51
|
Castaño AG, García-Díaz M, Torras N, Altay G, Comelles J, Martínez E. Dynamic photopolymerization produces complex microstructures on hydrogels in a moldless approach to generate a 3D intestinal tissue model. Biofabrication 2019; 11:025007. [DOI: 10.1088/1758-5090/ab0478] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
52
|
Saez-Tenorio M, Domenech J, García-Rodríguez A, Velázquez A, Hernández A, Marcos R, Cortés C. Assessing the relevance of exposure time in differentiated Caco-2/HT29 cocultures. Effects of silver nanoparticles. Food Chem Toxicol 2018; 123:258-267. [PMID: 30403969 DOI: 10.1016/j.fct.2018.11.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 10/30/2018] [Accepted: 11/03/2018] [Indexed: 12/24/2022]
Abstract
In vitro models of the intestinal barrier are being increasingly used to evaluate nanoparticles (NPs) exposure risk. Nevertheless, most of these studies have focused on short-term exposures lasting no more than 24 h of duration, which could underestimate the toxic effects of a given compound under a more realistic setting. Since the assessment of longer exposure time-points is crucial to evaluate the risk of cumulative exposure to NPs, we have analyzed the effects of AgNPs at different exposure time-points between 6 h and 4 days on the barrier model system constituted by Caco-2/HT29 cells. Our results indicate that i) the system is stable during this time frame; ii) AgNPs affect the barrier's integrity only at the highest concentration tested (100 μg/mL), and only after 96 h of exposure; iii) cellular uptake of AgNPs showed a time-dependent and concentration-dependent increase; iv) translocation through the barrier was only observed at the highest concentration and only after 96 h of exposure; v) the expression of genes involved in the barrier's structure differs depending on the exposure time analyzed. All these results reinforce our proposal of expanding exposure times beyond 24 h when performing assays for hazard assessment of NPs using in vitro models of the intestinal barrier.
Collapse
Affiliation(s)
- Miriam Saez-Tenorio
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Josefa Domenech
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Alba García-Rodríguez
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Antonia Velázquez
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Carlos III Institute of Health, Madrid, Spain
| | - Alba Hernández
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Carlos III Institute of Health, Madrid, Spain
| | - Ricard Marcos
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Carlos III Institute of Health, Madrid, Spain.
| | - Constanza Cortés
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| |
Collapse
|
53
|
De Gregorio V, Imparato G, Urciuolo F, Netti PA. Micro-patterned endogenous stroma equivalent induces polarized crypt-villus architecture of human small intestinal epithelium. Acta Biomater 2018; 81:43-59. [PMID: 30282052 DOI: 10.1016/j.actbio.2018.09.061] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/29/2018] [Accepted: 09/28/2018] [Indexed: 12/14/2022]
Abstract
The small intestine is the major site for digestion, drug and nutrient absorption, as well as a primary site for many diseases. Current in vitro gut models fail in reproducing the complex intestinal extracellular matrix (ECM) network of the lamina propria and the peculiar architecture of the crypt-villus axis. Here we proposed a novel in vitro human intestine model that mimics the intestinal stromal topography and composition and strictly reproduces the tissue polarity with the crypt-villus architecture. First we developed a 3D human intestinal stromal equivalent (3D-ISE) composed of human intestinal subepithelial myofibroblasts (ISEMFs) embedded in their own extracellular matrix. Then, we seeded human colon carcinoma-derived cells (Caco-2) onto flat or patterned cell-synthetized stromal equivalent structure and cultured them until the formation of a well-oriented epithelium. We demonstrated that the patterned stroma increases the absorbing surface area, the epithelial proliferation rate, and the density of microvilli. In addition it induces changes in the biological functions of the epithelial cells such as enzymes and mucus production, polarization and tightness showing a physiological cell-lineage compartmentalization along the crypt/villi axes with the undifferentiated phenotypes at the base. At last, we reproduced an inflamed intestinal tissue model in which we identified the contribution of the stromal microenvironment by molecular (cytokines release and MMPs production) and immunofluorescence analyses and the effects of the epithelial-stromal cross-talk in the intestinal innate immunity by multiphoton investigation that revealed differences in the collagen network architecture. STATEMENT OF SIGNIFICANCE: The intestinal stroma morphology and composition has a fundamental role in crypt-villus development and appropriate epithelial cell-lineage compartmentalization. On this base, here we develop an engineered organotypic model of human intestine equivalent in which a functional epithelial/ECM crosstalk is recapitulated. Due to its accessible luminal surface it provides a new platform for preclinical studies of mucosal immunology and bowel inflammation as well as the assessment of pharmaco-toxicity studies.
