"Лёгкое-на-чипе" как инструмент для изучения патофизиологии дыхания человека
Автор: Жукова О.А., Озерская Ю.В., Басманов Д.В., Столяров В.Ю., Богуш В.Г., Колесов В.В., Зыков К.А., Юсубалиева Г.М., Баклаушев В.П.
Журнал: Клиническая практика @clinpractice
Рубрика: Научные обзоры
Статья в выпуске: 4 т.15, 2024 года.
Бесплатный доступ
«Лёгкое-на-чипе» (от англ. Lung-on-a-Chip, LoC) - микрофлюидное устройство, имитирующее газожидкостный интерфейс лёгочной альвеолы человека и предназначенное для патофизиологических, фармакологических и молекулярно-биологических исследований гематоальвеолярного барьера in vitro. Устройство LoC включает систему жидкостных и газовых микроканалов, разделённых полупроницаемой эластичной мембраной, содержащей полимерную основу и клеточные элементы альвеолы. В зависимости от вида LoC (одно-, двух- и трёхканальное) на мембране могут находиться только альвеолоциты или альвеолоциты в сочетании с другими клетками - эндотелиоцитами, фибробластами, альвеолярными макрофагами, опухолевыми клетками. Некоторые модели LoC также включают белковую или гидрогелевую строму, имитирующую лёгочный интерстиций. Первый двухканальный вариант LoC, в котором с одной стороны мембраны находится монослой альвеолоцитов, а с другой - монослой эндотелиоцитов, был разработан в 2010 году группой учёных Гарвардского университета с целью максимально точного воспроизведения in vitro микроокружения и биомеханики работы альвеолы. Современные модификации LoC включают те же элементы и отличаются лишь конструкцией микрофлюидной системы, биоматериалом полупроницаемой мембраны, составом клеточных и стромальных элементов и решаемыми специальными задачами. Помимо LoC, воспроизводящих гематоальвеолярный барьер, существуют модификации для исследования определённых патофизиологических процессов, скрининга лекарственных препаратов, моделирования конкретных заболеваний, например рака лёгкого, хронической обструктивной болезни лёгких или астмы. В данном обзоре мы проанализировали существующие разновидности LoC, применяемые биоматериалы, методы детекции молекулярных процессов в микрофлюидных устройствах и основные направления исследований с помощью «лёгкого-на-чипе».
Лёгкое-на-чипе, гематоальвеолярный барьер, болезни органов дыхания, микрофлюидные устройства
Короткий адрес: https://sciup.org/143183766
IDR: 143183766 | DOI: 10.17816/clinpract637140
Список литературы "Лёгкое-на-чипе" как инструмент для изучения патофизиологии дыхания человека
- Федеральная служба государственной статистики [Интернет]. Здравоохранение. [Federal State Statistics Service (Internet). Health care. (In Russ.)] Режим доступа: https:// rosstat.gov.ru/folder/13721. Дата обращения: 15.12.2024.
