Organ-on-a-Chip for Studying Gut-Brain Interaction Mediated by Extracellular Vesicles in the Gut Microenvironment
Abstract
:1. Introduction
2. Involvement of Gut-Originated EVs in the GBA
2.1. Type and Functions of Gut-Originated EVs
2.2. Roles of EV in GBA
2.2.1. Microbial EVs
2.2.2. Gut-Released EVs
2.3. EVs from External Source
3. GBA-on-a-Chip for Research on EVs in Gut Microenvironment
3.1. Multi-Organ-on-a-Chip as a Novel In Vitro Platform for GBA Research
3.2. Considerations on the Design of GBA-on-a-Chip
3.2.1. BBB-on-a-Chip
3.2.2. Gut-on-a-Chip
3.2.3. GBA-on-a-Chip
3.3. GBA-on-a-Chip for Models of EV-Mediated Diseases
3.3.1. Investigating Organ Level Response Using Isolated EVs
3.3.2. Real-Time Tracking of Cell-to-Cell EV Delivery in an Organ-on-a-Chip
3.3.3. Studying EV Distribution and Tropism Using Multi-Organ-on-a-Chip
4. Limitation
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Yáñez-Mó, M.; Siljander, P.R.-M.; Andreu, Z.; Zavec, A.B.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef] [Green Version]
- Skotland, T.; Sagini, K.; Sandvig, K.; Llorente, A. An emerging focus on lipids in extracellular vesicles. Adv. Drug Deliv. Rev. 2020, 159, 308–321. [Google Scholar] [CrossRef]
- Raposo, G.; Nijman, H.W.; Stoorvogel, W.; Liejendekker, R.; Harding, C.V.; Melief, C.J.; Geuze, H.J. B lymphocytes secrete antigen-presenting vesicles. J. Exp. Med. 1996, 183, 1161–1172. [Google Scholar] [CrossRef]
- El Andaloussi, S.; Mäger, I.; Breakefield, X.O.; Wood, M.J.A. Extracellular vesicles: Biology and emerging therapeutic opportunities. Nat. Rev. Drug Discov. 2013, 12, 347–357. [Google Scholar] [CrossRef]
- Maas, S.L.N.; Breakefield, X.O.; Weaver, A.M. Extracellular Vesicles: Unique Intercellular Delivery Vehicles. Trends Cell Biol. 2017, 27, 172–188. [Google Scholar] [CrossRef] [Green Version]
- Cocucci, E.; Racchetti, G.; Meldolesi, J. Shedding microvesicles: Artefacts no more. Trends Cell Biol. 2009, 19, 43–51. [Google Scholar] [CrossRef]
- Neven, K.Y.; Nawrot, T.S.; Bollati, V. Extracellular Vesicles: How the External and Internal Environment Can Shape Cell-To-Cell Communication. Curr. Environ. Health Rep. 2017, 4, 30–37. [Google Scholar] [CrossRef]
- Pulliero, A.; Pergoli, L.; La Maestra, S.; Micale, R.T.; Camoirano, A.; Bollati, V.; Izzotti, A.; De Flora, S. Extracellular vesicles in biological fluids. A biomarker of exposure to cigarette smoke and treatment with chemopreventive drugs. J. Prev. Med. Hyg. 2019, 60, E327–E336. [Google Scholar]
- Stahl, P.D.; Raposo, G. Extracellular Vesicles: Exosomes and Microvesicles, Integrators of Homeostasis. Physiology 2019, 34, 169–177. [Google Scholar] [CrossRef]
- Robbins, P.D.; Morelli, A.E. Regulation of immune responses by extracellular vesicles. Nat. Rev. Immunol. 2014, 14, 195–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hill, A.F. Extracellular Vesicles and Neurodegenerative Diseases. J. Neurosci. 2019, 39, 9269–9273. [Google Scholar] [CrossRef] [PubMed]
- Raimondi, M.T.; Albani, D.; Giordano, C. An Organ-On-A-Chip Engineered Platform to Study the Microbiota–Gut–Brain Axis in Neurodegeneration. Trends Mol. Med. 2019, 25, 737–740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, M.-H.; Kim, D.; Sung, J.H. A Gut-Brain Axis-on-a-Chip for studying transport across epithelial and endothelial barriers. J. Ind. Eng. Chem. 2021, 101, 126–134. [Google Scholar] [CrossRef]
- Haas-Neill, S.; Forsythe, P. A Budding Relationship: Bacterial Extracellular Vesicles in the Microbiota–Gut–Brain Axis. Int. J. Mol. Sci. 2020, 21, 8899. [Google Scholar] [CrossRef]
- Sandhu, K.V.; Sherwin, E.; Schellekens, H.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Feeding the microbiota-gut-brain axis: Diet, microbiome, and neuropsychiatry. Transl. Res. 2016, 179, 223–244. [Google Scholar] [CrossRef] [PubMed]
- Zhu, S.; Jiang, Y.; Xu, K.; Cui, M.; Ye, W.; Zhao, G.; Jin, L.; Chen, X. The progress of gut microbiome research related to brain disorders. J. Neuroinflamm. 2020, 17, 25. [Google Scholar] [CrossRef] [Green Version]
- Cuesta, C.M.; Guerri, C.; Ureña, J.; Pascual, M. Role of Microbiota-Derived Extracellular Vesicles in Gut-Brain Communication. Int. J. Mol. Sci. 2021, 22, 4235. [Google Scholar] [CrossRef]
- Raimondi, I.; Izzo, L.; Tunesi, M.; Comar, M.; Albani, D.; Giordano, C. Organ-On-A-Chip in vitro Models of the Brain and the Blood-Brain Barrier and Their Value to Study the Microbiota-Gut-Brain Axis in Neurodegeneration. Front. Bioeng. Biotechnol. 2020, 7, 435. [Google Scholar] [CrossRef]
- Voelkl, B.; Altman, N.S.; Forsman, A.; Forstmeier, W.; Gurevitch, J.; Jaric, I.; Karp, N.A.; Kas, M.J.; Schielzeth, H.; Van de Casteele, T.; et al. Reproducibility of animal research in light of biological variation. Nat. Rev. Neurosci. 2020, 21, 384–393. [Google Scholar] [CrossRef]
