Microfluidic Organ-on-a-Chip System for Disease Modeling and Drug Development
Abstract
:1. Introduction
2. Design of an Organ-on-a-Chip
2.1. Lung-on-a-Chip
2.2. Liver-on-a-Chip
2.3. Gut-on-a-Chip
2.4. BBB-on-a-Chip
2.5. Heart-on-a-Chip
2.6. Skin-on-a-Chip
2.7. Other Single Organs-on-a-Chip
2.8. Multi-Organ-on-Chip Systems
3. Applications
3.1. Pharmacokinetic (PK) and Pharmacodynamic (PD) Analysis
3.2. Nano-Medicine
3.3. Dynamic Monitoring for Disease Modeling
3.4. Safety Assessment/Toxicity Evaluation
4. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Mitchell, P. Microfluidics—Downsizing large-scale biology. Nat. Biotechnol. 2001, 19, 717–721. [Google Scholar] [CrossRef] [PubMed]
- Mark, D.; Haeberle, S.; Roth, G.; von Stetten, F.; Zengerle, R. Microfluidic lab-on-a-chip platforms: Requirements, characteristics and applications. Chem. Soc. Rev. 2010, 39, 1153–1182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kartalov, E.P.; Scherer, A.; Quake, S.R.; Taylor, C.R.; Anderson, W.F. Experimentally validated quantitative linear model for the device physics of elastomeric microfluidic valves. J. Appl. Phys. 2007, 101, 64505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thorsen, T.A. Microfluidic tools for high-throughput screening. Biotechniques 2004, 36, 197–199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Manz, A.; Harrison, D.J.; Verpoorte, E.M.J.; Fettinger, J.C.; Paulus, A.; Lüdi, H.; Widmer, H.M. Planar chips technology for miniaturization and integration of separation techniques into monitoring systems. J. Chromatogr. A 1992, 593, 253–258. [Google Scholar] [CrossRef]
- Thorsen, T.; Maerkl, S.J.; Quake, S.R. Microfluidic large-scale integration. Science 2002, 298, 580–584. [Google Scholar] [CrossRef] [Green Version]
- Sung Kwon, C.; Hyejin, M.; Chang-Jin, K. Creating, transporting, cutting, and merging liquid droplets by electrowetting-based actuation for digital microfluidic circuits. J. Microelectromech. Syst. 2003, 12, 70–80. [Google Scholar] [CrossRef] [Green Version]
- Huh, D.; Matthews, B.D.; Mammoto, A.; Montoya-Zavala, M.; Hsin, H.Y.; Ingber, D.E. Reconstituting organ-level lung functions on a chip. Science 2010, 328, 1662–1668. [Google Scholar] [CrossRef] [Green Version]
- Zhang, L.; Ding, B.; Chen, Q.; Feng, Q.; Lin, L.; Sun, J. Point-of-care-testing of nucleic acids by microfluidics. TrAC Trends Anal. Chem. 2017, 94, 106–116. [Google Scholar] [CrossRef]
- Koh, A.; Kang, D.; Xue, Y.; Lee, S.; Pielak, R.M.; Kim, J.; Hwang, T.; Min, S.; Banks, A.; Bastien, P.; et al. A soft, wearable microfluidic device for the capture, storage, and colorimetric sensing of sweat. Sci. Transl. Med. 2016, 8, 366ra165. [Google Scholar] [CrossRef] [Green Version]
- Shehab, N.; Lovegrove, M.C.; Geller, A.I.; Rose, K.O.; Weidle, N.J.; Budnitz, D.S. US Emergency Department Visits for Outpatient Adverse Drug Events, 2013–2014. JAMA 2016, 316, 2115–2125. [Google Scholar] [CrossRef] [PubMed]
- Shanks, N.; Greek, R.; Greek, J. Are animal models predictive for humans? Philos. Ethics Humanit. Med. 2009, 4, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McLean, I.C.; Schwerdtfeger, L.A.; Tobet, S.A.; Henry, C.S. Powering ex vivo tissue models in microfluidic systems. Lab Chip 2018, 18, 1399–1410. [Google Scholar] [CrossRef] [PubMed]
- Zheng, F.; Fu, F.; Cheng, Y.; Wang, C.; Zhao, Y.; Gu, Z. Organ-on-a-Chip Systems: Microengineering to Biomimic Living Systems. Small 2016, 12, 2253–2282. [Google Scholar] [CrossRef] [PubMed]
- McConnell, H.M.; Owicki, J.C.; Parce, J.W.; Miller, D.L.; Baxter, G.T.; Wada, H.G.; Pitchford, S. The cytosensor microphysiometer: Biological applications of silicon technology. Science 1992, 257, 1906–1912. [Google Scholar] [CrossRef] [PubMed]
- Park, T.H.; Shuler, M.L. Integration of cell culture and microfabrication technology. Biotechnol. Prog. 2003, 19, 243–253. [Google Scholar] [CrossRef] [PubMed]
- Esch, M.B.; King, T.L.; Shuler, M.L. The role of body-on-a-chip devices in drug and toxicity studies. Annu. Rev. Biomed. Eng. 2011, 13, 55–72. [Google Scholar] [CrossRef] [Green Version]
- Nakao, Y.; Kimura, H.; Sakai, Y.; Fujii, T. Bile canaliculi formation by aligning rat primary hepatocytes in a microfluidic device. Biomicrofluidics 2011, 5, 22212. [Google Scholar] [CrossRef] [Green Version]
- Jang, K.J.; Mehr, A.P.; Hamilton, G.A.; McPartlin, L.A.; Chung, S.; Suh, K.Y.; Ingber, D.E. Human kidney proximal tubule-on-a-chip for drug transport and nephrotoxicity assessment. Integr. Biol. 2013, 5, 1119–1129. [Google Scholar] [CrossRef]