- Chen L, Rackley CR. Diagnosis and epidemiology of acute respiratory failure. Crit Care Clin. 2024;40(2):221–233. doi: 10.1016/j.ccc.2023.12.001
- Pan H, Deutsch GH, Wert SE; Ontology Subcommittee; NHLBI Molecular Atlas of Lung Development Program Consortium. Comprehensive anatomic ontologies for lung development: A comparison of alveolar formation and maturation within mouse and human lung. J Biomed Semantics. 2019;10(1):18. doi: 10.1186/s13326-019-0209-1
- Lagowala DA, Kwon S, Sidhaye VK, Kim DH. Human microphysiological models of airway and alveolar epithelia. Am J Physiol Lung Cell Mol Physiol. 2021;321(6):L1072–L1088. doi: 10.1152/ajplung.00103.2021
- Mullard A. Parsing clinical success rates. Nat Rev Drug Discov. 2016;15(7):447. doi: 10.1038/nrd.2016.136
- Zhu Y, Sun L, Wang Y, et al. A biomimetic human lung-on-a-chip with colorful display of microphysiological breath. Adv Mater. 2022;34(13):e2108972. doi: 10.1002/adma.202108972
- Huh D, Matthews BD, Mammoto A, et al. Reconstituting organ-level lung functions on a chip. Science. 2010;328(5986): 1662–1668. doi: 10.1126/science.1188302
- Varone A, Nguyen JK, Leng L, et al. A novel organ-chip system emulates three-dimensional architecture of the human epithelia and the mechanical forces acting on it. Biomaterials. 2021;275:120957. doi: 10.1016/j.biomaterials.2021.120957
- Doryab A, Amoabediny G, Salehi-Najafabadi A. Advances in pulmonary therapy and drug development: Lung tissue engineering to lung-on-a-chip. Biotechnol Adv. 2016;34(5): 588–596. doi: 10.1016/j.biotechadv.2016.02.006
- Jensen C, Teng Y. Is it time to start transitioning from 2D to 3D cell culture? Front Mol Biosci. 2020;7:33. doi: 10.3389/fmolb.2020.00033
- Wang H, Brown PC, Chow EC, et al. 3D cell culture models: Drug pharmacokinetics, safety assessment, and regulatory consideration. Clin Transl Sci. 2021;14(5):1659–1680. doi: 10.1111/cts.13066
- Duval K, Grover H, Han LH, et al. Modeling physiological events in 2D vs. 3D cell culture. Physiology (Bethesda). 2017;32(4):266–277. doi: 10.1152/physiol.00036.2016
- Martínez-Espuga M, Mata A, Ordóñez-Morán P. Intestinal cell differentiation and phenotype in 2D and 3D cell culture models. Methods Mol Biol. 2023;2650:235–243. doi: 10.1007/978-1-0716-3076-1_18
- Sisodia Y, Shah K, Ali Sayyed A, et al. Lung-on-chip microdevices to foster pulmonary drug discovery. Biomater Sci. 2023;11(3):777–790. doi: 10.1039/d2bm00951j
- Zhu L, Zhang J, Guo Q, et al. Advanced lung organoids and lung-on-a-chip for cancer research and drug evaluation: A review. Front Bioeng Biotechnol. 2023;11:1299033. doi: 10.3389/fbioe.2023.1299033
- Tan J, Guo Q, Tian L, et al. Biomimetic lung-on-a-chip to model virus infection and drug evaluation. Eur J Pharm Sci. 2023;180:106329. doi: 10.1016/j.ejps.2022.106329
- Kim HJ, Park S, Jeong S, et al. Lung organoid on a chip: A new ensemble model for preclinical studies. Int J Stem Cells. 2024;17(1):30–37. doi: 10.15283/ijsc23090
- Shrestha J, Razavi Bazaz S, Aboulkheyr EH, et al. Lungon- a-chip: The future of respiratory disease models and pharmacological studies. Crit Rev Biotechnol. 2020;40(2):213–230. doi: 10.1080/07388551.2019.1710458
