1
|
Moskal K, Khurana N, Siegert L, Lee YS, Clevers H, Elinav E, Puschhof J. Modeling cancer-microbiome interactions in vitro: A guide to co-culture platforms. Int J Cancer 2025; 156:2053-2067. [PMID: 39716471 PMCID: PMC11970552 DOI: 10.1002/ijc.35298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/10/2024] [Accepted: 10/29/2024] [Indexed: 12/25/2024]
Abstract
The biology of cancer is characterized by an intricate interplay of cells originating not only from the tumor mass, but also its surrounding environment. Different microbial species have been suggested to be enriched in tumors and the impacts of these on tumor phenotypes is subject to intensive investigation. For these efforts, model systems that accurately reflect human-microbe interactions are rapidly gaining importance. Here we present a guide for selecting a suitable in vitro co-culture platform used to model different cancer-microbiome interactions. Our discussion spans a variety of in vitro models, including 2D cultures, tumor spheroids, organoids, and organ-on-a-chip platforms, where we delineate their respective advantages, limitations, and applicability in cancer microbiome research. Particular focus is placed on methodologies that facilitate the exposure of cancer cells to microbes, such as organoid microinjections and co-culture on microfluidic devices. We highlight studies offering critical insights into possible cancer-microbe interactions and underscore the importance of in vitro models in those discoveries. We anticipate the integration of more complex microbial communities and the inclusion of immune cells into co-culture systems to more accurately simulate the tumor microenvironment. The advent of ever more sophisticated co-culture models will aid in unraveling the mechanisms of cancer-microbiome interplay and contribute to exploiting their potential in novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Kamil Moskal
- Junior Research Group Epithelium Microbiome Interactions (EMIL), German Cancer Research CenterHeidelbergGermany
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
- DKFZ Hector Cancer Institute at the University Medical CenterMannheimGermany
| | - Nimisha Khurana
- Junior Research Group Epithelium Microbiome Interactions (EMIL), German Cancer Research CenterHeidelbergGermany
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Luisa Siegert
- Junior Research Group Epithelium Microbiome Interactions (EMIL), German Cancer Research CenterHeidelbergGermany
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
| | - Ye Seul Lee
- Junior Research Group Epithelium Microbiome Interactions (EMIL), German Cancer Research CenterHeidelbergGermany
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Hans Clevers
- Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC UtrechtHubrecht InstituteUtrechtThe Netherlands
- Present address:
Roche Pharmaceutical Research and Early DevelopmentBaselSwitzerland
| | - Eran Elinav
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
- Systems Immunology DepartmentWeizmann Institute of ScienceRehovotIsrael
| | - Jens Puschhof
- Junior Research Group Epithelium Microbiome Interactions (EMIL), German Cancer Research CenterHeidelbergGermany
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
- DKFZ Hector Cancer Institute at the University Medical CenterMannheimGermany
| |
Collapse
|
2
|
Vats M, Rathod D, Patel H, Richards T, Patel K. Self-emulsifying Nano-PND oral delivery systems of PND1186: In silico modeling for bioavailability estimation. J Mol Liq 2025; 426:127161. [PMID: 40322757 PMCID: PMC12048016 DOI: 10.1016/j.molliq.2025.127161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Focal adhesion kinase (FAK) inhibitors have proven to aid the therapeutic potential of anti-cancer agents. PND1186 (PND) is a FAK inhibitor disrupting the oncogenic processes such as cell survival, proliferation, adhesion, migration and angiogenesis, as well as remodeling of tumor microenvironment. However, the pharmacological potential of PND is limited by its poor solubility and bioavailability due to rapid precipitation of weakly basic PND in the intestinal milieu. As a solution, we have developed a self-nanoemulsifying PND oral delivery system (NanoPODS) for rapid dissolution of PND while Soluplus containing system (NanoPODS-S) was prepared to prevent the precipitation of PND. Optimized NanoPODS-S depicted a particle size of 107.0 ± 3.6 nm, PDI of 0.223 ± 0.016, and a surface potential of -4.2 ± 0.007 mV, along with > 70% PND released at pH 6.8. In silico pharmacokinetics predicted 99% oral bioavailability for NanoPODS-S. This study evaluates the efficacy of NanoPODS and NanoPODS-S for improved oral bioavailability with better cytotoxicity efficacy on Pancreatic Ductal Adenocarcinoma (PDAC) cell lines. NanoPODS-S is the first of its kind, self-nanoemulsifying system containing a polymeric precipitation inhibitor mimicking a "spring-parachute effect". It will be a novel platform technology for rapid and enhanced dissolution of poorly soluble molecules.
Collapse
Affiliation(s)
- Mukti Vats
- College of Pharmacy and Health Sciences, St. John’s University, NY, USA
| | - Drishti Rathod
- College of Pharmacy and Health Sciences, St. John’s University, NY, USA
| | - Henis Patel
- College of Pharmacy and Health Sciences, St. John’s University, NY, USA
| | - Terjahna Richards
- College of Pharmacy and Health Sciences, St. John’s University, NY, USA
| | - Ketan Patel
- College of Pharmacy and Health Sciences, St. John’s University, NY, USA
| |
Collapse
|
3
|
Sanchez JM, Favaro MTP, López-Laguna H, Parladé E, Di Somma A, Casanova I, Unzueta U, Mangues R, Vazquez E, Voltà-Durán E, Villaverde A. Trans-Mediated, Cis-Inhibited Paradoxal Activity of Clostridium perfringens Enterotoxin (c-CPE) in Modulating Epithelial Permeability. Mol Pharm 2025; 22:1973-1982. [PMID: 40067325 DOI: 10.1021/acs.molpharmaceut.4c01205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2025]
Abstract
In the context of transdermal delivery, favoring the drug permeability of epithelia through convenient formulations would open new opportunities for local versus systemic drug delivery, envisaging higher patient comfort and an enhanced therapeutic effect. Ligands of tight junctions are interesting agents that enhance epithelial permeability by relaxing the protein complexes that form them. The C-terminal domain of Clostridium perfringens enterotoxin (c-CPE), which binds claudins, one of the tight junction (TJ) components, has been explored here as a functional domain in modular recombinant proteins, to evaluate its ability to self-promote its paracellular epithelial passage in a Caco-2 cell monolayer model. c-CPE-containing fusion proteins bind cells in the absence of internalization and cytotoxicity and support the passage, in trans, of other fusion proteins devoid of c-CPE. However, c-CPE-carrying proteins fail to cross the epithelia by themselves, probably because their affinity for TJs immobilizes them in the intercellular space. Therefore, while recombinant c-CPE versions have been here confirmed as convenient epithelial-permeabilizing agents, a paradoxical behavior has been observed where this effect is only successful when applied in trans, specifically on entities that lack c-CPE. Then, c-CPE itself inhibits the paracellular mobility of carrier molecules, not being suited as a self-driver (in c-CPE-drug complexes) for drug delivery through epithelia.
Collapse
Affiliation(s)
- Julieta M Sanchez
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, Barcelona 08193, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Barcelona 08034, Spain
- Departamento de Química, Cátedra de Química Biológica, Facultad de Ciencias Exactas, Físicas y Naturales, ICTA, Universidad Nacional de Córdoba, Av. Vélez Sársfield 1611, Córdoba 5016, Argentina
- Instituto de Investigaciones Biológicas y Tecnológicas (IIByT), CONICET-Universidad Nacional de Córdoba, Córdoba 5016, Argentina
| | - Marianna T P Favaro
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, Barcelona 08193, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Barcelona 08034, Spain
| | - Hèctor López-Laguna
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, Barcelona 08193, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Barcelona 08034, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Barcelona 08193, Spain
| | - Eloi Parladé
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, Barcelona 08193, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Barcelona 08034, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Barcelona 08193, Spain
| | - Angela Di Somma
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, Barcelona 08193, Spain
- Department of Chemical Sciences, University of Naples "Federico II", Vicinale Cupa Cintia 26, Naples 80126, Italy
| | - Isolda Casanova
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Barcelona 08034, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain
- Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
| | - Ugutz Unzueta
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Barcelona 08034, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Barcelona 08193, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain
- Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
| | - Ramón Mangues
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Barcelona 08034, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain
- Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
| | - Esther Vazquez
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, Barcelona 08193, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Barcelona 08034, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Barcelona 08193, Spain
| | - Eric Voltà-Durán
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, Barcelona 08193, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Barcelona 08034, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Barcelona 08193, Spain
| | - Antonio Villaverde
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, Barcelona 08193, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Barcelona 08034, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Barcelona 08193, Spain
| |
Collapse
|
4
|
Dash SK, Rahman MA, Yi B, Williams B, Lim GS, Zhou S, Zou P, Li Y, Mahler GJ, Zhang T. Microfluidic blood-milk barrier and physiologically based pharmacokinetic model to predict lofexidine secretion into breast milk. J Pharm Sci 2025; 114:103767. [PMID: 40113090 DOI: 10.1016/j.xphs.2025.103767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/15/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
INTRODUCTION Lofexidine (LUCEMYRA®) is the only FDA-approved, non-opioid, non-addictive treatment for opioid withdrawal symptoms, crucial for postpartum and pregnant women affected by the opioid crisis. Despite its clinical importance, data on its secretion into breast milk is limited. This study aims to develop a novel, microfluidic-based blood-milk-barrier on a chip model, a static human mammary cell transwell model, and a physiologically based pharmacokinetic (PBPK) lactation model to estimate the breast milk secretion of lofexidine, thereby ensuring maternal and infant safety and improving withdrawal management. METHODS A novel microfluidic device was developed to build a mammary epithelium-on-a-chip model, and a transwell plate was used to develop a static mammary epithelium using a human noncarcinogenic mammary epithelial cell (MEC) population that can form an integrated barrier with tight junctions. Both models were used to evaluate the transfer of lofexidine through the in vitro mammary cell barrier. The fraction of unbound lofexidine in the breast milk was determined by a Rapid Equilibrium Dialysis (RED) assay. Eleven approaches, including a novel, previously published in vitro to in vivo extrapolation (IVIVE) approach and various other approaches, were used to estimate milk-to-plasma (M/P) ratios of lofexidine. A whole-body lactation PBPK model was built using Simcyp® simulator v22 and used to predict the concentration-time profiles of lofexidine in both human plasma and breast milk. RESULTS A subpopulation of human normal mammary epithelial MCF10A cells (named MCF10A-TJ) was identified to form an integrated barrier that reaches trans-epithelial electrical resistance (TEER) values of over 1000 Ω·cm2 by culturing with in-house designed maintenance and boosting medium. The microfluidic device-based mammary epithelium-on-a-chip model generated slightly higher lofexidine permeability values than the static transwell mammary epithelial cell model. The predicted milk-to-plasma (M/P) ratio of lofexidine ranged from 0.40 to 15.88. Four approaches estimated an M/P ratio below 1, while seven predicted values above 1, mostly between 1.35 and 5.48. The whole-body lactation PBPK model predicted the concentration-time profile of lofexidine in breast milk, with an estimated M/P ratio of approximately 2.0. This value falls within the mid-range of the predictions obtained from all eleven methods. CONCLUSION This study introduces comprehensive and novel approaches to predict lofexidine secretion into breast milk. Most predictions suggest higher lofexidine concentration in milk than in plasma, raising potential safety concerns for opioid withdrawal management. Further pharmacokinetic clinical lactation studies are needed to validate these predictions.