Collapse
Affiliation(s)
- Vincenza De Gregorio
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy
| | - Giorgia Imparato
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy.
| | - Francesco Urciuolo
- Department of Chemical Materials and Industrial Production (DICMAPI) University of Naples Federico II, P.le Tecchio 80, 80125 Naples, Italy
| | - Paolo A Netti
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy; Department of Chemical Materials and Industrial Production (DICMAPI) University of Naples Federico II, P.le Tecchio 80, 80125 Naples, Italy; Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, P.le Tecchio 80, 80125 Naples, Italy
| |
Collapse
|
54
|
Jaworska K, Huc T, Gawrys M, Onyszkiewicz M, Samborowska E, Ufnal M. An In Vivo Method for Evaluating the Gut-Blood Barrier and Liver Metabolism of Microbiota Products. J Vis Exp 2018. [PMID: 30394384 DOI: 10.3791/58456] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The gut-blood barrier (GBB) controls the passage of nutrients, bacterial metabolites and drugs from intestinal lumen to the bloodstream. The GBB integrity is disturbed in gastrointestinal, cardiovascular and metabolic diseases, which may result in easier access of biologically active compounds, such as gut bacterial metabolites, to the bloodstream. Thus, the permeability of the GBB may be a marker of both intestinal and extraintestinal diseases. Furthermore, the increased penetration of bacterial metabolites may affect the functioning of the entire organism. Commonly used methods for studying the GBB permeability are performed ex vivo. The accuracy of those methods is limited, because the functioning of the GBB depends on intestinal blood flow. On the other hand, commonly used in vivo methods may be biased by liver and kidney performance, as those methods are based on evaluation of urine or/and peripheral blood concentrations of exogenous markers. Here, we present a direct measurement of GBB permeability in rats using an in vivo method based on portal blood sampling, which preserves intestinal blood flow and is virtually not affected by the liver and kidney function. Polyurethane catheters are inserted into the portal vein and inferior vena cava just above the hepatic veins confluence. Blood is sampled at baseline and after administration of a selected marker into a desired part of the gastrointestinal tract. Here, we present several applications of the method including (1) evaluation of the colon permeability to TMA, a gut bacterial metabolite, (2) evaluation of liver clearance of TMA, and (3) evaluation of a gut-portal blood-liver-peripheral blood pathway of gut bacteria-derived short-chain fatty acids. Furthermore, the protocol may also be used for tracking intestinal absorption and liver metabolism of drugs or for measurements of portal blood pressure.
Collapse
Affiliation(s)
- Kinga Jaworska
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw
| | - Tomasz Huc
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw
| | - Marta Gawrys
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw
| | - Maksymilian Onyszkiewicz
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw
| | - Emilia Samborowska
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences
| | - Marcin Ufnal
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw;
| |
Collapse
|
55
|
Gim-Krumm M, Donoso P, Zuñiga RN, Estay H, Troncoso E. A comprehensive study of glucose transfer in the human small intestine using an in vitro intestinal digestion system (i-IDS) based on a dialysis membrane process. J Memb Sci 2018. [DOI: 10.1016/j.memsci.2018.07.080] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
56
|
High degradation and no bioavailability of artichoke miRNAs assessed using an in vitro digestion/Caco-2 cell model. Nutr Res 2018; 60:68-76. [PMID: 30527261 DOI: 10.1016/j.nutres.2018.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 08/22/2018] [Accepted: 08/31/2018] [Indexed: 01/21/2023]
Abstract
Although the cross-kingdom transfer of vegetable miRNAs (miRNAs) in mammalian species, including humans, is still controversial, recent studies have rejected this theory. Based on these recent studies, we hypothesized that artichoke-derived miRNAs (cca-miRNAs) are not adsorbed into human intestinal cells after cooking and in vitro digestion. In order to test this hypothesis, we evaluated miRNA (cca-miRNAs) in the edible part of globe artichokes (head portion), after cooking and digestion by an in vitro digestion system. The cca-miRNA levels were analyzed by real-time PCR (RT-qPCR), and those that withstood cooking and digestion conditions were further analyzed for their bioavailability using an in vitro system (Caco-2/TC7 cell clone). We detected 20 cca-miRNAs after cooking, 5 of which were statistically down-regulated in comparison with uncooked samples. Only 4 cca-miRNAs were found after in vitro digestion. By using scanning electron microscopy (SEM), we also evaluated the extracellular vesicles (EVs) in homogenized artichoke as possible miRNA transporters. However, approximately 81% were degraded after cooking, while the remaining EVs had changed shape from round to elliptical. Finally, we detected no cell-free cca-miRNAs, miRNAs bound to protein complex, and no cca-miRNAs encapsulated in EVs inside Caco-2 cells or in basolateral medium after bioavailability experiments. In conclusion, the data from the present study agrees with recent findings that the human small intestine does not uptake dietary miRNAs from raw or cooked artichoke heads.