- Azizipour, N.; Avazpour, R.; Rosenzweig, D.H.; Sawan, M.; Ajji, A. Evolution of Biochip Technology: A Review from Lab-on-a-Chip to Organ-on-a-Chip. Micromachines 2020, 11, 599. [Google Scholar] [CrossRef] [PubMed]
- Liang, Y.; Lehrich, B.M.; Zheng, S.; Lu, M. Emerging methods in biomarker identification for extracellular vesicle-based liquid biopsy. J. Extracell. Vesicles 2021, 10, e12090. [Google Scholar] [CrossRef]
- Urbanelli, L.; Buratta, S.; Sagini, K.; Ferrara, G.; Lanni, M.; Emiliani, C. Exosome-based strategies for Diagnosis and Therapy. Recent Patents CNS Drug Discov. 2015, 10, 10–27. [Google Scholar] [CrossRef]
- Ko, J.; Carpenter, E.; Issadore, D. Detection and isolation of circulating exosomes and microvesicles for cancer monitoring and diagnostics using micro-/nano-based devices. Analyst 2015, 141, 450–460. [Google Scholar] [CrossRef] [Green Version]
- Meng, W.; He, C.; Hao, Y.; Wang, L.; Li, L.; Zhu, G. Prospects and challenges of extracellular vesicle-based drug delivery system: Considering cell source. Drug Deliv. 2020, 27, 585–598. [Google Scholar] [CrossRef] [Green Version]
- Ma, C.; Peng, Y.; Li, H.; Chen, W. Organ-on-a-Chip: A New Paradigm for Drug Development. Trends Pharmacol. Sci. 2020, 42, 119–133. [Google Scholar] [CrossRef]
- Sung, J.H.; Wang, Y.I.; Narasimhan Sriram, N.; Jackson, M.; Long, C.; Hickman, J.J.; Shuler, M.L. Recent Advances in Body-on-a-Chip Systems. Anal. Chem. 2019, 91, 330–351. [Google Scholar] [CrossRef]
- Macia, L.; Nanan, R.; Hosseini-Beheshti, E.; Grau, G.E. Host- and Microbiota-Derived Extracellular Vesicles, Immune Function, and Disease Development. Int. J. Mol. Sci. 2019, 21, 107. [Google Scholar] [CrossRef] [Green Version]
- Gill, S.; Catchpole, R.; Forterre, P. Extracellular membrane vesicles in the three domains of life and beyond. FEMS Microbiol. Rev. 2018, 43, 273–303. [Google Scholar] [CrossRef]
- Prez-Cruz, C.; Carrin, O.; Delgado, L.; Martinez, G.; Lpez-Iglesias, C.; Mercade, E. New Type of Outer Membrane Vesicle Produced by the Gram-Negative Bacterium Shewanella vesiculosa M7 T: Implications for DNA Content. Appl. Environ. Microbiol. 2013, 79, 1874–1881. [Google Scholar] [CrossRef] [Green Version]
- Pérez-Cruz, C.; Delgado, L.; López-Iglesias, C.; Mercade, E. Outer-Inner Membrane Vesicles Naturally Secreted by Gram-Negative Pathogenic Bacteria. PLoS ONE 2015, 10, e0116896. [Google Scholar] [CrossRef] [Green Version]
- Briaud, P.; Carroll, R.K. Extracellular Vesicle Biogenesis and Functions in Gram-Positive Bacteria. Infect. Immun. 2020, 88, e00433-20. [Google Scholar] [CrossRef]
- Ma, L.; Cao, Z. Membrane vesicles from periodontal pathogens and their potential roles in periodontal disease and systemic illnesses. J. Periodontal Res. 2021, 56, 646–655. [Google Scholar] [CrossRef]
- Toyofuku, M.; Nomura, N.; Eberl, L. Types and origins of bacterial membrane vesicles. Nat. Rev. Microbiol. 2019, 17, 13–24. [Google Scholar] [CrossRef]
- Szempruch, A.J.; Dennison, L.; Kieft, R.; Harrington, J.M.; Hajduk, S.L. Sending a message: Extracellular vesicles of pathogenic protozoan parasites. Nat. Rev. Genet. 2016, 14, 669–675. [Google Scholar] [CrossRef]
- Gonçalves, D.D.S.; Ferreira, M.D.S.; Guimarães, A.J. Extracellular Vesicles from the Protozoa Acanthamoeba castellanii: Their Role in Pathogenesis, Environmental Adaptation and Potential Applications. Bioengineering 2019, 6, 13. [Google Scholar] [CrossRef] [Green Version]
- Olajide, J.S.; Cai, J. Perils and Promises of Pathogenic Protozoan Extracellular Vesicles. Front. Cell. Infect. Microbiol. 2020, 10, 371. [Google Scholar] [CrossRef]
- Pérez, J.C. Fungi of the human gut microbiota: Roles and significance. Int. J. Med. Microbiol. 2021, 311, 151490. [Google Scholar] [CrossRef]
- Rizzo, J.; Rodrigues, M.L.; Janbon, G. Extracellular Vesicles in Fungi: Past, Present, and Future Perspectives. Front. Cell. Infect. Microbiol. 2020, 10, 346. [Google Scholar] [CrossRef]
- Zamith-Miranda, D.; Nimrichter, L.; Rodrigues, M.L.; Nosanchuk, J.D. Fungal extracellular vesicles: Modulating host–pathogen interactions by both the fungus and the host. Microbes Infect. 2018, 20, 501–504. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Liu, Y.; Liu, H.; Tang, W.H. Exosomes: Biogenesis, biologic function and clinical potential. Cell Biosci. 2019, 9, 19. [Google Scholar] [CrossRef]
- Akers, J.C.; Gonda, D.; Kim, R.; Carter, B.S.; Chen, C.C. Biogenesis of extracellular vesicles (EV): Exosomes, microvesicles, retrovirus-like vesicles, and apoptotic bodies. J. Neuro-Oncol. 2013, 113, 1–11. [Google Scholar] [CrossRef] [Green Version]