- An, F.; Qu, Y.; Luo, Y.; Fang, N.; Liu, Y.; Gao, Z.; Zhao, W.; Lin, B. A Laminated Microfluidic Device for Comprehensive Preclinical Testing in the Drug ADME Process. Sci. Rep. 2016, 6, 25022. [Google Scholar] [CrossRef] [Green Version]
- Ma, Z.; Wang, J.; Loskill, P.; Huebsch, N.; Koo, S.; Svedlund, F.L.; Marks, N.C.; Hua, E.W.; Grigoropoulos, C.P.; Conklin, B.R.; et al. Self-organizing human cardiac microchambers mediated by geometric confinement. Nat. Commun. 2015, 6, 7413. [Google Scholar] [CrossRef] [Green Version]
- Kim, H.J.; Huh, D.; Hamilton, G.; Ingber, D.E. Human gut-on-a-chip inhabited by microbial flora that experiences intestinal peristalsis-like motions and flow. Lab Chip 2012, 12, 2165–2174. [Google Scholar] [CrossRef] [PubMed]
- Franco, C.; Gerhardt, H. Tissue engineering: Blood vessels on a chip. Nature 2012, 488, 465–466. [Google Scholar] [CrossRef] [PubMed]
- Atac, B.; Wagner, I.; Horland, R.; Lauster, R.; Marx, U.; Tonevitsky, A.G.; Azar, R.P.; Lindner, G. Skin and hair on-a-chip: In vitro skin models versus ex vivo tissue maintenance with dynamic perfusion. Lab Chip 2013, 13, 3555–3561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Torisawa, Y.S.; Spina, C.S.; Mammoto, T.; Mammoto, A.; Weaver, J.C.; Tat, T.; Collins, J.J.; Ingber, D.E. Bone marrow-on-a-chip replicates hematopoietic niche physiology in vitro. Nat. Methods 2014, 11, 663–669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Griep, L.M.; Wolbers, F.; de Wagenaar, B.; ter Braak, P.M.; Weksler, B.B.; Romero, I.A.; Couraud, P.O.; Vermes, I.; van der Meer, A.D.; van den Berg, A. BBB on chip: Microfluidic platform to mechanically and biochemically modulate blood-brain barrier function. Biomed. Microdevices 2013, 15, 145–150. [Google Scholar] [CrossRef]
- Huh, D.; Kim, H.J.; Fraser, J.P.; Shea, D.E.; Khan, M.; Bahinski, A.; Hamilton, G.A.; Ingber, D.E. Microfabrication of human organs-on-chips. Nat. Protoc. 2013, 8, 2135–2157. [Google Scholar] [CrossRef]
- Park, S.E.; Georgescu, A.; Huh, D. Organoids-on-a-chip. Science 2019, 364, 960–965. [Google Scholar] [CrossRef]
- Lee, S.H.; Jun, B.-H. Advances in dynamic microphysiological organ-on-a-chip: Design principle and its biomedical application. J. Ind. Eng. Chem. 2019, 71, 65–77. [Google Scholar] [CrossRef]
- Mejías, J.C.; Nelson, M.R.; 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, 3601–3611. [Google Scholar] [CrossRef]
- Zhang, M.; Xu, C.; Jiang, L.; Qin, J. A 3D human lung-on-a-chip model for nanotoxicity testing. Toxicol. Res. 2018, 7, 1048–1060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zamprogno, P.; Wuthrich, S.; Achenbach, S.; Thoma, G.; Stucki, J.D.; Hobi, N.; Schneider-Daum, N.; Lehr, C.M.; Huwer, H.; Geiser, T.; et al. Second-generation lung-on-a-chip with an array of stretchable alveoli made with a biological membrane. Commun. Biol. 2021, 4, 168. [Google Scholar] [CrossRef] [PubMed]
- Gard, A.L.; Luu, R.J.; Miller, C.R.; Maloney, R.; Cain, B.P.; Marr, E.E.; Burns, D.M.; Gaibler, R.; Mulhern, T.J.; Wong, C.A.; et al. High-throughput human primary cell-based airway model for evaluating influenza, coronavirus, or other respiratory viruses in vitro. Sci. Rep. 2021, 11, 14961. [Google Scholar] [CrossRef] [PubMed]
- Toh, Y.C.; Lim, T.C.; Tai, D.; Xiao, G.; van Noort, D.; Yu, H. A microfluidic 3D hepatocyte chip for drug toxicity testing. Lab Chip 2009, 9, 2026–2035. [Google Scholar] [CrossRef]
- Du, Y.; Li, N.; Yang, H.; Luo, C.; Gong, Y.; Tong, C.; Gao, Y.; Lu, S.; Long, M. Mimicking liver sinusoidal structures and functions using a 3D-configured microfluidic chip. Lab Chip 2017, 17, 782–794. [Google Scholar] [CrossRef] [Green Version]
- Freag, M.S.; Namgung, B.; Reyna Fernandez, M.E.; Gherardi, E.; Sengupta, S.; Jang, H.L. Human Nonalcoholic Steatohepatitis on a Chip. Hepatol. Commun. 2020, 5, 217–233. [Google Scholar] [CrossRef]
- Shim, K.Y.; Lee, D.; Han, J.; Nguyen, N.T.; Park, S.; Sung, J.H. Microfluidic gut-on-a-chip with three-dimensional villi structure. Biomed. Microdevices 2017, 19, 37. [Google Scholar] [CrossRef] [Green Version]
- Henry, O.Y.F.; Villenave, R.; Cronce, M.J.; Leineweber, W.D.; Benz, M.A.; Ingber, D.E. Organs-on-chips with integrated electrodes for trans-epithelial electrical resistance (TEER) measurements of human epithelial barrier function. Lab Chip 2017, 17, 2264–2271. [Google Scholar] [CrossRef]
