Fang X, Dolganov G, Fremont RD, Bastarache JA, Ware LB, Matthay MA. Acute lung injury edema fluid decreases net fluid transport across human alveolar epithelial type II cells. J Biol Chem 282: 24109‐24119, 2007.
 129.Levin DL, Chen Q, Zhang M, Edelman RR, Hatabu H. Evaluation of regional pulmonary perfusion using ultrafast magnetic resonance imaging. Magn Reson Med 46: 166‐171, 2001.
 130.Lewis KJ, Hall JK, Kiyotake EA, Christensen T, Balasubramaniam V, Anseth KS. Epithelial‐mesenchymal crosstalk influences cellular behavior in a 3D alveolus‐fibroblast model system. Biomaterials 155: 124‐134, 2018.
 131.Low FN. Electron microscopy of the rat lung. Anat Rec 113: 437‐449, 1952.
 132.Lowe K, Alvarez DF, King JA, Stevens T. Perivascular fluid cuffs decrease lung compliance by increasing tissue resistance. Crit Care Med 38: 1458‐1466, 2010.
 133.Lumb AB. Elastic forces and lung volumes. In: Lumb AB, editor. Nunn's Applied Respiratory Physiology (8th ed). Edinburgh: Elsevier, 2017, p. 17‐32.e11.
 134.Lumb AB. Pulmonary vascular disease. In: Lumb AB, editor. Nunn's Applied Respiratory Physiology (8th ed). Edinburgh: Elsevier, 2017, p. 407‐418.e401.
 135.Luyts K, Napierska D, Dinsdale D, Klein SG, Serchi T, Hoet PH. A coculture model of the lung–blood barrier: The role of activated phagocytic cells. Toxicol In Vitro 29: 234‐241, 2015.
 136.Maina JN, West JB. Thin and strong! The bioengineering dilemma in the structural and functional design of the blood‐gas barrier. Physiol Rev 85: 811‐844, 2005.
 137.Maksym GN, Bates JH. A distributed nonlinear model of lung tissue elasticity. J Appl Physiol 82: 32‐41, 1997.
 138.Maloney JE, Castle BL. Pressure‐diameter relations of capillaries and small blood vessels in frog lung. Respir Physiol 7: 150‐162, 1969.
 139.Mason RJ, Voelker DR. Regulatory mechanisms of surfactant secretion. Biochim Biophys Acta 1408: 226‐240, 1998.
 140.Mathieu‐Costello O, Willford DC, Fu Z, Garden RM, West JB. Pulmonary capillaries are more resistant to stress failure in dogs than in rabbits. J Appl Physiol 79: 908‐917, 1995.
 141.Matsubara O, Tamura A, Ohdama S, Mark EJ. Alveolar basement membrane breaks down in diffuse alveolar damage: An immunohistochemical study. Pathol Int 45: 473‐482, 1995.
 142.Matthay MA, Folkesson HG, Clerici C. Lung epithelial fluid transport and the resolution of pulmonary edema. Physiol Rev 82: 569‐600, 2002.
 143.Matthay MA, Landolt CC, Staub NC. Differential liquid and protein clearance from the alveoli of anesthetized sheep. J Appl Physiol Respir Environ Exerc Physiol 53: 96‐104, 1982.
 144.Mazzuca E, Aliverti A, Miserocchi G. Computational micro‐scale model of control of extravascular water and capillary perfusion in the air blood barrier. J Theor Biol 400: 42‐51, 2016.
 145.Mead J, Takishima T, Leith D. Stress distribution in lungs: A model of pulmonary elasticity. J Appl Physiol (1985) 28: 596‐608, 1970.
 146.Mendenhall RM, Stokinger HE. Films from lung washings as a mechanism model for lung injury by ozone. J Appl Physiol 17: 28‐32, 1962.
 147.Mercer RR, Crapo JD. Spatial distribution of collagen and elastin fibers in the lungs. J Appl Physiol 69: 756‐765, 1990.
 148.Mercer RR, Laco JM, Crapo JD. Three‐dimensional reconstruction of alveoli in the rat lung for pressure‐volume relationships. J Appl Physiol 62: 1480‐1487, 1987.
 149.Mermoud Y, Felder M, Stucki J, Stucki A, Guenat OT. Microimpedance tomography system to monitor cell activity and membrane movements in a breathing lung‐on‐chip. Sens Actuators B 255: 3647‐3653, 2018.
 150.Miserocchi G, Negrini D, Del Fabbro M, Venturoli D. Pulmonary interstitial pressure in intact in situ lung: Transition to interstitial edema. J Appl Physiol 74: 1171‐1177, 1993.
 151.Miserocchi G, Negrini D, Gonano C. Direct measurement of interstitial pulmonary pressure in in situ lung with intact pleural space. J Appl Physiol 69: 2168‐2174, 1990.
 152.Mondrinos MJ, Yi Y‐S, Wu N‐K, Ding X, Huh D. Native extracellular matrix‐derived semipermeable, optically transparent, and inexpensive membrane inserts for microfluidic cell culture. Lab Chip 17: 3146‐3158, 2017.
 153.Moudgil R, Michelakis ED, Archer SL. Hypoxic pulmonary vasoconstriction. J Appl Physiol (1985) 98: 390‐403, 2005.
 154.Mutlu GM, Sznajder JI. Mechanisms of pulmonary edema clearance. Am J Physiol Lung Cell Mol Physiol 289: L685‐L695, 2005.
 155.Nardell EA, Brody JS. Determinants of mechanical properties of rat lung during postnatal development. J Appl Physiol 53: 140‐148, 1982.
 156.Network A. Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. N Engl J Med 342: 1301‐1308, 2000.
 157.Nichols JE, La Francesca S, Niles JA, Vega SP, Argueta LB, Frank L, Christiani DC, Pyles RB, Himes BE, Zhang R, Li S, Sakamoto J, Rhudy J, Hendricks G, Begarani F, Liu X, Patrikeev I, Pal R, Usheva E, Vargas G, Miller A, Woodson L, Wacher A, Grimaldo M, Weaver D, Mlcak R, Cortiella J. Production and transplantation of bioengineered lung into a large‐animal model. Sci Transl Med 10: eaao3926, 2018.
 158.Nichols JE, La Francesca S, Vega SP, Niles JA, Argueta LB, Riddle M, Sakamoto J, Vargas G, Pal R, Woodson L, Rhudy J, Lee D, Seanor D, Campbell G, Schnadig V, Cortiella J. Giving new life to old lungs: Methods to produce and assess whole human paediatric bioengineered lungs. J Tissue Eng Regen Med 11: 2136‐2152, 2017.