- Doyle, L.M.; Wang, M.Z. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [Green Version]
- Battistelli, M.; Falcieri, E. Apoptotic Bodies: Particular Extracellular Vesicles Involved in Intercellular Communication. Biology 2020, 9, 21. [Google Scholar] [CrossRef] [Green Version]
- Pirolli, N.H.; Bentley, W.E.; Jay, S.M. Bacterial Extracellular Vesicles and the Gut-Microbiota Brain Axis: Emerging Roles in Communication and Potential as Therapeutics. Adv. Biol. 2021, 5, 2000540. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.H.; Lee, J.; Park, J.; Gho, Y.S. Gram-negative and Gram-positive bacterial extracellular vesicles. Semin. Cell Dev. Biol. 2015, 40, 97–104. [Google Scholar] [CrossRef]
- Stanton, B.A. Extracellular Vesicles and Host–Pathogen Interactions: A Review of Inter-Kingdom Signaling by Small Noncoding RNA. Genes 2021, 12, 1010. [Google Scholar] [CrossRef]
- Bäuerl, C.; Coll-Marqués, J.M.; Tarazona-González, C.; Pérez-Martínez, G. Lactobacillus casei extracellular vesicles stimulate EGFR pathway likely due to the presence of proteins P40 and P75 bound to their surface. Sci. Rep. 2020, 10, 19237. [Google Scholar] [CrossRef]
- Liu, Y.; Defourny, K.A.Y.; Smid, E.J.; Abee, T. Gram-Positive Bacterial Extracellular Vesicles and Their Impact on Health and Disease. Front. Microbiol. 2018, 9, 1502. [Google Scholar] [CrossRef] [Green Version]
- Zaborowski, M.P.; Balaj, L.; Breakefield, X.O.; Lai, C.P. Extracellular Vesicles: Composition, Biological Relevance, and Methods of Study. Bioscience 2015, 65, 783–797. [Google Scholar] [CrossRef] [Green Version]
- Yu, L.-L.; Zhu, J.; Liu, J.-X.; Jiang, F.; Ni, W.-K.; Qu, L.-S.; Ni, R.-Z.; Lu, C.-H.; Xiao, M.-B. A Comparison of Traditional and Novel Methods for the Separation of Exosomes from Human Samples. BioMed Res. Int. 2018, 2018, 3634563. [Google Scholar] [CrossRef] [Green Version]
- Wiklander, O.P.B.; Brennan, M.; Lötvall, J.; Breakefield, X.O.; EL Andaloussi, S. Advances in therapeutic applications of extracellular vesicles. Sci. Transl. Med. 2019, 11, 492. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; He, X.; Li, Q.; Lai, H.; Zhang, H.; Hu, Z.; Li, Y.; Huang, S. EV-origin: Enumerating the tissue-cellular origin of circulating extracellular vesicles using exLR profile. Comput. Struct. Biotechnol. J. 2020, 18, 2851–2859. [Google Scholar] [CrossRef]
- Wang, Y.; Zhao, M.; Liu, S.; Guo, J.; Lu, Y.; Cheng, J.; Liu, J. Macrophage-derived extracellular vesicles: Diverse mediators of pathology and therapeutics in multiple diseases. Cell Death Dis. 2020, 11, 924. [Google Scholar] [CrossRef] [PubMed]
- Stentz, R.; Carvalho, A.L.; Jones, E.J.; Carding, S.R. Fantastic voyage: The journey of intestinal microbiota-derived microvesicles through the body. Biochem. Soc. Trans. 2018, 46, 1021–1027. [Google Scholar] [CrossRef]
- Jan, A.T. Outer Membrane Vesicles (OMVs) of Gram-negative Bacteria: A Perspective Update. Front. Microbiol. 2017, 8, 1053. [Google Scholar] [CrossRef] [PubMed]
- Wispelwey, B.; Hansen, E.J.; Scheld, W.M. Haemophilus influenzae outer membrane vesicle-induced blood-brain barrier permeability during experimental meningitis. Infect. Immun. 1989, 57, 2559–2562. [Google Scholar] [CrossRef] [Green Version]
- Wei, S.; Peng, W.; Mai, Y.; Li, K.; Wei, W.; Hu, L.; Zhu, S.; Zhou, H.; Jie, W.; Wei, Z.; et al. Outer membrane vesicles enhance tau phosphorylation and contribute to cognitive impairment. J. Cell. Physiol. 2019, 235, 4843–4855. [Google Scholar] [CrossRef]
- Al-Nedawi, K.; Mian, M.F.; Hossain, N.; Karimi, K.; Mao, Y.-K.; Forsythe, P.; Min, K.K.; Stanisz, A.M.; Kunze, W.A.; Bienenstock, J. Gut commensal microvesicles reproduce parent bacterial signals to host immune and enteric nervous systems. FASEB J. 2014, 29, 684–695. [Google Scholar] [CrossRef]
- Lee, K.-E.; Kim, J.-K.; Han, S.-K.; Lee, D.Y.; Lee, H.-J.; Yim, S.-V.; Kim, D.-H. The extracellular vesicle of gut microbial Paenalcaligenes hominis is a risk factor for vagus nerve-mediated cognitive impairment. Microbiome 2020, 8, 107. [Google Scholar] [CrossRef]
- Gonçalves, D.D.S.; Ferreira, M.D.S.; Liedke, S.C.; Gomes, K.X.; De Oliveira, G.A.; Leão, P.E.L.; Cesar, G.V.; Seabra, S.H.; Cortines, J.; Casadevall, A.; et al. Extracellular vesicles and vesicle-free secretome of the protozoa Acanthamoeba castellanii under homeostasis and nutritional stress and their damaging potential to host cells. Virulence 2018, 9, 818–836. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, P.; Cao, L.; Wang, X.; Dong, J.; Zhang, N.; Li, X.; Li, J.; Zhang, X.; Gong, P. Extracellular vesicles secreted by Giardia duodenalis regulate host cell innate immunity via TLR2 and NLRP3 inflammasome signaling pathways. PLOS Neglected Trop. Dis. 2021, 15, e0009304. [Google Scholar] [CrossRef]