- Jing, B.; Wang, Z.A.; Zhang, C.; Deng, Q.; Wei, J.; Luo, Y.; Zhang, X.; Li, J.; Du, Y. Establishment and Application of Peristaltic Human Gut-Vessel Microsystem for Studying Host-Microbial Interaction. Front. Bioeng. Biotechnol. 2020, 8, 272. [Google Scholar] [CrossRef]
- Wang, Y.I.; Abaci, H.E.; Shuler, M.L. Microfluidic blood-brain barrier model provides in vivo-like barrier properties for drug permeability screening. Biotechnol. Bioeng. 2017, 114, 184–194. [Google Scholar] [CrossRef]
- Wevers, N.R.; Kasi, D.G.; Gray, T.; Wilschut, K.J.; Smith, B.; van Vught, R.; Shimizu, F.; Sano, Y.; Kanda, T.; Marsh, G.; et al. A perfused human blood-brain barrier on-a-chip for high-throughput assessment of barrier function and antibody transport. Fluids Barriers CNS 2018, 15, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Papademetriou, I.; Vedula, E.; Charest, J.; Porter, T. Effect of flow on targeting and penetration of angiopep-decorated nanoparticles in a microfluidic model blood-brain barrier. PLoS ONE 2018, 13, e0205158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marsano, A.; Conficconi, C.; Lemme, M.; Occhetta, P.; Gaudiello, E.; Votta, E.; Cerino, G.; Redaelli, A.; Rasponi, M. Beating heart on a chip: A novel microfluidic platform to generate functional 3D cardiac microtissues. Lab Chip 2016, 16, 599–610. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.S.; Arneri, A.; Bersini, S.; Shin, S.R.; Zhu, K.; Goli-Malekabadi, Z.; Aleman, J.; Colosi, C.; Busignani, F.; Dell’Erba, V.; et al. Bioprinting 3D microfibrous scaffolds for engineering endothelialized myocardium and heart-on-a-chip. Biomaterials 2016, 110, 45–59. [Google Scholar] [CrossRef] [Green Version]
- Faulkner-Jones, A.; Zamora, V.; Hortigon-Vinagre, M.P.; Wang, W.; Ardron, M.; Smith, G.L.; Shu, W. A Bioprinted Heart-on-a-Chip with Human Pluripotent Stem Cell-Derived Cardiomyocytes for Drug Evaluation. Bioengineering 2022, 9, 32. [Google Scholar] [CrossRef]
- Jayne, R.K.; Karakan, M.C.; Zhang, K.; Pierce, N.; Michas, C.; Bishop, D.J.; Chen, C.S.; Ekinci, K.L.; White, A.E. Direct laser writing for cardiac tissue engineering: A microfluidic heart on a chip with integrated transducers. Lab Chip 2021, 21, 1724–1737. [Google Scholar] [CrossRef]
- Jeon, H.M.; Kim, K.; Choi, K.C.; Sung, G.Y. Side-effect test of sorafenib using 3-D skin equivalent based on microfluidic skin-on-a-chip. J. Ind. Eng. Chem. 2020, 82, 71–80. [Google Scholar] [CrossRef]
- Kim, K.; Jeon, H.M.; Choi, K.C.; Sung, G.Y. Testing the Effectiveness of Curcuma longa Leaf Extract on a Skin Equivalent Using a Pumpless Skin-on-a-Chip Model. Int. J. Mol. Sci. 2020, 21, 3898. [Google Scholar] [CrossRef]
- Zhang, J.; Chen, Z.; Zhang, Y.; Wang, X.; Ouyang, J.; Zhu, J.; Yan, Y.; Sun, X.; Wang, F.; Li, X.; et al. Construction of a high fidelity epidermis-on-a-chip for scalable in vitro irritation evaluation. Lab Chip 2021, 21, 3804–3818. [Google Scholar] [CrossRef]
- Llenas, M.; Paoli, R.; Feiner-Gracia, N.; Albertazzi, L.; Samitier, J.; Caballero, D. Versatile Vessel-on-a-Chip Platform for Studying Key Features of Blood Vascular Tumors. Bioengineering 2021, 8, 81. [Google Scholar] [CrossRef]
- Abudupataer, M.; Chen, N.; Yan, S.; Alam, F.; Shi, Y.; Wang, L.; Lai, H.; Li, J.; Zhu, K.; Wang, C. Bioprinting a 3D vascular construct for engineering a vessel-on-a-chip. Biomed. Microdevices 2019, 22, 10. [Google Scholar] [CrossRef] [PubMed]
- Zhou, K.; Dey, M.; Ayan, B.; Zhang, Z.; Ozbolat, V.; Kim, M.H.; Khristov, V.; Ozbolat, I.T. Fabrication of PDMS microfluidic devices using nanoclay-reinforced Pluronic F-127 as a sacrificial ink. Biomed. Mater. 2021, 16, 045005. [Google Scholar] [CrossRef] [PubMed]
- Tian, H.; Pang, J.; Qin, K.; Yuan, W.; Kong, J.; Ma, H.; He, J.; Yang, X.; Luo, Y.; Lu, Y.; et al. A Novel Tissue-Based Liver-Kidney-on-a-Chip Can Mimic Liver Tropism of Extracellular Vesicles Derived from Breast Cancer Cells. Biotechnol. J. 2020, 15, e1900107. [Google Scholar] [CrossRef] [PubMed]
- Vormann, M.K.; Tool, L.M.; Ohbuchi, M.; Gijzen, L.; van Vught, R.; Hankemeier, T.; Kiyonaga, F.; Kawabe, T.; Goto, T.; Fujimori, A.; et al. Modelling and Prevention of Acute Kidney Injury through Ischemia and Reperfusion in a Combined Human Renal Proximal Tubule/Blood Vessel-on-a-Chip. Kidney360 2022, 3, 217–231. [Google Scholar] [CrossRef] [PubMed]
- Selahi, A.; Fernando, T.; Chakraborty, S.; Muthuchamy, M.; Zawieja, D.C.; Jain, A. Lymphangion-chip: A microphysiological system which supports co-culture and bidirectional signaling of lymphatic endothelial and muscle cells. Lab Chip 2021, 22, 121–135. [Google Scholar] [CrossRef]