Collapse
Affiliation(s)
- Sanat Kumar Dash
- Department of Biomedical Engineering, SUNY-Binghamton University, Binghamton, P.O Box 6000, Binghamton, NY,13902, USA
| | - Mohammad Asikur Rahman
- Department of Pharmaceutical Sciences, SUNY-Binghamton University, 96 Corliss Ave, Johnson City, NY 13790, USA
| | - Bofang Yi
- Department of Pharmaceutical Sciences, SUNY-Binghamton University, 96 Corliss Ave, Johnson City, NY 13790, USA
| | - Brianna Williams
- Department of Pharmaceutical Sciences, SUNY-Binghamton University, 96 Corliss Ave, Johnson City, NY 13790, USA
| | - Gi S Lim
- Department of Pharmaceutical Sciences, SUNY-Binghamton University, 96 Corliss Ave, Johnson City, NY 13790, USA
| | - Sindi Zhou
- Department of Pharmaceutical Sciences, SUNY-Binghamton University, 96 Corliss Ave, Johnson City, NY 13790, USA
| | - Peng Zou
- Ultragenyx Pharmaceutical Inc. 5000 Marina Blvd, Brisbane, CA 94005, USA
| | - Yanyan Li
- Department of Pharmaceutical Sciences, SUNY-Binghamton University, 96 Corliss Ave, Johnson City, NY 13790, USA
| | - Gretchen J Mahler
- Department of Biomedical Engineering, SUNY-Binghamton University, Binghamton, P.O Box 6000, Binghamton, NY,13902, USA.
| | - Tao Zhang
- Department of Pharmaceutical Sciences, SUNY-Binghamton University, 96 Corliss Ave, Johnson City, NY 13790, USA.
| |
Collapse
|
5
|
Tofani LB, Avelino TM, de Azevedo RJ, Elias GB, Ganzerla MD, Terra MF, Rodrigues VKT, Rabelo RS, Harb SV, Figueira ACM. Biofabricated 3D Intestinal Models as an Alternative to Animal-Based Approaches for Drug Toxicity Assays. Tissue Eng Regen Med 2025; 22:181-194. [PMID: 39820960 PMCID: PMC11794730 DOI: 10.1007/s13770-024-00694-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND The main challenge in new drug development is accurately predicting the human response in preclinical models. METHODS In this study, we developed three different intestinal barrier models using advanced biofabrication techniques: (i) a manual model containing Caco-2 and HT-29 cells on a collagen bed, (ii) a manual model with a Caco-2/HT-29 layer on a HDFn-laden collagen layer, and (iii) a 3D bioprinted model incorporating both cellular layers. Each model was rigorously tested for its ability to simulate a functional intestinal membrane. RESULTS All models successfully replicated the structural and functional aspects of the intestinal barrier. The 3D bioprinted intestinal model, however, demonstrated superior epithelial barrier integrity enhanced tight junction formation, microvilli development, and increased mucus production. When subjected to Ibuprofen, the 3D bioprinted model provided a more predictive response, underscoring its potential as a reliable in vitro tool for drug toxicity testing. CONCLUSION Our 3D bioprinted intestinal model presents a robust and predictive platform for drug toxicity assessments, significantly reducing the need for animal testing. This model not only aligns with ethical testing protocols but also offers enhanced accuracy in predicting human responses, thereby advancing the field of drug development.
Collapse
Affiliation(s)
- Larissa Bueno Tofani
- Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-100, Brazil
| | - Thayná Mendonça Avelino
- Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-100, Brazil
| | - Rafael Júnior de Azevedo
- Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-100, Brazil
| | - Giovanna Blazutti Elias
- Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-100, Brazil
| | - Melissa Dibbernn Ganzerla
- Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-100, Brazil
| | - Maiara Ferreira Terra
- Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-100, Brazil
| | - Vanessa Kiraly Thomaz Rodrigues
- Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-100, Brazil
| | - Renata Santos Rabelo
- Brazilian Synchrotron Light Laboratory, Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-100, Brazil
| | - Samarah Vargas Harb
- Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-100, Brazil.
| | - Ana Carolina Migliorini Figueira
- Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-100, Brazil.
| |
Collapse
|
6
|
Guo Z, Tang S, Nie K, Liu J, Hu C. Studies on absorption mechanism and pharmacokinetic properties of albendazole-bile acid conjugate: In vivo and in vitro. Biomed Pharmacother 2024; 179:117400. [PMID: 39243427 DOI: 10.1016/j.biopha.2024.117400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/16/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024] Open
Abstract
PURPOSE To improve the oral bioavailability of albendazole (ABZ), a series of albendazole-bile acid conjugates (ABCs) were synthesized. ABC's transmembrane transport mechanism and in vivo pharmacokinetic properties were preliminarily studied. METHODS The transmembrane transport mechanism of ABCs was studied using the Caco-2 monolayer cell model and intestinal perfusion model. The concentration of ABCs and ABZ were evaluated using High-Performance Liquid Chromatography (HPLC) and HPLC-Mass Spectrometry (HPLC-MS/MS). RESULTS Compared to ABZ, better permeability was observed for different types and concentrations of ABCs using the Caco-2 monolayer cell model, with ABC-C8 showing the highest permeability. The transmembrane transport of ABCs was affected by ASBT inhibitors, indicating an ASBT-mediated active transport mechanism. Additionally, introducing cholic acid resulted in ABZ no longer being a substrate for P-gp, MRP2, and BCRP, effectively reversing ABZ efflux. In vivo unidirectional intestinal perfusion results in rats showed that ABCs altered the absorption site of ABZ from the jejunum to the ileum. The absorption efficiency of ABCs in each intestinal segment was higher than that of ABZ, and the transmembrane transport efficiency decreased with increasing concentrations of ASBT inhibitors. This further confirmed the presence of both passive diffusion and ASBT-mediated active transport mechanisms in the transport of ABCs. The solubility of ABCs in gastric juice and pharmacokinetics in rats showed that ABZ-C4 exhibited enhanced solubility. Moreover, ABCs significantly increased oral bioavailability compared to ABZ, with ABC-C4 showing an approximately 31-fold increase in bioavailability. CONCLUSION The transmembrane transport mechanism of ABCs involves a combination of ASBT-mediated active transport and passive diffusion. Moreover, the incorporation of BAs successfully reverses the efflux of ABZ by efflux proteins. Among the synthesized conjugates, ABC-C4 demonstrated superior dissolution behavior both in vitro and in vivo.
Collapse
Affiliation(s)
- Zhimei Guo
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, Qinghai 810001, PR China; Medical College, Qinghai University, Xining, Qinghai 810001, PR China
| | - Shizhen Tang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, Qinghai 810001, PR China; Medical College, Qinghai University, Xining, Qinghai 810001, PR China
| | - Kaili Nie
- College of Life Science and Technology, Beijing University of Chemical of Technology, Beijing 100086, PR China
| | - Jingshuai Liu
- College of Life Science and Technology, Beijing University of Chemical of Technology, Beijing 100086, PR China
| | - Chunhui Hu
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, Qinghai 810001, PR China.
| |
Collapse
|
7
|
Sah SK, Alam K, Kumari M, Malootty R, Nath S, Ravichandiran V, Roy S, Kaity S. A 3D in-vitro biomimicking Caco-2 intestinal permeability model-based assessment of physically modified telmisartan towards an alkalizer-free formulation development. Eur J Pharm Biopharm 2024; 203:114480. [PMID: 39222674 DOI: 10.1016/j.ejpb.2024.114480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 08/26/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
Efficient telmisartan delivery for hypertension management requires the incorporation of meglumine and/or sodium hydroxide as an alkalizer in the formulation. Long-term use of powerful alkalis with formulation as part of chronic therapy can cause metabolic alkalosis, ulcers, diarrhea, and body pain. Here, we aimed to design a telmisartan formulation without alkalizers. Telmisartan properties were tailor-made by microfluidizer-based physical modification. After microfluidization, telmisartan nanosuspension was lyophilized to obtain telmisartan premix powder. The optimized telmisartan nanosuspension had an average particle size of 579.85 ± 32.14 nm. The lyophilized premix was characterized by FT-IR, DSC, and PXRD analysis to ensure its physicochemical characteristics. The solubility analysis of premix showed 2.2 times, 2.3 times, and 6 times solubility improvement in 0.1 N HCl, phosphate buffer pH 7.5, and pH 6.8 compared to pure telmisartan. A 3D in-vitro Caco-2 model was developed to compare apparent permeability of API and powder premix. It showed that the powder premix was more permeable than pure API. The tablet formulation prepared from the telmisartan premix showed a dissolution profile comparable to that of the marketed formulation. The technique present herein can be used as a platform technology for solubility and permeability improvement of similar classes of molecules.