- Nievas, Y.R.; Lizarraga, A.; Salas, N.; Cóceres, V.M.; De Miguel, N. Extracellular vesicles released by anaerobic protozoan parasites: Current situation. Cell. Microbiol. 2020, 22, e13257. [Google Scholar] [CrossRef]
- Van Niel, G.; Raposo, G.; Candalh, C.; Boussac, M.; Hershberg, R.; Cerf-Bensussan, N.; Heyman, M. Intestinal epithelial cells secrete exosome–like vesicles. Gastroenterology 2001, 121, 337–349. [Google Scholar] [CrossRef]
- Zhang, Y.; Yan, Y.; Meng, J.; Girotra, M.; Ramakrishnan, S.; Roy, S. Immune modulation mediated by extracellular vesicles of intestinal organoids is disrupted by opioids. Mucosal Immunol. 2021, 14, 887–898. [Google Scholar] [CrossRef]
- Sugihara, Y.; Onoue, S.; Tashiro, K.; Sato, M.; Hasegawa, T.; Katakura, Y. Carnosine induces intestinal cells to secrete exosomes that activate neuronal cells. PLoS ONE 2019, 14, e0217394. [Google Scholar] [CrossRef]
- Inotsuka, R.; Uchimura, K.; Yamatsu, A.; Kim, M.; Katakura, Y. γ-Aminobutyric acid (GABA) activates neuronal cells by inducing the secretion of exosomes from intestinal cells. Food Funct. 2020, 11, 9285–9290. [Google Scholar] [CrossRef] [PubMed]
- Inotsuka, R.; Udono, M.; Yamatsu, A.; Kim, M.; Katakura, Y. Exosome-Mediated Activation of Neuronal Cells Triggered by γ-Aminobutyric Acid (GABA). Nutrients 2021, 13, 2544. [Google Scholar] [CrossRef]
- Hepsomali, P.; Groeger, J.A.; Nishihira, J.; Scholey, A. Effects of Oral Gamma-Aminobutyric Acid (GABA) Administration on Stress and Sleep in Humans: A Systematic Review. Front. Neurosci. 2020, 14. [Google Scholar] [CrossRef]
- Mazzoli, R.; Pessione, E. The Neuro-endocrinological Role of Microbial Glutamate and GABA Signaling. Front. Microbiol. 2016, 7, 1934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ayyar, K.K.; Moss, A.C. Exosomes in Intestinal Inflammation. Front. Pharmacol. 2021, 12, 1295. [Google Scholar] [CrossRef] [PubMed]
- Deng, Z.; Mu, J.; Tseng, M.; Wattenberg, B.; Zhuang, X.; Egilmez, N.K.; Wang, Q.; Zhang, L.; Norris, J.D.; Guo, H.; et al. Enterobacteria-secreted particles induce production of exosome-like S1P-containing particles by intestinal epithelium to drive Th17-mediated tumorigenesis. Nat. Commun. 2015, 6, 6956. [Google Scholar] [CrossRef] [Green Version]
- Hu, G.; Gong, A.-Y.; Roth, A.L.; Huang, B.Q.; Ward, H.D.; Zhu, G.; LaRusso, N.F.; Hanson, N.D.; Chen, X.-M. Release of Luminal Exosomes Contributes to TLR4-Mediated Epithelial Antimicrobial Defense. PLoS Pathog. 2013, 9, e1003261. [Google Scholar] [CrossRef] [Green Version]
- Chen, Y.; Sun, H.; Bai, Y.; Zhi, F. Gut dysbiosis-derived exosomes trigger hepatic steatosis by transiting HMGB1 from intestinal to liver in mice. Biochem. Biophys. Res. Commun. 2019, 509, 767–772. [Google Scholar] [CrossRef]
- Rolhion, N.; Barnich, N.; Claret, L.; Darfeuille-Michaud, A. Strong Decrease in Invasive Ability and Outer Membrane Vesicle Release in Crohn’s Disease-Associated Adherent-Invasive Escherichia coli Strain LF82 with the yfgL Gene Deleted. J. Bacteriol. 2005, 187, 2286–2296. [Google Scholar] [CrossRef] [Green Version]
- Rolhion, N.; Barnich, N.; Bringer, M.-A.; Glasser, A.-L.; Ranc, J.; Hébuterne, X.; Hofman, P.; Darfeuille-Michaud, A. Abnormally expressed ER stress response chaperone Gp96 in CD favours adherent-invasive Escherichia coli invasion. Gut 2010, 59, 1355–1362. [Google Scholar] [CrossRef] [PubMed]
- Carrière, J.; Bretin, A.; Darfeuille-Michaud, A.; Barnich, N.; Nguyen, H.T.T. Exosomes Released from Cells Infected with Crohn’s Disease–associated Adherent-Invasive Escherichia coli Activate Host Innate Immune Responses and Enhance Bacterial Intracellular Replication. Inflamm. Bowel Dis. 2016, 22, 516–528. [Google Scholar] [CrossRef] [Green Version]
- Larabi, A.; Dalmasso, G.; Delmas, J.; Barnich, N.; Nguyen, H.T.T. Exosomes transfer miRNAs from cell-to-cell to inhibit autophagy during infection with Crohn’s disease-associated adherent-invasive E. coli. Gut Microbes 2020, 11, 1677–1694. [Google Scholar] [CrossRef]
- Wang, Y.; Wang, M.-D.; Xia, Y.-P.; Gao, Y.; Zhu, Y.-Y.; Chen, S.-C.; Mao, L.; He, Q.-W.; Yue, Z.-Y.; Hu, B. MicroRNA-130a regulates cerebral ischemia-induced blood-brain barrier permeability by targeting Homeobox A5. FASEB J. 2018, 32, 935–944. [Google Scholar] [CrossRef] [Green Version]
- Wang, M.-D.; Wang, Y.; Xia, Y.-P.; Dai, J.-W.; Gao, L.; Wang, S.-Q.; Wang, H.-J.; Mao, L.; Li, M.; Yu, S.-M.; et al. High Serum MiR-130a Levels Are Associated with Severe Perihematomal Edema and Predict Adverse Outcome in Acute ICH. Mol. Neurobiol. 2015, 53, 1310–1321. [Google Scholar] [CrossRef]
