AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (6.9 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Review Article | Open Access

Perspectives on organ-on-a-chip technology for natural products evaluation

Xin WangYu-Hang MiaoXiao-Min ZhaoXin LiuYan-Wei HuDa-Wei Deng( )
Department of Biomedical Engineering/Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
Show Author Information

Highlights

(1) Explored the mechanisms and principles of organ-on-chip technology, which simulates human organs' physiological environments and functions.

(2) Organ-on-a-chip technology offers significant advantages in evaluating drug activity, physicochemical properties, and safety, bridging the gap between in vitro assessment models and the complex pathophysiology in vivo.

(3) Outlined the research progress on various organ-on-a-chip models used to simulate complex human diseases, offering a new paradigm for establishing the next generation of natural drug evaluation platforms.

Graphical Abstract

Organ-on-a-chip (OoC) is an emerging technology based on microfluidic platforms and in vitro cell culture, capable of mimicking the physiological environment and functions of human organs, for disease modeling and drug assessment. As illustrated in Figure 1, this review outlines the research progress of various organ models on chips for simulating complex human disease conditions, providing new paradigms for establishing the next generation of natural drug assessment platforms.

Abstract

Natural products have always been a treasure trove for clinical drug development and a source of inspiration for lead compounds in the process of new drug discovery. However, two-dimensional cell cultures and animal models in the traditional drug development model have serious limitations in generalizing human physiopathology and cannot accurately predict the real clinical response of the human body to drugs, which brings obstacles and challenges to drug evaluation. Organ-on-a-chip (OoC) is an emerging technology based on microfluidic platforms and in vitro cell culture that can mimic the physiological environment and function of human organs for disease modeling and drug evaluation. In this review, we explore several major examples of how human single-OoC systems can be used to simulate complex disease models and outline recent advances in organoids for natural drug screening. Finally, we summarize the challenges and future trends that OoCs must overcome in drug discovery and development. Overall, this review highlights that OoCs, instead of animal models, open new avenues for natural drug development and evaluation, therapeutic innovation, and in vivo embodiment of personalized medicine.

References

[1]

Wishart, D. S., Girod, S., Peters, H., et al. ChemFOnt: the chemical functional ontology resource. Nucleic Acids Research, 2023, 51: D1220–D1229. https://doi.org/10.1093/nar/gkac919

[2]

Jiamu, M., Jianling, Y., Xueyang, R., et al. Machine learning-assisted data-driven optimization and understanding of the multiple stage process for extraction of polysaccharides and secondary metabolites from natural products. Green Chemistry, 2023, 25: 3057–3068. https://doi.org/10.1039/d2gc04574e

[3]

Lu, X., Friedrich, L. J., Efferth, T. Natural products targeting tumour angiogenesis. British Journal of Pharmacology, 2023, 2023: 1–43. https://doi.org/10.1111/bph.16232

[4]

Grigalunas, M., Brakmann, S., Waldmann, H. Chemical evolution of natural product structure. Journal of the American Chemical Society, 2022, 144: 3314–3329. https://doi.org/10.1021/jacs.1c11270

[5]

Zhang, L., Song, J., Kong, L., et al. The strategies and techniques of drug discovery from natural products. Pharmacology & Therapeutics, 2020, 216: 107686. https://doi.org/10.1016/j.pharmthera.2020.107686

[6]

Grigalunas, M., Burhop, A., Zinken, S., et al. Natural product fragment combination to performance-diverse pseudo-natural products. Nature Communications, 2021, 12: 1883. https://doi.org/10.1038/s41467-021-22174-4

[7]

Roope, L. S. J. The economic challenges of new drug development. Journal of Controlled Release, 2022, 345: 275–277. https://doi.org/10.1016/j.jconrel.2022.03.023

[8]

Loscalzo, J. Multi-Omics and single-cell omics: new tools in drug target discovery. Arteriosclerosis Thrombosis and Vascular Biology, 2024, 44: 759–762. https://doi.org/10.1161/ATVBAHA.124.320686

[9]

Sarvas, H., Carlisle, B., Dolter, S., et al. Impact of precision medicine on efficiencies of novel drug development in cancer. Journal of the National Cancer Institute, 2020, 112: 859–862. https://doi.org/10.1093/jnci/djz212

