Applications of Omics Technologies for Three-Dimensional In Vitro Disease Models
Publication: Tissue Engineering Part C: Methods
Volume 27, Issue Number 3
Abstract
Omics technologies, such as genomics, epigenomics, transcriptomics, proteomics, metabolomics, lipidomics, multiomics, and integrated modalities, have greatly contributed to our understanding of various diseases by enabling researchers to probe the molecular wiring of cellular systems in a high-throughput and precise manner. With the development of tissue-engineered three-dimensional (3D) in vitro disease models, such as organoids and spheroids, there is potential of integrating omics technologies with 3D disease models to elucidate the complex links between genotype and phenotype. These 3D disease models have been used to model cancer, infectious disease, toxicity, neurological disorders, and others. In this review, we provide an overview of omics technologies, highlight current and emerging studies, discuss the associated experimental design considerations, barriers and challenges of omics technologies, and provide an outlook on the future applications of omics technologies with 3D models. Overall, this review aims to provide a valuable resource for tissue engineers seeking to leverage omics technologies for diving deeper into biological discovery.
Impact statement
With the emergence of three-dimensional (3D) in vitro disease models, tissue engineers are increasingly interested to investigate these systems to address biological questions related to disease mechanism, drug target discovery, therapy resistance, and more. Omics technologies are a powerful and high-throughput approach, but their application for 3D disease models is not maximally utilized. This review illustrates the achievements and potential of using omics technologies to leverage the full potential of 3D in vitro disease models. This will improve the quality of such models, advance our understanding of disease, and contribute to therapy development.
Get full access to this article
View all available purchase options and get full access to this article.
References
1. Karczewski, K.J., and Snyder, M.P. Integrative omics for health and disease. Nat Rev Genet 19, 299, 2018.
2. Olivier, M., Asmis, R., Hawkins, G.A., et al. The need for multi-omics biomarker signatures in precision medicine. Int J Mol Sci 20, 4781, 2019.
3. McShane, L.M., Cavenagh, M.M., Lively, T.G., et al. Criteria for the use of omics-based predictors in clinical trials: explanation and elaboration. BMC Med 11, 220, 2013.
4. Ibrahim, R., Pasic, M., and Yousef, G.M. Omics for personalized medicine: defining the current we swim in. Expert Rev Mol Diagn 16, 719, 2016.
5. Heather, J.M., and Chain, B. The sequence of sequencers: the history of sequencing DNA. Genomics 107, 1, 2016.
6. Hood, L., and Rowen, L. The human genome project: big science transforms biology and medicine. Genome Med 5, 79, 2013.
7. Klemm, S.L., Shipony, Z., and Greenleaf, W.J. Chromatin accessibility and the regulatory epigenome. Nat Rev Genet 20, 207, 2019.
8. Wang, K.C., and Chang, H.Y. Epigenomics technologies and applications. Circ Res 122, 1191, 2018.
9. Wang, Z., Gerstein, M., and Snyder, M. RNA-Seq: a revolutionary tool for transcriptomics. Nat Rev Genet 10, 57, 2009.
10. Nagalakshmi, U., Wang, Z., Waern, K., et al. The transcriptional landscape of the yeast genome defined by RNA sequencing. Science (80-) 320, 1344, 2008.
11. Wilhelm, B.T., Marguerat, S., Watt, S., et al. Dynamic repertoire of a eukaryotic transcriptome surveyed at single-nucleotide resolution. Nature 453, 1239, 2008.
12. Tang, F., Barbacioru, C., Wang, Y., et al. mRNA-Seq whole-transcriptome analysis of a single cell. Nat Methods 6, 377, 2009.
13. Grindberg, R.V., Yee-Greenbaum, J.L., McConnell, M.J., et al. RNA-sequencing from single nuclei. Proc Natl Acad Sci U S A 110, 19802, 2013.
14. Ståhl, P.L., Salmén, F., Vickovic, S., et al. Visualization and analysis of gene expression in tissue sections by spatial transcriptomics. Science (80-) 353, 78, 2016.