 159.Nichols JEE, Niles J, Riddle M, Vargas G, Schilagard T, Ma L, Edward K, Lafrancesca S, Sakamoto J, Vega S, Ogedegbe M, Mlcak R, Deyo D, Woodson L, McQuitty C, Lick S, Beckles D, Melo E, Cortiella J. Production and assessment of decellularized pig and human lung scaffolds. Tissue Eng, 2013. DOI: 10.1089/ten.TEA.2012.0250.
 160.Nicolaysen G, Waaler BA, Aarseth P. On the existence of stretchable pores in the exchange vessels of the isolated rabbit lung preparation. Lymphology 12: 201‐207, 1979.
 161.Nieman GF, Bredenberg CE. High surface tension pulmonary edema induced by detergent aerosol. J Appl Physiol 58: 129‐136, 1985.
 162.Ochoa CD, Stevens T. Studies on the cell biology of interendothelial cell gaps. Am J Physiol Lung Cell Mol Physiol 302: L275‐L286, 2012.
 163.Ochs M, Nyengaard JR, Jung A, Knudsen L, Voigt M, Wahlers T, Richter J, Gundersen HJ. The number of alveoli in the human lung. Am J Respir Crit Care Med 169: 120‐124, 2004.
 164.Odijk M, van der Meer AD, Levner D, Kim HJ, van der Helm MW, Segerink LI, Frimat JP, Hamilton GA, Ingber DE, van den Berg A. Measuring direct current trans‐epithelial electrical resistance in organ‐on‐a‐chip microsystems. Lab Chip 15: 745‐752, 2015.
 165.Okada O, Presson R Jr, Kirk K, Godbey P, Capen R, Wagner W Jr. Capillary perfusion patterns in single alveolar walls. J Appl Physiol 72: 1838‐1844, 1992.
 166.O'Neill JD, Guenthart BA, Kim J, Chicotka S, Queen D, Fung K, Marboe C, Romanov A, Huang SXL, Chen Y‐W, Snoeck H‐W, Bacchetta M, Vunjak‐Novakovic G. Cross‐circulation for extracorporeal support and recovery of the lung. Nat Biomed Eng 1: 0037, 2017.
 167.Oostveen E, MacLeod D, Lorino H, Farré R, Hantos Z, Desager K, Marchal F, ERS Task Force on Respiratory Impedance Measurements. The forced oscillation technique in clinical practice: Methodology, recommendations and future developments. Eur Respir J 22: 1026‐1041, 2003.
 168.Ott HC, Clippinger B, Conrad C, Schuetz C, Pomerantseva I, Ikonomou L, Kotton D, Vacanti JP. Regeneration and orthotopic transplantation of a bioartificial lung. Nat Med 16: 927‐933, 2010.
 169.Overgaard CE, Mitchell LA, Koval M. Roles for claudins in alveolar epithelial barrier function. Ann N Y Acad Sci 1257: 167‐174, 2012.
 170.Parthasarathi K. The pulmonary vascular barrier: Insights into structure, function, and regulatory mechanisms. In: Parthasarathi K, editor, Molecular and Functional Insights into the Pulmonary Vasculature. Advances in Anatomy, Embryology, and Cell Biology. Cham, Switzerland: Springer International Publishing, 2018, vol. 228, p. 41‐61.
 171.Pattle RE. Properties, function and origin of the alveolar lining layer. Nature 175: 1125‐1126, 1955.
 172.Pattle RE. Surface lining of lung alveoli. Physiol Rev 45: 48‐79, 1965.
 173.Perlman CE, Bhattacharya J. Alveolar expansion imaged by optical sectioning microscopy. J Appl Physiol 103: 1037‐1044, 2007.
 174.Permutt S, Bromberger‐Barnea B, Bane HN. Alveolar pressure, pulmonary venous pressure, and the vascular waterfall. Med Thorac 19: 239‐260, 1962.
 175.Petersen TH, Calle EA, Colehour MB, Niklason LE. Matrix composition and mechanics of decellularized lung scaffolds. Cells Tissues Organs 195: 222‐231, 2012.
 176.Petersen TH, Calle EA, Zhao L, Lee EJ, Gui L, Raredon MB, Gavrilov K, Yi T, Zhuang ZW, Breuer C, Herzog E, Niklason LE. Tissue‐engineered lungs for in vivo implantation. Science (New York, NY) 329: 538‐541, 2010.
 177.Pezzulo AA, Starner TD, Scheetz TE, Traver GL, Tilley AE, Harvey BG, Crystal RG, McCray PB Jr, Zabner J. The air‐liquid interface and use of primary cell cultures are important to recapitulate the transcriptional profile of in vivo airway epithelia. Am J Physiol Lung Cell Mol Physiol 300: L25‐L31, 2011.
 178.Pietra GG, Szidon JP, Leventhal MM, Fishman AP. Hemoglobin as a tracer in hemodynamic pulmonary edema. Science (New York, NY) 166: 1643‐1646, 1969.
 179.Platz J, Bonenfant NR, Uhl FE, Coffey AL, McKnight T, Parsons C, Sokocevic D, Borg ZD, Lam Y‐W, Deng B, Fields JG, DeSarno M, Loi R, Hoffman AM, Bianchi J, Dacken B, Petersen T, Wagner DE, Weiss DJ. Comparative decellularization and recellularization of wild‐type and alpha 1,3 galactosyltransferase knockout pig lungs: A model for ex vivo xenogeneic lung bioengineering and transplantation. Tissue Eng Part C Methods 22: 725‐739, 2016.
 180.Prange HD. Laplace's law and the alveolus: A misconception of anatomy and a misapplication of physics. Adv Physiol Educ 27: 34‐40, 2003.
 181.Presson RG Jr, Baumgartner WA Jr, Peterson AJ, Glenny RW, Wagner WW Jr. Pulmonary capillaries are recruited during pulsatile flow. J Appl Physiol (1985) 92: 1183‐1190, 2002.
 182.Price AP, England KA, Matson AM, Blazar BR, Panoskaltsis‐Mortari A. Development of a decellularized lung bioreactor system for bioengineering the lung: The matrix reloaded. Tissue Eng Part A 16: 2581‐2591, 2010.
 183.Price AP, Godin LM, Domek A, Cotter T, D'Cunha J, Taylor DA, Panoskaltsis‐Mortari A. Automated decellularization of intact, human‐sized lungs for tissue engineering. Tissue Eng Part C Methods 21: 94‐103, 2015.
 184.Protti A, Cressoni M, Santini A, Langer T, Mietto C, Febres D, Chierichetti M, Coppola S, Conte G, Gatti S, Leopardi O, Masson S, Lombardi L, Lazzerini M, Rampoldi E, Cadringher P, Gattinoni L. Lung stress and strain during mechanical ventilation: Any safe threshold? Am J Respir Crit Care Med 183: 1354‐1362, 2011.
 185.Punde TH, Wu W‐H, Lien P‐C, Chang Y‐L, Kuo P‐H, Chang MD‐T, Lee K‐Y, Huang C‐D, Kuo H‐P, Chan Y‐F. A biologically inspired lung‐on‐a‐chip device for the study of protein‐induced lung inflammation. Integr Biol 7: 162‐169, 2015.