Collapse
Affiliation(s)
- Sunil Kumar Sah
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700054, India
| | - Kamare Alam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700054, India
| | - Mamta Kumari
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700054, India
| | - R Malootty
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700054, India
| | - Subham Nath
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700054, India
| | - Velayutham Ravichandiran
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700054, India
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700054, India.
| | - Santanu Kaity
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700054, India.
| |
Collapse
|
8
|
Parente IA, Chiara L, Bertoni S. Exploring the potential of human intestinal organoids: Applications, challenges, and future directions. Life Sci 2024; 352:122875. [PMID: 38942359 DOI: 10.1016/j.lfs.2024.122875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/13/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
The complex and dynamic environment of the gastrointestinal tract shapes one of the fastest renewing tissues in the human body, the intestinal epithelium. Considering the lack of human preclinical studies, reliable models that mimic the intestinal environment are increasingly explored. Patient-derived intestinal organoids are powerful tools that recapitulate in vitro many pathophysiological features of the human intestine. In this review, the possible applications of human intestinal organoids in different research fields are highlighted. From physiologically relevant to intestinal disease modeling, regenerative medicine, and toxicology studies, the potential of intestinal organoids will be here presented and discussed. Despite the remarkable opportunities offered, limitations related to ethical concerns, tissue collection, reproducibility, and methodologies may hinder the full exploitation of this cell-based model into high throughput studies and clinical practice. Currently, distinct approaches can be used to overcome the numerous challenges found along the way and to allow the full implementation of this ground-breaking technology.
Collapse
Affiliation(s)
- Inês A Parente
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Linda Chiara
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Simona Bertoni
- Department of Food and Drug, University of Parma, Parma, Italy.
| |
Collapse
|
9
|
Sorokina L, Solberg NT, Koga S, Rønning SB, Afseth NK, Wilson SR, Rieder A, Wubshet SG. In vitro gastrointestinal stability and intestinal absorption of ACE-1 and DPP4 inhibitory peptides from poultry by-product hydrolysates. Food Funct 2024; 15:7364-7374. [PMID: 38912915 DOI: 10.1039/d4fo01214c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Bioactive peptides derived from food are promising health-promoting ingredients that can be used in functional foods and nutraceutical formulations. In addition to the potency towards the selected therapeutic target, the bioavailability of bioactive peptides is a major factor regarding clinical efficacy. We have previously shown that a low molecular weight peptide fraction (LMWPF) from poultry by-product hydrolysates possesses angiotensin-1-converting enzyme (ACE-1) and dipeptidyl-peptidase 4 (DPP4) inhibitory activities. The present study aimed to investigate the bioavailability of the bioactive peptides in the LMWPF. Prior to the investigation of bioavailability, a dipeptide YA was identified from this fraction as a dual inhibitor of ACE-1 and DPP4. Gastrointestinal (GI) stability and intestinal absorption of the bioactive peptides (i.e., YA as well as two previously reported bioactive dipeptides (VL and IY)) in the LMWPF were evaluated using the INFOGEST static in vitro digestion model and intestinal Caco-2 cell monolayer, respectively. Analysis of peptides after in vitro digestion confirmed that the dipeptides were resistant to the simulated GI conditions. After 4 hours of incubation, the concentration of the peptide from the apical side of the Caco-2 cell monolayer showed a significant decrease. However, the corresponding absorbed peptides were not detected on the basolateral side, suggesting that the peptides were not transported across the intestinal monolayer but rather taken up or metabolized by the Caco2 cells. Furthermore, when analyzing the gene expression of the Caco-2 cells upon peptide stimulation, a down-regulation of peptide transporters, the transcription factor CDX2, and the tight junction protein-1 (TJP1) was observed, suggesting the specific effects of the peptides on the Caco-2 cells. The study demonstrated that bioactive dipeptides found in the LMWPF were stable through in vitro GI digestion; however, the overall bioavailability may be hindered by inadequate uptake across the intestinal barrier.
Collapse
Affiliation(s)
- Liudmila Sorokina
- Nofima AS-Norwegian Institute of Food, Fisheries and Aquaculture Research, PB 210, N-1431 Ås, Norway.
- Department of Chemistry, University of Oslo, Oslo, Norway
| | - Nina Therese Solberg
- Nofima AS-Norwegian Institute of Food, Fisheries and Aquaculture Research, PB 210, N-1431 Ås, Norway.
| | - Shiori Koga
- Nofima AS-Norwegian Institute of Food, Fisheries and Aquaculture Research, PB 210, N-1431 Ås, Norway.
| | - Sissel Beate Rønning
- Nofima AS-Norwegian Institute of Food, Fisheries and Aquaculture Research, PB 210, N-1431 Ås, Norway.
| | - Nils Kristian Afseth
- Nofima AS-Norwegian Institute of Food, Fisheries and Aquaculture Research, PB 210, N-1431 Ås, Norway.
| | | | - Anne Rieder
- Nofima AS-Norwegian Institute of Food, Fisheries and Aquaculture Research, PB 210, N-1431 Ås, Norway.
| | - Sileshi Gizachew Wubshet
- Nofima AS-Norwegian Institute of Food, Fisheries and Aquaculture Research, PB 210, N-1431 Ås, Norway.
| |
Collapse
|
10
|
Pires CL, Moreno MJ. Improving the Accuracy of Permeability Data to Gain Predictive Power: Assessing Sources of Variability in Assays Using Cell Monolayers. MEMBRANES 2024; 14:157. [PMID: 39057665 PMCID: PMC11278619 DOI: 10.3390/membranes14070157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024]
Abstract
The ability to predict the rate of permeation of new compounds across biological membranes is of high importance for their success as drugs, as it determines their efficacy, pharmacokinetics, and safety profile. In vitro permeability assays using Caco-2 monolayers are commonly employed to assess permeability across the intestinal epithelium, with an extensive number of apparent permeability coefficient (Papp) values available in the literature and a significant fraction collected in databases. The compilation of these Papp values for large datasets allows for the application of artificial intelligence tools for establishing quantitative structure-permeability relationships (QSPRs) to predict the permeability of new compounds from their structural properties. One of the main challenges that hinders the development of accurate predictions is the existence of multiple Papp values for the same compound, mostly caused by differences in the experimental protocols employed. This review addresses the magnitude of the variability within and between laboratories to interpret its impact on QSPR modelling, systematically and quantitatively assessing the most common sources of variability. This review emphasizes the importance of compiling consistent Papp data and suggests strategies that may be used to obtain such data, contributing to the establishment of robust QSPRs with enhanced predictive power.
Collapse
Affiliation(s)
- Cristiana L. Pires
- Coimbra Chemistry Center—Institute of Molecular Sciences (CQC-IMS), University of Coimbra, 3004-535 Coimbra, Portugal
- Chemistry Department, Faculty of Science and Technology, University of Coimbra, 3004-535 Coimbra, Portugal
| | - Maria João Moreno
- Coimbra Chemistry Center—Institute of Molecular Sciences (CQC-IMS), University of Coimbra, 3004-535 Coimbra, Portugal
- Chemistry Department, Faculty of Science and Technology, University of Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
11
|
Ren J, Ren X, Ma L, Liu J, Yuan S, Wang G. Pharmacokinetics and antioxidant activity of dihydrocaffeic acid grafted chitosan nanomicelles loaded with chicoric acid in broilers. Poult Sci 2024; 103:103776. [PMID: 38688136 PMCID: PMC11077034 DOI: 10.1016/j.psj.2024.103776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/06/2024] [Accepted: 04/13/2024] [Indexed: 05/02/2024] Open
Abstract
Chicoric acid (CA) is a natural nutrient found in plants, showcasing diverse biological activities, including anti-inflammatory and antioxidant properties. Despite its valuable properties, CA faces limitations in bioavailability and susceptibility to oxidative breakdown during utilization. Previous research introduced synthesized dihydrocaffeic acid grafted chitosan self-assembled nanomicelles (DA-g-CS), demonstrating its potential to enhance CA absorption. This study aims to investigate the pharmacokinetics, tissue distribution, and antioxidant activity of both CA and DA-g-CS loaded CA (DA-g-CS/CA) in broilers. An IPEC-J2 cell model was established and evaluated to delve deeper into the transport mechanism and antioxidant potential. The in vivo pharmacokinetic analysis in broilers highlighted a substantial difference: the maximum plasma concentration (Cmax) of DA-g-CS/CA exceeded CA by 2.6-fold, yielding a notable increased relative bioavailability to 214%. This evidence underscores the significant enhancement in CA's oral absorption, facilitated by DA-g-CS. The collective evaluation outcomes affirm the successful development of the cell model, indicating its suitability for drug transporter experiments. The findings from the intestinal transit analysis revealed that both CA and DA-g-CS/CA underwent passive entry into IPEC-J2 cells. Notably, the cellular uptake rate of DA-g-CS loaded with CA was significantly amplified, reaching 2.1 times higher than that of CA alone. Intracellular transport mechanisms involved microtubules, lysosomes, and the endoplasmic reticulum, with an additional pathway involving the endoplasmic reticulum observed specifically for DA-g-CS/CA, distinguishing it from CA. Moreover, the results from both in vivo and in vitro antioxidant assessments highlight the potent antioxidant activity of DA-g-CS/CA, showcasing its efficacy in preventing and treating cellular damage induced by oxidative stress. In summary, these findings underscore the significant enhancement of CA's efficacy facilitated by DA-g-CS, establishing a robust theoretical foundation for the prospective application of CA within livestock and poultry farming.
