Acta Med. 2011, 54: 3-8

https://doi.org/10.14712/18059694.2016.9

Caco-2 Cells, Biopharmaceutics Classification System (BCS) and Biowaiter

Libuše Smetanová, Věra Štětinová, Zbyněk Svoboda, Jaroslav Květina

Institute of Experimental Biopharmaceutics, Joint Research Centre of PRO.MED.CS Praha a.s. and the Academy of Sciences of the Czech Republic, Heyrovského 1207, Hradec Králové, Czech Republic

Received June 30, 2010
Accepted December 6, 2010

Crossref Cited-by Linking

  • Liu Tiange, Gu Jia, Fu Caili, Su Lingshan: Three-Dimensional Scaffolds for Intestinal Cell Culture: Fabrication, Utilization, and Prospects. Tissue Engineering Part B: Reviews 2024, 30, 158. <https://doi.org/10.1089/ten.teb.2023.0124>
  • Cavalcante Keila N., Feitor Jessica, Morais Sinara T.B., Nassu Renata T., Ahrné Lilia, Cardoso Daniel R.: Impact of UV-C pretreatment on β-lactoglobulin hydrolysis by trypsin: production and bioavailability of bioactive peptides. International Dairy Journal 2023, 105650. <https://doi.org/10.1016/j.idairyj.2023.105650>
  • Liu Tiange, Gu Jia, Fu Caili, Su Lingshan: Three-Dimensional (3D) Scaffolds for Intestinal Cell Culture: Fabrication, Utilization and Prospects. Tissue Engineering Part B: Reviews 2023. <https://doi.org/10.1089/ten.TEB.2023.0124>
  • Li Rong, Chen Yuwen, Jia Mi, Jiang Xuehua, Wang Ling: Pharmacokinetics and absorption mechanism of tandospirone citrate. Front. Pharmacol. 2023, 14. <https://doi.org/10.3389/fphar.2023.1283103>
  • Panse Nimishraj, Gerk Phillip M.: The Caco-2 Model: Modifications and enhancements to improve efficiency and predictive performance. International Journal of Pharmaceutics 2022, 624, 122004. <https://doi.org/10.1016/j.ijpharm.2022.122004>
  • Kikuchi Takuya, Hayashi Akane, Ikeda Naohiro, Morita Osamu, Tasaki Junichi: Multidrug resistance-associated protein 2 (MRP2) is an efflux transporter of EGCG and its metabolites in the human small intestine. The Journal of Nutritional Biochemistry 2022, 107, 109071. <https://doi.org/10.1016/j.jnutbio.2022.109071>
  • Jung Sang-Myung, Kim Seonghun: In vitro Models of the Small Intestine for Studying Intestinal Diseases. Front. Microbiol. 2022, 12. <https://doi.org/10.3389/fmicb.2021.767038>
  • Rodrigues Elsa T., Nascimento Susana F., Pires Cristiana L., Godinho Lia P., Churro Catarina, Moreno Maria João, Pardal Miguel A.: Determination of intestinal absorption of the paralytic shellfish toxin GTX-5 using the Caco-2 human cell model. Environ Sci Pollut Res 2021, 28, 67256. <https://doi.org/10.1007/s11356-021-15342-y>
  • Papaj Katarzyna, Kasprzycka Anna, Góra Artur, Grajoszek Aniela, Rzepecka Grażyna, Stojko Jerzy, Barski Jarosław-Jerzy, Szeja Wiesław, Rusin Aleksandra: Structure–bioavailability relationship study of genistein derivatives with antiproliferative activity on human cancer cell. Journal of Pharmaceutical and Biomedical Analysis 2020, 185, 113216. <https://doi.org/10.1016/j.jpba.2020.113216>
  • Kulthong Kornphimol, Duivenvoorde Loes, Sun Huiyi, Confederat Samuel, Wu Jiaqing, Spenkelink Bert, de Haan Laura, Marin Victor, van der Zande Meike, Bouwmeester Hans: Microfluidic chip for culturing intestinal epithelial cell layers: Characterization and comparison of drug transport between dynamic and static models. Toxicology in Vitro 2020, 65, 104815. <https://doi.org/10.1016/j.tiv.2020.104815>