 186.Ramasamy SK, Kusumbe AP, Adams RH. Regulation of tissue morphogenesis by endothelial cell‐derived signals. Trends Cell Biol 25: 148‐157, 2015.
 187.Raredon MSB, Rocco KA, Gheorghe CP, Sivarapatna A, Ghaedi M, Balestrini JL, Raredon TL, Calle EA, Niklason LE. Biomimetic culture reactor for whole‐lung engineering. BioResearch Open Access 5: 72‐83, 2016.
 188.Ravasio A, Hobi N, Bertocchi C, Jesacher A, Dietl P, Haller T. Interfacial sensing by alveolar type II cells: A new concept in lung physiology? Am J Physiol Cell Physiol 300: C1456‐C1465, 2011.
 189.Ren X, Moser PT, Gilpin SE, Okamoto T, Wu T, Tapias LF, Mercier FE, Xiong L, Ghawi R, Scadden DT, Mathisen DJ, Ott HC. Engineering pulmonary vasculature in decellularized rat and human lungs. Nat Biotechnol 33: 1097‐1102, 2015.
 190.Rokkam D, Lafemina MJ, Lee JW, Matthay MA, Frank JA. Claudin‐4 levels are associated with intact alveolar fluid clearance in human lungs. Am J Pathol 179: 1081‐1087, 2011.
 191.Roughton F, Forster R. Relative importance of diffusion and chemical reaction rates in determining rate of exchange of gases in the human lung, with special reference to true diffusing capacity of pulmonary membrane and volume of blood in the lung capillaries. J Appl Physiol 11: 290‐302, 1957.
 192.Rutili G, Kvietys P, Martin D, Parker JC, Taylor AE. Increased pulmonary microvasuclar permeability induced by alpha‐naphthylthiourea. J Appl Physiol 52: 1316‐1323, 1982.
 193.Scarritt ME, Bonvillain RW, Burkett BJ, Wang G, Glotser EY, Zhang Q, Sammarco MC, Betancourt AM, Sullivan DE, Bunnell BA. Hypertensive rat lungs retain hallmarks of vascular disease upon decellularization but support the growth of mesenchymal stem cells. Tissue Eng Part A 20: 1426‐1443, 2014.
 194.Scarritt ME, Pashos NC, Motherwell JM, Eagle ZR, Burkett BJ, Gregory AN, Mostany R, Weiss DJ, Alvarez DF, Bunnell BA. Re‐endothelialization of rat lung scaffolds through passive, gravity‐driven seeding of segment‐specific pulmonary endothelial cells. J Tissue Eng Regen Med 12: e786‐e806, 2018.
 195.Schittny JC, Mund SI, Stampanoni M. Evidence and structural mechanism for late lung alveolarization. Am J Physiol Lung Cell Mol Physiol 294: L246‐L254, 2008.
 196.Schlingmann B, Molina SA, Claudins KM. Gatekeepers of lung epithelial function. Semin Cell Dev Biol 42: 47‐57, 2015.
 197.Schneeberger EEL, Robert D. Tight junctions: Their structure, composition, and function. Circ Res 55: 723‐733, 1984.
 198.Schneeberger‐Keeley EE, Karnovsky MJ. The ultrastructural basis of alveolar‐capillary membrane permeability to peroxidase used as a tracer. J Cell Biol 37: 781‐793, 1968.
 199.Schürch S. Surface tension at low lung volumes: Dependence on time and alveolar size. Respir Physiol 48: 339‐355, 1982.
 200.Schürch S, Goerke J, Clements JA. Direct determination of surface tension in the lung. Proc Natl Acad Sci 73: 4698‐4702, 1976.
 201.Singhal S, Henderson R, Horsfield K, Harding K, Cumming G. Morphometry of the human pulmonary arterial tree. Circ Res 33: 190‐197, 1973.
 202.Skardal A, Murphy SV, Devarasetty M, Mead I, Kang H‐W, Seol Y‐J, Zhang YS, Shin S‐R, Zhao L, Aleman J. Multi‐tissue interactions in an integrated three‐tissue organ‐on‐a‐chip platform. Sci Rep 7: 8837, 2017.
 203.Slutsky AS, Ranieri VM. Ventilator‐induced lung injury. N Engl J Med 369: 2126‐2136, 2013.
 204.Sobin SS, Fung Y‐C, Lindal RG, Tremer HM, Clark L. Topology of pulmonary arterioles, capillaries, and venules in the cat. Microvasc Res 19: 217‐233, 1980.
 205.Sobin SS, Fung YC, Tremer HM. Collagen and elastin fibers in human pulmonary alveolar walls. J Appl Physiol 64: 1659‐1675, 1988.
 206.Sobin SS, Fung YC, Tremer HM, Rosenquist TH. Elasticity of the pulmonary alveolar microvascular sheet in the cat. Circ Res 30: 440‐450, 1972.
 207.Sobin SS, Tremer HM, Fung Y. Morphometric basis of the sheet‐flow concept of the pulmonary alveolar microcirculation in the cat. Circ Res 26: 397‐414, 1970.
 208.Sokocevic D, Bonenfant NR, Wagner DE, Borg ZD, Lathrop MJ, Lam Y‐W, Deng B, DeSarno MJ, Ashikaga T, Loi R, Hoffman AM, Weiss DJ. The effect of age and emphysematous and fibrotic injury on the re‐cellularization of de‐cellularized lungs. Biomaterials 34: 3256‐3269, 2013.
 209.Song JJ, Kim SS, Liu Z, Madsen JC, Mathisen DJ, Vacanti JP, Ott HC. Enhanced in vivo function of bioartificial lungs in rats. Ann Thorac Surg 92: 998‐1005; discussion 1005‐1006, 2011.
 210.Srinivasan B, Kolli AR, Esch MB, Abaci HE, Shuler ML, Hickman JJ. TEER measurement techniques for in vitro barrier model systems. J Lab Autom 20: 107‐126, 2015.
 211.Stabler CT, Caires LC Jr, Mondrinos MJ, Marcinkiewicz C, Lazarovici P, Wolfson MR, Lelkes PI. Enhanced re‐endothelialization of decellularized rat lungs. Tissue Eng Part C Methods 22: 439‐450, 2016.
 212.Stahl EC, Bonvillain RW, Skillen CD, Burger BL, Hara H, Lee W, Trygg CB, Didier PJ, Grasperge BF, Pashos NC, Bunnell BA, Bianchi J, Ayares DL, Guthrie KI, Brown BN, Petersen TH. Evaluation of the host immune response to decellularized lung scaffolds derived from α‐Gal knockout pigs in a non‐human primate model. Biomaterials 187: 93‐104, 2018.
 213.Staub NC, Nagano H, Pearce ML. Pulmonary edema in dogs, especially the sequence of fluid accumulation in lungs. J Appl Physiol 22: 227‐240, (1985) 1967.
 214.Staub NC, Schultz EL. Pulmonary capillary length in dogs, cat and rabbit. Respir Physiol 5: 371‐378, 1968.