Collapse
Affiliation(s)
- Juan Ren
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei 071000, People's Republic of China
| | - Xin Ren
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei 071000, People's Republic of China
| | - Leying Ma
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei 071000, People's Republic of China
| | - Juxiang Liu
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei 071000, People's Republic of China
| | - Sikun Yuan
- Baoding Institute for Food and Drug Control, Baoding, Hebei 071000, People's Republic of China
| | - Gengnan Wang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei 071000, People's Republic of China.
| |
Collapse
|
12
|
Grégoire S, Moustié A, Lereaux G, Roussel-Berlier L, Hewitt N. Use of in vitro ADME methods to identify suitable analogs of homosalate and octisalate for use in a read-across safety assessment. J Appl Toxicol 2024; 44:1067-1083. [PMID: 38539266 DOI: 10.1002/jat.4603] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/07/2024] [Accepted: 03/09/2024] [Indexed: 07/21/2024]
Abstract
Case studies are needed to demonstrate the use of human-relevant New Approach Methodologies in cosmetics ingredient safety assessments. For read-across assessments, it is crucial to compare the target chemical with the most appropriate analog; therefore, reliable analog selection should consider physicochemical properties, bioavailability, metabolism, as well as the bioactivity of potential analogs. To complement in vitro bioactivity assays, we evaluated the suitability of three potential analogs for the UV filters, homosalate and octisalate, according to their in vitro ADME properties. We describe how technical aspects of conducting assays for these highly lipophilic chemicals were addressed and interpreted. There were several properties that were common to all five chemicals: they all had similar stability in gastrointestinal fluids (in which no hydrolysis to salicylic occurred); were not substrates of the P-glycoprotein efflux transporter; were highly protein bound; and were hydrolyzed to salicylic acid (which was also a major metabolite). The main properties differentiating the chemicals were their permeability in Caco-2 cells, plasma stability, clearance in hepatic models, and the extent of hydrolysis to salicylic acid. Cyclohexyl salicylate, octisalate, and homosalate were identified suitable analogs for each other, whereas butyloctyl salicylate exhibited ADME properties that were markedly different, indicating it is unsuitable. Isoamyl salicylate can be a suitable analog with interpretation for octisalate. In conclusion, in vitro ADME properties of five chemicals were measured and used to pair target and potential analogs. This study demonstrates the importance of robust ADME data for the selection of analogs in a read-across safety assessment.
Collapse
Affiliation(s)
| | - Anne Moustié
- L'Oréal Research & Innovation, Aulnay-sous Bois, France
| | | | | | | |
Collapse
|
13
|
Du C, Wang P, Li Y, Cong X, Huang D, Chen S, Zhu S. Investigation of selenium and selenium species in Cardamine violifolia using in vitro digestion coupled with a Caco-2 cell monolayer model. Food Chem 2024; 444:138675. [PMID: 38335688 DOI: 10.1016/j.foodchem.2024.138675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 01/16/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024]
Abstract
Inadequate Se intake can enhance vulnerability to certain health risks, with supplementation lessening these risks. This study investigated the bioavailability of Se and Se species in five Se compounds and in Se-rich Cardamine violifolia using in vitro digestion coupled with a Caco-2 cell monolayer model, which enabled the study of Se transport and uptake. Translocation results showed that SeCys2 and MeSeCys had high translocation rates in C. violifolia leaves (CVLs). The uptake rate of organic Se increased with time, and MeSeCys exhibited a higher uptake rate than that for SeCys2 and SeMet. The translocation mechanisms of SeMet, Se(IV), and Se(VI) were passive transport, whereas those of SeCys2 and MeSeCys were active transport. The bioavailability of organic Se was higher than that of inorganic Se, with a total Se bioavailability in CVLs of 49.11 %. This study would provide a theoretical basis for the application of C. violifolia in the functional food.
Collapse
Affiliation(s)
- Chaodong Du
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Peiyu Wang
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yue Li
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xin Cong
- Enshi Se-Run Material Engineering Technology Co., Ltd., Enshi, Hubei 445000, China; National R&D Center for Se-Rich Agricultural Products Processing, Wuhan Polytechnic University, Wuhan 430023, China
| | - Dejian Huang
- Department of Food Science and Technology, National University of Singapore, Singapore 117543, Singapore
| | - Shangwei Chen
- Analysis and Testing Center, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Song Zhu
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
14
|
Olaizola-Rodrigo C, Castro-Abril H, Perisé-Badía I, Pancorbo L, Ochoa I, Monge R, Oliván S. Reducing Inert Materials for Optimal Cell-Cell and Cell-Matrix Interactions within Microphysiological Systems. Biomimetics (Basel) 2024; 9:262. [PMID: 38786472 PMCID: PMC11118140 DOI: 10.3390/biomimetics9050262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/19/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
In the pursuit of achieving a more realistic in vitro simulation of human biological tissues, microfluidics has emerged as a promising technology. Organ-on-a-chip (OoC) devices, a product of this technology, contain miniature tissues within microfluidic chips, aiming to closely mimic the in vivo environment. However, a notable drawback is the presence of inert material between compartments, hindering complete contact between biological tissues. Current membranes, often made of PDMS or plastic materials, prevent full interaction between cell types and nutrients. Furthermore, their non-physiological mechanical properties and composition may induce unexpected cell responses. Therefore, it is essential to minimize the contact area between cells and the inert materials while simultaneously maximizing the direct contact between cells and matrices in different compartments. The main objective of this work is to minimize inert materials within the microfluidic chip while preserving proper cellular distribution. Two microfluidic devices were designed, each with a specific focus on maximizing direct cell-matrix or cell-cell interactions. The first chip, designed to increase direct cell-cell interactions, incorporates a nylon mesh with regular pores of 150 microns. The second chip minimizes interference from inert materials, thereby aiming to increase direct cell-matrix contact. It features an inert membrane with optimized macropores of 1 mm of diameter for collagen hydrogel deposition. Biological validation of both devices has been conducted through the implementation of cell migration and cell-to-cell interaction assays, as well as the development of epithelia, from isolated cells or spheroids. This endeavor contributes to the advancement of microfluidic technology, aimed at enhancing the precision and biological relevance of in vitro simulations in pursuit of more biomimetic models.
Collapse
Affiliation(s)
- Claudia Olaizola-Rodrigo
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (C.O.-R.); (H.C.-A.); (I.P.-B.); (S.O.)
- BEOnChip S.L., 50018 Zaragoza, Spain; (L.P.); (R.M.)
| | - Héctor Castro-Abril
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (C.O.-R.); (H.C.-A.); (I.P.-B.); (S.O.)
- Laboratorio de Biomiméticos, Universidad Nacional de Colombia, Bogotá 111321, Colombia
| | - Ismael Perisé-Badía
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (C.O.-R.); (H.C.-A.); (I.P.-B.); (S.O.)
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Lara Pancorbo
- BEOnChip S.L., 50018 Zaragoza, Spain; (L.P.); (R.M.)
| | - Ignacio Ochoa
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (C.O.-R.); (H.C.-A.); (I.P.-B.); (S.O.)
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Rosa Monge
- BEOnChip S.L., 50018 Zaragoza, Spain; (L.P.); (R.M.)
| | - Sara Oliván
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (C.O.-R.); (H.C.-A.); (I.P.-B.); (S.O.)
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
| |
Collapse
|
15
|
Ding F, Wang H, Li Y, Leng X, Gao J, Huang D. Polystyrene microplastics with absorbed nonylphenol induce intestinal dysfunction in human Caco-2 cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 107:104426. [PMID: 38527597 DOI: 10.1016/j.etap.2024.104426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 03/21/2024] [Indexed: 03/27/2024]
Abstract
Due to the massive production and use of plastic, the chronic and evolving exposure to microplastics in our daily lives is omnipresent. Nonylphenol (NP), a persistent organic pollutant, may change toxicity when it co-exists with microplastics. In this study, polystyrene microplastics (PS-MPs), either alone or with pre-absorbed NP, generated oxidative stress and inflammatory lesions to Caco-2 cells, as well as affecting proliferation via the MAPK signaling pathway and causing apoptosis. Damage to cell membrane integrity and intestinal barrier (marked by lower transepithelial electric resistance, greater bypass transport, and tight junction structural changes) leads to enhanced internalization risk of PS-MPs. Some important intestinal functions including nutrient absorption and xenobiotic protection were also harmed. It is worth noting that the exposure of PS-MPs with a diameter of 0.1 μm improved intestinal functions quickly but acted as a chemosensitizer for a long time, inhibiting cell perception of other toxic substances and making the cells more vulnerable.