  • Polet Madeleine, Laloux Laurie, Cambier Sébastien, Ziebel Johanna, Gutleb Arno C., Schneider Yves-Jacques: Soluble silver ions from silver nanoparticles induce a polarised secretion of interleukin-8 in differentiated Caco-2 cells. Toxicology Letters 2020, 325, 14. <https://doi.org/10.1016/j.toxlet.2020.02.004>
  • Ibrahim Mohamed Moustafa, Maria Doaa Nabih, Wang XiangDi, Simpson Raven N., Hollingsworth T.J., Jablonski Monica M.: Enhanced Corneal Penetration of a Poorly Permeable Drug Using Bioadhesive Multiple Microemulsion Technology. Pharmaceutics 2020, 12, 704. <https://doi.org/10.3390/pharmaceutics12080704>
  • Matsuzaki Takumi, Nakamura Masao, Nogita Takehide, Sato Atsushi: Cellular Uptake and Release of Intact Lactoferrin and Its Derivatives in an Intestinal Enterocyte Model of Caco-2 Cells. Biological & Pharmaceutical Bulletin 2019, 42, 989. <https://doi.org/10.1248/bpb.b19-00011>
  • Costa Joana, Ahluwalia Arti: Advances and Current Challenges in Intestinal in vitro Model Engineering: A Digest. Front. Bioeng. Biotechnol. 2019, 7. <https://doi.org/10.3389/fbioe.2019.00144>
  • Domenger Dorothée, Cudennec Benoit, Kouach Mostafa, Touche Véronique, Landry Christophe, Lesage Jean, Gosselet Fabien, Lestavel Sophie, Goossens Jean-François, Dhulster Pascal, Ravallec Rozenn: Food-Derived Hemorphins Cross Intestinal and Blood–Brain Barriers In Vitro. Front. Endocrinol. 2018, 9. <https://doi.org/10.3389/fendo.2018.00159>
  • Ressaissi Asma, Attia Nebil, Falé Pedro Luis, Pacheco Rita, Victor Bruno L., Machuqueiro Miguel, Serralheiro Maria Luísa M.: Isorhamnetin derivatives and piscidic acid for hypercholesterolemia: cholesterol permeability, HMG-CoA reductase inhibition, and docking studies. Arch. Pharm. Res. 2017, 40, 1278. <https://doi.org/10.1007/s12272-017-0959-1>
  • Zhang Qisong, Zhu Lijun, Gong Xia, Ruan Yanjiao, Yu Jia, Jiang Huangyu, Wang Ying, Qi XiaoXiao, Lu Linlin, Liu Zhongqiu: Sulfonation Disposition of Acacetin: In Vitro and in Vivo. J. Agric. Food Chem. 2017, 65, 4921. <https://doi.org/10.1021/acs.jafc.7b00854>
  • Majima Atsushi, Handa Osamu, Naito Yuji, Suyama Yosuke, Onozawa Yuriko, Higashimura Yasuki, Mizushima Katsura, Morita Mayuko, Uehara Yukiko, Horie Hideki, Iida Takaya, Fukui Akifumu, Dohi Osamu, Okayama Tetsuya, Yoshida Naohisa, Kamada Kazuhiro, Katada Kazuhiro, Uchiyama Kazuhiko, Ishikawa Takeshi, Takagi Tomohisa, Konishi Hideyuki, Yasukawa Zenta, Tokunaga Makoto, Okubo Tsutomu, Itoh Yoshito: Real‐time monitoring of trans‐epithelial electrical resistance in cultured intestinal epithelial cells: the barrier protection of water‐soluble dietary fiber. J of Digest Diseases 2017, 18, 151. <https://doi.org/10.1111/1751-2980.12456>
  • Jantarajit Walailak, Lertsuwan Kornkamon, Teerapornpuntakit Jarinthorn, Krishnamra Nateetip, Charoenphandhu Narattaphol: CFTR-mediated anion secretion across intestinal epithelium-like Caco-2 monolayer under PTH stimulation is dependent on intermediate conductance K+ channels. American Journal of Physiology-Cell Physiology 2017, 313, C118. <https://doi.org/10.1152/ajpcell.00010.2017>
  • Sánchez-Díaz Ana María, Romero-Hernández Beatriz, Conde-Moreno Elisa, Kwak Young-Keun, Zamora Javier, Colque-Navarro Patricia, Möllby Roland, Ruiz-Garbajosa Patricia, Cantón Rafael, García-Bermejo Laura, del Campo Rosa, Lemos José A: New Insights into the Enterococcus faecium and Streptococcus gallolyticus subsp. gallolyticus Host Interaction Mechanisms. PLoS ONE 2016, 11, e0159159. <https://doi.org/10.1371/journal.pone.0159159>