- Nawroth JC, Barrile R, Conegliano D, et al. Stem cell-based lung-on-chips: The best of both worlds? Adv Drug Deliv Rev. 2019;140:12–32. doi: 10.1016/j.addr.2018.07.005
- Zarrintaj P, Saeb MR, Stadler FJ, et al. Human organs-onchips: A review of the state-of-the-art, current prospects, and future challenges. Adv Biol (Weinh). 2022;6(1):e2000526. doi: 10.1002/adbi.202000526
- Travaglini KJ, Nabhan AN, Penland L, et al. A molecular cell atlas of the human lung from single-cell RNA sequencing. Nature. 2020;587(7835):619–625. doi: 10.1038/s41586-020-2922-4
- Hartmann B, Fleischhauer L, Nicolau M, et al. Profiling native pulmonary basement membrane stiffness using atomic force microscopy. Nat Protoc. 2024;19(5):1498–1528. doi: 10.1038/s41596-024-00955-7
- Doryab A, Tas S, Taskin MB, et al. Evolution of bioengineered lung models: Recent advances and challenges in tissue mimicry for studying the role of mechanical forces in cell biology. Adv Functional Materials. 2019;29(39). doi: 10.1002/adfm.201903114
- Bai H, Ingber DE. What can an organ-on-a-chip teach us about human lung pathophysiology? Physiology (Bethesda). 2022;37(5):0. doi: 10.1152/physiol.00012.2022
- Whitsett JA, Weaver TE. Hydrophobic surfactant proteins in lung function and disease. N Engl J Med. 2002;347(26): 2141–2148. doi: 10.1056/NEJMra022387
- Doryab A, Groll J. Biomimetic in vitro lung models: Current challenges and future perspective. Adv Mater. 2023;35(13): e2210519. doi: 10.1002/adma.202210519
- Shah DD, Raghani NR, Chorawala MR, et al. Harnessing three-dimensional (3D) cell culture models for pulmonary infections: State of the art and future directions. Naunyn Schmiedebergs Arch Pharmacol. 2023;396(11):2861–2880. doi: 10.1007/s00210-023-02541-2
- Fang Y, Eglen RM. Three-dimensional cell cultures in drug discovery and development. SLAS Discov. 2017;22(5):456–472. doi: 10.1177/1087057117696795
- Morin JP, Baste JM, Gay A, et al. Precision cut lung slices as an efficient tool for in vitro lung physiopharmacotoxicology studies. Xenobiotica. 2012;43(1):63–72. doi: 10.3109/00498254.2012.727043
- Wyss Institute. Human organs-on-chips [2021 Mar 28]. Режим доступа: https://wyss.harvard.edu/. Дата обращения: 15.12.2024.
- Novik E, Maguire TJ, Chao P, et al. A microfluidic hepatic coculture platform for cell-based drug metabolism studies. Biochem Pharmacol. 2010;79(7):1036–1044. doi: 10.1016/j.bcp.2009.11.010
- Jang KJ, Mehr AP. Hamilton GA, et al. Human kidney proximal tubule-on-a-chip for drug transport and nephrotoxicity assessment. Integr Biol (Camb). 2013;5(9):1119–1129. doi: 10.1039/c3ib40049b
- Kim HJ, Ingber DE. Gut-on-a-chip microenvironment induces human intestinal cells to undergo villus differentiation. Integr Biol (Camb). 2013;5(9):1130–1140. doi: 10.1039/c3ib40126j
- Zhang W, Lee WY, Siegel DS, et al. Patient-specific 3D microfluidic tissue model for multiple myeloma. Tissue Engin Part C Methods. 2014;20(8):663–670. doi: 10.1089/ten.TEC.2013.0490
- Van der Meer AD, Orlova VV, ten Dijke P, et al. Three-dimensional co-cultures of human endothelial cells and embryonic stem cell-derived pericytes inside a microfluidic device. Lab Chip. 2013;13(18):3562–3568. doi: 10.1039/c3lc50435b