Collapse
Affiliation(s)
- Fangfang Ding
- State Key Laboratory of Food Science and Resource, International Institute of Food Innovation Co., Ltd., China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Huimei Wang
- State Key Laboratory of Food Science and Resource, International Institute of Food Innovation Co., Ltd., China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Yingzhi Li
- State Key Laboratory of Food Science and Resource, International Institute of Food Innovation Co., Ltd., China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Xueping Leng
- State Key Laboratory of Food Science and Resource, International Institute of Food Innovation Co., Ltd., China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Jiaming Gao
- State Key Laboratory of Food Science and Resource, International Institute of Food Innovation Co., Ltd., China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Danfei Huang
- State Key Laboratory of Food Science and Resource, International Institute of Food Innovation Co., Ltd., China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| |
Collapse
|
16
|
Jaber N, Billet S. How to use an in vitro approach to characterize the toxicity of airborne compounds. Toxicol In Vitro 2024; 94:105718. [PMID: 37871865 DOI: 10.1016/j.tiv.2023.105718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/13/2023] [Accepted: 09/21/2023] [Indexed: 10/25/2023]
Abstract
As part of the development of new approach methodologies (NAMs), numerous in vitro methods are being developed to characterize the potential toxicity of inhalable xenobiotics (gases, volatile organic compounds, polycyclic aromatic hydrocarbons, particulate matter, nanoparticles). However, the materials and methods employed are extremely diverse, and no single method is currently in use. Method standardization and validation would raise trust in the results and enable them to be compared. This four-part review lists and compares biological models and exposure methodologies before describing measurable biomarkers of exposure or effect. The first section emphasizes the importance of developing alternative methods to reduce, if not replace, animal testing (3R principle). The biological models presented are mostly to cultures of epithelial cells from the respiratory system, as the lungs are the first organ to come into contact with air pollutants. Monocultures or cocultures of primary cells or cell lines, as well as 3D organotypic cultures such as organoids, spheroids and reconstituted tissues, but also the organ(s) model on a chip are examples. The exposure methods for these biological models applicable to airborne compounds are submerged, intermittent, continuous either static or dynamic. Finally, within the restrictions of these models (i.e. relative tiny quantities, adhering cells), the mechanisms of toxicity and the phenotypic markers most commonly examined in models exposed at the air-liquid interface (ALI) are outlined.
Collapse
Affiliation(s)
- Nour Jaber
- UR4492, Unité de Chimie Environnementale et Interactions sur le Vivant, Université du Littoral Côte d'Opale, Dunkerque, France
| | - Sylvain Billet
- UR4492, Unité de Chimie Environnementale et Interactions sur le Vivant, Université du Littoral Côte d'Opale, Dunkerque, France.
| |
Collapse
|
17
|
Chomanicova N, Adamickova A, Valaskova S, Gazova A, Kyselovic J. Caco-2 Monolayer as a Model of the Intestinal Barrier: Permeability of Magnesium Salts. Methods Mol Biol 2024; 2835:301-306. [PMID: 39105925 DOI: 10.1007/978-1-0716-3995-5_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
Magnesium, an essential mineral for various physiological functions, is subject to tight regulation within the body. Understanding its absorption across epithelial cell monolayers is crucial for optimizing dietary magnesium intake and therapeutic strategies. The Caco-2 monolayer model, widely recognized for its relevance to the human intestinal epithelium, provides a suitable platform for this investigation. This protocol covers the step-by-step procedures for the cultivation of Caco-2 monolayer preparation of transwell systems. It provides guidance on the setup of magnesium transport experiments, which involve the application of magnesium salts to the apical side of the Caco-2 monolayer and monitoring their transport to the basolateral side.
Collapse
Affiliation(s)
- Nikola Chomanicova
- International Laser Centre, Slovak Centre of Scientifc and Technical Information, Bratislava, Slovakia
| | - Adriana Adamickova
- Faculty of Medicine, 5th Department of Internal Medicine, Comenius University Bratislava, Bratislava, Slovakia
| | - Simona Valaskova
- International Laser Centre, Slovak Centre of Scientifc and Technical Information, Bratislava, Slovakia
| | - Andrea Gazova
- Faculty of Medicine, Institute of Pharmacology and Clinical Pharmacology, Comenius University Bratislava, Bratislava, Slovakia
| | - Jan Kyselovic
- Faculty of Medicine, 5th Department of Internal Medicine, Comenius University Bratislava, Bratislava, Slovakia
| |
Collapse
|
18
|
Kus M, Ibragimow I, Piotrowska-Kempisty H. Caco-2 Cell Line Standardization with Pharmaceutical Requirements and In Vitro Model Suitability for Permeability Assays. Pharmaceutics 2023; 15:2523. [PMID: 38004503 PMCID: PMC10674574 DOI: 10.3390/pharmaceutics15112523] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/19/2023] [Accepted: 10/19/2023] [Indexed: 11/26/2023] Open
Abstract
The Caco-2 cell line derived from human colon carcinoma is commonly used to assess the permeability of compounds in in vitro conditions. Due to the significant increase in permeability studies using the Caco-2 cell line in recent years, the need to standardize this biological model seems necessary. The pharmaceutical requirements define only the acceptance criteria for the validation of the Caco-2 cell line and do not specify the protocol for its implementation. Therefore, the aim of this study is to review the conditions for permeability studies across the Caco-2 monolayer reported in the available literature concerning validation guidelines. We summarized the main aspects affecting the validation process of the Caco-2 cell line, including the culture conditions, cytotoxicity, cell differentiation process, and monolayer transport conditions, and the main conclusions may be useful in developing individual methods for preparing the cell line for validation purposes and further permeability research.
Collapse
Affiliation(s)
- Marta Kus
- Department of Toxicology, Poznan University of Medical Sciences, 30 Dojazd St., 60-631 Poznan, Poland;
- Research and Development Department of Ethifarm, Ethifarm Manufacturing Plant, 9 Stefana Zeromskiego St., 60-544 Poznan, Poland;
| | - Izabela Ibragimow
- Research and Development Department of Ethifarm, Ethifarm Manufacturing Plant, 9 Stefana Zeromskiego St., 60-544 Poznan, Poland;
| | - Hanna Piotrowska-Kempisty
- Department of Toxicology, Poznan University of Medical Sciences, 30 Dojazd St., 60-631 Poznan, Poland;
- Department of Basic and Preclinical Science, Institute of Veterinary Medicine, Nicolaus Copernicus University in Toruń, 7 Gagarina St., 87-100 Torun, Poland
| |
Collapse
|
19
|
Jabor Z, Sutton SC. Effects of Digestion, Cell Culture Media, and Mucous on the Physical Properties, Cellular Effects, and Translocation of Polystyrene and Polymethacrylate Nanoparticles. TOXICS 2023; 11:708. [PMID: 37624213 PMCID: PMC10458608 DOI: 10.3390/toxics11080708] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/27/2023] [Accepted: 08/12/2023] [Indexed: 08/26/2023]
Abstract
The discovery of plastic and metal nanoparticles in organisms, foods, and beverages has generated numerous studies on the effects of these particles on the barrier cells and their subsequent absorption into the body. Following ingestion, nanoparticles travel down the gastrointestinal tract (GIT), and their physicochemical characteristics change in response to the change in proteins and pH during their digestion. We measured the translocation of digested nanoparticles across a co-culture monolayer of Caco-2 and various combinations (1:9, 5:5, and 9:1) of HT29-MTX-E12. The in vitro model of the intestine was used to determine the translocation of digested 20 nm polymethacrylate (PMA) particles and the accompanying monolayer barrier effects after a 72 h exposure. The in vitro digestion increased the agglomeration and hydrodynamic diameters and decreased the surface charge of the nanoparticles. For NH2-functionalized polymethacrylate nanoparticles (PMA-NH2), the diameters increased from 57 nm (water) to 3800 nm (media), or 2660 nm (chyme). These nanoparticles compromised the integrity of the monolayer (trans-epithelial electrical resistance, Lucifer yellow translocation) and translocated across all the cell ratio configurations. Digestion can have a large effect on nanoparticle agglomeration and surface charge. Excess mucous was not seen as a barrier to the translocation of PMA-NH2.
Collapse
Affiliation(s)
- Zainab Jabor
- School of Pharmacy, Westbrook College of Health Professions, University of New England, 716 Stevens Ave, Portland, ME 04103, USA
| | - Steven C. Sutton
- Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Westbrook College of Health Professions, University of New England, 716 Stevens Ave, Portland, ME 04103, USA
| |
Collapse
|
20
|
Masloh S, Culot M, Gosselet F, Chevrel A, Scapozza L, Zeisser Labouebe M. Challenges and Opportunities in the Oral Delivery of Recombinant Biologics. Pharmaceutics 2023; 15:pharmaceutics15051415. [PMID: 37242657 DOI: 10.3390/pharmaceutics15051415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Recombinant biological molecules are at the cutting-edge of biomedical research thanks to the significant progress made in biotechnology and a better understanding of subcellular processes implicated in several diseases. Given their ability to induce a potent response, these molecules are becoming the drugs of choice for multiple pathologies. However, unlike conventional drugs which are mostly ingested, the majority of biologics are currently administered parenterally. Therefore, to improve their limited bioavailability when delivered orally, the scientific community has devoted tremendous efforts to develop accurate cell- and tissue-based models that allow for the determination of their capacity to cross the intestinal mucosa. Furthermore, several promising approaches have been imagined to enhance the intestinal permeability and stability of recombinant biological molecules. This review summarizes the main physiological barriers to the oral delivery of biologics. Several preclinical in vitro and ex vivo models currently used to assess permeability are also presented. Finally, the multiple strategies explored to address the challenges of administering biotherapeutics orally are described.