[10]

Chesney, J., Puzanov, I., Ross, M. The importance of outcome and precise evaluation in economic analysis of cancer drugs. JAMA Dermatology, 2019, 155: 862–863. https://doi.org/10.1001/jamadermatol.2019.0594

[11]

Caiado, J., Castells, M. C. Drug desensitizations for chemotherapy: safety and efficacy in preventing anaphylaxis. Current Allergy and Asthma Reports, 2021, 21: 37. https://doi.org/10.1007/s11882-021-01014-x

[12]

Xu, D., Xiao, H., Wang, S., et al. Universal and sensitive drug assessment biosensing platform using optimal mechanical beating detection of single cardiomyocyte. ACS Nano, 2022, 16: 15484–15494. https://doi.org/10.1021/acsnano.2c08049

[13]

Hang, X., Huang, Z., He, S., et al. A nano-electroporation-DNA tensioner platform enhances intracellular delivery and mechanical analysis toward rapid drug assessment. Small Methods, 2024, 8: e2300915. https://doi.org/10.1002/smtd.202300915

[14]

Henderson, K., Lewis, Sloan, C. E., et al. Effectiveness and safety of drugs for obesity. The British Medical Journal, 2024, 384: e072686. https://doi.org/10.1136/bmj-2022-072686

[15]

Adhyaru, B. B., Jacobson, T. A. Safety and efficacy of statin therapy. Nature Reviews Cardiology, 2018, 15: 757–769. https://doi.org/10.1038/s41569-018-0098-5

[16]

Jenei, K., Aziz, Z., Booth, C., et al. Cancer medicines on the WHO model list of essential medicines: processes, challenges, and a way forward. Lancet Global Health, 2022, 10: e1860–e1866. https://doi.org/10.1016/S2214-109X(22)00376-X

[17]

Khalil, A. S., Jaenisch, R., Mooney, D. J. Engineered tissues and strategies to overcome challenges in drug development. Advanced Drug Delivery Reviews, 2020, 158: 116–139. https://doi.org/10.1016/j.addr.2020.09.012

[18]

Begley, C. G., Ellis, L. M. Drug development: raise standards for preclinical cancer research. Nature, 2012, 483: 531–533. https://doi.org/10.1038/483531a

[19]
Brancato, V., Oliveira, J. M., Correlo, V. M., et al. Could 3D models of cancer enhance drug screening? Biomaterials, 2020 , 232: 119744. https://doi.org/10.1016/j.biomaterials.2019.119744
[20]

Zhao, Y., Demirci, U., Chen, Y., et al. Multiscale brain research on a microfluidic chip. Lab on a Chip, 2020, 20: 1531–1543. https://doi.org/10.1039/c9lc01010f

[21]

Jang, K. J., Otieno, M. A., Ronxhi, J., et al. Reproducing human and cross-species drug toxicities using a liver-chip. Science Translational Medicine, 2019, 11: eaax5516. https://doi.org/10.1126/scitranslmed.aax5516

[22]

Donowitz, M., Turner, J. R., Verkman, A. S., et al. Current and potential future applications of human stem cell models in drug development. Journal of Clinical Investigation, 2020, 130: 3342–3344. https://doi.org/10.1172/JCI138645

[23]

Jalili-Firoozinezhad, S., Miranda, C. C., Cabral, J. M. S. Modeling the human body on microfluidic chips. Trends in Biotechnology, 2021, 39: 838–852. https://doi.org/10.1016/j.tibtech.2021.01.004

[24]

Utada, A. S., Lorenceau, E., Link, D. R., et al. Monodisperse double emulsions generated from a microcapillary device. Science, 2005, 308: 537–541. https://doi.org/10.1126/science.1109164

[25]

Valencia, P. M., Farokhzad, O. C., Karnik, R., et al. Microfluidic technologies for accelerating the clinical translation of nanoparticles. Nature Nanotechnology, 2012, 7: 623–629. https://doi.org/10.1038/nnano.2012.168

[26]