Collapse
|
57
|
Maares M, Keil C, Thomsen S, Günzel D, Wiesner B, Haase H. Characterization of Caco-2 cells stably expressing the protein-based zinc probe eCalwy-5 as a model system for investigating intestinal zinc transport. J Trace Elem Med Biol 2018; 49:296-304. [PMID: 29395783 DOI: 10.1016/j.jtemb.2018.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/22/2017] [Accepted: 01/17/2018] [Indexed: 12/30/2022]
Abstract
Intestinal zinc resorption, in particular its regulation and mechanisms, are not yet fully understood. Suitable intestinal cell models are needed to investigate zinc uptake kinetics and the role of labile zinc in enterocytes in vitro. Therefore, a Caco-2 cell clone was produced, stably expressing the genetically encoded zinc biosensor eCalwy-5. The aim of the present study was to reassure the presence of characteristic enterocyte-specific properties in the Caco-2-eCalwy clone. Comparison of Caco-2-WT and Caco-2-eCalwy cells revealed only slight differences regarding subcellular localization of the tight junction protein occludin and alkaline phosphatase activity, which did not affect basic integrity of the intestinal barrier or the characteristic brush border membrane morphology. Furthermore, introduction of the additional zinc-binding protein in Caco-2 cells did not alter mRNA expression of the major intestinal zinc transporters (zip4, zip5, znt-1 and znt-5), but increased metallothionein 1a-expression and cellular resistance to higher zinc concentrations. Moreover, this study examines the effect of sensor expression level on its saturation with zinc. Fluorescence cell imaging indicated considerable intercellular heterogeneity in biosensor-expression. However, FRET-measurements confirmed that these differences in expression levels have no effect on fractional zinc-saturation of the probe.
Collapse
Affiliation(s)
- Maria Maares
- Department of Food Chemistry and Toxicology, Berlin Institute of Technology, Berlin, Germany
| | - Claudia Keil
- Department of Food Chemistry and Toxicology, Berlin Institute of Technology, Berlin, Germany
| | - Susanne Thomsen
- Department of Food Chemistry and Toxicology, Berlin Institute of Technology, Berlin, Germany
| | - Dorothee Günzel
- Institute of Clinical Physiology, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | | | - Hajo Haase
- Department of Food Chemistry and Toxicology, Berlin Institute of Technology, Berlin, Germany; TraceAge-DFG Research Unit on Interactions of essential trace elements in healthy and diseased elderly, Potsdam-Berlin-Jena, Germany.