Collapse
Affiliation(s)
- Solene Masloh
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, University of Artois, UR 2465, Rue Jean Souvraz, 62300 Lens, France
- Affilogic, 24 Rue de la Rainière, 44300 Nantes, France
- School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
| | - Maxime Culot
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, University of Artois, UR 2465, Rue Jean Souvraz, 62300 Lens, France
| | - Fabien Gosselet
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, University of Artois, UR 2465, Rue Jean Souvraz, 62300 Lens, France
| | - Anne Chevrel
- Affilogic, 24 Rue de la Rainière, 44300 Nantes, France
| | - Leonardo Scapozza
- School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
| | - Magali Zeisser Labouebe
- School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
| |
Collapse
|
21
|
Ta W, Li X, Song J, Hua R, Zheng Y, Lu W. Customizable Dual-Fluorescent Nanoparticles for Tracing and Quantifying of Cell Transport. Int J Nanomedicine 2023; 18:1823-1834. [PMID: 37041817 PMCID: PMC10083028 DOI: 10.2147/ijn.s394953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 03/07/2023] [Indexed: 04/08/2023] Open
Abstract
Purpose Nanotechnology-based drug delivery systems (nano-DDS) have been developed to be a promising strategy to improve the efficacy, safety, physicochemical and pharmacokinetic/pharmacodynamics properties of drugs. It is very necessary to elucidate the delivery process in vivo or in cells for the rational design and accurate preparation of nano-DDS. The aim of this study was to construct a nano-DDS to visualize and quantify the intracellular behavior of the loaded cargo and carrier in such a system. Methods A carboxyl-terminal end of poly(lactic-co-glycolic acid) polymer was fluorescently labeled with rhodamine B by conjugation of ethylenediamine. Dual-fluorescent nanoparticles (DFPs) were prepared from this fluorescently labeled polymer to encapsulate a fluorescent cargo, coumarin 6. The carrier and cargo of DFPs were monitored by confocal fluorescence microscopy during cellular uptake. Furthermore, the transcellular transportation of DFPs was evaluated quantitatively by measuring the fluorescence intensity. Results The obtained fluorescent polymer showed stable and quantifiable characteristics. DFPs could be customized in terms of coumarin 6 content (97.7±1.0%), size (367.3±1.7 nm) and dual-emission fluorescence (green cargo and red carrier). DFPs did not significantly affect cell viability, the integrity of cell monolayers and the microscopic morphology at concentrations below 0.7 mg/mL within 3 h of co-incubation with Caco-2 cells. Multichannel fluorescence monitoring revealed that the fluorescence intensity of the carrier and cargo increased with time, but not synchronously. By calculating the residual, intracellular, and transport amounts of DFPs, the material balance between the total amount of cellular transport and the dose administered was obtained. Conclusion Based on the advantages of dual fluorescent labeling, the differential behavior of cell trafficking can be visualized and quantitatively analyzed for the cargo and carrier of DFPs. These results provide insights into the cellular transport process of holistic nanoparticles and complement our understanding of the biological behaviors of nano-DDS.
Collapse
Affiliation(s)
- Wenjing Ta
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Xingyue Li
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Jihong Song
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Ruochen Hua
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Yuting Zheng
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Wen Lu
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, People’s Republic of China
- Correspondence: Wen Lu, School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China, Email
| |
Collapse
|
22
|
Huang S, Zhai B, Fan Y, Sun J, Cheng J, Zou J, Zhang X, Shi Y, Guo D. Development of Paeonol Liposomes: Design, Optimization, in vitro and in vivo Evaluation. Int J Nanomedicine 2022; 17:5027-5046. [PMID: 36303804 PMCID: PMC9594912 DOI: 10.2147/ijn.s363135] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 09/07/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is one of the intractable diseases recognized by the World Health Organization, and paeonol has been proven to have therapeutic effects. However, the low solubility of paeonol limits its clinical application. To prepare and optimize paeonol liposome, study its absorption mechanism and the anti-inflammatory activity in vitro and in vivo, in order to provide experimental basis for the further development of paeonol into an anti-inflammatory drug in the future. METHODS Paeonol loaded liposomes were prepared and optimized by thin film dispersion-ultrasonic method. The absorption mechanism of paeonol-loaded liposomes was studied by pharmacokinetics, in situ single-pass intestinal perfusion and Caco-2 cell monolayer model, the anti-inflammatory activity was studied in a mouse ulcerative model. RESULTS Box-Behnken response surface methodology permits to screen the best formulations. The structural and morphological characterization showed that paeonol was entrapped inside the bilayer in liposomes. Pharmacokinetic studies found that the AUC0-t of Pae-Lips was 2.78 times than that of paeonol suspension, indicating that Pae-Lips significantly improved the absorption of paeonol. In situ single intestinal perfusion and Caco-2 monolayer cell model results showed that paeonol was passively transported and absorbed, and was the substrate of P-gp, MRP2 and BCRP, and the Papp value of Pae-Lips was significantly higher than that of paeonol. In vitro and in vivo anti-inflammatory experiments showed that compared with paeonol, Pae-Lips exhibited excellent anti-inflammatory activity. CONCLUSION In this study, Pae-Lips were successfully prepared to improve the oral absorption of paeonol. Absorption may involve passive diffusion and efflux transporters. Moreover, Pae-Lips have excellent anti-inflammatory activity in vitro and in vivo, which preliminarily clarifies the feasibility of further development of Pae-Lips into oral anti-inflammatory drugs.
Collapse
Affiliation(s)
- Shan Huang
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, People’s Republic of China
| | - Bingtao Zhai
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, People’s Republic of China
| | - Yu Fan
- School of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, People’s Republic of China
| | - Jing Sun
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, People’s Republic of China
| | - Jiangxue Cheng
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, People’s Republic of China
| | - Junbo Zou
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, People’s Republic of China
| | - Xiaofei Zhang
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, People’s Republic of China
| | - Yajun Shi
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, People’s Republic of China
| | - Dongyan Guo
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, People’s Republic of China,Correspondence: Dongyan Guo, Tel +86-029-38185180, Email
| |
Collapse
|
23
|
Ribeiro-Oliveira R, Martins ZE, Faria MÂ, Sousa JB, Ferreira IMPLVO, Diniz C. Protein Hydrolysates from Brewing By-Products as Natural Alternatives to ACE-Inhibitory Drugs for Hypertension Management. LIFE (BASEL, SWITZERLAND) 2022; 12:life12101554. [PMID: 36294989 PMCID: PMC9605471 DOI: 10.3390/life12101554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/28/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022]
Abstract
Simple Summary Hypertension is the predominant risk factor for cardiovascular disease, which is the leading cause of mortality and morbidity worldwide. The search for natural compounds with antihypertensive properties, such as bioactive peptides from brewing by-products (spent grain and yeast), which are less likely to cause severe side effects compared with anti-hypertensive drugs, is of major importance to reduce cardiovascular events. Since oral intake of these peptides may modify their expected effects, the aim of the present study was to simulate oral administration and evaluate the impact of gastrointestinal digestion, intestinal absorption, and liver metabolism on the effectiveness of those bioactive peptides and determine their potential to be used as supplements or nutraceuticals as well as anti-hypertensive drugs before moving forward to animal studies. Results showed that peptides derived from the brewing industry maintain or present higher antihypertensive activity after simulation of oral administration, validating the usefulness of these peptides to reduce the risk, ameliorate, or treat primary hypertension. In conclusion, this study reinforces, through in vitro studies, the benefits of oral administrated brewing bioactive peptides to directly manage hypertension by lowering blood pressure, thus being promising compounds. Abstract The treatment of hypertension is of major importance to reduce the risk of cardiovascular disease, the leading cause of death worldwide. Angiotensin-converting enzyme (ACE) inhibitors are anti-hypertensive drugs associated with several side effects. Natural products, namely bioactive peptides from brewing by-products, brewers’ spent grain (BSG), and yeast (BSY), are promising alternatives since they can inhibit ACE in vitro. However, the oral intake of these peptides may modify their expected inhibitory effect owing to possible changes in active peptides’ bioavailability, which have not been assessed so far. The goal of this study was to simulate oral administration to evaluate BSG/BSY peptides’ effectiveness by submitting protein hydrolysates sequentially to simulated gastrointestinal digestion, intestinal absorption (Caco-2 cells), and liver metabolism (HepG2 cells). MTT assay was used to assess BSG/BSY protein hydrolysates safeness. The ACE-inhibitory potential of initial and final protein hydrolysates (BSY, BSG, and a new product, MIX) were tested using a fluorometric assay and compared with captopril (1 µM, an ACE-inhibitory drug). Simulation of oral administration greatly increased BSY and MIX protein hydrolysates’ ACE-inhibitory capacity, though final MIX and BSG revealed greater ACE-inhibitory potential than captopril. Notwithstanding, all final protein hydrolysates presented ACE-inhibitory capacity, thus being promising compounds to manage hypertension.
Collapse
Affiliation(s)
- Rita Ribeiro-Oliveira
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Zita E. Martins
- LAQV/REQUIMTE, Laboratory of Bromatology and Hydrology, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Miguel Ângelo Faria
- LAQV/REQUIMTE, Laboratory of Bromatology and Hydrology, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Joana Beatriz Sousa
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Isabel M. P. L. V. O. Ferreira
- LAQV/REQUIMTE, Laboratory of Bromatology and Hydrology, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Correspondence: (I.M.P.L.V.O.F.); (C.D.)
| | - Carmen Diniz
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Correspondence: (I.M.P.L.V.O.F.); (C.D.)
| |
Collapse
|
24
|
Effect of Coffee on the Bioavailability of Sterols. Foods 2022; 11:foods11192935. [PMID: 36230011 PMCID: PMC9563500 DOI: 10.3390/foods11192935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/12/2022] [Accepted: 09/16/2022] [Indexed: 11/24/2022] Open
Abstract
Absorption at the intestinal epithelium is a major determinant of cholesterol levels in the organism, influencing the entry of dietary cholesterol and the excretion of endogenous cholesterol. Several strategies are currently being followed to reduce cholesterol absorption, using both pharmacological agents or food ingredients with hypocholesterolemic properties. Coffee has recently been shown to affect cholesterol bioaccessibility, although it has not been shown if this translates into a decrease on cholesterol bioavailability. In this work, coffee obtained with different commercial roasting (light and dark) and grinding (finer and coarser) was evaluated regarding their effect on cholesterol absorption through Caco-2 monolayers, mimicking the intestinal epithelium. The fluorescent dehydroergosterol was used as a sterol model, which was shown to permeate Caco-2 monolayers with a low-to-moderate permeability coefficient depending on its concentration. In the presence of coffee extracts, a 50% decrease of the sterol permeability coefficient was observed, showing their potential to affect sterol bioavailability. This was attributed to an increased sterol precipitation and its deposition on the apical epithelial surface. A higher hypocholesterolemic effect was observed for the dark roasting and finer grinding, showing that the modulation of these technological processing parameters may produce coffees with optimized hypocholesterolemic activity.