 215.Stone KC, Mercer RR, Freeman BA, Chang LY, Crapo JD. Distribution of lung cell numbers and volumes between alveolar and nonalveolar tissue. Am Rev Respir Dis 146: 454‐456, 1992.
 216.Stone KC, Mercer RR, Gehr P, Stockstill B, Crapo JD. Allometric relationships of cell numbers and size in the mammalian lung. Am J Respir Cell Mol Biol 6: 235‐243, 1992.
 217.Stucki AO, Stucki JD, Hall SR, Felder M, Mermoud Y, Schmid RA, Geiser T, Guenat OT. A lung‐on‐a‐chip array with an integrated bio‐inspired respiration mechanism. Lab Chip 15: 1302‐1310, 2015.
 218.Suki B. Assessing the functional mechanical properties of bioengineered organs with emphasis on the lung. J Cell Physiol 229: 1134‐1140, 2014.
 219.Suki B, Stamenovic D, Hubmayr R. Lung parenchymal mechanics. Compr Physiol 1: 1317‐1351, 2011.
 220.Sweet DG, Carnielli V, Greisen G, Hallman M, Ozek E, Plavka R, Saugstad OD, Simeoni U, Speer CP, Vento M, Halliday HL, European Association of Perinatal Medicine. European Consensus Guidelines on the Management of Neonatal Respiratory Distress Syndrome in Preterm Infants—2013 Update. Basel: Karger Publishers, 2013, p. 353‐368.
 221.Szidon JP, Pietra GG, Fishman AP. The alveolar‐capillary membrane and pulmonary edema. N Engl J Med 286: 1200‐1204, 1972.
 222.Takahashi A, Majumdar A, Parameswaran H, Bartolák‐Suki E, Suki B. Proteoglycans maintain lung stability in an elastase‐treated mouse model of emphysema. Am J Respir Cell Mol Biol 51: 26‐33, 2014.
 223.Taylor AE. Capillary fluid filtration. Starling forces and lymph flow. Circ Res 49: 557‐575, 1981.
 224.Taylor AE, Gaar KA. Estimation of equivalent pore radii of pulmonary capillary and alveolar membranes. Am J Physiol 218: 1133‐1140, 1970.
 225.Thompson BT, Chambers RC, Liu KD. Acute respiratory distress syndrome. N Engl J Med 377: 1904‐1905, 2017.
 226.Toepke MW, Beebe DJ. PDMS absorption of small molecules and consequences in microfluidic applications. Lab Chip 6: 1484‐1486, 2006.
 227.Tong MZ, Johnston DR, Pettersson GB. The role of bronchial artery revascularization in lung transplantation. Thorac Surg Clin 25: 77‐85, 2015.
 228.Toshima M, Ohtani Y, Ohtani O. Three‐dimensional architecture of elastin and collagen fiber networks in the human and rat lung. Arch Histol Cytol 67: 31‐40, 2004.
 229.Tschanz SA, Salm LA, Roth‐Kleiner M, Barré SF, Burri PH, Schittny JC. Rat lungs show a biphasic formation of new alveoli during postnatal development. J Appl Physiol (1985) 117: 89‐95, 2014.
 230.Tschumperlin DJ, Margulies SS. Alveolar epithelial surface area‐volume relationship in isolated rat lungs. J Appl Physiol 86: 2026‐2033, 1999.
 231.Tsukimoto K, Mathieu‐Costello O, Prediletto R, Elliott AR, West JB. Ultrastructural appearances of pulmonary capillaries at high transmural pressures. J Appl Physiol 71: 573‐582, 1991.
 232.Vaccaro CA, Brody JS. Structural features of alveolar wall basement membrane in the adult rat lung. J Cell Biol 91: 427‐437, 1981.
 233.Wagner DE, Bonenfant NR, Parsons CS, Sokocevic D, Brooks EM, Borg ZD, Lathrop MJ, Wallis JD, Daly AB, Lam Y‐W, Deng B, DeSarno MJ, Ashikaga T, Loi R, Weiss DJ. Comparative decellularization and recellularization of normal versus emphysematous human lungs. Biomaterials 35: 3281‐3297, 2014.
 234.Wagner DE, Bonenfant NR, Sokocevic D, DeSarno MJ, Borg ZD, Parsons CS, Brooks EM, Platz JJ, Khalpey ZI, Hoganson DM, Deng B, Lam YW, Oldinski RA, Ashikaga T, Weiss DJ. Three‐dimensional scaffolds of acellular human and porcine lungs for high throughput studies of lung disease and regeneration. Biomaterials 35: 2664‐2679, 2014.
 235.Wagner W Jr, Latham L. Pulmonary capillary recruitment during airway hypoxia in the dog. J Appl Physiol 39: 900‐905, 1975.
 236.Wagner WW Jr, Latham LP, Capen RL. Capillary recruitment during airway hypoxia: Role of pulmonary artery pressure. J Appl Physiol 47: 383‐387, 1979.
 237.Wagner W, Bennett RD, Ackermann M, Ysasi AB, Belle J, Valenzuela CD, Pabst A, Tsuda A, Konerding MA, Mentzer SJ. Elastin cables define the axial connective tissue system in the murine lung. Anat Rec (Hoboken) 298: 1960‐1968, 2015.
 238.Wallis JM, Borg ZD, Daly AB, Deng B, Ballif BA, Allen GB, Jaworski DM, Weiss DJ. Comparative assessment of detergent‐based protocols for mouse lung de‐cellularization and re‐cellularization. Tissue Eng Part C Methods 18: 420‐432, 2012.
 239.Walter FR, Valkai S, Kincses A, Petneházi A, Czeller T, Veszelka S, Ormos P, Deli MA, Dér A. A versatile lab‐on‐a‐chip tool for modeling biological barriers. Sens Actuators B 222: 1209‐1219, 2016.
 240.Wang JD, Douville NJ, Takayama S, ElSayed M. Quantitative analysis of molecular absorption into PDMS microfluidic channels. Ann Biomed Eng 40: 1862‐1873, 2012.
 241.Wangensteen OD, Wittmers LE, Johnson JA. Permeability of the mammalian blood‐gas barrier and its components. Am J Physiol 216: 719‐727, 1969.
 242.Warburton D, El‐Hashash A, Carraro G, Tiozzo C, Sala F, Rogers O, De Langhe S, Kemp PJ, Riccardi D, Torday J, Bellusci S, Shi W, Lubkin SR, Jesudason E. Lung organogenesis. Curr Top Dev Biol 90: 73‐158, 2010.
 243.Ware LB, Matthay MA. Alveolar fluid clearance is impaired in the majority of patients with acute lung injury and the acute respiratory distress syndrome. Am J Respir Crit Care Med 163: 1376‐1383, 2001.
 244.Weibel E. Lung morphometry and models in respiratory physiology. In: Chang HK, Paiva M, editors. Respiratory Physiology: An Analytical Approach. New York: Marcel Dekker, 1989, p. 1‐56.