Shih, S. C., Gach, P. C., Sustarich, J., et al. A droplet-to-digital (D2D) microfluidic device for single cell assays. Lab on a Chip, 2015, 15: 225–236. https://doi.org/10.1039/c4lc00794h

[27]

Hou, X., Zhang, Y. S., Santiago, G. T. D., et al. Interplay between materials and microfluidics. Nature Reviews Materials, 2017, 2: 17016. https://doi.org/10.1038/natrevmats.2017.16

[28]

Wang, B., Prinsen, P., Wang, H., et al. Macroporous materials: microfluidic fabrication, functionalization and applications. Chemical Society Reviews, 2017, 46: 855–914. https://doi.org/10.1039/c5cs00065c

[29]

Shang, L., Cheng, Y., Zhao, Y. Emerging droplet microfluidics. Chemical Reviews, 2017, 117: 7964–8040. https://doi.org/10.1021/acs.chemrev.6b00848

[30]

Sivagnanam, V., Gijs, M. A. Exploring living multicellular organisms, organs, and tissues using microfluidic systems. Chemical Reviews, 2013, 113: 3214–3247. https://doi.org/10.1021/cr200432q

[31]

Fuchs, S., Rieger, V., Tjell, A. O., et al. Optical glucose sensor for microfluidic cell culture systems. Biosensors & Bioelectronics, 2023, 237: 115491. https://doi.org/10.1016/j.bios.2023.115491

[32]

Si, L., Bai, H., Rodas, M., et al. A human-airway-on-a-chip for the rapid identification of candidate antiviral therapeutics and prophylactics. Nature Biomedical Engineering, 2021, 5: 815–829. https://doi.org/10.1038/s41551-021-00718-9

[33]

Zhang, B., Korolj, A., Lai, B. F. L., et al. Advances in organ-on-a-chip engineering. Nature Reviews Materials, 2018, 3: 257–278. https://doi.org/10.1038/s41578-018-0034-7

[34]

Park, S. E., Georgescu, A., Huh, D. Organoids-on-a-chip. Science, 2019, 364: 960–965. https://doi.org/10.1126/science.aaw7894

[35]
Shoji, J. Y., Davis, R. P., Mummery, C. L., et al. Global meta-analysis of organoid and organ-on-chip research. Advanced Healthcare Materials, 2023 : e2301067. https://doi.org/10.1002/adhm.202301067
[36]

Huang, Y., Liu, T., Huang, Q., et al. From Organ-on-a-Chip to Human-on-a-Chip: A review of research progress and latest applications. ACS Sens, 2024, 9: 3466–3488. https://doi.org/10.1021/acssensors.4c00004

[37]

Monteduro, A. G., Rizzato, S., Caragnano, G., et al. Organs-on-chips technologies-a guide from disease models to opportunities for drug development. Biosensors & Bioelectronics, 2023, 231: 115271. https://doi.org/10.1016/j.bios.2023.115271

[38]

Sun, W., Luo, Z., Lee, J., et al. Organ-on-a-chip for cancer and immune organs modeling. Advanced Healthcare Materials, 2019, 8: e1801363. https://doi.org/10.1002/adhm.201801363

[39]

Huh, D., Matthews, B. D., Mammoto, A., et al. Reconstituting organ-level lung functions on a chip. Science, 2010, 328: 1662–1668. https://doi.org/10.1126/science.1188302

[40]

Vunjak-Novakovic, G., Ronaldson-Bouchard, K., Radisic, M. Organs-on-a-chip models for biological research. Cell, 2021, 184: 4597–4611. https://doi.org/10.1016/j.cell.2021.08.005

[41]

Strelez, C., Jiang, H. Y., Mumenthaler, S. M. Organs-on-chips: a decade of innovation. Trends in Biotechnology, 2023, 41: 278–280. https://doi.org/10.1016/j.tibtech.2023.01.004

[42]

Yan, J., Li, Z., Guo, J., et al. Organ-on-a-chip: a new tool for in vitro research. Biosensors and Bioelectronics, 2022, 216: 114626. https://doi.org/10.1016/j.bios.2022.114626

[43]