15. Lowe, R., Shirley, N., Bleackley, M., et al. Transcriptomics technologies. PLoS Comput Biol 13, e1005457, 2017.
16. Hanash, S. Disease proteomics. Nature 422, 226, 2003.
17. Cox, J., and Mann, M. Is Proteomics the New Genomics? Cell 130, 395, 2007.
18. Dunbar, S.A. Applications of Luminex® xMAP™ technology for rapid, high-throughput multiplexed nucleic acid detection. Clin Chim Acta 363, 71, 2006.
19. Melton, L. Proteomics in multiplex. Nature 429, 101, 2004.
20. Aebersold, R., and Mann, M. Mass spectrometry-based proteomics. Nature 422, 198, 2003.
21. Bandura, D.R., Baranov, V.I., Ornatsky, O.I., et al. Mass cytometry: technique for real time single cell multitarget immunoassay based on inductively coupled plasma time-of-flight mass spectrometry. Anal Chem 81, 6813, 2009.
22. Smith, L., Villaret-Cazadamont, J., Claus, S.P., et al. Important considerations for sample collection in metabolomics studies with a special focus on applications to liver functions. Metabolites 10, 104, 2020.
23. Wishart, D.S., Feunang, Y.D., Marcu, A., et al. HMDB 4.0: the human metabolome database for 2018. Nucleic Acids Res 46, D608, 2018.
24. Broadhurst, D., Goodacre, R., Reinke, S.N., et al. Guidelines and considerations for the use of system suitability and quality control samples in mass spectrometry assays applied in untargeted clinical metabolomic studies. Metabolomics 14, 72, 2018.
25. Eyster, K.M. The membrane and lipids as integral participants in signal transduction: lipid signal transduction for the non-lipid biochemist. Adv Physiol Educ 31, 5, 2007.
26. Jurowski, K., Kochan, K., Walczak, J., et al. Analytical techniques in lipidomics: state of the art. Crit Rev Anal Chem 47, 418, 2017.
27. Stoeckius, M., Hafemeister, C., Stephenson, W., et al. Simultaneous epitope and transcriptome measurement in single cells. Nat Methods 14, 865, 2017.
28. Yan, H.H.N., Siu, H.C., Law, S., et al. A Comprehensive Human Gastric Cancer Organoid Biobank Captures Tumor Subtype Heterogeneity and Enables Therapeutic Screening. Cell Stem Cell 23, 882, 2018.
29. Van De Wetering, M., Francies, H.E., Francis, J.M., et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell 161, 933, 2015.
30. Maru, Y., Tanaka, N., Itami, M., et al. Efficient use of patient-derived organoids as a preclinical model for gynecologic tumors. Gynecol Oncol 154, 189, 2019.
31. Li, X., Francies, H.E., Secrier, M., et al. Organoid cultures recapitulate esophageal adenocarcinoma heterogeneity providing a model for clonality studies and precision therapeutics. Nat Commun 9, 1, 2018.
32. Neal, J.T., Li, X., Zhu, J., et al. Organoid modeling of the tumor immune microenvironment. Cell 175, 1972, 2018.
33. Kopper, O., de Witte, C.J., Lõhmussaar, K., et al. An organoid platform for ovarian cancer captures intra- and interpatient heterogeneity. Nat Med 25, 838, 2019.
34. Árnadóttir, S.S., Jeppesen, M., Lamy, P., et al. Characterization of genetic intratumor heterogeneity in colorectal cancer and matching patient-derived spheroid cultures. Mol Oncol 12, 132, 2018.
35. Tiriac, H., Belleau, P., Engle, D.D., et al. Organoid profiling identifies common responders to chemotherapy in pancreatic cancer. Cancer Discov 8, 1112, 2018.
36. Jabs, J., Zickgraf, F.M., Park, J., et al. Screening drug effects in patient-derived cancer cells links organoid responses to genome alterations. Mol Syst Biol 13, 955, 2017.
37. Roerink, S.F., Sasaki, N., Lee-Six, H., et al. Intra-tumour diversification in colorectal cancer at the single-cell level. Nature 556, 437, 2018.