 245.Weibel ER. Geometric and dimensional airway models of conductive, transitory and respiratory zones of the human lung. In: Morphometry of the Human Lung. Berlin, Heidelberg: Springer, 1963, p. 136‐142.
 246.Weibel ER. Morphometric estimation of pulmonary diffusion capacity: I. Model and method. Respir Physiol 11: 54‐75, 1970.
 247.Weibel ER. The mystery of “non‐nucleated plates” in the alveolar epithelium of the lung explained. Acta Anat 78: 425‐443, 1971.
 248.Weibel ER. Morphometric estimation of pulmonary diffusion capacity: V. Comparative morphometry of alveolar lungs. Respir Physiol 14: 26‐43, 1972.
 249.Weibel ER. Morphological basis of alveolar‐capillary gas exchange. Physiol Rev 53: 419‐495, 1973.
 250.Weibel ER. On the tricks alveolar epithelial cells play to make a good lung. Am J Respir Crit Care Med 191: 504‐513, 2015.
 251.Weibel ER, Federspiel WJ, Fryder‐Doffey F, Hsia CC, König M, Stalder‐Navarro V, Vock R. Morphometric model for pulmonary diffusing capacity I. Membrane diffusing capacity. Respir Physiol 93: 125‐149, 1993.
 252.Weibel ER, Knight BW. A morphometric study on the thickness of the pulmonary air‐blood barrier. J Cell Biol 21: 367‐384, 1964.
 253.Welling LW, Grantham JJ. Physical properties of isolated perfused renal tubules and tubular basement membranes. J Clin Invest 51: 1063‐1075, 1972.
 254.West JB. Regional differences in gas exchange in the lung of erect man. J Appl Physiol 17: 893‐898, 1962.
 255.West JB. Thoughts on the pulmonary blood‐gas barrier. Am J Physiol Lung Cell Mol Physiol 285: L501‐L513, 2003.
 256.West JB, Dollery CT. Distribution of blood flow and ventilation‐perfusion ratio in the lung, measured with radioactive carbon dioxide. J Appl Physiol 15: 405‐410, 1960.
 257.West JB, Dollery CT, Naimark A. Distribution of blood flow in isolated lung; relation to vascular and alveolar pressures. J Appl Physiol 19: 713‐724, 1964.
 258.West JB, Mathieu‐Costello O. Strength of the pulmonary blood‐gas barrier. Respir Physiol 88: 141‐148, 1992.
 259.West JB, Mathieu‐Costello O. Stress failure of pulmonary capillaries: Role in lung and heart disease. Lancet 340: 762‐767, 1992.
 260.West JB, Mathieu‐Costello O, Jones JH, Birks EK, Logemann RB, Pascoe JR, Tyler WS. Stress failure of pulmonary capillaries in racehorses with exercise‐induced pulmonary hemorrhage. J Appl Physiol 75: 1097‐1109, 1993.
 261.West JB, Tsukimoto K, Mathieu‐Costello O, Prediletto R. Stress failure in pulmonary capillaries. J Appl Physiol 70: 1731‐1742, 1991.
 262.West JBL, Andrew M. Mechanics of breathing. In: West's Respiratory Physiology. Philadelphia: Wolters Kluwer, 2016, p. 108‐141.
 263.Wilson TA. Relations among recoil pressure, surface area, and surface tension in the lung. J Appl Physiol Respir Environ Exerc Physiol 50: 921‐930, 1981.
 264.Wilson TA, Bachofen H. A model for mechanical structure of the alveolar duct. J Appl Physiol 52: 1064‐1070, 1982.
 265.Wirtz HR, Dobbs LG. Calcium mobilization and exocytosis after one mechanical stretch of lung epithelial cells. Science (New York, NY) 250: 1266‐1269, 1990.
 266.Wright JR, Dobbs LG. Regulation of pulmonary surfactnt secretion and clearance. Annu Rev Physiol 53: 395‐414, 1991.
 267.Xu Z, Li E, Guo Z, Yu R, Hao H, Xu Y, Sun Z, Li X, Lyu J, Wang Q. Design and construction of a multi‐organ microfluidic chip mimicking the in vivo microenvironment of lung cancer metastasis. ACS Appl Mater Interfaces 8: 25840‐25847, 2016.
 268.Yang X, Li K, Zhang X, Liu C, Guo B, Wen W, Gao X. Nanofiber membrane supported lung‐on‐a‐chip microdevice for anti‐cancer drug testing. Lab Chip 18: 486‐495, 2018.
 269.Yao J, Guihard PJ, Wu X, Blazquez‐Medela AM, Spencer MJ, Jumabay M, Tontonoz P, Fogelman AM, Boström KI, Yao Y. Vascular endothelium plays a key role in directing pulmonary epithelial cell differentiation. J Cell Biol 216: 3369‐3385, 2017.
 270.Yen R, Sobin S. Pulmonary blood flow in the cat: Correlation between theory and experiment. In: Schmid‐Schönbein GW, Woo SL‐Y, Zweifach BW, editors. Frontiers in Biomechanics. New York: Springer‐Verlag, 1986, p. 365‐376.
 271.Yen R, Zhuang F, Fung Y, Ho H, Tremer H, Sobin S. Morphometry of cat pulmonary venous tree. J Appl Physiol 55: 236‐242, 1983.
 272.Yen R, Zhuang F, Fung Y, Ho H, Tremer H, Sobin S. Morphometry of cat's pulmonary arterial tree. J Biomech Eng 106: 131‐136, 1984.
 273.Yuan H, Kononov S, Cavalcante FS, Lutchen KR, Ingenito EP, Suki B. Effects of collagenase and elastase on the mechanical properties of lung tissue strips. J Appl Physiol 89: 3‐14, 2000.
 274.Zacharias WJ, Frank DB, Zepp JA, Morley MP, Alkhaleel FA, Kong J, Zhou S, Cantu E, Morrisey EE. Regeneration of the lung alveolus by an evolutionarily conserved epithelial progenitor. Nature 555: 251, 2018.
 275.Zeltner TB, Burri PH. The postnatal development and growth of the human lung. II. Morphology. Respir Physiol 67: 269‐282, 1987.
 276.Zeltner TB, Caduff JH, Gehr P, Pfenninger J, Burri PH. The postnatal development and growth of the human lung. I. Morphometry. Respir Physiol 67: 247‐267, 1987.
 277.Zhou H, Kitano K, Ren X, Rajab TK, Wu M, Gilpin SE, Wu T, Baugh L, Black LD, Mathisen DJ, Ott HC. Bioengineering human lung grafts on porcine matrix. Ann Surg 267: 590‐598, 2018.
 278.Zhuang F, Yen M, Fung Y, Sobin S. How many pulmonary alveoli are supplied by a single arteriole and drained by a single venule? Microvasc Res 29: 18‐31, 1985.