Picollet-D'hahan, N., Zuchowska, A., Lemeunier, I., et al. Multiorgan-on-a-chip: a systemic approach to model and decipher inter-organ communication. Trends in Biotechnology, 2021, 39: 788–810. https://doi.org/10.1016/j.tibtech.2020.11.014

[44]

Joseph, X., Akhil, V., Arathi, A., et al. Comprehensive development in organ-on-a-chip technology. Journal of Pharmaceutical Sciences, 2022, 111: 18–31. https://doi.org/10.1016/j.xphs.2021.07.014

[45]

Li, X., Zhu, H., Gu, B., et al. Advancing intelligent organ-on-a-chip systems with comprehensive in situ bioanalysis. Advanced Materials, 2023, 36: e2305268. https://doi.org/10.1002/adma.202305268

[46]

Wang, Y. I., Carmona, C., Hickman, J. J., et al. Multiorgan microphysiological systems for drug development: strategies, advances, and challenges. Advanced Healthcare Materials, 2018, 7: 1701000. https://doi.org/10.1002/adhm.201701000

[47]

Sasserath, T., Rumsey, J. W., McAleer, C. W., et al. Differential monocyte actuation in a three-organ functional innate immune system-on-a-chip. Advanced Science, 2020, 7: 2000323. https://doi.org/10.1002/advs.202000323

[48]

Kawakita, S., Mandal, K., Mou, L., et al. Organ-on-a-chip models of the blood-brain barrier: recent advances and future prospects. Small, 2022, 18: e2201401. https://doi.org/10.1002/smll.202201401

[49]

Oddo, A., Peng, B., Tong, Z., et al. Advances in microfluidic blood-brain barrier (BBB) models. Trends in Biotechnology, 2019, 37: 1295–1314. https://doi.org/10.1016/j.tibtech.2019.04.006

[50]

Xu, J., Ma, C., Hua, M., et al. CNS and CNS diseases in relation to their immune system. Frontiers in Immunology, 2022, 13: 1063928. https://doi.org/10.3389/fimmu.2022.1063928

[51]

Bi, W., Cai, S., Lei, T., et al. Implementation of blood-brain barrier on microfluidic chip: recent advance and future prospects. Ageing Research Reviews, 2023, 87: 101921. https://doi.org/10.1016/j.arr.2023.101921

[52]

Chung, M., Lee, S., Lee, B. J., et al. Wet-AMD on a chip: modeling outer blood-retinal barrier in vitro. Advanced Healthcare Materials, 2018, 7: 1700028. https://doi.org/10.1002/adhm.201700028

[53]

Booth, R., Kim, H. Characterization of a microfluidic in vitro model of the blood-brain barrier (muBBB). Lab on a Chip, 2012, 12: 1784–1792. https://doi.org/10.1039/c2lc40094d

[54]

Zhang, X. Z., Qian, S. S., Zhang, Y. J., et al. Salvia miltiorrhiza: a source for anti-Alzheimer's disease drugs. Pharmaceutical Biology, 2016, 54: 18–24. https://doi.org/10.3109/13880209.2015.1027408

[55]

Fang, J., He, Y., Cao, Y., et al. Effect of P-glycoprotein on the blood-brain barrier transport of the major active constituents of salvia miltiorrhiza based on the MDCK-MDR1 cell model. ACS Chemical Neuroscience, 2023, 14: 766–772. https://doi.org/10.1021/ acschemneuro.2c00757

[56]

Bhise, N. S., Manoharan, V., Massa, S., et al. A liver-on-a-chip platform with bioprinted hepatic spheroids. Biofabrication, 2016, 8: 014101. https://doi.org/10.1088/1758-5090/8/1/014101

[57]

Li, Z., Li, J., Sun, M., et al. Analysis of metabolites and metabolism-mediated biological activity assessment of ginsenosides on microfluidic co-culture system. Frontiers in Pharmacolog, 2023, 14: 1046722. https://doi.org/10.3389/fphar.2023.1046722

[58]

Farooqi, H. M. U., Kang, B., Khalid, M. A. U., et al. Real-time monitoring of liver fibrosis through embedded sensors in a microphysiological system. Nano Convergence, 2021, 8: 3. https://doi.org/10.1186/s40580-021-00253-y