- van Meer, B.J.; de Vries, H.; Firth, K.S.A.; van Weerd, J.; Tertoolen, L.G.J.; Karperien, H.B.J.; Jonkheijm, P.; Denning, C.; AP, I.J.; Mummery, C.L. Small molecule absorption by PDMS in the context of drug response bioassays. Biochem. Biophys. Res. Commun. 2017, 482, 323–328. [Google Scholar] [CrossRef] [Green Version]
- Zhou, Y.; Horowitz, J.C.; Naba, A.; Ambalavanan, N.; Atabai, K.; Balestrini, J.; Bitterman, P.B.; Corley, R.A.; Ding, B.S.; Engler, A.J.; et al. Extracellular matrix in lung development, homeostasis and disease. Matrix Biol. 2018, 73, 77–104. [Google Scholar] [CrossRef] [Green Version]
- Xu, C.; Zhang, M.; Chen, W.; Jiang, L.; Chen, C.; Qin, J. Assessment of Air Pollutant PM2.5 Pulmonary Exposure Using a 3D Lung-on-Chip Model. ACS Biomater. Sci. Eng. 2020, 6, 3081–3090. [Google Scholar] [CrossRef]
- Moradi, E.; Jalili-Firoozinezhad, S.; Solati-Hashjin, M. Microfluidic organ-on-a-chip models of human liver tissue. Acta Biomater. 2020, 116, 67–83. [Google Scholar] [CrossRef]
- Younossi, Z.M.; Koenig, A.B.; Abdelatif, D.; Fazel, Y.; Henry, L.; Wymer, M. Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 2016, 64, 73–84. [Google Scholar] [CrossRef] [Green Version]
- Hassan, S.; Sebastian, S.; Maharjan, S.; Lesha, A.; Carpenter, A.M.; Liu, X.; Xie, X.; Livermore, C.; Zhang, Y.S.; Zarrinpar, A. Liver-on-a-Chip Models of Fatty Liver Disease. Hepatology 2020, 71, 733–740. [Google Scholar] [CrossRef] [PubMed]
- Jeon, J.W.; Lee, S.H.; Kim, D.; Sung, J.H. In vitro hepatic steatosis model based on gut-liver-on-a-chip. Biotechnol. Prog. 2021, 37, e3121. [Google Scholar] [CrossRef] [PubMed]
- Chen, W.L.K.; Edington, C.; Suter, E.; Yu, J.; Velazquez, J.J.; Velazquez, J.G.; Shockley, M.; Large, E.M.; Venkataramanan, R.; Hughes, D.J.; et al. Integrated gut/liver microphysiological systems elucidates inflammatory inter-tissue crosstalk. Biotechnol. Bioeng. 2017, 114, 2648–2659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, S.Y.; Sung, J.H. Gut–liver on a chip toward an in vitro model of hepatic steatosis. Biotechnol. Bioeng. 2018, 115, 2817–2827. [Google Scholar] [CrossRef]
- Valles-Colomer, M.; Falony, G.; Darzi, Y.; Tigchelaar, E.F.; Wang, J.; Tito, R.Y.; Schiweck, C.; Kurilshikov, A.; Joossens, M.; Wijmenga, C.; et al. The neuroactive potential of the human gut microbiota in quality of life and depression. Nat. Microbiol. 2019, 4, 623–632. [Google Scholar] [CrossRef]
- van Schaik, W. The human gut resistome. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2015, 370, 20140087. [Google Scholar] [CrossRef] [Green Version]
- Compare, D.; Coccoli, P.; Rocco, A.; Nardone, O.M.; De Maria, S.; Carteni, M.; Nardone, G. Gut--liver axis: The impact of gut microbiota on non alcoholic fatty liver disease. Nutr. Metab. Cardiovasc. Dis. 2012, 22, 471–476. [Google Scholar] [CrossRef]
- Kim, H.J.; Ingber, D.E. Gut-on-a-Chip microenvironment induces human intestinal cells to undergo villus differentiation. Integr. Biol. 2013, 5, 1130–1140. [Google Scholar] [CrossRef] [Green Version]
- Sung, J.H.; Yu, J.; Luo, D.; Shuler, M.L.; March, J.C. Microscale 3-D hydrogel scaffold for biomimetic gastrointestinal (GI) tract model. Lab Chip 2011, 11, 389–392. [Google Scholar] [CrossRef]
- Benson, K.; Cramer, S.; Galla, H.J. Impedance-based cell monitoring: Barrier properties and beyond. Fluids Barriers CNS 2013, 10, 5. [Google Scholar] [CrossRef] [Green Version]
- Thuenauer, R.; Rodriguez-Boulan, E.; Romer, W. Microfluidic approaches for epithelial cell layer culture and characterisation. Analyst 2014, 139, 3206–3218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- van der Helm, M.W.; Henry, O.Y.F.; Bein, A.; Hamkins-Indik, T.; Cronce, M.J.; Leineweber, W.D.; Odijk, M.; van der Meer, A.D.; Eijkel, J.C.T.; Ingber, D.E.; et al. Non-invasive sensing of transepithelial barrier function and tissue differentiation in organs-on-chips using impedance spectroscopy. Lab Chip 2019, 19, 452–463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hawkins, B.T.; Davis, T.P. The blood-brain barrier/neurovascular unit in health and disease. Pharm. Rev. 2005, 57, 173–185. [Google Scholar] [CrossRef] [PubMed]
- Cardoso, F.L.; Brites, D.; Brito, M.A. Looking at the blood-brain barrier: Molecular anatomy and possible investigation approaches. Brain Res. Rev. 2010, 64, 328–363. [Google Scholar] [CrossRef]