| |
Collapse
|
58
|
Garcia J, Fernández-Blanco Á, Teixidó M, Sánchez-Navarro M, Giralt E. d-Polyarginine Lipopeptides as Intestinal Permeation Enhancers. ChemMedChem 2018; 13:2045-2052. [DOI: 10.1002/cmdc.201800428] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 07/30/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Josep Garcia
- Institute for Research in Biomedicine (IRB Barcelona); The Barcelona Institute of Science and Technology (BIST); Baldiri Reixac 10 08028 Barcelona Spain
| | - Álvaro Fernández-Blanco
- Institute for Research in Biomedicine (IRB Barcelona); The Barcelona Institute of Science and Technology (BIST); Baldiri Reixac 10 08028 Barcelona Spain
- Cellular & Systems Neurobiology, Systems Biology Program, Centre for Genomic Regulation (CRG); The Barcelona Institute of Science and Technology; Dr. Aiguader 88 08003 Barcelona Spain
| | - Meritxell Teixidó
- Institute for Research in Biomedicine (IRB Barcelona); The Barcelona Institute of Science and Technology (BIST); Baldiri Reixac 10 08028 Barcelona Spain
| | - Macarena Sánchez-Navarro
- Institute for Research in Biomedicine (IRB Barcelona); The Barcelona Institute of Science and Technology (BIST); Baldiri Reixac 10 08028 Barcelona Spain
| | - Ernest Giralt
- Institute for Research in Biomedicine (IRB Barcelona); The Barcelona Institute of Science and Technology (BIST); Baldiri Reixac 10 08028 Barcelona Spain
- University of Barcelona; Department of Inorganic and Organic Chemistry; Spain
| |
Collapse
|
59
|
Warrier A, Gunosewoyo H, Crowe A. Efflux transporters and tight junction expression changes in human gastrointestinal cell lines cultured in defined medium vs serum supplemented medium. Life Sci 2018; 207:138-144. [PMID: 29857072 DOI: 10.1016/j.lfs.2018.05.053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 11/25/2022]
Abstract
AIMS Many gastrointestinal cell lines including Caco-2, LS174T and RKO require foetal calf serum (FCS) in culture medium. However, when isolating secreted product from conditioned medium (CM), after cell exposure to a trigger, it is better to remove FCS in the culture medium for identification of secreted products of interest. However, it is unknown whether defined medium adversely affects active efflux protein expression and tight junction formation. MATERIALS AND METHODS Using different gastrointestinal cell lines chosen with different levels of efflux transporter expression, fully defined components, such as using transferrin, insulin, selenium and ethanolamine without FCS or with a reduced percentage of FCS (2%) were tested as an optimal choice for cell growth. In addition to morphological characteristics, the expression of the ABC efflux transporters, ABCB1 (P-glycoprotein [P-gp]), ABCC2 (multidrug resistance associated protein 2), ABCG2 (breast cancer resistance protein) and occludin was determined. KEY FINDINGS The cells required a minimum of 2% FCS for expression of transporters. Fully defined medium with no serum adversely affected the expression of transporters, especially P-gp. An important characteristic of Caco-2 cells is its ability to form tight junctions. Caco-2 did not form adequate tight junctions without 10% FCS added in the medium, as evidenced by low TEER values and reduced occluding immunohistochemistry. SIGNIFICANCE FCS is required for efflux protein expression and tight junction generation. Nevertheless, it is possible to use 5 fold less FCS which assists with low molecular weight secretion isolation. Passage number also contributes significantly to the presence of these transporters.
Collapse
Affiliation(s)
- Aparna Warrier
- School of Pharmacy and biomedical sciences, Curtin University, Perth, Western Australia, 6102, Australia; Curtin Health Innovation Research Institute (CHIRI), Curtin University, Perth, Western Australia, 6102, Australia
| | - Hendra Gunosewoyo
- School of Pharmacy and biomedical sciences, Curtin University, Perth, Western Australia, 6102, Australia
| | - Andrew Crowe
- School of Pharmacy and biomedical sciences, Curtin University, Perth, Western Australia, 6102, Australia; Curtin Health Innovation Research Institute (CHIRI), Curtin University, Perth, Western Australia, 6102, Australia.
| |
Collapse
|
60
|
iPSC-Derived Enterocyte-like Cells for Drug Absorption and Metabolism Studies. Trends Mol Med 2018; 24:696-708. [PMID: 29945758 DOI: 10.1016/j.molmed.2018.06.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/27/2018] [Accepted: 06/01/2018] [Indexed: 12/14/2022]
Abstract
Intestinal cell models have been widely studied and used to evaluate absorption and metabolism of drugs in the small intestine, constituting valuable tools as a first approach to evaluate the behavior of new drugs. However, such cell models might not be able to fully predict the absorption mechanisms and metabolic pathways of the tested compounds. In recent years, induced pluripotent stem cells (iPSCs) differentiated into enterocyte-like cells have been proposed as more biorelevant intestinal models. In this review, we describe mechanisms underlying the differentiation of iPSCs into enterocyte-like cells, appraise the usefulness of these cells in tridimensional intestinal models, and discuss their suitability to be used in the future for drug screening.