[59]

Marmol, I., Abizanda-Campo, S., Ayuso, J. M., et al. Towards novel biomimetic in vitro models of the blood-brain barrier for drug permeability evaluation. Bioengineering, 2023, 10: 572. https://doi.org/10.3390/bioengineering10050572

[60]
Vatine, G. D., Barrile, R., Workman, M. J., et al. Human iPSC-derived blood-brain barrier chips enable disease modeling and personalized medicine applications. Cell Stem Cell, 2019 , 24: 995-1005 e1006. https://doi.org/10.1016/j.stem.2019.05.011
[61]

Jiang, X., Du, B., Zheng, J. Glutathione-mediated biotransformation in the liver modulates nanoparticle transport. Nature Nanotechnology, 2019, 14: 874–882. https://doi.org/10.1038/s41565-019-0499-6

[62]

Mancio-Silva, L., Fleming, H. E., Miller, A. B., et al. Improving drug discovery by nucleic acid delivery in engineered human microlivers. Cell Metabolism, 2019, 29: 727–735. https://doi.org/10.1016/j.cmet.2019.02.003

[63]

Moradi, E., Jalili-Firoozinezhad, S., Solati-Hashjin, M. Microfluidic organ-on-a-chip models of human liver tissue. Acta Biomaterialia, 2020, 116: 67–83. https://doi.org/10.1016/j.actbio.2020.08.041

[64]

Messelmani, T., Morisseau, L., Sakai, Y., et al. Liver organ-on-chip models for toxicity studies and risk assessment. Lab on a Chip, 2022, 22: 2423–2450. https://doi.org/10.1039/d2lc00307d

[65]

van Midwoud, P. M., Groothuis, G. M., Merema, M. T., et al. Microfluidic biochip for the perifusion of precision-cut rat liver slices for metabolism and toxicology studies. Biotechnology And Bioengineering, 2010, 105: 184–194. https://doi.org/10.1002/bit.22516

[66]

Zuchowska, A., Kwapiszewska, K., Chudy, M., et al. Studies of anticancer drug cytotoxicity based on long-term HepG2 spheroid culture in a microfluidic system. Electrophoresis, 2017, 38: 1206–1216. https://doi.org/10.1002/elps.201600417

[67]

Sun, Y. K., Zhang, Y. F., Xie, L., et al. Progress in the treatment of drug-induced liver injury with natural products. Pharmacological Research, 2022, 183: 106361. https://doi.org/10.1016/j.phrs.2022.106361

[68]

Perl, A. J., Schuh, M. P., Kopan, R. Regulation of nephron progenitor cell lifespan and nephron endowment. Nature Reviews Nephrology, 2022, 18: 683–695. https://doi.org/10.1038/s41581-022-00620-w

[69]

Qu, Y., An, F., Luo, Y., et al. A nephron model for study of drug-induced acute kidney injury and assessment of drug-induced nephrotoxicity. Biomaterials, 2018, 155: 41–53. https://doi.org/10.1016/j.biomaterials.2017.11.010

[70]

Perazella, M. A., Rosner, M. H. Drug-induced acute kidney injury. Clinical Journal of the American Society of Nephrology, 2022, 17: 1220–1233. https://doi.org/10.2215/CJN.11290821

[71]

Oxburgh, L. Kidney nephron determination. Annual Review of Cell and Developmental Biology, 2018, 34: 427–450. https://doi.org/10.1146/annurev-cellbio-100616-060647

[72]

Vize, P. D. A beautiful, complex simplicity: the origins of nephron segmentation uncovered by single-cell sequencing of the pronephros. Kidney International, 2023, 103: 23–25. https://doi.org/ 10.1016/j.kint.2022.09.013

[73]

Li, X., Tian, T. Phytochemical characterization of Mentha spicata L. under differential dried-conditions and associated nephrotoxicity screening of main compound with organ-on-a-chip. Frontiers in Pharmacology, 2018, 9: 1067. https://doi.org/10.3389/fphar.2018.01067

[74]