- Grosberg A, Nesmith AP, Goss JA, et al. Muscle on a chip: In vitro contractility assays for smooth and striated muscle. J Pharmacol Toxicol Methods. 2012;65(3):126–135. doi: 10.1016/j.vascn.2012.04.001
- Ingber DE. Developmentally inspired human ‘organs on chips’. Development. 2018;145(16):dev156125. doi: 10.1242/dev.156125
- Sone N, Konishi S, Igura K, et al. Multicellular modeling of ciliopathy by combining iPS cells and microfluidic airway-ona- chip technology. Sci Transl Med. 2021;13(601):eabb1298. doi: 10.1126/scitranslmed.abb1298
- Wang H, Yin F, Li Z, et al. Advances of microfluidic lung chips for assessing atmospheric pollutants exposure. Environ Int. 2023;172:107801. doi: 10.1016/j.envint.2023.107801
- Sengupta A, Roldan N, Kiener M, et al. A new immortalized human alveolar epithelial cell model to study lung injury and toxicity on a breathing lung-on-chip system. Front Toxicol. 2022;4:840606. doi: 10.3389/ftox.2022.840606
- Fisher CR, Mba Medie F, Luu RJ, et al. A high-throughput, high-containment human primary epithelial airway organ-onchip platform for SARS-CoV-2 therapeutic screening. Cells. 2023;12(22):2639. doi: 10.3390/cells12222639
- Sengupta A, Dorn A, Jamshidi M, et al. A multiplex inhalation platform to model in situ like aerosol delivery in a breathing lung-on-chip. Front Pharmacol. 2023;14:1114739. doi: 10.3389/fphar.2023.1114739
- Dey M, Ozbolat IT. 3D bioprinting of cells, tissues and organs. Sci Rep. 2020;10(1):14023. doi: 10.1038/s41598-020-70086-y
- Matai I, Kaur G, Seyedsalehi A, et al. Progress in 3D bioprinting technology for tissue/organ regenerative engineering. Biomaterials. 2020;226:119536. doi: 10.1016/j.biomaterials.2019.119536
- Kim W, Lee Y, Kang D, et al. 3D inkjet-bioprinted lungon- a-chip. ACS Biomater Sci Eng. 2023;9(5):2806–2815. doi: 10.1021/acsbiomaterials.3c00089
- Baptista D, Moreira Teixeira L, Barata D, et al. 3D lung-onchip model based on biomimetically microcurved culture membranes. ACS Biomater Sci Eng. 2022;8(6):2684–2699. doi: 10.1021/acsbiomaterials.1c01463
- Li K, Yang X, Xue C, et al. Biomimetic human lung-on-achip for modeling disease investigation. Biomicrofluidics. 2019;13(3):031501. doi: 10.1063/1.5100070
- Regehr KJ, Domenech M, Koepsel JT, et al. Biological implications of polydimethylsiloxane-based microfluidic cell culture. Lab Chip. 2009;9(15):2132–2139. doi: 10.1039/b903043c
- Jo BH, van Lerberghe LM, Motsegood KM, Beebe DJ. Threedimensional micro-channel fabrication in polydimethylsiloxane (PDMS) elastomer. J Microelectromech Syst. 2000;9(1):76–81. doi: 10.1109/84.825780
- Gaio N, van Meer B, Quirós Solano W, et al. Cytostretch, an organ-on-chip platform. Micromachines (Basel). 2016;7(7):120. doi: 10.3390/mi7070120
- Bennet TJ, Randhawa A, Hua J, Cheung KC. Airway-on-a-chip: Designs and applications for lung repair and disease. Cells. 2021;10(7):1602. doi: 10.3390/cells10071602
- Park SE, Georgescu A, Oh JM, et al. Polydopamine-based interfacial engineering of extracellular matrix hydrogels for the construction and long-term maintenance of living three-dimensional tissues. ACS Appl Mater Interfaces. 2019;11(27):23919–23925. doi: 10.1021/acsami.9b07912
- Toepke MW, Beebe DJ. PDMS absorption of small molecules and consequences in microfluidic applications. Lab Chip. 2006;6(12):1484–1486. doi: 10.1039/b612140c