Collapse
|
61
|
Heller AA, Lockwood SY, Janes TM, Spence DM. Technologies for Measuring Pharmacokinetic Profiles. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2018; 11:79-100. [PMID: 29324183 DOI: 10.1146/annurev-anchem-061417-125611] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
The creation of a pharmacokinetic (PK) curve, which follows the plasma concentration of an administered drug as a function of time, is a critical aspect of the drug development process and includes such information as the drug's bioavailability, clearance, and elimination half-life. Prior to a drug of interest gaining clearance for use in human clinical trials, research is performed during the preclinical stages to establish drug safety and dosing metrics from data obtained from the PK studies. Both in vivo animal models and in vitro platforms have limitations in predicting human reaction to a drug due to differences in species and associated simplifications, respectively. As a result, in silico experiments using computer simulation have been implemented to accurately predict PK parameters in human studies. This review assesses these three approaches (in vitro, in vivo, and in silico) when establishing PK parameters and evaluates the potential for in silico studies to be the future gold standard of PK preclinical studies.
Collapse
Affiliation(s)
- A A Heller
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, USA;
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, USA
| | - S Y Lockwood
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, USA
| | - T M Janes
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, USA;
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, USA
| | - D M Spence
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, USA
| |
Collapse
|
62
|
Pajoumshariati SR, Azizi M, Wesner D, Miller PG, Shuler ML, Abbaspourrad A. Microfluidic-Based Cell-Embedded Microgels Using Nonfluorinated Oil as a Model for the Gastrointestinal Niche. ACS APPLIED MATERIALS & INTERFACES 2018; 10:9235-9246. [PMID: 29474057 DOI: 10.1021/acsami.7b16916] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Microfluidic-based cell encapsulation has promising potential in therapeutic applications. It also provides a unique approach for studying cellular dynamics and interactions, though this concept has not yet been fully explored. No in vitro model currently exists that allows us to study the interaction between crypt cells and Peyer's patch immune cells because of the difficulty in recreating, with sufficient control, the two different microenvironments in the intestine in which these cell types belong. However, we demonstrate that a microfluidic technique is able to provide such precise control and that these cells can proliferate inside microgels. Current microfluidic-based cell microencapsulation techniques primarily use fluorinated oils. Herein, we study the feasibility and biocompatibility of different nonfluorinated oils for application in gastrointestinal cell encapsulation and further introduce a model for studying intercellular chemical interactions with this approach. Our results demonstrate that cell viability is more affected by the solidification and purification processes that occur after droplet formation rather than the oil type used for the carrier phase. Specifically, a shorter polymer cross-linking time and consequently lower cell exposure to the harsh environment (e.g., acidic pH) results in a high cell viability of over 90% within the protected microgels. Using nonfluorinated oils, we propose a model system demonstrating the interplay between crypt and Peyer's patch cells using this microfluidic approach to separately encapsulate the cells inside distinct alginate/gelatin microgels, which allow for intercellular chemical communication. We observed that the coculture of crypt cells alongside Peyer's patch immune cells improves the growth of healthy organoids inside these microgels, which contain both differentiated and undifferentiated cells over 21 days of coculture. These results indicate the possibility of using droplet-based microfluidics for culturing organoids to expand their applicability in clinical research.