- Van Langenberg, D.R.; Yelland, G.W.; Robinson, S.R.; Gibson, P.R. Cognitive impairment in Crohn’s disease is associated with systemic inflammation, symptom burden and sleep disturbance. United Eur. Gastroenterol. J. 2017, 5, 579–587. [Google Scholar] [CrossRef]
- Kojima, M.; Costantini, T.W.; Eliceiri, B.P.; Chan, T.W.; Baird, A.; Coimbra, R. Gut epithelial cell-derived exosomes trigger posttrauma immune dysfunction. J. Trauma Acute Care Surg. 2018, 84, 257–264. [Google Scholar] [CrossRef]
- Chen, X.-D.; Zhao, J.; Yang, X.; Zhou, B.-W.; Yan, Z.; Liu, W.-F.; Li, C.; Liu, K.-X. Gut-Derived Exosomes Mediate Memory Impairment After Intestinal Ischemia/Reperfusion via Activating Microglia. Mol. Neurobiol. 2021, 58, 4828–4841. [Google Scholar] [CrossRef] [PubMed]
- Ocansey, D.K.W.; Zhang, L.; Wang, Y.; Yan, Y.; Qian, H.; Zhang, X.; Xu, W.; Mao, F. Exosome-mediated effects and applications in inflammatory bowel disease. Biol. Rev. 2020, 95, 1287–1307. [Google Scholar] [CrossRef]
- Salvo-Romero, E.; Stokes, P.; Gareau, M.G. Microbiota-immune interactions: From gut to brain. LymphoSign J. 2020, 7, 1–23. [Google Scholar] [CrossRef]
- Hawkins, K.G.; Casolaro, C.; Brown, J.A.; Edwards, D.A.; Wikswo, J.P. The Microbiome and the Gut-Liver-Brain Axis for Central Nervous System Clinical Pharmacology: Challenges in Specifying and Integrating In Vitro and In Silico Models. Clin. Pharmacol. Ther. 2020, 108, 929–948. [Google Scholar] [CrossRef] [PubMed]
- Yang, T.; Richards, E.M.; Pepine, C.J.; Raizada, M.K. The gut microbiota and the brain–gut–kidney axis in hypertension and chronic kidney disease. Nat. Rev. Nephrol. 2018, 14, 442–456. [Google Scholar] [CrossRef]
- Filipović, B.; Marković, O.; Đurić, V.; Filipović, B. Cognitive Changes and Brain Volume Reduction in Patients with Nonalcoholic Fatty Liver Disease. Can. J. Gastroenterol. Hepatol. 2018, 2018, 2097435. [Google Scholar] [CrossRef]
- Rogers, G.B.; Keating, D.J.; Young, R.L.; Wong, M.-L.; Licinio, J.; Wesselingh, S. From gut dysbiosis to altered brain function and mental illness: Mechanisms and pathways. Mol. Psychiatry 2016, 21, 738–748. [Google Scholar] [CrossRef] [Green Version]
- Carobolante, G.; Mantaj, J.; Ferrari, E.; Vllasaliu, D. Cow Milk and Intestinal Epithelial Cell-Derived Extracellular Vesicles as Systems for Enhancing Oral Drug Delivery. Pharmaceutics 2020, 12, 226. [Google Scholar] [CrossRef] [Green Version]
- Wolf, T.; Baier, S.R.; Zempleni, J. The Intestinal Transport of Bovine Milk Exosomes Is Mediated by Endocytosis in Human Colon Carcinoma Caco-2 Cells and Rat Small Intestinal IEC-6 Cells. J. Nutr. 2015, 145, 2201–2206. [Google Scholar] [CrossRef] [Green Version]
- Izumi, H.; Tsuda, M.; Sato, Y.; Kosaka, N.; Ochiya, T.; Iwamoto, H.; Namba, K.; Takeda, Y. Bovine milk exosomes contain microRNA and mRNA and are taken up by human macrophages. J. Dairy Sci. 2015, 98, 2920–2933. [Google Scholar] [CrossRef] [Green Version]
- Matic, S.; D’Souza, D.H.; Wu, T.; Pangloli, P.; Dia, V.P. Bovine Milk Exosomes Affect Proliferation and Protect Macrophages against Cisplatin-Induced Cytotoxicity. Immunol. Investig. 2020, 49, 711–725. [Google Scholar] [CrossRef]
- Munir, J.; Lee, M.; Ryu, S. Exosomes in Food: Health Benefits and Clinical Relevance in Diseases. Adv. Nutr. 2019, 11, 687–696. [Google Scholar] [CrossRef]
- Teng, Y.; Ren, Y.; Sayed, M.; Hu, X.; Lei, C.; Kumar, A.; Hutchins, E.; Mu, J.; Deng, Z.; Luo, C.; et al. Plant-Derived Exosomal MicroRNAs Shape the Gut Microbiota. Cell Host Microbe 2018, 24, 637–652.e8. [Google Scholar] [CrossRef] [Green Version]
- Manca, S.; Upadhyaya, B.; Mutai, E.; Desaulniers, A.T.; Cederberg, R.A.; White, B.R.; Zempleni, J. Milk exosomes are bioavailable and distinct microRNA cargos have unique tissue distribution patterns. Sci. Rep. 2018, 8, 11321. [Google Scholar] [CrossRef] [Green Version]
- Mutai, E.; Zhou, F.; Zempleni, J. Depletion of Dietary Bovine Milk Exosomes Impairs Sensorimotor Gating and Spatial Learning in C57BL/6 Mice. FASEB J. 2017, 31, 150.4. [Google Scholar] [CrossRef]
- Zempleni, J.; Sukreet, S.; Zhou, F.; Wu, D.; Mutai, E. Milk-Derived Exosomes and Metabolic Regulation. Annu. Rev. Anim. Biosci. 2019, 7, 245–262. [Google Scholar] [CrossRef] [PubMed]
- Maheshwari, R.; Gupta, A.; Ganeshpurkar, A.; Chourasiya, Y.; Tekade, M.; Tekade, R.K. Chapter 18—Guiding principles for human and animal research during pharmaceutical product development. In Advances in Pharmaceutical Product Development and Research; Tekade, R.K., Ed.; Academic Press: Cambridge, MA, USA, 2018; pp. 621–664. ISBN 978-0-12-814421-3. [Google Scholar]
- Zhang, B.; Korolj, A.; Lai, B.F.L.; Radisic, M. Advances in organ-on-a-chip engineering. Nat. Rev. Mater. 2018, 3, 257–278. [Google Scholar] [CrossRef]