- Wang JD, Douville NJ, Takayama S, El-Sayed M. Quantitative analysis of molecular absorption into PDMS microfluidic channels. Ann Biomed Eng. 2012;40(9):1862–1873. doi: 10.1007/s10439-012-0562-z
- Ingber DE. Human organs-on-chips for disease modelling, drug development and personalized medicine. Nat Rev Genet. 2022;23(8):467–491. doi: 10.1038/s41576-022-00466-9
- Ewart L, Apostolou A, Briggs S, et al. Qualifying a human liver-chip for predictive toxicology: Performance assessment and economic implications. bioRxiv. 2021. doi: 10.1101/2021.12.14.472674
- Van Meer BJ, de Vries H, Firth KS, et al. Small molecule absorption by PDMS in the context of drug response bioassays. Biochem Biophys Res Commun. 2017;482(2):323–328. doi: 10.1016/j.bbrc.2016.11.062
- Domansky K, Leslie DC, McKinney J, et al. Clear castable polyurethane elastomer for fabrication of microfluidic devices. Lab Chip. 2013;13(19):3956–3964. doi: 10.1039/c3lc50558h
- Domansky K, Sliz JD, Wen N, et al. SEBS elastomers for fabrication of microfluidic devices with reduced drug absorption by injection molding and extrusion. Microfluid Nanofluid. 2017;21(6):107. doi: 10.1007/s10404-017-1941-4
- Schneider S, Brás EJ, Schneider O, et al. Facile patterning of thermoplastic elastomers and robust bonding to glass and thermoplastics for microfluidic cell culture and organ-on-chip. Micromachines. 2021;12:575. doi: 10.3390/mi12050575
- Gomez-Sjoberg R, Leyrat AA, Houseman BT, et al. Biocompatibility and reduced drug absorption of solgel- treated poly (dimethyl siloxane) for microfluidic cell culture applications. Anal Chem. 2010;82(21):8954–8960. doi: 10.1021/ac101870s
- Sasaki H, Onoe H, Osaki T, et al. Sensors and actuators B: Chemical parylene-coating in PDMS microfluidic channels prevents the absorption of fluorescent dyes. Sensors Actuators B Chem. 2010;150(1):478–482. doi: 10.1016/j.snb.2010.07.021
- Sicard D, Haak AJ, Choi KM, et al. Aging and anatomical variations in lung tissue stiffness. Am J Physiol Lung Cell Mol Physiol. 2018;314(6):L946–L955. doi: 10.1152/ajplung.00415.2017
- Polio SR, Kundu AN, Dougan CE, et al. Cross-platform mechanical characterization of lung tissue. PLoS One. 2018;13(10):e0204765. doi: 10.1371/journal.pone.0204765
- Kumar V, Madhurakkat Perikamana SK, Tata A, et al. an in vitro microfluidic alveolus model to study lung biomechanics. Front Bioeng Biotechnol. 2022;10:848699. doi: 10.3389/fbioe.2022.848699
- Mohammed MI, Haswell S, Gibson I. Lab-on-a-chip or chip-in-alab: Challenges of commercialization lost in translation. Procedia Technol. 2015;20:54–59. doi: 10.1016/j.protcy.2015.07.010
- Ongaro AE, di Giuseppe D, Kermanizadeh A, et al. Polylactic is a sustainable, low absorption, low autofluorescence alternative to other plastics for microfluidic and organon- chip applications. Anal Chem. 2020;92(9):6693–6701. doi: 10.1021/acs.analchem.0c00651
- Guan M, Tang S, Chang H, et al. Development of alveolarcapillary- exchange (ACE) chip and its application for assessment of PM2.5-induced toxicity. Ecotoxicol Environ Saf. 2021;223:112601. doi: 10.1016/j.ecoenv.2021.112601