Weber, E. J., Lidberg, K. A., Wang, L., et al. Human kidney on a chip assessment of polymyxin antibiotic nephrotoxicity. JCI Insight, 2018, 3: e123673. https://doi.org/10.1172/jci.insight.123673

[75]

Tabibzadeh, N., Morizane, R. Advancements in therapeutic development: kidney organoids and organs on a chip. Kidney International, 2024, 105: 702–708. https://doi.org/10.1016/j.kint.2023.11.035

[76]

Grosberg, A., Alford, P. W., McCain, M. L., et al. Ensembles of engineered cardiac tissues for physiological and pharmacological study: heart on a chip. Lab on a Chip, 2011, 11: 4165–4173. https://doi.org/10.1039/c1lc20557a

[77]

Caluori, G., Pribyl, J., Pesl, M., et al. Non-invasive electromechanical cell-based biosensors for improved investigation of 3D cardiac models. Biosensors & Bioelectronics, 2019, 124/125: 129–135. https://doi.org/10.1016/j.bios.2018.10.021

[78]

Telles-Silva, K. A., Pacheco, L., Komatsu, S., et al. Applied hepatic bioengineering: modeling the human liver using organoid and liver-on-a-chip technologies. Frontiers in Bioengineering and Biotechnology, 2022, 10: 845360. https://doi.org/10.3389/fbioe.2022.845360

[79]

Ashammakhi, N., Wesseling-Perry, K., Hasan, A., et al. Kidney-on-a-chip: untapped opportunities. Kidney International, 2018, 94: 1073–1086. https://doi.org/10.1016/j.kint.2018.06.034

[80]

Chen, X., Liu, S., Han, M., et al. Engineering cardiac tissue for advanced heart-on-a-chip platforms. Advanced Healthcare Materials, 2024, 13: e2301338. https://doi.org/10.1002/adhm.202301338

[81]

Butler, D., Reyes, D. R. Heart-on-a-chip systems: disease modeling and drug screening applications. Lab on a Chip, 2024, 24: 1494–1528. https://doi.org/10.1039/d3lc00829k

[82]

Zhao, Y., Rafatian, N., Wang, E. Y., et al. Towards chamber specific heart-on-a-chip for drug testing applications. Advanced Drug Delivery Reviews, 2020, 165-166: 60–76. https://doi.org/10.1016/j.addr.2019.12.002

[83]

Eschenhagen, T., Bolli, R., Braun, T., et al. Cardiomyocyte regeneration: a consensus statement. Circulation, 2017, 136: 680–686. https://doi.org/10.1161/CIRCULATIONAHA.117.029343

[84]

Deir, S., Mozhdehbakhsh Mofrad, Y., Mashayekhan, S., et al. Step-by-step fabrication of heart-on-chip systems as models for cardiac disease modeling and drug screening. Talanta, 2024, 266: 124901. https://doi.org/10.1016/j.talanta.2023.124901

[85]

Zhang, Y. S., Arneri, A., Bersini, S., et al. Bioprinting 3D microfibrous scaffolds for engineering endothelialized myocardium and heart-on-a-chip. Biomaterials, 2016, 110: 45–59. https://doi.org/ 10.1016/j.biomaterials.2016.09.003

[86]

Deng, S., Li, C., Cao, J., et al. Organ-on-a-chip meets artificial intelligence in drug evaluation. Theranostics, 2023, 13: 4526–4558. https://doi.org/10.7150/thno.87266

[87]

Bein, A., Fadel, C. W., Swenor, B., et al. Nutritional deficiency in an intestine-on-a-chip recapitulates injury hallmarks associated with environmental enteric dysfunction. Nature Biomedical Engineering, 2022, 6: 1236–1247. https://doi.org/10.1038/s41551-022-00899-x

[88]
Shin, W., Hackley, L. A., Kim, H. J. “Good fences make good neighbors”: how does the human gut microchip unravel mechanism of intestinal inflammation? Gut Microbes, 2020 , 11: 581–586. https://doi.org/10.1080/19490976.2019.1626684
[89]

Moossavi, S., Arrieta, M. C., Sanati-Nezhad, A., et al. Gut-on-chip for ecological and causal human gut microbiome research. Trends in Microbiology, 2022, 30: 710–721. https://doi.org/10.1016/j.tim.2022.01.014