38. Blokzijl, F., De Ligt, J., Jager, M., et al. Tissue-specific mutation accumulation in human adult stem cells during life. Nature 538, 260, 2016.
39. Michels, B.E., Mosa, M.H., Streibl, B.I., et al. Pooled in vitro and in vivo CRISPR-Cas9 screening identifies tumor suppressors in human colon organoids. Cell Stem Cell 26, 782, 2020.
40. Ringel, T., Frey, N., Ringnalda, F., et al. Genome-scale CRISPR screening in human intestinal organoids identifies drivers of TGF-β resistance. Cell Stem Cell 26, 431, 2020.
41. Berger, A., Brady, N.J., Bareja, R., et al. N-Myc-mediated epigenetic reprogramming drives lineage plasticity in advanced prostate cancer. J Clin Invest 129, 3924, 2019.
42. Bose, R., Karthaus, W.R., Armenia, J., et al. ERF mutations reveal a balance of ETS factors controlling prostate oncogenesis. Nature 546, 671, 2017.
43. Sarvestani, S.K., Signs, S.A., Lefebvre, V., et al. Cancer-predicting transcriptomic and epigenetic signatures revealed for ulcerative colitis in patient-derived epithelial organoids. Oncotarget 9, 28717, 2018.
44. Tung, K.L., Chen, K.Y., Negrete, M., et al. Integrated chromatin and transcriptomic profiling of patient-derived colon cancer organoids identifies personalized drug targets to overcome oxaliplatin resistance. Genes Dis 2019 [Epub ahead of print]; https://doi.org/10.1016/j.gendis.2019.10.012.
45. Sherman, M.H., Yu, R.T., Tseng, T.W., et al. Stromal cues regulate the pancreatic cancer epigenome and metabolome. Proc Natl Acad Sci U S A 114, 1129, 2017.
46. Verheijen, M., Lienhard, M., Schrooders, Y., et al. DMSO induces drastic changes in human cellular processes and epigenetic landscape in vitro. Sci Rep 9, 1, 2019.
47. Puca, L., Bareja, R., Prandi, D., et al. Patient derived organoids to model rare prostate cancer phenotypes. Nat Commun 9, 1, 2018.
48. Roe, J.S., Hwang, C. Il., Somerville, T.D.D., et al. Enhancer reprogramming promotes pancreatic cancer metastasis. Cell 170, 875, 2017.
49. McCracken, K.W., Catá, E.M., Crawford, C.M., et al. Modelling human development and disease in pluripotent stem-cell-derived gastric organoids. Nature 516, 400, 2014.
50. Crespo, M., Vilar, E., Tsai, S.Y., et al. Colonic organoids derived from human induced pluripotent stem cells for modeling colorectal cancer and drug testing. Nat Med 23, 878, 2017.
51. Romero-Calvo, I., Weber, C.R., Ray, M., et al. Human organoids share structural and genetic features with primary pancreatic adenocarcinoma tumors. Mol Cancer Res 17, 70, 2019.
52. Sachs, N., de Ligt, J., Kopper, O., et al. A living biobank of breast cancer organoids captures disease heterogeneity. Cell 172, 373, 2018.
53. Bian, S., Repic, M., Guo, Z., et al. Erratum to: genetically engineered cerebral organoids model brain tumor formation (Nature Methods, (2018), 15, 8, (631–639), 10.1038/s41592-018-0070-7). Nat Methods 15, 748, 2018.
54. Mullenders, J., de Jongh, E., Brousali, A., et al. Mouse and human urothelial cancer organoids: a tool for bladder cancer research. Proc Natl Acad Sci U S A 116, 4567, 2019.
55. Gao, D., Vela, I., Sboner, A., et al. Organoid cultures derived from patients with advanced prostate cancer. Cell 159, 176, 2014.
56. Ogawa, J., Pao, G.M., Shokhirev, M.N., et al. Glioblastoma model using human cerebral organoids. Cell Rep 23, 1220, 2018.