Collapse
|
63
|
Ayehunie S, Landry T, Stevens Z, Armento A, Hayden P, Klausner M. Human Primary Cell-Based Organotypic Microtissues for Modeling Small Intestinal Drug Absorption. Pharm Res 2018; 35:72. [PMID: 29476278 PMCID: PMC6599640 DOI: 10.1007/s11095-018-2362-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 02/02/2018] [Indexed: 12/11/2022]
Abstract
PURPOSE The study evaluates the use of new in vitro primary human cell-based organotypic small intestinal (SMI) microtissues for predicting intestinal drug absorption and drug-drug interaction. METHODS The SMI microtissues were reconstructed using human intestinal fibroblasts and enterocytes cultured on a permeable support. To evaluate the suitability of the intestinal microtissues to model drug absorption, the permeability coefficients across the microtissues were determined for a panel of 11 benchmark drugs with known human absorption and Caco-2 permeability data. Drug-drug interactions were examined using efflux transporter substrates and inhibitors. RESULTS The 3D-intestinal microtissues recapitulate the structural features and physiological barrier properties of the human small intestine. The microtissues also expressed drug transporters and metabolizing enzymes found on the intestinal wall. Functionally, the SMI microtissues were able to discriminate between low and high permeability drugs and correlated better with human absorption data (r2 = 0.91) compared to Caco-2 cells (r2 = 0.71). Finally, the functionality of efflux transporters was confirmed using efflux substrates and inhibitors which resulted in efflux ratios of >2.0 fold and by a decrease in efflux ratios following the addition of inhibitors. CONCLUSION The SMI microtissues appear to be a useful pre-clinical tool for predicting drug bioavailability of orally administered drugs.
Collapse
Affiliation(s)
- Seyoum Ayehunie
- MatTek Corporation, 200 Homer Avenue, Ashland, Massachusetts, USA.
| | - Tim Landry
- MatTek Corporation, 200 Homer Avenue, Ashland, Massachusetts, USA
| | - Zachary Stevens
- MatTek Corporation, 200 Homer Avenue, Ashland, Massachusetts, USA
| | - Alex Armento
- MatTek Corporation, 200 Homer Avenue, Ashland, Massachusetts, USA
| | - Patrick Hayden
- MatTek Corporation, 200 Homer Avenue, Ashland, Massachusetts, USA
| | | |
Collapse
|
64
|
De Gregorio V, Imparato G, Urciuolo F, Netti PA. 3D stromal tissue equivalent affects intestinal epithelium morphogenesis in vitro. Biotechnol Bioeng 2018; 115:1062-1075. [DOI: 10.1002/bit.26522] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 12/12/2017] [Accepted: 12/14/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Vincenza De Gregorio
- Center for Advanced Biomaterials for HealthCare@CRIBIstituto Italiano di TecnologiaNaplesItaly
| | - Giorgia Imparato
- Center for Advanced Biomaterials for HealthCare@CRIBIstituto Italiano di TecnologiaNaplesItaly
| | - Francesco Urciuolo
- Center for Advanced Biomaterials for HealthCare@CRIBIstituto Italiano di TecnologiaNaplesItaly
| | - Paolo A. Netti
- Center for Advanced Biomaterials for HealthCare@CRIBIstituto Italiano di TecnologiaNaplesItaly
- Interdisciplinary Research Centre on Biomaterials (CRIB)University of NaplesNaplesItaly
- Department of Chemical Materials and Industrial Production (DICMAPI)University of NaplesNaplesItaly
| |
Collapse
|
65
|
Bhatt AP, Gunasekara DB, Speer J, Reed MI, Peña AN, Midkiff BR, Magness ST, Bultman SJ, Allbritton NL, Redinbo MR. Nonsteroidal Anti-Inflammatory Drug-Induced Leaky Gut Modeled Using Polarized Monolayers of Primary Human Intestinal Epithelial Cells. ACS Infect Dis 2018; 4:46-52. [PMID: 29094594 DOI: 10.1021/acsinfecdis.7b00139] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The intestinal epithelium provides a critical barrier that separates the gut microbiota from host tissues. Nonsteroidal anti-inflammatory drugs (NSAIDs) are efficacious analgesics and antipyretics and are among the most frequently used drugs worldwide. In addition to gastric damage, NSAIDs are toxic to the intestinal epithelium, causing erosions, perforations, and longitudinal ulcers in the gut. Here, we use a unique in vitro human primary small intestinal cell monolayer system to pinpoint the intestinal consequences of NSAID treatment. We found that physiologically relevant doses of the NSAID diclofenac (DCF) are cytotoxic because they uncouple mitochondrial oxidative phosphorylation and generate reactive oxygen species. We also find that DCF induces intestinal barrier permeability, facilitating the translocation of compounds from the luminal to the basolateral side of the intestinal epithelium. The results we outline here establish the utility of this novel platform, representative of the human small intestinal epithelium, to understand NSAID toxicity, which can be applied to study multiple aspects of gut barrier function including defense against infectious pathogens and host-microbiota interactions.