- Kim, J.A.; Kim, H.N.; Im, S.K.; Chung, S.; Kang, J.Y.; Choi, N. Collagen-based brain microvasculature model in vitro using three-dimensional printed template. Biomicrofluidics 2015, 9, 024115. [Google Scholar] [CrossRef] [Green Version]
- Abbott, N.J.; Ronnback, L.; Hansson, E. Astrocyte-endothelial interactions at the blood-brain barrier. Nat. Rev. Neurosci. 2006, 7, 41–53. [Google Scholar] [CrossRef]
- Jeong, S.; Kim, S.; Buonocore, J.; Park, J.; Welsh, C.J.; Li, J.; Han, A. A Three-Dimensional Arrayed Microfluidic Blood-Brain Barrier Model With Integrated Electrical Sensor Array. IEEE Trans. Biomed. Eng. 2018, 65, 431–439. [Google Scholar] [CrossRef]
- Pardridge, W.M. Drug transport across the blood-brain barrier. J. Cereb. Blood Flow. Metab. 2012, 32, 1959–1972. [Google Scholar] [CrossRef]
- Park, T.E.; Mustafaoglu, N.; Herland, A.; Hasselkus, R.; Mannix, R.; FitzGerald, E.A.; Prantil-Baun, R.; Watters, A.; Henry, O.; Benz, M.; et al. Hypoxia-enhanced Blood-Brain Barrier Chip recapitulates human barrier function and shuttling of drugs and antibodies. Nat. Commun. 2019, 10, 2621. [Google Scholar] [CrossRef]
- Hirose, K.; Payumo, A.Y.; Cutie, S.; Hoang, A.; Zhang, H.; Guyot, R.; Lunn, D.; Bigley, R.B.; Yu, H.; Wang, J.; et al. Evidence for hormonal control of heart regenerative capacity during endothermy acquisition. Science 2019, 364, 184–188. [Google Scholar] [CrossRef]
- Benjamin, E.J.; Muntner, P.; Alonso, A.; Bittencourt, M.S.; Callaway, C.W.; Carson, A.P.; Chamberlain, A.M.; Chang, A.R.; Cheng, S.; Das, S.R.; et al. Heart Disease and Stroke Statistics-2019 Update: A Report From the American Heart Association. Circulation 2019, 139, e56–e528. [Google Scholar] [CrossRef] [PubMed]
- Visone, R.; Gilardi, M.; Marsano, A.; Rasponi, M.; Bersini, S.; Moretti, M. Cardiac Meets Skeletal: What’s New in Microfluidic Models for Muscle Tissue Engineering. Molecules 2016, 21, 1128. [Google Scholar] [CrossRef] [PubMed]
- Grosberg, A.; Alford, P.W.; McCain, M.L.; Parker, K.K. Ensembles of engineered cardiac tissues for physiological and pharmacological study: Heart on a chip. Lab Chip 2011, 11, 4165–4173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Agarwal, A.; Goss, J.A.; Cho, A.; McCain, M.L.; Parker, K.K. Microfluidic heart on a chip for higher throughput pharmacological studies. Lab Chip 2013, 13, 3599–3608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Colosi, C.; Shin, S.R.; Manoharan, V.; Massa, S.; Costantini, M.; Barbetta, A.; Dokmeci, M.R.; Dentini, M.; Khademhosseini, A. Microfluidic Bioprinting of Heterogeneous 3D Tissue Constructs Using Low-Viscosity Bioink. Adv. Mater. 2016, 28, 677–684. [Google Scholar] [CrossRef]
- Sun, L.; Chen, Z.; Xu, D.; Zhao, Y. Electroconductive and Anisotropic Structural Color Hydrogels for Visual Heart-on-a-Chip Construction. Adv. Sci. 2022, e2105777. [Google Scholar] [CrossRef]
- Gotz, C.; Pfeiffer, R.; Tigges, J.; Blatz, V.; Jackh, C.; Freytag, E.M.; Fabian, E.; Landsiedel, R.; Merk, H.F.; Krutmann, J.; et al. Xenobiotic metabolism capacities of human skin in comparison with a 3D epidermis model and keratinocyte-based cell culture as in vitro alternatives for chemical testing: Activating enzymes (Phase I). Exp. Derm. 2012, 21, 358–363. [Google Scholar] [CrossRef]
- He, R.; Liu, H.; Fang, T.; Niu, Y.; Zhang, H.; Han, F.; Gao, B.; Li, F.; Xu, F. A Colorimetric Dermal Tattoo Biosensor Fabricated by Microneedle Patch for Multiplexed Detection of Health-Related Biomarkers. Adv. Sci. 2021, 8, e2103030. [Google Scholar] [CrossRef]
- Lukacs, B.; Bajza, A.; Kocsis, D.; Csorba, A.; Antal, I.; Ivan, K.; Laki, A.J.; Erdo, F. Skin-on-a-Chip Device for Ex Vivo Monitoring of Transdermal Delivery of Drugs-Design, Fabrication, and Testing. Pharmaceutics 2019, 11, 455. [Google Scholar] [CrossRef] [Green Version]
- Autier, J.; Escudier, B.; Wechsler, J.; Spatz, A.; Robert, C. Prospective study of the cutaneous adverse effects of sorafenib, a novel multikinase inhibitor. Arch. Derm. 2008, 144, 886–892. [Google Scholar] [CrossRef]
- Lee, E.J.; Jain, M.; Alimperti, S. Bone Microvasculature: Stimulus for Tissue Function and Regeneration. Tissue Eng. Part B Rev. 2021, 27, 313–329. [Google Scholar] [CrossRef] [PubMed]
- Nagy, J.A.; Chang, S.H.; Dvorak, A.M.; Dvorak, H.F. Why are tumour blood vessels abnormal and why is it important to know? Br. J. Cancer 2009, 100, 865–869. [Google Scholar] [CrossRef] [PubMed]