57. Kathuria, A., Lopez-Lengowski, K., Jagtap, S.S., et al. Transcriptomic landscape and functional characterization of induced pluripotent stem cell-derived cerebral organoids in schizophrenia. JAMA Psychiatry 77, 745, 2020.
58. Kathuria, A., Lopez-Lengowski, K., Vater, M., et al. Transcriptome analysis and functional characterization of cerebral organoids in bipolar disorder. Genome Med 12, 34, 2020.
59. Wang, P., Mokhtari, R., Pedrosa, E., et al. CRISPR/Cas9-mediated heterozygous knockout of the autism gene CHD8 and characterization of its transcriptional networks in cerebral organoids derived from iPS cells. Mol Autism 8, 11, 2017.
60. Dang, J., Tiwari, S.K., Lichinchi, G., et al. Zika virus depletes neural progenitors in human cerebral organoids through activation of the innate immune receptor TLR3. Cell Stem Cell 19, 258, 2016.
61. Heo, I., Dutta, D., Schaefer, D.A., et al. Modelling Cryptosporidium infection in human small intestinal and lung organoids. Nat Microbiol 3, 814, 2018.
62. Kallman, A., Capowski, E.E., Wang, J., et al. Investigating cone photoreceptor development using patient-derived NRL null retinal organoids. Commun Biol 3, 82, 2020.
63. Guo, Y., Wang, P., Ma, J.H., et al. Modeling retinitis pigmentosa: retinal organoids generated from the iPSCs of a patient with the USH2A mutation show early developmental abnormalities. Front Cell Neurosci 13, 361, 2019.
64. Verheijen, M., Schrooders, Y., Gmuender, H., et al. Bringing in vitro analysis closer to in vivo: studying doxorubicin toxicity and associated mechanisms in 3D human microtissues with PBPK-based dose modelling. Toxicol Lett 294, 184, 2018.
65. Schulte, L.N., Schweinlin, M., Westermann, A.J., et al. An advanced human intestinal coculture model reveals compartmentalized host and pathogen strategies during Salmonella infection. mBio 11, 1, 2020.
66. Boj, S.F., Hwang, C. I.l., Baker, L.A., et al. Organoid models of human and mouse ductal pancreatic cancer. Cell 160, 324, 2015.
67. Aref, A.R., Campisi, M., Ivanova, E., et al. 3D microfluidic: ex vivo culture of organotypic tumor spheroids to model immune checkpoint blockade. Lab Chip 18, 3129, 2018.
68. Vlachogiannis, G., Hedayat, S., Vatsiou, A., et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science (80-) 359, 920, 2018.
69. Nakaoka, T., Saito, Y., Shimamoto, Y., et al. Cluster microRNAs miR-194 and miR-215 suppress the tumorigenicity of intestinal tumor organoids. Cancer Sci 108, 678, 2017.
70. Ke, X., Yan, R., Sun, Z., et al. Esophageal adenocarcinoma–derived extracellular vesicle microRNAs induce a neoplastic phenotype in gastric organoids. Neoplasia 19, 941, 2017.
71. Diermeier, S.D., Chang, K.C., Freier, S.M., et al. Mammary tumor-associated RNAs impact tumor cell proliferation, invasion, and migration. Cell Rep 17, 261, 2016.
72. Zeöld, A., Sándor, G.O., Kiss, A., et al. Shared extracellular vesicle miRNA profiles of matched ductal pancreatic adenocarcinoma organoids and blood plasma samples show the power of organoid technology. Cell Mol Life Sci 2020 [Epub ahead of print]; https://doi.org/10.1007/s00018-020-03703-8.
73. Vernon, M., Lambert, B., Meryet-Figuière, M., et al. Functional miRNA screening identifies wide-ranging antitumor properties of miR-3622b-5p and reveals a new therapeutic combination strategy in ovarian tumor organoids. Mol Cancer Ther 19, 1506, 2020.
74. Jones, E.J., Matthews, Z.J., Gul, L., et al. Integrative analysis of Paneth cell proteomic and transcriptomic data from intestinal organoids reveals functional processes dependent on autophagy. Dis Model Mech 12, 2019 [Epub ahead of print];.