- Mejias JC, Nelson MR, Liseth O, Roy K. A 96-well format microvascularized human lung-on-a-chip platform for microphysiological modeling of fibrotic diseases. Lab Chip. 2020;20(19):3601–3611. doi: 10.1039/d0lc00644k
- Carius P, Dubois A, Ajdarirad M, et al. PerfuPul-A versatile perfusable platform to assess permeability and barrier function of air exposed pulmonary epithelia. Front Bioeng Biotechnol. 2021;9:743236. doi: 10.3389/fbioe.2021.743236
- Grigoriev TE, Bukharova TB, Vasilyev AV, et al. Effect of molecular characteristics and morphology on mechanical performance and biocompatibility of PLA-based spongious scaffolds. BioNanoSci. 2018;8(4):977–983. doi: 10.1007/s12668-018-0557-9
- Da Silva D, Kaduri M, Poley M, et al. Biocompatibility, biodegradation and excretion of polylactic acid (PLA) in medical implants and theranostic systems. Chem Eng J. 2018;340:9–14. doi: 10.1016/j.cej.2018.01.010
- Ramot Y, Haim-Zada M, Domb AJ, Nyska A. Biocompatibility and safety of PLA and its copolymers. Adv Drug Deliv Rev. 2016;107:153–162. doi: 10.1016/j.addr.2016.03.012
- Yang X, Li K, Zhang X, et al. Nanofiber membrane supported lung-on-a-chip microdevice for anti-cancer drug testing. Lab Chip. 2018;18(3):486–495. doi: 10.1039/c7lc01224a
- Li W, Sun X, Ji B, et al. PLGA Nanofiber/PDMS microporous composite membrane-sandwiched microchip for drug testing. Micromachines (Basel). 2020;11(12):1054. doi: 10.3390/mi11121054
- Carlborg CF, Haraldsson T, Öberg K, et al. Beyond PDMS: Off-stoichiometry thiol-ene (OSTE) based soft lithography for rapid prototyping of microfluidic devices. Lab Chip. 2011;11(18):3136–3147. doi: 10.1039/c1lc20388f
- Rimsa R, Galvanovskis A, Plume J, et al. Lung on a chip development from off-stoichiometry thiol-ene polymer. Micromachines (Basel). 2021;12(5):546. doi: 10.3390/mi12050546
- Huang D, Liu T, Liao J, et al. Reversed-engineered human alveolar lung-on-a-chip model. Proc Natl Acad Sci USA. 2021;118(19):e2016146118. doi: 10.1073/pnas.2016146118
- Ochs M, Nyengaard JR, Jung A, et al. The number of alveoli in the human lung. Am J Respir Crit Care Med. 2004;169(1):120–124. doi: 10.1164/rccm.200308-1107OC
- Zamprogno P, Wüthrich S, Achenbach S, et al. Secondgeneration lung-on-a-chip with an array of stretchable alveoli made with a biological membrane. Commun Biol. 2021;4(1):168. doi: 10.1038/s42003-021-01695-0
- Park S, Newton J, Hidjir T, Young EW. Bidirectional airflow in lung airway-on-a-chip with matrix-derived membrane elicits epithelial glycocalyx formation. Lab Chip. 2023;23(16): 3671–3682. doi: 10.1039/d3lc00259d
- Licciardello M, Traldi C, Cicolini M, et al. A miniaturized multicellular platform to mimic the 3D structure of the alveolarcapillary barrier. Frontiers Bioeng Biotechnol. 2024;12:1346660. doi: 10.3389/fbioe.2024.1346660
- Carvalho M, Ribeiro V, Caballero D, et al. Biomimetic and soft lab-on-a-chip platform based on enzymatic-crosslinked silk fibroin hydrogel for colorectal tumor model. Authorea. 2022. doi: 10.22541/au.167232609.96998643/v1
- Богуш В.Г., Давыдова Л.И., Шуляков В.С., и др. Разработка биоадгезивов на основе рекомбинантных аналогов белков паутины // Биотехнология. 2021. Т. 37, № 2. С. 20–33. [Bogush VG, Davydova LI, Shulyakov VS, et al. Development of bioadhesives based on recombinant analogues of spider web proteins. Biotechnology. 2021;37(2):20–33]. EDN: ZHUEDS doi: 10.21519/0234-2758-2021-37-2-20-33