Collapse
|
25
|
Visigalli R, Rotoli BM, Ferrari F, Di Lascia M, Riccardi B, Puccini P, Dall’Asta V, Barilli A. Expression and Function of ABC Transporters in Human Alveolar Epithelial Cells. Biomolecules 2022; 12:biom12091260. [PMID: 36139099 PMCID: PMC9496151 DOI: 10.3390/biom12091260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 12/02/2022] Open
Abstract
ATP-binding cassette (ABC) transporters are a large superfamily of membrane transporters that facilitate the translocation of different substrates. While ABC transporters are clearly expressed in various tumor cells where they can play a role in drug extrusion, the presence of these transporters in normal lung tissues is still controversial. Here, we performed an analysis of ABC transporters in EpiAlveolarTM, a recently developed model of human alveoli, by defining the expression and activity of MDR1, BCRP, and MRPs. Immortalized primary epithelial cells hAELVi (human alveolar epithelial lentivirus-immortalized cells) were employed for comparison. Our data underline a close homology between these two models, where none of the ABC transporters here studied are expressed on the apical membrane and only MRP1 is clearly detectable and functional at the basolateral side. According to these findings, we can conclude that other thus-far-unidentified transporter/s involved in drug efflux from alveolar epithelium deserve investigations.
Collapse
Affiliation(s)
- Rossana Visigalli
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy
| | - Bianca Maria Rotoli
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy
| | - Francesca Ferrari
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy
| | - Maria Di Lascia
- Preclinical Pharmacokinetics, Biochemistry & Metabolism Department, Chiesi Farmaceutici, 43122 Parma, Italy
| | - Benedetta Riccardi
- Preclinical Pharmacokinetics, Biochemistry & Metabolism Department, Chiesi Farmaceutici, 43122 Parma, Italy
| | - Paola Puccini
- Preclinical Pharmacokinetics, Biochemistry & Metabolism Department, Chiesi Farmaceutici, 43122 Parma, Italy
| | - Valeria Dall’Asta
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy
| | - Amelia Barilli
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy
- Correspondence:
| |
Collapse
|
26
|
Przybylla R, Mullins CS, Krohn M, Oswald S, Linnebacher M. Establishment and Characterization of Novel Human Intestinal In Vitro Models for Absorption and First-Pass Metabolism Studies. Int J Mol Sci 2022; 23:9861. [PMID: 36077251 PMCID: PMC9456142 DOI: 10.3390/ijms23179861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/26/2022] [Accepted: 08/26/2022] [Indexed: 11/23/2022] Open
Abstract
Commonly used intestinal in vitro models are limited in their potential to predict oral drug absorption. They either lack the capability to form a tight cellular monolayer mimicking the intestinal epithelial barrier or the expression of cytochrome P450 3A4 (CYP3A4). The aim of this study was to establish a platform of colorectal cancer patient-derived cell lines for evaluation of human intestinal drug absorption and metabolism. We characterized ten 2D cell lines out of our collection with confluent outgrowth and long-lasting barrier forming potential as well as suitability for high throughput applications with special emphasis on expression and inducibility of CYP3A4. By assessment of the transepithelial electrical resistance (TEER) the cells barrier function capacity can be quantified. Very high TEER levels were detected for HROC60. A high basal CYP3A4 expression and function was found for HROC32. Eight cell lines showed higher CYP3A4 induction by stimulation via the vitamin D receptor compared to Caco-2 cells (5.1- to 16.8-fold change). Stimulation of the pregnane X receptor led to higher CYP3A4 induction in two cell lines. In sum, we identified the two cell lines HROC183 T0 M2 and HROC217 T1 M2 as useful tools for in vitro drug absorption studies. Due to their high TEER values and inducibility by drug receptor ligands, they may be superior to Caco-2 cells to analyze oral drug absorption and intestinal drug-drug interactions. Significance statement: Selecting appropriate candidates is important in preclinical drug development. Therefore, cell models to predict absorption from the human intestine are of the utmost importance. This study revealed that the human cell lines HROC183 T0 M2 and HROC217 T1 M2 may be better suited models and possess higher predictive power of pregnane X receptor- and vitamin D-mediated drug metabolism than Caco-2 cells. Consequently, they represent useful tools for predicting intestinal absorption and simultaneously enable assessment of membrane permeability and first-pass metabolism.
Collapse
Affiliation(s)
- Randy Przybylla
- Molecular Oncology and Immunotherapy, Clinic of General Surgery, 18057 Rostock, Germany
| | | | - Mathias Krohn
- Molecular Oncology and Immunotherapy, Clinic of General Surgery, 18057 Rostock, Germany
| | - Stefan Oswald
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Michael Linnebacher
- Molecular Oncology and Immunotherapy, Clinic of General Surgery, 18057 Rostock, Germany
| |
Collapse
|
27
|
Zheng M, Guo Y, Li W, Wu M, Xu M, Shao M, He G, Liu Y. Medium Chain Triglycerides Promote the Uptake of β-Carotene in O/W Emulsions via Intestinal Transporter SR-B1 in Caco-2 Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:9377-9387. [PMID: 35861437 DOI: 10.1021/acs.jafc.2c02660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
This study aimed to elucidate the impacts of carrier oil types (long chain triglycerides (LCT), medium chain triglycerides (MCT), and orange oil (indigestible oil)) on the micellization and cellular uptake of β-carotene (BC) formulated in O/W emulsions, with an emphasis on the role of intestinal transporters. The micellization and cellular uptake of BC in the gastrointestinal tract were evaluated via an in vitro digestion model and a Caco-2 cell monolayer. And the interactions between lipids and intestinal transporters were monitored by nontargeted lipidomics, RT-PCR, and Western blot. The BC micellization rates followed a decreasing trend in emulsions: corn oil (69.47 ± 4.19%) > MCT (22.22 ± 0.89%) > orange oil (11.01 ± 2.86%), whereas the cellular uptake rate of BC was significantly higher in MCT emulsion (56.30 ± 20.13%) than in corn oil emulsion (14.01 ± 1.04%, p < 0.05). The knockdown of SR-B1 led to a 31.63% loss of BC cellular uptake from MCT micelles but had no effect on corn oil micelles. Lipidomics and transporter analysis revealed that TG (10:0/10:0/12:0) and TG (10:0/12:0/12:0) might be the fingerprint lipids that promoted the cellular absorption of BC-MCT micelles via stimulating the mRNA expression of SR-B1.
Collapse
Affiliation(s)
- Mengman Zheng
- School of Public Health, Fudan University/Key Laboratory of Public Health Safety, Ministry of Education, Shanghai 200032, China
- Department of Nutriology, Affiliated Hospital of Shaoxing University (Shaoxing Municipal Hospital), Shaoxing, Zhejiang 312000, China
| | - Yi Guo
- School of Public Health, Fudan University/Key Laboratory of Public Health Safety, Ministry of Education, Shanghai 200032, China
| | - Wenyun Li
- School of Public Health, Fudan University/Key Laboratory of Public Health Safety, Ministry of Education, Shanghai 200032, China
| | - Min Wu
- School of Public Health, Fudan University/Key Laboratory of Public Health Safety, Ministry of Education, Shanghai 200032, China
| | - Mingjing Xu
- School of Public Health, Fudan University/Key Laboratory of Public Health Safety, Ministry of Education, Shanghai 200032, China
| | - Manman Shao
- School of Public Health, Fudan University/Key Laboratory of Public Health Safety, Ministry of Education, Shanghai 200032, China
| | - Gengsheng He
- School of Public Health, Fudan University/Key Laboratory of Public Health Safety, Ministry of Education, Shanghai 200032, China
| | - Yuwei Liu
- School of Public Health, Fudan University/Key Laboratory of Public Health Safety, Ministry of Education, Shanghai 200032, China
| |
Collapse
|
28
|
Wang Y, Zhang X, Zhuang W, Yu Y, Sun X, Wang H, Li F, Li Q. Investigation of the Uptake and Transport of Two Novel Camptothecin Derivatives in Caco-2 Cell Monolayers. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27123669. [PMID: 35744795 PMCID: PMC9230870 DOI: 10.3390/molecules27123669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 02/07/2023]
Abstract
Irinotecan and Topotecan are two Camptothecin derivatives (CPTs) whose resistance is associated with the high expression of breast cancer resistance protein (BCRP) and P-glycoprotein (P-gp). To reverse this resistance, two novel CPTs, FL77-28 (7-(3-Fluoro-4-methylphenyl)-10,11-methylenedioxy-20(S)-CPT) and FL77-29 (7-(4-Fluoro-3-methylphenyl)-10,11-methylenedioxy-20(S)-CPT), were synthesized by our group. In this study, the anti-tumor activities of FL77-28, FL77-29, and their parent, FL118 (10,11-methylenedioxy-20(S)-CPT), were evaluated and the results showed that FL77-28 and FL77-29 had stronger anti-tumor activities than FL118. The transport and uptake of FL118, FL77-28, and FL77-29 were investigated in Caco-2 cells for the preliminary prediction of intestinal absorption. The apparent permeability coefficient from apical to basolateral (Papp AP-BL) values of FL77-28 and FL77-29 were (2.32 ± 0.04) × 10−6 cm/s and (2.48 ± 0.18) × 10−6 cm/s, respectively, suggesting that the compounds had moderate absorption. Since the transport property of FL77-28 was passive diffusion and the efflux ratio (ER) was less than 2, two chemical inhibitors were added to further confirm the involvement of efflux proteins. The results showed that FL77-28 was not a substrate of P-gp or BCRP, but FL77-29 was mediated by P-gp. In conclusion, FL77-28 might be a promising candidate to overcome drug resistance induced by multiple efflux proteins.