75. Kuzmanov, U., Wang, E.Y., Vanderlaan, R., et al. Mapping signalling perturbations in myocardial fibrosis via the integrative phosphoproteomic profiling of tissue from diverse sources. Nat Biomed Eng 4, 889, 2020.
76. Čermák, V., Gandalovičová, A., Merta, L., et al. High-throughput transcriptomic and proteomic profiling of mesenchymal-amoeboid transition in 3D collagen. Sci Data 7, 1, 2020.
77. Thul, P.J., Akesson, L., Wiking, M., et al. A subcellular map of the human proteome. Science (80-) 356, 820, 2017.
78. Bojkova, D., Klann, K., Koch, B., et al. Proteomics of SARS-CoV-2-infected host cells reveals therapy targets. Nature 583, 469, 2020.
79. Kim, D.K., Park, J., Han, D., et al. Molecular and functional signatures in a novel Alzheimer's disease mouse model assessed by quantitative proteomics. Mol Neurodegener 13, 2, 2018.
80. Geyer, P.E., Wewer Albrechtsen, N.J., Tyanova, S., et al. Proteomics reveals the effects of sustained weight loss on the human plasma proteome. Mol Syst Biol 12, 901, 2016.
81. Doll, S., Gnad, F., and Mann, M. The case for proteomics and phospho-proteomics in personalized cancer medicine. Proteomics Clin Appl 13, e1800113, 2019.
82. He, W., He, W., Kuang, Y., et al. Proteomic comparison of 3D and 2D glioma models reveals increased HLA-E expression in 3D models is associated with resistance to NK cell-mediated cytotoxicity. J Proteome Res 13, 2272, 2014.
83. Tölle, R.C., Gaggioli, C., and Dengjel, J. Three-dimensional cell culture conditions affect the proteome of cancer-associated fibroblasts. J Proteome Res 17, 2780, 2018.
84. Lee, S.Y., Park, S.B., Kim, Y.E., et al. iTRAQ-based quantitative proteomic comparison of 2D and 3D adipocyte cell models co-cultured with macrophages using online 2D-nanoLC-ESI-MS/MS. Sci Rep 9, 1, 2019.
85. Hale, L.J., Howden, S.E., Phipson, B., et al. 3D organoid-derived human glomeruli for personalised podocyte disease modelling and drug screening. Nat Commun 9, 1, 2018.
86. Yue, X., Lukowski, J.K., Weaver, E.M., et al. Quantitative proteomic and phosphoproteomic comparison of 2D and 3D colon cancer cell culture models. J Proteome Res 15, 4265, 2016.
87. Levin, V.A., Panchabhai, S., Shen, L., et al. Protein and phosphoprotein levels in glioma and adenocarcinoma cell lines grown in normoxia and hypoxia in monolayer and three-dimensional cultures. Proteome Sci 10, 5, 2012.
88. Labonia, G.J., Ludwig, K.R., Mousseau, C.B., et al. ITRAQ quantitative proteomic profiling and MALDI-MSI of colon cancer spheroids treated with combination chemotherapies in a 3D printed fluidic device. Anal Chem 90, 1423, 2018.
89. Lee, H.K., Velazquez Sanchez, C., Chen, M., et al. Three dimensional human neuro-spheroid model of Alzheimer's disease based on differentiated induced pluripotent stem cells. PLoS One 11, 1, 2016.
90. Rosenbluth, J.M., Schackmann, R.C.J., Gray, G.K., et al. Organoid cultures from normal and cancer-prone human breast tissues preserve complex epithelial lineages. Nat Commun 11, 1, 2020.
91. Al-Juboori, A.A.A., Ghosh, A., Jamaluddin, M.F. Bin., et al. Proteomic analysis of stromal and epithelial cell communications in human endometrial cancer using a unique 3D co-culture model. Proteomics 19, 1, 2019.
92. Cristobal, A., van den Toorn, H.W.P., van de Wetering, M., et al. Personalized proteome profiles of healthy and tumor human colon organoids reveal both individual diversity and basic features of colorectal cancer. Cell Rep 18, 263, 2017.