[90]

Margolis, K. G., Gershon, M. D. Enteric neuronal regulation of intestinal inflammation. Trends in Neurosciences, 2016, 39: 614–624. https://doi.org/10.1016/j.tins.2016.06.007

[91]

Marrero, D., Pujol-Vila, F., Vera, D., et al. Gut-on-a-chip: Mimicking and monitoring the human intestine. Biosensors & Bioelectronics, 2021, 181: 113156. https://doi.org/10.1016/j.bios.2021.113156

[92]
Kim, R., Sung, J. H. Recent advances in gut- and gut-organ-axis-on-a-chip models. Advanced Healthcare Materials, 2024 : e2302777. https://doi.org/10.1002/adhm.202302777
[93]

Guo, Y., Luo, R., Wang, Y., et al. SARS-CoV-2 induced intestinal responses with a biomimetic human gut-on-chip. Science Bulletin, 2021, 66: 783–793. https://doi.org/10.1016/j.scib.2020.11.015

[94]

Jalili-Firoozinezhad, S., Gazzaniga, F. S., Calamari, E. L., et al. A complex human gut microbiome cultured in an anaerobic intestine-on-a-chip. Nature Biomedical Engineering, 2019, 3: 520–531. https://doi.org/10.1038/s41551-019-0397-0

[95]

Kim, H. J., Huh, D., Hamilton, G., et al. Human gut-on-a-chip inhabited by microbial flora that experiences intestinal peristalsis-like motions and flow. Lab on a Chip, 2012, 12: 2165–2174. https://doi.org/10.1039/c2lc40074j

[96]

Kim, H. J., Li, H., Collins, J. J., et al. Contributions of microbiome and mechanical deformation to intestinal bacterial overgrowth and inflammation in a human gut-on-a-chip. Proceedings of the National Academy of Sciences of the United States of America, 2016, 113: E7–E15. https://doi.org/10.1073/pnas.1522193112

[97]

Jeon, M. S., Choi, Y. Y., Mo, S. J., et al. Contributions of the microbiome to intestinal inflammation in a gut-on-a-chip. Nano Convergence, 2022, 9: 8. https://doi.org/10.1186/s40580-022-00299-6

[98]

Pocock, K., Delon, L., Bala, V., et al. Intestine-on-a-chip microfluidic model for efficient in vitro screening of oral chemotherapeutic uptake. ACS Biomaterials Science & Engineering, 2017, 3: 951–959. https://doi.org/10.1021/acsbiomaterials.7b00023

[99]

Chen, L., Dai, Z., Ge, C., et al. Specific metabolic response of patient-derived organoids to curcumin of colorectal cancer. Journal of Chromatography B, 2022, 1203: 123260. https://doi.org/10.1016/ j.jchromb.2022.123260

[100]

Yang, S., Zhang, T., Ge, Y., et al. Sentinel supervised lung-on-a-chip: a new environmental toxicology platform for nanoplastic-induced lung injury. Journal of Hazardous Materials, 2023, 458: 131962. https://doi.org/10.1016/j.jhazmat.2023.131962

[101]

Gkatzis, K., Taghizadeh, S., Huh, D., et al. Use of three-dimensional organoids and lung-on-a-chip methods to study lung development, regeneration and disease. European Respiratory Journal, 2018, 52: 1800876. https://doi.org/10.1183/13993003.00876-2018

[102]

Dasgupta, Q., Jiang, A., Wen, A. M., et al. A human lung alveolus-on-a-chip model of acute radiation-induced lung injury. Nature Communications, 2023, 14: 6506. https://doi.org/10.1038/s41467-023-42171-z

[103]

Zhu, Y., Sun, L., Wang, Y., et al. A biomimetic human lung-on-a-chip with colorful display of microphysiological breath. Advanced Materials, 2022, 34: e2108972. https://doi.org/10.1002/adma.202108972

[104]

Zhang, M., Xu, C., Jiang, L., et al. A 3D human lung-on-a-chip model for nanotoxicity testing. Toxicology Research, 2018, 7: 1048–1060. https://doi.org/10.1039/c8tx00156a