Teaching Material

Katherine L. Leiby, Micha Sam Brickman Raredon, and Laura E. Niklason. Bioengineering the Blood-gas Barrier. Compr Physiol 10 : 2020, 415-452.

Didactic Synopsis

Major teaching points:

1. Key structural features of the blood-gas barrier:

    a. Extreme thinness
    b. Large alveolar surface area
    c. High density of vessels within the pulmonary capillary "sheet"

2. Lungs have a complex pressure-volume relationship principally governed by tissue stretch and surface tension.

3. Biological aspects of blood-gas separation:

    a. Cell-cell junctions, particularly epithelial tight junctions
    b. Alveolar fluid clearance
    c. Surfactant, which aids fluid balance across the pulmonary capillaries

4. Collagen I, elastin, and proteoglycans provide the bulk of lung parenchymal structural integrity. Collagen IV is the primary source of alveolar-capillary membrane strength.

5. Lung-on-a-chip:

    a. Advances: Chips with multiple cell types, air-liquid interface, and cyclic strain; multi-organ chip systems
    b. Challenges: Physiologically-appropriate membrane, resistance measurements comparable across platforms

6. Engineered whole lungs:

    a. Advances: Decellularization and culture of human-sized lungs, improved endothelial recellularization
    b. Challenges: Achieving blood-gas separation, developing/implementing methodologies for assessing barrier function

Didactic Legends

The following legends to the figures that appear throughout the article are written to be useful for teaching.

Figure 1. Teaching points: This figure illustrates the principle phases of early lung development, beginning in the fetal period. While aspects of blood-gas barrier development (noted in the figure) occur as early as the pseudoglandular stage, the most significant restructuring of the alveolar membrane takes place during alveolarization and microvascular maturation. These phases are characterized by a dramatic increase in alveolar surface area (which cannot be explained by an increase in lung volume alone) with simultaneous thinning of the alveolar membrane. The result is a substantial increase in lung diffusing capacity. It is still debated whether septation, or the subdivision of existing alveoli into new airspaces, occurs past early childhood in human.