93. Ortega-Prieto, A.M., Skelton, J.K., Wai, S.N., et al. 3D microfluidic liver cultures as a physiological preclinical tool for hepatitis B virus infection. Nat Commun 9, 1, 2018.
94. Kuen, J., Darowski, D., Kluge, T., et al. Pancreatic cancer cell/fibroblast co-culture induces M2 like macrophages that influence therapeutic response in a 3D model. PLoS One 12, e0182039, 2017.
95. Marx, V. A dream of single-cell proteomics. Nat Methods 16, 809, 2019.
96. Lundberg, E., and Borner, G.H.H. Spatial proteomics: a powerful discovery tool for cell biology. Nat Rev Mol Cell Biol 20, 285, 2019.
97. Sato, M., Kawana, K., Adachi, K., et al. Spheroid cancer stem cells display reprogrammed metabolism and obtain energy by actively running the tricarboxylic acid (TCA) cycle. Oncotarget 7, 33297, 2016.
98. Fujisawa, K., Takami, T., Sasai, N., et al. Metabolic alterations in spheroid-cultured hepatic stellate cells. Int J Mol Sci 21, 3451, 2020.
99. Vorrink, S.U., Ullah, S., Schmidt, S., et al. Endogenous and xenobiotic metabolic stability of primary human hepatocytes in long-term 3D spheroid cultures revealed by a combination of targeted and untargeted metabolomics. FASEB J 31, 2696, 2017.
100. Ramachandran, G.K., and Yeow, C.H. Proton NMR characterization of intact primary and metastatic melanoma cells in 2D & 3D cultures. Biol Res 50, 12, 2017.
101. Palma, A., Grande, S., Luciani, A.M., et al. Metabolic study of breast MCF-7 tumor spheroids after gamma irradiation by 1H NMR spectroscopy and microimaging. Front Oncol 6, 105, 2016.
102. Fan, T.W.M., El-Amouri, S.S., Macedo, J.K.A., et al. Stable isotope-resolved metabolomics shows metabolic resistance to anti-cancer selenite in 3D spheroids versus 2D cell cultures. Metabolites 8, 40, 2018.
103. Lu, X., Lodi, A., Konopleva, M., et al. Three-dimensional leukemia co-culture system for in vitro high-content metabolomics screening. SLAS Discov 24, 817, 2019.
104. Choi, U.Y., Lee, J.J., Park, A., et al. Oncogenic human herpesvirus hijacks proline metabolism for tumorigenesis. Proc Natl Acad Sci U S A 117, 8083, 2020.
105. Shah, P., Fritz, J.V., Glaab, E., et al. A microfluidics-based in vitro model of the gastrointestinal human-microbe interface. Nat Commun 7, 1, 2016.
106. Priyadarsini, S., Sarker-Nag, A., Rowsey, T.G., et al. Establishment of a 3D in vitro model to accelerate the development of human therapies against corneal diabetes. PLoS One 11, 1, 2016.
107. Coloff, J.L., Murphy, J.P., Braun, C.R., et al. Differential glutamate metabolism in proliferating and quiescent mammary epithelial cells. Cell Metab 23, 867, 2016.
108. Vermeersch, K.A., Wang, L., Mezencev, R., et al. OVCAR-3 spheroid-derived cells display distinct metabolic profiles. PLoS One 10, 1, 2015.
109. Jansen, J., Jansen, K., Neven, E., et al. Remote sensing and signaling in kidney proximal tubules stimulates gut microbiome-derived organic anion secretion. Proc Natl Acad Sci U S A 116, 16105, 2019.
110. van Gorsel, M., Elia, I., and Fendt, S.M. 13C tracer analysis and metabolomics in 3D cultured cancer cells. Methods Mol Biol 1862, 53, 2019.
111. Aguilar, E., Marin De Mas, I., Zodda, E., et al. Metabolic reprogramming and dependencies associated with epithelial cancer stem cells independent of the epithelial-mesenchymal transition program. Stem Cells 34, 1163, 2016.