- Asaumi, Y.; Sasaki, N. Photolithography-free Vessel-on-a-chip to Simulate Tumor Cell Extravasation. Sens. Mater. 2021, 33, 241. [Google Scholar] [CrossRef]
- Salipante, P.F.; Hudson, S.D.; Alimperti, S. Blood vessel-on-a-chip examines the biomechanics of microvasculature. Soft Matter 2021, 18, 117–125. [Google Scholar] [CrossRef]
- Basile, D.P.; Anderson, M.D.; Sutton, T.A. Pathophysiology of acute kidney injury. Compr. Physiol. 2012, 2, 1303–1353. [Google Scholar] [CrossRef] [Green Version]
- Wilmer, M.J.; Ng, C.P.; Lanz, H.L.; Vulto, P.; Suter-Dick, L.; Masereeuw, R. Kidney-on-a-Chip Technology for Drug-Induced Nephrotoxicity Screening. Trends Biotechnol. 2016, 34, 156–170. [Google Scholar] [CrossRef]
- Xiao, S.; Coppeta, J.R.; Rogers, H.B.; Isenberg, B.C.; Zhu, J.; Olalekan, S.A.; McKinnon, K.E.; Dokic, D.; Rashedi, A.S.; Haisenleder, D.J.; et al. A microfluidic culture model of the human reproductive tract and 28-day menstrual cycle. Nat. Commun. 2017, 8, 14584. [Google Scholar] [CrossRef]
- Tsamandouras, N.; Chen, W.L.K.; Edington, C.D.; Stokes, C.L.; Griffith, L.G.; Cirit, M. Integrated Gut and Liver Microphysiological Systems for Quantitative In Vitro Pharmacokinetic Studies. AAPS J. 2017, 19, 1499–1512. [Google Scholar] [CrossRef] [Green Version]
- Edington, C.D.; Chen, W.L.K.; Geishecker, E.; Kassis, T.; Soenksen, L.R.; Bhushan, B.M.; Freake, D.; Kirschner, J.; Maass, C.; Tsamandouras, N.; et al. Interconnected Microphysiological Systems for Quantitative Biology and Pharmacology Studies. Sci. Rep. 2018, 8, 4530. [Google Scholar] [CrossRef] [Green Version]
- Novak, R.; Ingram, M.; Marquez, S.; Das, D.; Delahanty, A.; Herland, A.; Maoz, B.M.; Jeanty, S.S.F.; Somayaji, M.R.; Burt, M.; et al. Robotic fluidic coupling and interrogation of multiple vascularized organ chips. Nat. Biomed. Eng. 2020, 4, 407–420. [Google Scholar] [CrossRef]
- Herland, A.; Maoz, B.M.; Das, D.; Somayaji, M.R.; Prantil-Baun, R.; Novak, R.; Cronce, M.; Huffstater, T.; Jeanty, S.S.F.; Ingram, M.; et al. Quantitative prediction of human pharmacokinetic responses to drugs via fluidically coupled vascularized organ chips. Nat. Biomed. Eng. 2020, 4, 421–436. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.S.; Aleman, J.; Shin, S.R.; Kilic, T.; Kim, D.; Mousavi Shaegh, S.A.; Massa, S.; Riahi, R.; Chae, S.; Hu, N.; et al. Multisensor-integrated organs-on-chips platform for automated and continual in situ monitoring of organoid behaviors. Proc. Natl. Acad. Sci. USA 2017, 114, E2293–E2302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wagner, I.; Materne, E.M.; Brincker, S.; Sussbier, U.; Fradrich, C.; Busek, M.; Sonntag, F.; Sakharov, D.A.; Trushkin, E.V.; Tonevitsky, A.G.; et al. A dynamic multi-organ-chip for long-term cultivation and substance testing proven by 3D human liver and skin tissue co-culture. Lab Chip 2013, 13, 3538–3547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bauer, S.; Wennberg Huldt, C.; Kanebratt, K.P.; Durieux, I.; Gunne, D.; Andersson, S.; Ewart, L.; Haynes, W.G.; Maschmeyer, I.; Winter, A.; et al. Functional coupling of human pancreatic islets and liver spheroids on-a-chip: Towards a novel human ex vivo type 2 diabetes model. Sci. Rep. 2017, 7, 14620. [Google Scholar] [CrossRef] [PubMed]
- Baert, Y.; Ruetschle, I.; Cools, W.; Oehme, A.; Lorenz, A.; Marx, U.; Goossens, E.; Maschmeyer, I. A multi-organ-chip co-culture of liver and testis equivalents: A first step toward a systemic male reprotoxicity model. Hum. Reprod. 2020, 35, 1029–1044. [Google Scholar] [CrossRef] [PubMed]
- Maschmeyer, I.; Hasenberg, T.; Jaenicke, A.; Lindner, M.; Lorenz, A.K.; Zech, J.; Garbe, L.A.; Sonntag, F.; Hayden, P.; Ayehunie, S.; et al. Chip-based human liver-intestine and liver-skin co-cultures—A first step toward systemic repeated dose substance testing in vitro. Eur. J. Pharm. Biopharm. 2015, 95, 77–87. [Google Scholar] [CrossRef] [Green Version]
- Lave, T.; Parrott, N.; Grimm, H.P.; Fleury, A.; Reddy, M. Challenges and opportunities with modelling and simulation in drug discovery and drug development. Xenobiotica 2007, 37, 1295–1310. [Google Scholar] [CrossRef]
- Rowland, M.; Peck, C.; Tucker, G. Physiologically-based pharmacokinetics in drug development and regulatory science. Annu. Rev. Pharm. Toxicol. 2011, 51, 45–73. [Google Scholar] [CrossRef]
- Prantil-Baun, R.; Novak, R.; Das, D.; Somayaji, M.R.; Przekwas, A.; Ingber, D.E. Physiologically Based Pharmacokinetic and Pharmacodynamic Analysis Enabled by Microfluidically Linked Organs-on-Chips. Annu. Rev. Pharm. Toxicol. 2018, 58, 37–64. [Google Scholar] [CrossRef]