Collapse
Affiliation(s)
- Yi Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310032, China; (Y.W.); (X.Z.); (W.Z.); (Y.Y.); (X.S.); (H.W.)
| | - Xiangli Zhang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310032, China; (Y.W.); (X.Z.); (W.Z.); (Y.Y.); (X.S.); (H.W.)
| | - Wenya Zhuang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310032, China; (Y.W.); (X.Z.); (W.Z.); (Y.Y.); (X.S.); (H.W.)
| | - Yanlei Yu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310032, China; (Y.W.); (X.Z.); (W.Z.); (Y.Y.); (X.S.); (H.W.)
| | - Xuanrong Sun
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310032, China; (Y.W.); (X.Z.); (W.Z.); (Y.Y.); (X.S.); (H.W.)
| | - Hong Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310032, China; (Y.W.); (X.Z.); (W.Z.); (Y.Y.); (X.S.); (H.W.)
| | - Fengzhi Li
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Qingyong Li
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310032, China; (Y.W.); (X.Z.); (W.Z.); (Y.Y.); (X.S.); (H.W.)
- Correspondence: ; Tel./Fax: +86-571-8832-0984
| |
Collapse
|
29
|
Effects of CB2 Receptor Modulation on Macrophage Polarization in Pediatric Celiac Disease. Biomedicines 2022; 10:biomedicines10040874. [PMID: 35453624 PMCID: PMC9029516 DOI: 10.3390/biomedicines10040874] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/04/2022] [Accepted: 04/07/2022] [Indexed: 01/27/2023] Open
Abstract
Celiac Disease (CD) represents an autoimmune disorder triggered by the exposure to gluten in genetically susceptible individuals. Recent studies suggest the involvement of macrophages in CD pathogenesis. Macrophages are immune cells, present as pro-inflammatory classically activated macrophages (M1) or as anti-inflammatory alternatively activated macrophages (M2). The Cannabinoid Receptor 2 (CB2) has important anti-inflammatory and immunoregulatory properties. We previously demonstrated that a common CB2 functional variant, Q63R, causing CB2 reduced function, is associated with several inflammatory and autoimmune diseases The first aim of this study was to investigate the phenotype of macrophages isolated from peripheral blood of CD patients and CB2 expression. The second aim was to evaluate the effects of CB2 pharmacological modulation on CD macrophage polarization. Moreover, by an in vitro model of “immunocompetent gut” we investigated the role of CD macrophages in inducing intestinal barrier damage and the possibility to restore its functionality modulating their polarization. We found an increased expression of M1 macrophages and a CB2 reduced expression. We also demonstrated CD M1 macrophages in inducing the typical mucosal barrier damage of CD. CB2 stimulation switches macrophage polarization towards the anti-inflammatory M2 phenotype thus reducing inflammation but also limiting the epithelial dysfunction. Therefore, we suggest CB2 receptor as a possible novel therapeutic target for CD by regulating macrophages polarization and by preventing mucosal barrier damage.
Collapse
|
30
|
Cordeiro MM, Salvador A, Moreno MJ. Calculation of Permeability Coefficients from Solute Equilibration Dynamics: An Assessment of Various Methods. MEMBRANES 2022; 12:membranes12030254. [PMID: 35323728 PMCID: PMC8951150 DOI: 10.3390/membranes12030254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 02/01/2023]
Abstract
Predicting the rate at which substances permeate membrane barriers in vivo is crucial for drug development. Permeability coefficients obtained from in vitro studies are valuable for this goal. These are normally determined by following the dynamics of solute equilibration between two membrane-separated compartments. However, the correct calculation of permeability coefficients from such data is not always straightforward. To address these problems, here we develop a kinetic model for solute permeation through lipid membrane barriers that includes the two membrane leaflets as compartments in a four-compartment model. Accounting for solute association with the membrane allows assessing various methods in a wide variety of conditions. The results showed that the often-used expression Papp= β × r/3 is inapplicable to very large or very small vesicles, to moderately or highly lipophilic solutes, or when the development of a significant pH gradient opposes the solute’s flux. We establish useful relationships that overcome these limitations and allow predicting permeability in compartmentalised in vitro or in vivo systems with specific properties. Finally, from the parameters for the interaction of the solute with the membrane barrier, we defined an intrinsic permeability coefficient that facilitates quantitative comparisons between solutes.
Collapse
Affiliation(s)
- Margarida M. Cordeiro
- Coimbra Chemistry Centre-Institute of Molecular Sciences (CQC-IMS), University of Coimbra, 3004-535 Coimbra, Portugal;
- Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal
| | - Armindo Salvador
- Coimbra Chemistry Centre-Institute of Molecular Sciences (CQC-IMS), University of Coimbra, 3004-535 Coimbra, Portugal;
- CNC—Centre for Neuroscience Cell Biology, University of Coimbra, UC-Biotech, Parque Tecnológico de Cantanhede, Núcleo 04, Lote 8, 3060-197 Cantanhede, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Casa Costa Alemão, 3030-789 Coimbra, Portugal
- Correspondence: (A.S.); (M.J.M.)
| | - Maria João Moreno
- Coimbra Chemistry Centre-Institute of Molecular Sciences (CQC-IMS), University of Coimbra, 3004-535 Coimbra, Portugal;
- Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal
- Correspondence: (A.S.); (M.J.M.)
| |
Collapse
|
31
|
Chen X, Huang M, Liu D, Li Y, Luo Q, Pham K, Wang M, Zhang J, Zhang R, Peng Z, Wu X. Absorption and Transport Characteristics and Mechanisms of Carnosic Acid. BIOLOGY 2021; 10:biology10121278. [PMID: 34943193 PMCID: PMC8698657 DOI: 10.3390/biology10121278] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/03/2021] [Accepted: 12/03/2021] [Indexed: 01/08/2023]
Abstract
Simple Summary Carnosic acid (CA), a phenolic diterpenoid mainly found in rosemary and sage, has been reported to possess various health-beneficial activities. However, detailed information about the absorption characteristics and mechanisms of CA and its tissue distribution still remains unclear. It has been well-recognized that the absorption, transport, and metabolism of dietary bioactive compounds are closely related to their biological functions. Herein, a mouse study and Caco-2 cell monolayer model of the intestinal epithelial barrier were used to understand the absorption and transport characteristics of CA. First, we determined the tissue distribution of CA in mice following oral gavage at a physiologically relevant dose. We found that CA was bioavailable systemically and present locally in the digestive tract, especially in the cecum and colon. Next, in Caco-2 cell monolayers, CA exhibited a moderate permeability and was subjected to mild efflux. Moreover, the apparent permeability coefficient of CA transported across Caco-2 cell monolayers was significantly changed when the inhibitors of specific active transporter and passive diffusion were added, suggesting that the absorption and transport of CA involved both passive and active transportation. The present study is an important first step towards understanding the absorption, transport, and metabolic mechanisms of CA. Abstract Carnosic acid (CA) is a phenolic diterpenoid mainly found in rosemary and sage. CA has been reported to possess health-beneficial effects in various experimental settings. Herein, a mouse experiment and Caco-2 single-cell model were used to understand the absorption and transport characteristics of CA. First, we determined the tissue distribution of CA in mice, following an oral gavage at a physiologically relevant dose. We found that CA was bioavailable systemically and present locally in the digestive tract, especially in the cecum and colon. Next, we thought to characterize the absorption and transport of CA in the Caco-2 cell monolayer model of the intestinal epithelial barrier. In the Caco-2 cell model, CA exhibited a moderate permeability and was subjected to a mild efflux. Moreover, the apparent permeability coefficient (Papp) of CA transported across Caco-2 cell monolayers was significantly changed when the inhibitors of specific active transporter and passive diffusion were added to cells, suggesting that the absorption and transport of CA involved both passive and active transportation. The present study is an important first step towards understanding the absorption, transport, and metabolic mechanisms of CA. This could provide the scientific basis for developing CA-containing functional foods or dietary supplements with improved bioavailability.
Collapse
Affiliation(s)
- Xuexiang Chen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, China; (D.L.); (Y.L.); (Q.L.); (M.W.); (J.Z.); (R.Z.); (Z.P.)
- Correspondence: (X.C.); (X.W.)
| | - Meigui Huang
- Department of Food Science and Engineering, College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing 210037, China;
| | - Dongmei Liu
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, China; (D.L.); (Y.L.); (Q.L.); (M.W.); (J.Z.); (R.Z.); (Z.P.)
| | - Yongze Li
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, China; (D.L.); (Y.L.); (Q.L.); (M.W.); (J.Z.); (R.Z.); (Z.P.)
| | - Qiu Luo
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, China; (D.L.); (Y.L.); (Q.L.); (M.W.); (J.Z.); (R.Z.); (Z.P.)
| | - Katherine Pham
- Department of Kinesiology, Nutrition, and Health, Miami University, Oxford, OH 45056, USA;
| | - Minghong Wang
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, China; (D.L.); (Y.L.); (Q.L.); (M.W.); (J.Z.); (R.Z.); (Z.P.)
| | - Jing Zhang
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, China; (D.L.); (Y.L.); (Q.L.); (M.W.); (J.Z.); (R.Z.); (Z.P.)
| | - Runbin Zhang
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, China; (D.L.); (Y.L.); (Q.L.); (M.W.); (J.Z.); (R.Z.); (Z.P.)
| | - Zhixi Peng
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, China; (D.L.); (Y.L.); (Q.L.); (M.W.); (J.Z.); (R.Z.); (Z.P.)
| | - Xian Wu
- Department of Kinesiology, Nutrition, and Health, Miami University, Oxford, OH 45056, USA;
- Correspondence: (X.C.); (X.W.)
| |
Collapse
|