112. Rodenhizer, D., Gaude, E., Cojocari, D., et al. Erratum: a three-dimensional engineered tumour for spatial snapshot analysis of cell metabolism and phenotype in hypoxic gradients (Nature Materials (2015) 10.1038/nmat4482). Nat Mater 15, 244, 2016.
113. Palubeckaitė, I., Crooks, L., Smith, D.P., et al. Mass spectrometry imaging of endogenous metabolites in response to doxorubicin in a novel 3D osteosarcoma cell culture model. J Mass Spectrom 55, e4461, 2020.
114. Tucker, L.H., Hamm, G.R., Sargeant, R.J.E., et al. Untargeted metabolite mapping in 3D cell culture models using high spectral resolution FT-ICR Mass Spectrometry Imaging. Anal Chem 91, 9522, 2019.
115. Vidavsky, N., Kunitake, J.A.M.R., Diaz-Rubio, M.E., et al. Mapping and profiling lipid distribution in a 3D model of breast cancer progression. ACS Cent Sci 5, 768, 2019.
116. Feldman, A., Mukha, D., Maor, I.I., et al. Blimp1+ cells generate functional mouse sebaceous gland organoids in vitro. Nat Commun 10, 1, 2019.
117. Kozyra, M., Johansson, I., Nordling, Å., et al. Human hepatic 3D spheroids as a model for steatosis and insulin resistance. Sci Rep 8, 1, 2018.
118. Jones, D.T., Valli, A., Haider, S., et al. 3D growth of cancer cells elicits sensitivity to kinase inhibitors but not lipid metabolism modifiers. Mol Cancer Ther 18, 376, 2019.
119. Pisanu, M.E., Maugeri-Saccà, M., Fattore, L., et al. Inhibition of Stearoyl-CoA desaturase 1 reverts BRAF and MEK inhibition-induced selection of cancer stem cells in BRAF-mutated melanoma. J Exp Clin Cancer Res 37, 318, 2018.
120. Ahlf, D.R., Masyuko, R.N., Hummon, A.B., et al. Correlated mass spectrometry imaging and confocal Raman microscopy for studies of three-dimensional cell culture sections. Analyst 139, 4578, 2014.
121. Grabowski, P., and Rappsilber, J. A primer on data analytics in functional genomics: how to move from data to insight? Trends Biochem Sci 44, 21, 2019.
122. Hasin, Y., Seldin, M., and Lusis, A. Multi-omics approaches to disease. Genome Biol 18, 83, 2017.
123. Seidlitz, T., Merker, S.R., Rothe, A., et al. Human gastric cancer modelling using organoids. Gut 68, 207, 2019.
124. Guo, F., Li, L., Li, J., et al. Single-cell multi-omics sequencing of mouse early embryos and embryonic stem cells. Cell Res 27, 967, 2017.
125. Weeber, F., Van De Wetering, M., Hoogstraat, M., et al. Preserved genetic diversity in organoids cultured from biopsies of human colorectal cancer metastases. Proc Natl Acad Sci U S A 112, 13308, 2015.
126. Fong, E.L.S., Toh, T.B., Lin, X., et al. Generation of matched patient-derived xenograft in vitro-in vivo models using 3D macroporous hydrogels for the study of liver cancer. Biomaterials 159, 229, 2018.
127. Feaver, R.E., Cole, B.K., Lawson, M.J., et al. Development of an in vitro human liver system for interrogating nonalcoholic steatohepatitis. JCI Insight 1, e90954, 2016.
128. Blandin, A.F., Durand, A., Litzler, M., et al. Hypoxic environment and paired hierarchical 3D and 2D models of pediatric H3.3-mutated gliomas recreate the patient tumor complexity. Cancers (Basel) 11, 1875, 2019.
129. Wang, J., Chen, F., Liu, L., et al. Engineering EMT using 3D micro-scaffold to promote hepatic functions for drug hepatotoxicity evaluation. Biomaterials 91, 11, 2016.