- Chi, C.W.; Lao, Y.H.; Ahmed, A.H.R.; Benoy, E.C.; Li, C.; Dereli-Korkut, Z.; Fu, B.M.; Leong, K.W.; Wang, S. High-Throughput Tumor-on-a-Chip Platform to Study Tumor-Stroma Interactions and Drug Pharmacokinetics. Adv. Healthc. Mater. 2020, 9, e2000880. [Google Scholar] [CrossRef]
- Liu, D.; Jiao, S.; Wei, J.; Zhang, X.; Pei, Y.; Pei, Z.; Li, J.; Du, Y. Investigation of absorption, metabolism and toxicity of ginsenosides compound K based on human organ chips. Int. J. Pharm. 2020, 587, 119669. [Google Scholar] [CrossRef] [PubMed]
- Shinha, K.; Nihei, W.; Ono, T.; Nakazato, R.; Kimura, H. A pharmacokinetic-pharmacodynamic model based on multi-organ-on-a-chip for drug-drug interaction studies. Biomicrofluidics 2020, 14, 044108. [Google Scholar] [CrossRef] [PubMed]
- Skardal, A.; Aleman, J.; Forsythe, S.; Rajan, S.; Murphy, S.; Devarasetty, M.; Pourhabibi Zarandi, N.; Nzou, G.; Wicks, R.; Sadri-Ardekani, H.; et al. Drug compound screening in single and integrated multi-organoid body-on-a-chip systems. Biofabrication 2020, 12, 025017. [Google Scholar] [CrossRef] [PubMed]
- Gokaltun, A.; Yarmush, M.L.; Asatekin, A.; Usta, O.B. Recent advances in nonbiofouling PDMS surface modification strategies applicable to microfluidic technology. Technology 2017, 5, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wagner, V.; Dullaart, A.; Bock, A.K.; Zweck, A. The emerging nanomedicine landscape. Nat. Biotechnol. 2006, 24, 1211–1217. [Google Scholar] [CrossRef] [PubMed]
- Chen, X.; Zhang, Y.S.; Zhang, X.; Liu, C. Organ-on-a-chip platforms for accelerating the evaluation of nanomedicine. Bioact. Mater. 2021, 6, 1012–1027. [Google Scholar] [CrossRef]
- Lu, R.X.Z.; Radisic, M. Organ-on-a-chip platforms for evaluation of environmental nanoparticle toxicity. Bioact. Mater. 2021, 6, 2801–2819. [Google Scholar] [CrossRef]
- Davila, S.; Cacheux, J.; Rodriguez, I. Microvessel-on-Chip Fabrication for the In Vitro Modeling of Nanomedicine Transport. ACS Omega 2021, 6, 25109–25115. [Google Scholar] [CrossRef]
- Kim, S.; Chung, M.; Ahn, J.; Lee, S.; Jeon, N.L. Interstitial flow regulates the angiogenic response and phenotype of endothelial cells in a 3D culture model. Lab Chip 2016, 16, 4189–4199. [Google Scholar] [CrossRef]
- Lee, S.; Kim, S.; Koo, D.J.; Yu, J.; Cho, H.; Lee, H.; Song, J.M.; Kim, S.Y.; Min, D.H.; Jeon, N.L. 3D Microfluidic Platform and Tumor Vascular Mapping for Evaluating Anti-Angiogenic RNAi-Based Nanomedicine. ACS Nano 2021, 15, 338–350. [Google Scholar] [CrossRef]
- Egorov, E.; Pieters, C.; Korach-Rechtman, H.; Shklover, J.; Schroeder, A. Robotics, microfluidics, nanotechnology and AI in the synthesis and evaluation of liposomes and polymeric drug delivery systems. Drug Deliv. Transl. Res. 2021, 11, 345–352. [Google Scholar] [CrossRef] [PubMed]
- Fang, G.; Lu, H.; Al-Nakashli, R.; Chapman, R.; Zhang, Y.; Ju, L.A.; Lin, G.; Stenzel, M.H.; Jin, D. Enabling peristalsis of human colon tumor organoids on microfluidic chips. Biofabrication 2021, 14, 015006. [Google Scholar] [CrossRef] [PubMed]
- Aleman, J.; Kilic, T.; Mille, L.S.; Shin, S.R.; Zhang, Y.S. Microfluidic integration of regeneratable electrochemical affinity-based biosensors for continual monitoring of organ-on-a-chip devices. Nat. Protoc. 2021, 16, 2564–2593. [Google Scholar] [CrossRef] [PubMed]
- Shang, W.; Chen, C.-Y.; Lo, K.; Payne, G.F.; Bentley, W.E. Chip modularity enables molecular information access from organ-on-chip devices with quality control. Sens. Actuators B Chem. 2019, 295, 30–39. [Google Scholar] [CrossRef]
- Farooqi, H.M.U.; Khalid, M.A.U.; Kim, K.H.; Lee, S.R.; Choi, K.H. Real-time physiological sensor-based liver-on-chip device for monitoring drug toxicity. J. Micromech. Microeng. 2020, 30, 115013. [Google Scholar] [CrossRef]
- Jang, K.-J.; Otieno, M.A.; Ronxhi, J.; Lim, H.-K.; Ewart, L.; Kodella, K.R.; Petropolis, D.B.; Kulkarni, G.; Rubins, J.E.; Conegliano, D.; et al. Reproducing human and cross-species drug toxicities using a Liver-Chip. Sci. Transl. Med. 2019, 11, eaax5516. [Google Scholar] [CrossRef]
- Bovard, D.; Sandoz, A.; Luettich, K.; Frentzel, S.; Iskandar, A.; Marescotti, D.; Trivedi, K.; Guedj, E.; Dutertre, Q.; Peitsch, M.C.; et al. A lung/liver-on-a-chip platform for acute and chronic toxicity studies. Lab Chip 2018, 18, 3814–3829. [Google Scholar] [CrossRef] [Green Version]