- Zhang Y, Wang X, Yang Y, et al. Recapitulating essential pathophysiological characteristics in lung-on-a-chip for disease studies. Front Immunol. 2023;14:1093460. doi: 10.3389/fimmu.2023.1093460
- Moreira A, Müller M, Costa PF, Kohl Y. Advanced in vitro lung models for drug and toxicity screening: The promising role of induced pluripotent stem cells. Adv Biol (Weinh). 2022;6(2):e2101139. doi: 10.1002/adbi.202101139
- Nawroth JC, Roth D, van Schadewijk A, et al. Breathing on chip: Dynamic flow and stretch accelerate mucociliary maturation of airway epithelium in vitro. Mater Today Bio. 2023;21:100713. doi: 10.1016/j.mtbio.2023.100713
- Heinen N, Klöhn M, Steinmann E, Pfaender S. In vitro lung models and their application to study SARS-CoV-2 pathogenesis and disease. Viruses. 2021;13(5):792. doi: 10.3390/v13050792
- Petpiroon N, Netkueakul W, Sukrak K, et al. Development of lung tissue models and their applications. Life Sci. 2023;334:122208. doi: 10.1016/j.lfs.2023.122208
- Damle EB, Yamaguchi E, Yao JE, Gaver DP. Preparation and structural evaluation of epithelial cell monolayers in a physiologically sized microfluidic culture device. J Vis Exp. 2022;(185):10.3791/64148. doi: 10.3791/64148
- Maoz BM, Herland A, Henry OY, et al. Organs-on-chips with combined multi-electrode array and transepithelial electrical resistance measurement capabilities. Lab Chip. 2017;17:2294–2302. doi: 10.1039/c7lc00412e
- Niemeyer BF, Zhao P, Tuder RM, Benam KH. Advanced microengineered lung models for translational drug discovery. SLAS Discov. 2018;23(8):777–789. doi: 10.1177/2472555218760217
- Yousafzai MS, Hammer JA. Using biosensors to study organoids, spheroids and organs-on-a-chip: A mechanobiology perspective. Biosensors (Basel). 2023;13(10):905. doi: 10.3390/bios13100905
- Kwon A, Lee NY, Yu JH, et al. Mitochondrial stress activates YAP/TAZ through RhoA oxidation to promote liver injury. Cell Death Dis. 2024;15(1):51. doi: 10.1038/s41419-024-06448-5
- Smirnov AV, Anisimkin VI, Shamsutdinova ES, et al. Acoustiс waves in piezoelectric layered structure for selective detection of liqiod viscosity. Sensors. 2023;23:7329. doi: 10.3390/s23177329
- Lee SH, Cha B, Yi HG, et al. Acoustofluidic separation of bacteria from platelets using tilted-angle standing surface acoustic wave. Sensors Actuators B Chem. 2024;417:136161. doi: 10.1016/j.snb.2024.136161
- Vachon P, Merugu S, Sharma J, et al. Cavity-agnostic acoustofluidic manipulations enabled by guided flexural waves on a membrane acoustic waveguide actuator. Microsystems Nanoengineering. 2024;10(1):33. doi: 10.1038/s41378-023-00643-8
- Huh D, Leslie DC, Matthews BD, et al. A human disease model of drug toxicity-induced pulmonary edema in a lung-on-a-chip microdevice. Sci Transl Med. 2012;4(159):159ra147. Erratum in: Sci Transl Med. 2018;10(449). doi: 10.1126/scitranslmed.3004249
- Jain A, Barrile R, van der Meer AD, et al. Primary human lung alveolus-on-a-chip model of intravascular thrombosis for assessment of therapeutics. Clin Pharmacol Ther. 2018; 103(2):332–340. doi: 10.1002/cpt.742