Collapse
Affiliation(s)
- Aadra P. Bhatt
- Department of Chemistry, University of North Carolina, 250 Bell Tower Drive, Chapel
Hill, North Carolina 27599-3290, United States
| | - Dulan B. Gunasekara
- Department of Chemistry, University of North Carolina, 250 Bell Tower Drive, Chapel
Hill, North Carolina 27599-3290, United States
| | - Jennifer Speer
- Department of Chemistry, University of North Carolina, 250 Bell Tower Drive, Chapel
Hill, North Carolina 27599-3290, United States
| | - Mark I. Reed
- Department of Chemistry, University of North Carolina, 250 Bell Tower Drive, Chapel
Hill, North Carolina 27599-3290, United States
| | - Alexis N. Peña
- Department of Chemistry, University of North Carolina, 250 Bell Tower Drive, Chapel
Hill, North Carolina 27599-3290, United States
| | - Bentley R. Midkiff
- Translational Pathology Laboratory, Lineberger
Comprehensive Cancer Center, University of North Carolina, 160
North Medical Drive, Chapel Hill, North Carolina 27599-7525, United States
| | - Scott T. Magness
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, United States, and North Carolina State University, Raleigh, North Carolina 27607, United States
- Departments of Medicine, Cell Biology and
Physiology, University of North Carolina, 111 Mason Farm Road, Chapel Hill, North Carolina 27599-7032, United States
| | - Scott J. Bultman
- Department of Genetics, University of North Carolina, 120 Mason Farm Road, Chapel
Hill, North Carolina 27599-7264, United States
- Lineberger
Comprehensive Cancer Center, University of North Carolina, 450
West Drive, Chapel Hill, North Carolina 27599, United States
| | - Nancy L. Allbritton
- Department of Chemistry, University of North Carolina, 250 Bell Tower Drive, Chapel
Hill, North Carolina 27599-3290, United States
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, United States, and North Carolina State University, Raleigh, North Carolina 27607, United States
- Lineberger
Comprehensive Cancer Center, University of North Carolina, 450
West Drive, Chapel Hill, North Carolina 27599, United States
| | - Matthew R. Redinbo
- Department of Chemistry, University of North Carolina, 250 Bell Tower Drive, Chapel
Hill, North Carolina 27599-3290, United States
- Lineberger
Comprehensive Cancer Center, University of North Carolina, 450
West Drive, Chapel Hill, North Carolina 27599, United States
- Departments of Biochemistry and Biophysics,
and Microbiology and Immunology, and the Integrated Program for Biological
and Genome Science, University of North Carolina, 250 Bell Tower
Drive, Chapel Hill, North
Carolina 27599-3290, United States
| |
Collapse
|
66
|
Hymery N, Puel O, Tadrist S, Canlet C, Le Scouarnec H, Coton E, Coton M. Effect of PR toxin on THP1 and Caco-2 cells: an in vitro study. WORLD MYCOTOXIN J 2017. [DOI: 10.3920/wmj2017.2196] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Penicillium roqueforti produces mycotoxins including PR toxin, which is a food and feed contaminant. In this study, PR toxin was purified from culture material of the Penicillium roqueforti F43-1 strain. Toxic effects were evaluated in undifferentiated human Caco-2 intestinal epithelial cells and THP-1 monocytic immune cells. To understand the mechanisms involved in PR-toxin toxicity, cell death and pro-inflammatory gene expression were studied. In addition, PR toxin degradation was assessed. Cytotoxicity studies showed a dose-dependent effect of PR toxin and the calculated mean cytotoxic concentration (IC50) concentrations were for Caco-2 and THP-1 cells >12.5 and 0.83 μM, respectively. Gene expression studies showed that tumour necrosis factor-α expression was significantly increased after 24 h exposure to 312 μM PR toxin. PR toxin induced necrosis on THP-1 cells after 3 h exposure. In the cell culture system, the PR toxin showed a 10-fold reduction in PR toxin concentration within 48 h, indicating that PR toxin was degraded by THP-1. To conclude, PR toxin appears to be one of the most cytotoxic P. roqueforti mycotoxins on Caco-2 and/or THP-1 cells and induces in THP-1 cells both necrosis and an inflammatory response.