- Ambrosini, Y.M.; Borcherding, D.; Kanthasamy, A.; Kim, H.J.; Willette, A.A.; Jergens, A.; Allenspach, K.; Mochel, J.P. The Gut-Brain Axis in Neurodegenerative Diseases and Relevance of the Canine Model: A Review. Front. Aging Neurosci. 2019, 11, 130. [Google Scholar] [CrossRef] [PubMed]
- Jeon, J.; 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]
- Wu, Q.; Liu, J.; Wang, X.; Feng, L.; Wu, J.; Zhu, X.; Wen, W.; Gong, X. Organ-on-a-chip: Recent breakthroughs and future prospects. Biomed. Eng. Online 2020, 19, 1–19. [Google Scholar] [CrossRef] [Green Version]
- Kilic, T.; Navaee, F.; Stradolini, F.; Renaud, P.; Carrara, S. Organs-on-chip monitoring: Sensors and other strategies. Microphysiol. Syst. 2018, 1, 1–32. [Google Scholar] [CrossRef]
- Fuchs, S.; Johansson, S.; Tjell, A.; Werr, G.; Mayr, T.; Tenje, M. In-Line Analysis of Organ-on-Chip Systems with Sensors: Integration, Fabrication, Challenges, and Potential. ACS Biomater. Sci. Eng. 2021, 7, 2926–2948. [Google Scholar] [CrossRef] [PubMed]
- van Noort, D.; Park, S.; Nguyen, N.-T. Towards Human on a Chip: Recent Progress and Future Perspective. Micro Nanosyst. 2014, 6, 215–231. [Google Scholar]
- Park, D.; Lee, J.; Chung, J.J.; Jung, Y.; Kim, S.H. Integrating Organs-on-Chips: Multiplexing, Scaling, Vascularization, and Innervation. Trends Biotechnol. 2019, 38, 99–112. [Google Scholar] [CrossRef]
- Sung, J.H. Multi-organ-on-a-chip for pharmacokinetics and toxicokinetic study of drugs. Expert Opin. Drug Metab. Toxicol. 2021, 17, 969–986. [Google Scholar] [CrossRef]
- Kimura, H.; Sakai, Y.; Fujii, T. Organ/body-on-a-chip based on microfluidic technology for drug discovery. Drug Metab. Pharmacokinet. 2018, 33, 43–48. [Google Scholar] [CrossRef]
- Sung, J.H.; Wang, Y.; Shuler, M.L. Strategies for using mathematical modeling approaches to design and interpret multi-organ microphysiological systems (MPS). APL Bioeng. 2019, 3, 021501. [Google Scholar] [CrossRef]
- Sung, J.H.; Wang, Y.I.; Kim, J.H.; Lee, J.M.; Shuler, M.L. Application of chemical reaction engineering principles to “body-on-a-chip” systems. AIChE J. 2018, 64, 4351–4360. [Google Scholar] [CrossRef]
- Picollet-D’Hahan, N.; Zuchowska, A.; Lemeunier, I.; Le Gac, S. Multiorgan-on-a-Chip: A Systemic Approach To Model and Decipher Inter-Organ Communication. Trends Biotechnol. 2021, 39, 788–810. [Google Scholar] [CrossRef]
- Liu, W.; Song, J.; Du, X.; Zhou, Y.; Li, Y.; Li, R.; Lyu, L.; He, Y.; Hao, J.; Ben, J.; et al. AKR1B10 (Aldo-keto reductase family 1 B10) promotes brain metastasis of lung cancer cells in a multi-organ microfluidic chip model. Acta Biomater. 2019, 91, 195–208. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Hou, Y.; Ai, X.; Sun, J.; Xu, B.; Meng, X.; Zhang, Y.; Zhang, S. Potential applications of microfluidics based blood brain barrier (BBB)-on-chips for in vitro drug development. Biomed. Pharmacother. 2020, 132, 110822. [Google Scholar] [CrossRef]
- Martin, C.R.; Osadchiy, V.; Kalani, A.; Mayer, E.A. The Brain-Gut-Microbiome Axis. Cell. Mol. Gastroenterol. Hepatol. 2018, 6, 133–148. [Google Scholar] [CrossRef] [Green Version]
- Obrenovich, M.E.M. Leaky Gut, Leaky Brain? Microorganisms 2018, 6, 107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shimizu, F.; Nishihara, H.; Kanda, T. Blood–brain barrier dysfunction in immuno-mediated neurological diseases. Immunol. Med. 2018, 41, 120–128. [Google Scholar] [CrossRef] [Green Version]
- Jiang, L.; Li, S.; Zheng, J.; Li, Y.; Huang, H. Recent Progress in Microfluidic Models of the Blood-Brain Barrier. Micromachines 2019, 10, 375. [Google Scholar] [CrossRef] [Green Version]
- Sharma, G.; Sharma, A.R.; Lee, S.-S.; Bhattacharya, M.; Nam, J.-S.; Chakraborty, C. Advances in nanocarriers enabled brain targeted drug delivery across blood brain barrier. Int. J. Pharm. 2019, 559, 360–372. [Google Scholar] [CrossRef] [PubMed]
- Seo, S.; Kim, H.; Sung, J.H.; Choi, N.; Lee, K.; Kim, H.N. Microphysiological systems for recapitulating physiology and function of blood-brain barrier. Biomaterials 2019, 232, 119732. [Google Scholar] [CrossRef]
- Elbakary, B.; Badhan, R.K.S. A dynamic perfusion based blood-brain barrier model for cytotoxicity testing and drug permeation. Sci. Rep. 2020, 10, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Lee, C.S.; Leong, K.W. Advances in microphysiological blood-brain barrier (BBB) models towards drug delivery. Curr. Opin. Biotechnol. 2020, 66, 78–87. [Google Scholar] [CrossRef]
- Shin, Y.; Choi, S.H.; Kim, E.; Bylykbashi, E.; Kim, J.A.; Chung, S.; Kim, D.Y.; Kamm, R.D.; Tanzi, R.E. Blood–Brain Barrier Dysfunction in a 3D In Vitro Model of Alzheimer’s Disease. Adv. Sci. 2019, 6, 1900962. [Google Scholar] [CrossRef] [Green Version]