  • Catalán Úrsula, López de las Hazas Maria-Carmen, Rubió Laura, Fernández-Castillejo Sara, Pedret Anna, de la Torre Rafael, Motilva Maria-José, Solà Rosa: Protective effect of hydroxytyrosol and its predominant plasmatic human metabolites against endothelial dysfunction in human aortic endothelial cells. Mol. Nutr. Food Res. 2015, 59, 2523. <https://doi.org/10.1002/mnfr.201500361>
  • Goodwin Daryn, Varamini Pegah, Simerska Pavla, Toth Istvan: Stability, Permeability and Growth-Inhibitory Properties of Gonadotropin-Releasing Hormone Liposaccharides. Pharm Res 2015, 32, 1570. <https://doi.org/10.1007/s11095-014-1558-1>
  • Soto A.M., Morales P., Haza A.I., García M.L., Selgas M.D.: Bioavailability of calcium from enriched meat products using Caco-2 cells. Food Research International 2014, 55, 263. <https://doi.org/10.1016/j.foodres.2013.10.038>
  • Araújo Francisca, Sarmento Bruno: Towards the characterization of an in vitro triple co-culture intestine cell model for permeability studies. International Journal of Pharmaceutics 2013, 458, 128. <https://doi.org/10.1016/j.ijpharm.2013.10.003>
  • Baruwati Babita, Simmons Steven O., Varma Rajendar S., Veronesi Bellina: “Green” Synthesized and Coated Nanosilver Alters the Membrane Permeability of Barrier (Intestinal, Brain Endothelial) Cells and Stimulates Oxidative Stress Pathways in Neurons. ACS Sustainable Chem. Eng. 2013, 1, 753. <https://doi.org/10.1021/sc400024a>
  • Cai Zheng, Huang Juan, Luo Hui, Lei Xiaolu, Yang Zhaoxiang, Mai Yang, Liu Zhongqiu: Role of glucose transporters in the intestinal absorption of gastrodin, a highly water-soluble drug with good oral bioavailability. Journal of Drug Targeting 2013, 21, 574. <https://doi.org/10.3109/1061186X.2013.778263>
  • Shanmugam Srinivasan, Im Ho Taek, Sohn Young Taek, Kim Kyung Soo, Kim Yong-Il, Yong Chul Soon, Kim Jong Oh, Choi Han-Gon, Woo Jong Soo: Zanamivir Oral Delivery: Enhanced Plasma and Lung Bioavailability in Rats. Biomolecules and Therapeutics 2013, 21, 161. <https://doi.org/10.4062/biomolther.2013.010>
  • Varamini Pegah, Hussein Waleed M., Mansfeld Friederike M., Toth Istvan: Synthesis, biological activity and structure–activity relationship of endomorphin-1/substance P derivatives. Bioorganic & Medicinal Chemistry 2012, 20, 6335. <https://doi.org/10.1016/j.bmc.2012.09.003>
  • Mohamed Hala F: Molecular analysis and anticancer properties of two identified isolates, Fusarium solani and Emericella nidulans isolated from Wady El–Natron soil in Egypt against Caco–2 (ATCC) cell line. Asian Pacific Journal of Tropical Biomedicine 2012, 2, 863. <https://doi.org/10.1016/S2221-1691(12)60244-5>
  • Varamini Pegah, Mansfeld Friederike M., Blanchfield Joanne T., Wyse Bruce D., Smith Maree T., Toth Istvan: Synthesis and Biological Evaluation of an Orally Active Glycosylated Endomorphin-1. J. Med. Chem. 2012, 55, 5859. <https://doi.org/10.1021/jm300418d>
  • Pusch Jacqueline, Votteler Miriam, Göhler Stella, Engl Jasmin, Hampel Martina, Walles Heike, Schenke-Layland Katja: The physiological performance of a three-dimensional model that mimics the microenvironment of the small intestine. Biomaterials 2011, 32, 7469. <https://doi.org/10.1016/j.biomaterials.2011.06.035>
Crossref Cited-by Linking logo