- Hassell BA, Goyal G, Lee E, et al. Human organ chip models recapitulate orthotopic lung cancer growth, therapeutic responses, and tumor dormancy in vitro. Cell Rep. 2017; 21(2):508–516. doi: 10.1016/j.celrep.2017.09.043
- Benam KH, Villenave R, Lucchesi C, et al. Small airway-ona- chip enables analysis of human lung inflammation and drug responses in vitro. Nat Methods. 2016;13(2):151–157. doi: 10.1038/nmeth.3697
- Si L, Bai H, Rodas M, et al. A human-airway-on-a-chip for the rapid identification of candidate antiviral therapeutics and prophylactics. Nat Biomed Eng. 2021;5(8):815–829. doi: 10.1038/s41551-021-00718-9
- Danahay H, Pessotti AD, Coote J, et al. Notch2 is required for inflammatory cytokine-driven goblet cell metaplasia in the lung. Cell Rep. 2015;10(2):239–252. doi: 10.1016/j.celrep.2014.12.017
- Ordonez CL, Khashayar R, Wong HH, et al. Mild and moderate asthma is associated with airway goblet cell hyperplasia and abnormalities in mucin gene expression. Am J Respir Crit Care Med. 2001;163(2):517–523. doi: 10.1164/ajrccm.163.2.2004039
- Papi A, Bellettato CM, Braccioni F, et al. Infections and airway inflammation in chronic obstructive pulmonary disease severe exacerbations. Am J Respir Crit Care Med. 2006;173(10):1114–1121. doi: 10.1164/rccm.200506-859OC
- Jain A, van der Meer A, Papa AL, et al. Assessment of whole blood thrombosis in a microfluidic device lined by fixed human endothelium. Biomed Microdevices. 2016;18(4):73. doi: 10.1007/s10544-016-0095-6
- Сooley BC. In vivo fluorescence imaging of large-vessel thrombosis in mice. Arterioscler Thromb Vasc Biol. 2011;31(6):1351–1356. doi: 10.1161/ATVBAHA.111.225334
- Li L, Bo W, Wang G, et al. Progress and application of lung-on-a-chip for lung cancer. Front Bioeng Biotechnol. 2024;12:1378299. doi: 10.3389/fbioe.2024.1378299
- Assaad S, Kratzert WB, Shelley B, et al. Assessment of pulmonary edema: Principles and practice. J Cardiothorac Vasc Anesth. 2018;32(2):901–914. doi: 10.1053/j.jvca.2017.08.028
- Melencio L, McKallip RJ, Guan H, et al. Role of CD4(+)CD25(+) T regulatory cells in IL-2-induced vascular leak. Int Immunol. 2006;18(10):1461–1471. doi: 10.1093/intimm/dxl079
- Batool I, Qadir A, Levermore JM, Kelly FJ. Dynamics of airborne microplastics, appraisal and distributional behaviour in atmosphere: A review. Sci Total Environ. 2022;806(Pt 4):150745. doi: 10.1016/j.scitotenv.2021.150745
- Tokunaga Y, Okochi H, Tani Y, et al. Airborne microplastics detected in the lungs of wild birds in Japan. Chemosphere. 2023;321:138032. doi: 10.1016/j.chemosphere.2023.138032
- Baeza-Martínez C, Olmos S, González-Pleiter M, et al. First evidence of microplastics isolated in European citizens’ lower airway. J Hazard Mater. 2022;438:129439. doi: 10.1016/j.jhazmat.2022.129439
- Yang S, Zhang T, Ge Y, et al. Sentinel supervised lung-on-achip: A new environmental toxicology platform for nanoplasticinduced lung injury. J Hazard Mater. 2023;458:131962. doi: 10.1016/j.jhazmat.2023.131962
- Zhang M, Xu C, Jiang L, Qin J. A 3D human lung-on-achip model for nanotoxicity testing. Toxicol Res (Camb). 2018;7(6):1048–1060. doi: 10.1039/c8tx00156a
- Van den Berg A, Mummery CL, Passier R, et al. Personalised organs-on-chips: Functional testing for precision medicine. Lab Chip. 2019;19(2):198–205. doi: 10.1039/c8lc00827b