Human Organ Chip | Materials | Cell Types | Technology for Fabrications | Disease Model or Clinical Mimicry | Ref |
---|---|---|---|---|---|
Lung | PDMS | Epi and endo | Soft lithography | Nanoparticle transport and toxicity | [8] |
PDMS, collagen, fibrin gel | Endo (HUVEC), fibroblast (HLF and iPF-HLF), epi (SAEC) | Polyester track etch, soft lithography | Fibrotic αSMA-positive disease | [30] | |
PDMS 3D Matrigel | Epi (HPAEpiC), endo (HUVEC) | Soft lithography | Nanoparticle toxicity | [31] | |
Soft collagen–elastin gel, thin gold mesh, PDMS, polycarbonate | Epi (HAEpC), endo (VeraVec) | Collagen molecules self-assembled, soft lithography | The air–blood barrier and alveoli network | [32] | |
Plastic | Epi | Laser-cut, polyester track etch | Viral infectivity, inflammation | [33] | |
Liver | PDMS | Hep | Soft lithography | Drug toxicity | [34] |
PDMS, PE membrane | Hep, endo, hepatic stellate, Kupffer | Soft lithography | Liver-specific functions | [35] | |
PDMS, ECM gel | Hep, endo, hepatic stellate, Kupffer | Soft lithography | Drug efficacy | [36] | |
Gut | PDMS | Epi (Caco-2) | Soft lithography | Intestinal barrier function | [22] |
PDMS, PET membrane | Epi (Caco-2) | Soft lithography, photolithography | Differentiation, drug absorption, and metabolism | [37] | |
Polycarbonate, PDMS, titanium, gold, PET membrane | Epi (hAEC) | Laser-cut, soft lithography | Real-time measurements of barrier function | [38] | |
PDMS, PMMA frame | Epi (Caco-2), endo (HUVEC) | Soft lithography | Injury of intestine villus and inflammatory reactions | [39] | |
BBB | PDMS, polycarbonate membrane, Pt | Endo (HCMEC) | Soft lithography | Effect of inflammation cytokine | [26] |
PDMS, Ag/AgCl, polycarbonate membrane, silicone gasket | Endo (iPS) | 3D print | Drug permeability | [40] | |
Glass and polymers (OrganoPlate), ECM gel | Endo, pericyte, astrocyte | - | High-throughput drug screening | [41] | |
PDMS, polycarbonate membrane | Endo (bEnd.3) | Soft lithography | Angiopep-2 coupled liposome transport | [42] | |
Heart | PDMS | Cardiomyocyte/iPS | Soft lithography | Drug concentration-response | [43] |
Bioink, PMMA, PDMS | Endo(HUVEC), cardiomyocyte | Bioprinting, soft lithography | Drug screening | [44] | |
Super aligned carbon nanotube sheets, hydrogel, conductive methacrylated gelatin, PDMS | Cardiomyocyte | Polymerization, ultraviolet (UV) irradiation, soft lithography | Dynamic cardiomyocyte sensing and drug screening | [45] | |
PDMS | Cardiomyocyte (iPS) | Direct laser writing (DLW) lithography and soft lithography | Response of cardiac under mechanical loading and pacing. | [46] | |
Skin | PDMS | Fibroblast, keratinocyte | Soft lithography | Skin side effects of sorafenib | [47] |
PDMS, polyester membrane | Fibroblast, keratinocyte | Soft lithography | Drug testing | [48] | |
PMMA, PET membrane | Keratinocyte | Micromilling, track etch | Skin irritation and drug toxicity | [49] | |
Vessel | PDMS | Endo (HUVEC) | Soft lithography | Thrombi and hemorrhage | [50] |
Hydrogel, PMMA | Endo (HAEC), aortic smooth muscle | Bioprinting, numerical control engraving | Physiologic and pathologic process in vascular wall | [51] | |
Pluronic, nanoclay, PDMS | Endo (HUVEC) | 3D printed, soft lithography | Endothelial cell morphology | [52] | |
Kidney | PDMS, polycarbonate membrane | Liver and kidney precision-cut tissue slices, Endo (HUVEC), breast cancer | Soft lithography | Extracellular vesicles organotropism | [53] |
Glass and polymers (OrganoPlate), ECM gel | Epi (RPTEC), Endo (HUVEC) | - | Renal ischemia and reperfusion injury | [54] | |
Lymphangion | PDMS | Endo (LEC), muscle (LMC) | Soft lithography | Lymphatic inflammation | [55] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Li, Z.; Hui, J.; Yang, P.; Mao, H. Microfluidic Organ-on-a-Chip System for Disease Modeling and Drug Development. Biosensors 2022, 12, 370. https://doi.org/10.3390/bios12060370
Li Z, Hui J, Yang P, Mao H. Microfluidic Organ-on-a-Chip System for Disease Modeling and Drug Development. Biosensors. 2022; 12(6):370. https://doi.org/10.3390/bios12060370
Chicago/Turabian StyleLi, Zening, Jianan Hui, Panhui Yang, and Hongju Mao. 2022. "Microfluidic Organ-on-a-Chip System for Disease Modeling and Drug Development" Biosensors 12, no. 6: 370. https://doi.org/10.3390/bios12060370
APA StyleLi, Z., Hui, J., Yang, P., & Mao, H. (2022). Microfluidic Organ-on-a-Chip System for Disease Modeling and Drug Development. Biosensors, 12(6), 370. https://doi.org/10.3390/bios12060370