130. Hart, P.C., Kenny, H.A., Grassl, N., et al. Mesothelial cell HIF1α expression is metabolically downregulated by metformin to prevent oncogenic tumor-stromal crosstalk. Cell Rep 29, 4086.e6, 2019.
131. Arai, K., Sakamoto, R., Kubota, D., et al. Proteomic approach toward molecular backgrounds of drug resistance of osteosarcoma cells in spheroid culture system. Proteomics 13, 2351, 2013.
132. Tian, X., Zhang, G., Zou, Z., et al. Anticancer drug affects metabolomic profiles in multicellular spheroids: studies using mass spectrometry imaging combined with machine learning. Anal Chem 91, 5802, 2019.
133. Pamies, D., Block, K., Lau, P., et al. Rotenone exerts developmental neurotoxicity in a human brain spheroid model. Toxicol Appl Pharmacol 354, 101, 2018.
134. Ishikawa, S., Matsumura, K., Kitamura, N., et al. Multi-omics analysis: repeated exposure of a 3D bronchial tissue culture to whole-cigarette smoke. Toxicol In Vitro 54, 251, 2019.
135. Haswell, L.E., Baxter, A., Banerjee, A., et al. Reduced biological effect of e-cigarette aerosol compared to cigarette smoke evaluated in vitro using normalized nicotine dose and RNA-seq-based toxicogenomics. Sci Rep 7, 888, 2017.
136. Elias, M.S., Wright, S.C., Nicholson, W.V., et al. Functional and proteomic analysis of a full thickness filaggrin-deficient skin organoid model. Wellcome Open Res 4, 134, 2019.
137. Braun, L.M., Lagies, S., Klar, R.F.U., et al. Metabolic profiling of early and late recurrent pancreatic ductal adenocarcinoma using patient-derived organoid cultures. Cancers (Basel) 12, 1440, 2020.
138. Lu, M., Sanderson, S.M., Zessin, A., et al. Exercise inhibits tumor growth and central carbon metabolism in patient-derived xenograft models of colorectal cancer. Cancer Metab 6, 14, 2018.
139. Kang, Y.P., Yoon, J.H., Long, N.P., et al. Spheroid-induced epithelial-mesenchymal transition provokes global alterations of breast cancer lipidome: a multi-layered omics analysis. Front Oncol 9, 145, 2019.
140. Worthington Biochemical Corporation. 2020. Cell Isolation Optimizing System, Available at www.worthington-biochem.com/CIT/CIT_Product%20Insert_2012.pdf on October 1, 2020.
141. Dutta, D., Heo, I., and Clevers, H. Disease modeling in stem cell-derived 3D organoid systems. Trends Mol Med 23, 393, 2017.
142. Zhao, Y., Rafatian, N., Feric, N.T., et al. A platform for generation of chamber-specific cardiac tissues and disease modeling. Cell 176, 913, 2019.
143. Berkers, G., Van Mourik, P., Vonk, A.M., et al. Rectal organoids enable personalized treatment of cystic fibrosis. Cell Rep 26, 1701, 2019.
Information & Authors
Information
Published In
Tissue Engineering Part C: Methods
Volume 27 • Issue Number 3 • March 2021
Pages: 183 - 199
PubMed: 33406987
Copyright
Copyright 2021, Mary Ann Liebert, Inc., publishers.
History
Published online: 15 March 2021
Published in print: March 2021
Published ahead of print: 22 February 2021
Published ahead of production: 6 January 2021
Accepted: 5 January 2021
Received: 2 October 2020
Topics
Authors
Disclosure Statement
The authors have no conflicts of interests to declare.
Funding Information
This work was funded by a Natural Sciences and Engineering Research Council of Canada (NSERC) Discovery grant to A.P.M. (Fund #RGPIN-2019-06486) and an NSERC CREATE Fellowship to NW.
Metrics & Citations
Metrics
Citations
Export Citation
Export citation
Select the format you want to export the citations of this publication.
View Options
Get Access
Access content
To read the fulltext, please use one of the options below to sign in or purchase access.⚠ Society Access
If you are a member of a society that has access to this content please log in via your society website and then return to this publication.