Figure 2. Teaching points: These hematoxylin and eosin (H&E) images of native rat lung illustrate the dramatic changes that occur in airspace structure during later lung development, from the late saccular period (A) (postnatal day 1 (P1)), through alveolarization (B)-(E) (P4-P21), to adult (F) (P60). (A) The P1 rat lung is comprised of large saccules with thick walls. (B) Beginning at P4, the onset of alveolarization, secondary septa subdivide the airspaces into smaller alveoli, a process that continues through P7 (C), P14 (D), and P21 (E). Secondary septation leads to a significant increase in alveolar surface area. Note over this time period that there is a simultaneous thinning of the alveolar walls, due to both tissue remodeling and to a slowing of cellular proliferation near the end of alveolarization. The P21 lung has an appearance similar to that of adult (F), though airspace size may change with growth of the animal.

Figure 3. Teaching points: This figure illustrates several key features of the blood-gas barrier, as revealed by different length scales. In the top of each panel is an image of native lung taken by light or electron microscopy. In the bottom of each panel is a corresponding schematic for clarity. (A) The distal lung parenchyma is made up of alveoli packed together as if in a foam. This organization allows for an extremely large surface area for gas exchange to be packed into a relatively small volume. (B) This light micrograph and corresponding schematic illustrate the structure of individual alveolar septa. This image was taken from a saline-filled lung; it is the absence of surface tension that causes the capillaries to bulge into the airspaces. Note that the septa are rich with capillaries (open spaces bulging to either side of the septa in the top image; "blood-filled" round capillaries in the schematic). This high density of alveolar capillaries serves to create a very large capillary surface area, to facilitate gas exchange. (C) This transmission electron micrograph and accompanying schematic zooms in further, on a cross-section of a single alveolar capillary. Note that one side of the capillary (upper right) is "thin," comprising only of a thin epithelial cell process, a fused basement membrane, and a thin endothelial cell process. This thin side is the site of gas exchange. The "thick" side (lower left), on the other hand, contains additional interstitial components and provides support to the adjacent capillary and to the septum. (D) This panel provides a closer look at the thin gas-exchanging part of the blood gas barrier.

Figure 4. Teaching points: This schematic illustrates in more detail the layers comprising the blood-gas barrier, through which gas must diffuse to pass from the airspaces to the blood, or vice-versa. By Fick's law, an increase in thickness or a decrease in the oxygen permeation coefficient of any of these layers will decrease the diffusion capacity of gas across the barrier.

Figure 5. Teaching points: This figure illustrates several key features of the alveolar capillary network. (A) Different regions of capillaries are either supplied by an arteriole, or drained by a venule. Those regions drained by venules comprise a slightly larger area of continuous tissue surrounding "islands" of alveoli supplied by individual arterioles. (B,C) The alveolar capillary network is extremely dense, with many different capillary "paths" crossing the distance from arteriole to vein and forming a dense meshwork of vessels in the alveolar walls. (D,E) This dense meshwork may be modeled as a "sheet" of blood that is very short (distance between arteriole and venule) but extremely wide due to the large number of capillaries.

Figure 6. Teaching points: This figure illustrates the unique morphology of alveolar epithelial type I cells (AEC1s). (A) These cells cover the vast majority of the alveolar surface. (B,C) AEC1s can have a complex morphology that includes thin cell stalks passing between capillaries, and multiple flat plates of cytoplasm lining both sides of a single septum.

Figure 7. Teaching points: This graph shows the lung pressure-volume curve in the setting of either air or saline inflation. In the air-filled lung, due to the presence of surface tension at the air-liquid interface, there is prominent hysteresis between the inflation and deflation limbs of the curve (i.e., the volume at a given transpulmonary pressure depends on whether that volume was reached by inflation or deflation). In a saline-filled lung, which does not have surface tension, hysteresis is nearly absent. Note as well how the curve for the air-filled lung is nearly flat at low transpulmonary pressures, corresponding to very low compliance (low change in volume in response to a given change in pressure) at low lung volumes.

Figure 8. Teaching points: Several forces act upon the alveolar wall at the microscopic level, with important consequences for alveolar morphology and architecture as discussed in the text. 1) Surface tension acts tangential to the surface of the alveolar lining layer (ALL). It generally results in a net lumen-directed force, as is the case in the alveolar corner at the left of this figure; however it may also produce a compressive force on convex structures that bulge into the alveolar space such as the alveolar type II cell (AEC2) in this illustration. 2) Wall tension (a tissue force) arises due to stretching of the alveolar wall with lung inflation. 3) Circumferential wall tension in the capillary wall arises due to capillary transmural pressure. If this tension leads to stretch of neighboring septal components it may also contribute to tissue forces.

Figure 9. Teaching points: This figure, a transmission electron micrograph and accompanying line drawing of an alveolar septum, illustrates how the surface area of the alveolar lumen (arrow heads in line drawing) may be less than the surface area of the alveolar epithelial basement membrane (dotted line) due to extensive septal/basement membrane pleating. As a result, expansion of the lung from low volumes occurs primarily by unfolding of these tissue pleats, rather than by stretching.

Figure 10. Teaching points: These images, taken by scanning electron microscope (SEM), depict how alveolar architecture is affected by surface tension and by lung volume. In an air-filled lung with normal surface tension, the alveolar septa appear "crumpled" due to low tissue stretch at lower lung volumes, with redundant tissue pleating at the corners (A). Note how the alveolar surfaces appear smoother, and the capillaries flatter, at higher lung volumes in the setting of higher surface tension (B). By comparison, in a saline-filled lung with no surface tension, capillaries bulge at both low (C) and high (D) lung volumes, lending a bumpy appearance to the alveolar surfaces, and the alveolar septa are wavy.

Figure 11. Teaching points: Pulmonary capillary blood flow is affected by location within the lung, due in part to the effects of gravity. A capillary in Zone 1 (apex of the lung), compared to a capillary in Zone 3 (base of the lung), is relatively more compressed, with associated lower blood flow, due to relatively higher alveolar pressures and lower arterial and venous pressures higher up in the lung.