- Vatine, G.D.; Barrile, R.; Workman, M.J.; Sances, S.; Barriga, B.K.; Rahnama, M.; Barthakur, S.; Kasendra, M.; Lucchesi, C.; Kerns, J.; et al. Human iPSC-Derived Blood-Brain Barrier Chips Enable Disease Modeling and Personalized Medicine Applications. Cell Stem Cell 2019, 24, 995–1005.e6. [Google Scholar] [CrossRef]
- Ashammakhi, N.; Nasiri, R.; de Barros, N.R.; Tebon, P.; Thakor, J.; Goudie, M.; Shamloo, A.; Martin, M.G.; Khademhosseini, A. Gut-on-a-chip: Current progress and future opportunities. Biomaterials 2020, 255, 120196. [Google Scholar] [CrossRef] [PubMed]
- Donkers, J.M.; Amirabadi, H.E.; van de Steeg, E. Intestine-on-a-chip: Next level in vitro research model of the human intestine. Curr. Opin. Toxicol. 2020, 25, 6–14. [Google Scholar] [CrossRef]
- 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 a Chip 2010, 11, 389–392. [Google Scholar] [CrossRef]
- Yu, J.; Peng, S.; Luo, D.; March, J.C. In vitro 3D human small intestinal villous model for drug permeability determination. Biotechnol. Bioeng. 2012, 109, 2173–2178. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.H.; Chi, M.; Yi, B.; Kim, S.H.; Oh, S.; Kim, Y.; Park, S.; Sung, J.H. Three-dimensional intestinal villi epithelium enhances protection of human intestinal cells from bacterial infection by inducing mucin expression. Integr. Biol. 2014, 6, 1122–1131. [Google Scholar] [CrossRef] [PubMed]
- Yi, B.; Shim, K.Y.; Ha, S.K.; Han, J.; Hoang, H.-H.; Choi, I.; Park, S.; Sung, J.H. Three-dimensional in vitro gut model on a villi-shaped collagen scaffold. BioChip J. 2017, 11, 219–231. [Google Scholar] [CrossRef]
- Kim, S.H.; Lee, J.W.; Choi, I.; Kim, Y.-C.; Lee, J.B.; Sung, J.H. A microfluidic device with 3-d hydrogel villi scaffold to simulate intestinal absorption. J. Nanosci. Nanotechnol. 2013, 13, 7220–7228. [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] [PubMed] [Green Version]
- Rudmann, D.G. The Emergence of Microphysiological Systems (Organs-on-chips) as Paradigm-changing Tools for Toxicologic Pathology. Toxicol. Pathol. 2018, 47, 4–10. [Google Scholar] [CrossRef]
- 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]
- Lee, Y.; Kim, M.-H.; Alves, D.R.; Kim, S.; Lee, L.P.; Sung, J.H.; Park, S. Gut–Kidney Axis on Chip for Studying Effects of Antibiotics on Risk of Hemolytic Uremic Syndrome by Shiga Toxin-Producing Escherichia coli. Toxins 2021, 13, 775. [Google Scholar] [CrossRef] [PubMed]
- Trapecar, M.; Wogram, E.; Svoboda, D.; Communal, C.; Omer, A.; Lungjangwa, T.; Sphabmixay, P.; Velazquez, J.; Schneider, K.; Wright, C.W.; et al. Human physiomimetic model integrating microphysiological systems of the gut, liver, and brain for studies of neurodegenerative diseases. Sci. Adv. 2021, 7, eabd1707. [Google Scholar] [CrossRef] [PubMed]
- Maschmeyer, I.; Lorenz, A.K.; Schimek, K.; Hasenberg, T.; Ramme, A.P.; Hübner, J.; Lindner, M.; Drewell, C.; Bauer, S.; Thomas, A.; et al. A four-organ-chip for interconnected long-term co-culture of human intestine, liver, skin and kidney equivalents. Lab Chip 2015, 15, 2688–2699. [Google Scholar] [CrossRef] [Green Version]
- Wagner, K.T.; Nash, T.R.; Liu, B.; Vunjak-Novakovic, G.; Radisic, M. Extracellular Vesicles in Cardiac Regeneration: Potential Applications for Tissues-on-a-Chip. Trends Biotechnol. 2020, 39, 755–773. [Google Scholar] [CrossRef] [PubMed]
- Ural, E.E.; Toomajian, V.; Apu, E.H.; Veletic, M.; Balasingham, I.; Ashammakhi, N.; Kanada, M.; Contag, C.H. Visualizing Extracellular Vesicles and Their Function in 3D Tumor Microenvironment Models. Int. J. Mol. Sci. 2021, 22, 4784. [Google Scholar] [CrossRef]
- Yadid, M.; Lind, J.U.; Ardoña, H.A.M.; Sheehy, S.P.; Dickinson, L.E.; Eweje, F.; Bastings, M.M.C.; Pope, B.; O’Connor, B.B.; Straubhaar, J.R.; et al. Endothelial extracellular vesicles contain protective proteins and rescue ischemia-reperfusion injury in a human heart-on-chip. Sci. Transl. Med. 2020, 12, eaax8005. [Google Scholar] [CrossRef]
- Woo, H.-K.; Sunkara, V.; Park, J.; Kim, T.-H.; Han, J.-R.; Kim, C.-J.; Choi, H.-I.; Kim, Y.-K.; Cho, Y.-K. Exodisc for Rapid, Size-Selective, and Efficient Isolation and Analysis of Nanoscale Extracellular Vesicles from Biological Samples. ACS Nano 2017, 11, 1360–1370. [Google Scholar] [CrossRef]
- Kim, J.; Lee, C.; Kim, I.; Ro, J.; Kim, J.; Min, Y.; Park, J.; Sunkara, V.; Park, Y.-S.; Michael, I.; et al. Three-Dimensional Human Liver-Chip Emulating Premetastatic Niche Formation by Breast Cancer-Derived Extracellular Vesicles. ACS Nano 2020, 14, 14971–14988. [Google Scholar] [CrossRef]