[105]

Yang, X., Li, K., Zhang, X., et al. Nanofiber membrane supported lung-on-a-chip microdevice for anti-cancer drug testing. Lab on a Chip, 2018, 18: 486–495. https://doi.org/10.1039/c7lc01224a

[106]

Huh, D., Leslie, D. C., Matthews, B. D., et al. A human disease model of drug toxicity-induced pulmonary edema in a lung-on-a-chip microdevice. Science Translational Medicine, 2012, 4: 159ra147. https://doi.org/10.1126/scitranslmed.3004249

[107]

Louhivuori, L., Kanatani, S., Uhlen, P. Predicting a tumour's drug uptake. Nature Biomedical Engineering, 2018, 2: 717–718. https://doi.org/10.1038/s41551-018-0311-1

[108]
Samadian, H., Jafari, S., Sepand, M. R., et al. 3D bioprinting technology to mimic the tumor microenvironment: tumor-on-a-chip concept. Materials Today Advances, 2021 , 12: 2590–0498. https://doi.org/10.1016/j.mtadv.2021.100160
[109]
Mi, X., Su, Z., Yue, X., et al. 3D bioprinting tumor models mimic the tumor microenvironment for drug screening. Biomaterials Science, 2023 , 11: 3813–3827. https://doi.org/10.1039/d3bm00159h
[110]
Kumari, S., Advani, D., Sharma, S., et al. Combinatorial therapy in tumor microenvironment: where do we stand? Biochimica et Biophysica Acta-Reviews on Cancer, 2021 , 1876: 188585. https://doi.org/10.1016/j.bbcan.2021.188585
[111]

Zhang, J., Tavakoli, H., Ma, L., et al. Immunotherapy discovery on tumor organoid-on-a-chip platforms that recapitulate the tumor microenvironment. Advanced Drug Delivery Reviews, 2022, 187: 114365. https://doi.org/10.1016/j.addr.2022.114365

[112]

Li, C., Holman, J. B., Shi, Z., et al. On-chip modeling of tumor evolution: advances, challenges and opportunities. Mater Today Bio, 2023, 21: 100724. https://doi.org/10.1016/j.mtbio.2023.100724

[113]

Wang, H. F., Ran, R., Liu, Y., et al. Tumor-vasculature-on-a-chip for investigating nanoparticle extravasation and tumor accumulation. ACS Nano, 2018, 12: 11600–11609. https://doi.org/10.1021/acsnano.8b06846

[114]

Khalid, M. A. U., Kim, Y. S., Ali, M., et al. A lung cancer-on-chip platform with integrated biosensors for physiological monitoring and toxicity assessment. Biochemical Engineering Journal, 2020, 155: 107469. https://doi.org/10.1016/j.bej.2019.107469

[115]

Xie, M., Gao, Q., Fu, J., et al. Bioprinting of novel 3D tumor array chip for drug screening. Bio-Design and Manufacturing, 2020, 2020: 175–188. https://doi.org/10.1007/s42242-020-00078-4

[116]

Kamei, K. I., Kato, Y., Hirai, Y., et al. Integrated heart/cancer on a chip to reproduce the side effects of anti-cancer drugs in vitro. RSC Advances, 2017, 7: 36777. https://doi.org/10.1039/c7ra07716e

[117]

Vidi, P. A., Maleki, T., Ochoa, M., et al. Disease-on-a-chip: mimicry of tumor growth in mammary ducts. Lab on a Chip, 2013, 14: 172. https://doi.org/10.1039/c3lc50819f

Food & Medicine Homology
Article number: 9420013
Cite this article:
Wang X, Miao Y-H, Zhao X-M, et al. Perspectives on organ-on-a-chip technology for natural products evaluation. Food & Medicine Homology, 2024, 1(2): 9420013. https://doi.org/10.26599/FMH.2024.9420013

578

Views

172

Downloads

0

Crossref

Altmetrics

Received: 24 April 2024
Revised: 03 July 2024
Accepted: 04 July 2024
Published: 07 August 2024
© National R & D Center for Edible Fungus Processing Technology 2024. Published by Tsinghua University Press.

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return