Figure 12. Teaching points: This figure illustrates how the alveolar septum is uniquely designed to protect against edema that may lead to thickening of the blood-gas barrier and, in severe cases, flooding of the alveoli, significantly impairing gas exchange. (A) In homeostasis, three of the Starling forces (capillary hydrostatic pressure Pc, the negative interstitial hydrostatic pressure Pis, and interstitial oncotic pressure πis favor filtration from the capillaries, whereas capillary oncotic pressure πc favors reabsorption; the net balance of these forces leads to a small amount of transudated fluid entering the lymphatic circulation in the space around the small vessels. (B) Several protective mechanisms act to prevent and/or limit edema. Illustrated here is the case of hydrostatic pulmonary edema in setting of increased Pc: 1) Pis increases (becomes less negative) as fluid enters the interstitium due to the low compliance of this space. 2) πis decreases due to dilution of the interstitial space by transudated fluid. 3) The pulmonary capillaries are particularly impermeable (effective pore size approximately 5 nm), compared to systemic capillaries and to extraalveolar pulmonary vessels. 4) Alveolar epithelial tight junctions provide a very tight barrier, at 1 nm. 5) Both alveolar epithelial type I (AEC1s) and type II (AEC2s) cells participate in alveolar fluid clearance, transporting ions from apical to basolateral membrane to create a driving force for fluid movement out of the alveolar lumen. 6) Surfactant secreted by AEC2s, which forms the surface film of the alveolar lining layer (ALL), reduces surface tension, thereby decreasing the contribution of this force to the negative Pis. 7) Lymph flow may increase significantly.

Figure 13. Teaching points: This figure illustrates the major supporting structures of the alveolar wall. A thin basement membrane composed primarily of collagen IV and laminin, underlies the alveolar epithelium and endothelium. Note that a fused basement membrane is the only supporting structure on the "thin" side of the capillaries. However, in the space between the capillaries, and in the "thick" portions of the wall, additional support is provided by collagen I (illustrated here in both cross-sectional and longitudinal orientations) and elastin, and to a lesser degree by fibroblasts. Proteoglycans, not illustrated here, have important interactions with collagen and elastin to further stabilize the delicate alveolar walls.

Figure 14. Teaching points: This figure illustrates one model of the behavior of individual collagen and elastin fibers in the lung, and shows the stress/strain behavior of the lung parenchyma as a whole. According to the "string-spring" model of collagen and elastin in the lung, at low volumes (A), collagen fiber "strings" are wavy and incompletely distended, whereas the elastin "springs" bear the stress within the tissue. At high volumes (B) the collagen fibers eventually reach their stop lengths (beyond which they cannot be extended without rupture), and limit further lung expansion. This behavior of collagen may at least partially explain the non-linear behavior of the lung stress-strain curve at higher strain values (C). Note as well in (C) that there is a threshold strain (approximately 1.5) above which there is a risk for ventilator-associated lung injury.

Figure 15. Teaching points: This figure illustrates several published lung-on-a-chip platforms from the past 15 years. While all models involve the co-culture of different cell types on either side of a membrane, more recent chips have incorporated additional physiologically relevant features and/or technological advancements in barrier assessment or visualization. These improvements include cyclic stretch (B); media flow (B,C); simultaneous microscopy and resistance measurements by trans-epithelial electrical resistance (TEER) (C); and microimpedance tomography electrodes, which provide more accurate resistance measurements than TEER and are compatible with application of cyclic stretch.

Figure 16. Teaching points: This figure illustrates the overall paradigm of whole lung engineering. In (A), (B), and (D), the bottom part of the panel provides a schematic of the alveolar septum during each stage of the process. (A) A native lung (either from an animal model, or a human lung that is unsuitable for transplantation) serves as the initial starting material. (B) The lung is decellularized, typically via detergents rinsed through the vasculature, to create an acellular scaffold. This process is designed to preserve the underlying extracellular matrix and native lung architecture while removing as much cellular material as possible. This process is facilitated by cannulas providing access to the airways (via the trachea) and the vasculature (via the pulmonary artery, through the heart). (C) The decellularized scaffold is next repopulated with cells that are typically derived either from primary isolation, or from the differentiation of induced pluripotent stem cells (iPSCs). Cells may be seeded into the airways and/or the vasculature. (D) The reseeded lung is transferred to a bioreactor that provides a sterile environment for lung culture, and which is designed to mimic physiological conditions by providing stimuli such as vascular perfusion and ventilation. (E) Following culture, engineered lungs may be analyzed ex vivo, before ultimate transplantation into a recipient (F).

 

Figure 17. Teaching points: Successful introduction of endothelial cells into the microvasculature of decellularized lung scaffolds has been a significant challenge for the field of lung engineering. (A) Due to the leaky nature of vessels within the decellularized lung (due to removal of endothelial cells and potential damage to the underlying basement membrane), fluid preferentially leaks out, leaving cells (which are relatively large in diameter compared to the caliber of the alveolar capillaries) stuck in the initial vascular segments. (B) Recent recognition of this "sieving" effect has led to strategies to more effectively introduce cells for revascularization. Seeding a dilute cell suspension helps to counteract the leakage of fluid, delaying the time until cell clumping, while seeding from a greater initial pressure allows cells to travel further into the capillary bed. Reaching the most distal segments of the capillaries is aided by seeding from both ends (i.e. from both the pulmonary artery and the pulmonary veins). Finally, seeding under pulsatile flow aids in recruitment of collapsed capillaries, facilitating entry of cells into more vessels.


Related Articles:

Functional Morphology of Lung Parenchyma
Lung Parenchymal Mechanics
Lung Structure and the Intrinsic Challenges of Gas Exchange
Tissue Engineering of the Microvasculature
Teaching Material

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Katherine L. Leiby, Micha Sam Brickman Raredon, Laura E. Niklason. Bioengineering the Blood‐gas Barrier. Compr Physiol 2020, 10: 415-452. doi: 10.1002/cphy.c190026