- Mondal, A.; Ashiq, K.A.; Phulpagar, P.; Singh, D.K.; Shiras, A. Effective Visualization and Easy Tracking of Extracellular Vesicles in Glioma Cells. Biol. Proced. Online 2019, 21, 4. [Google Scholar] [CrossRef] [Green Version]
- Liu, T.; Zhu, Y.; Zhao, R.; Wei, X.; Xin, X. Visualization of exosomes from mesenchymal stem cells in vivo by magnetic resonance imaging. Magn. Reson. Imaging 2020, 68, 75–82. [Google Scholar] [CrossRef]
- Morad, G.; Carman, C.V.; Hagedorn, E.J.; Perlin, J.R.; Zon, L.I.; Mustafaoglu, N.; Park, T.-E.; Ingber, D.E.; Daisy, C.C.; Moses, M.A. Tumor-Derived Extracellular Vesicles Breach the Intact Blood–Brain Barrier via Transcytosis. ACS Nano 2019, 13, 13853–13865. [Google Scholar] [CrossRef] [PubMed]
- Oh, H.J.; Shin, Y.; Chung, S.; Hwang, D.W.; Lee, D.S. Convective exosome-tracing microfluidics for analysis of cell-non-autonomous neurogenesis. Biomaterials 2017, 112, 82–94. [Google Scholar] [CrossRef]
- Oh, H.J.; Kim, J.; Park, H.; Chung, S.; Hwang, D.W.; Lee, D.S. Graphene-oxide quenching-based molecular beacon imaging of exosome-mediated transfer of neurogenic miR-193a on microfluidic platform. Biosens. Bioelectron. 2018, 126, 647–656. [Google Scholar] [CrossRef]
- Wiklander, O.P.B.; Nordin, J.Z.; O’Loughlin, A.; Gustafsson, Y.; Corso, G.; Mäger, I.; Vader, P.; Lee, Y.; Sork, H.; Seow, Y.; et al. Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting. J. Extracell. Vesicles 2015, 4, 26316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lara, P.; Chan, A.B.; Cruz, L.J.; Quest, A.F.G.; Kogan, M.J. Exploiting the Natural Properties of Extracellular Vesicles in Targeted Delivery towards Specific Cells and Tissues. Pharmaceutics 2020, 12, 1022. [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. 2019, 15, e1900107. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Liu, F.; Vermesh, O.; Mani, V.; Ge, T.J.; Madsen, S.J.; Sabour, A.; Hsu, E.-C.; Gowrishankar, G.; Kanada, M.; Jokerst, J.V.; et al. The Exosome Total Isolation Chip. ACS Nano 2017, 11, 10712–10723. [Google Scholar] [CrossRef]
- Lim, C.Z.J.; Zhang, L.; Zhang, Y.; Sundah, N.R.; Shao, H. New Sensors for Extracellular Vesicles: Insights on Constituent and Associated Biomarkers. ACS Sens. 2019, 5, 4–12. [Google Scholar] [CrossRef] [PubMed]
Origin | Eukaryotes | Gram-Negative Bacteria | Gram-Positive Bacteria | |||
---|---|---|---|---|---|---|
Type | Exosome | MV | AB | OMV | O-IMV | CMV |
Surface compounds | CD63, TSG101, alix, flotillin | Integrin, selectin, flotillin-2 | Thrombospondin, C3b | LPS | LPS | LTA |
Contents | Protein, lipid, RNA and DNA | Protein, lipid, RNA, and DNA | Cytosolic content (protein, RNAs, fragmented DNA) and cellular organelles | DNA, RNA, periplasmic proteins, peptidoglycans, enzymes, and toxins | DNA, RNA, ATP, periplasmic proteins, cytoplasmic proteins, phages, endolysin, and toxins | DNA, RNA, cytoplasmic proteins, phages, endolysin, enzymes, and toxins |
Biological purposes | Packing cargoes to deliver them to distant cells, involving cell–cell communication, cell maintenance, cell proliferation, and tumor progression | Similar to exosomes | Little known | Bacteria adhesion/invasion, modulation of host cell pathophysiology, host immune evasion, killing competing bacterial species, and antibiotics resistance | Role in interdomain signaling is unclear | Bacteria adhesion/invasion, modulation of host cell pathophysiology, killing competing bacterial species, and antibiotics resistance |
References | [40,41,42] | [40,42] | [40,41,43] | [17,33,44,45] | [30,33,46] | [31,32,33,45,47,48] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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
Kim, M.-H.; van Noort, D.; Sung, J.H.; Park, S. Organ-on-a-Chip for Studying Gut-Brain Interaction Mediated by Extracellular Vesicles in the Gut Microenvironment. Int. J. Mol. Sci. 2021, 22, 13513. https://doi.org/10.3390/ijms222413513
Kim M-H, van Noort D, Sung JH, Park S. Organ-on-a-Chip for Studying Gut-Brain Interaction Mediated by Extracellular Vesicles in the Gut Microenvironment. International Journal of Molecular Sciences. 2021; 22(24):13513. https://doi.org/10.3390/ijms222413513
Chicago/Turabian StyleKim, Min-Hyeok, Danny van Noort, Jong Hwan Sung, and Sungsu Park. 2021. "Organ-on-a-Chip for Studying Gut-Brain Interaction Mediated by Extracellular Vesicles in the Gut Microenvironment" International Journal of Molecular Sciences 22, no. 24: 13513. https://doi.org/10.3390/ijms222413513
APA StyleKim, M. -H., van Noort, D., Sung, J. H., & Park, S. (2021). Organ-on-a-Chip for Studying Gut-Brain Interaction Mediated by Extracellular Vesicles in the Gut Microenvironment. International Journal of Molecular Sciences, 22(24), 13513. https://doi.org/10.3390/ijms222413513