Collapse
Affiliation(s)
- N. Hymery
- Université de Brest, EA 3882, Laboratoire Universitaire de Biodiversité et Ecologie Microbienne, IBSAM, ESIAB, Technopôle Brest-Iroise, 29280 Plouzané, France
| | - O. Puel
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | - S. Tadrist
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | - C. Canlet
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
- MetaToul-MetaboHUB, National Infrastructure of Metabolomics and Fluxomics, 31027 Toulouse Cedex, France
| | - H. Le Scouarnec
- Université de Brest, EA 3882, Laboratoire Universitaire de Biodiversité et Ecologie Microbienne, IBSAM, ESIAB, Technopôle Brest-Iroise, 29280 Plouzané, France
| | - E. Coton
- Université de Brest, EA 3882, Laboratoire Universitaire de Biodiversité et Ecologie Microbienne, IBSAM, ESIAB, Technopôle Brest-Iroise, 29280 Plouzané, France
| | - M. Coton
- Université de Brest, EA 3882, Laboratoire Universitaire de Biodiversité et Ecologie Microbienne, IBSAM, ESIAB, Technopôle Brest-Iroise, 29280 Plouzané, France
| |
Collapse
|
67
|
Eissa N, Hussein H, Kermarrec L, Grover J, Metz-Boutigue MHE, Bernstein CN, Ghia JE. Chromofungin Ameliorates the Progression of Colitis by Regulating Alternatively Activated Macrophages. Front Immunol 2017; 8:1131. [PMID: 28951733 PMCID: PMC5599789 DOI: 10.3389/fimmu.2017.01131] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 08/28/2017] [Indexed: 12/24/2022] Open
Abstract
Ulcerative colitis (UC) is characterized by a functional dysregulation of alternatively activated macrophage (AAM) and intestinal epithelial cells (IECs) homeostasis. Chromogranin-A (CHGA) secreted by neuroendocrine cells is implicated in intestinal inflammation and immune dysregulation. CHGA undergoes proteolytic processing to generate CHGA-derived peptides. Chromofungin (CHR: CHGA47–66) is a short CHGA-derived peptide encoded by CHGA Exon-IV and is involved in innate immune regulation, but the basis is poorly investigated. We investigated the expression of CHR in colonic tissue of patients with active UC and assessed the effects of the CHR in dextran sulfate sodium (DSS) colitis in mice and on macrophages and human colonic epithelial cells. We found that mRNA expression of CHR correlated positively with mRNA levels of AAM markers and gene expression of tight junction (TJ) proteins and negatively with mRNA levels of interleukin (IL)-8, IL-18, and collagen in patients with active UC. Moreover, AAM markers correlated positively with gene expression of TJ proteins and negatively with IL-8, IL-18, and collagen gene expression. Experimentally, intracolonic administration of CHR protected against DSS-induced colitis by priming macrophages into AAM, reducing colonic collagen deposition, and maintaining IECs homeostasis. This effect was associated with a significant increase of AAM markers, reduction of colonic IL-18 release and conservation of gene expression of TJ proteins. In vitro, CHR enhanced AAM polarization and increased the production of anti-inflammatory mediators. CHR-treated AAM conditioned medium increased Caco-2 cell migration, viability, proliferation, and mRNA levels of TJ proteins, and decreased oxidative stress-induced apoptosis and proinflammatory cytokines release. Direct CHR treatments had the same effect. In conclusion, CHR treatment reduces the severity of colitis and the inflammatory process via enhancing AAM functions and maintaining IECs homeostasis. CHR is involved in the pathogenesis of inflammation in experimental colitis. These findings provide insight into the mechanisms of colonic inflammation and could lead to new therapeutic strategies for UC.
Collapse
Affiliation(s)
- Nour Eissa
- Immunology Department, University of Manitoba, Winnipeg, MB, Canada.,Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada
| | - Hayam Hussein
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Ohio State University, Columbus, OH, United States
| | | | - Jasmine Grover
- Immunology Department, University of Manitoba, Winnipeg, MB, Canada
| | - Marie-Hélène Et Metz-Boutigue
- INSERM U977, Biomatériaux et Ingéniérie tissulaire, Institut Leriche 2éme étage, Hôpital Civil, Porte de l'Hôpital, Strasbourg, France
| | - Charles N Bernstein
- Rady Faculty of Health Sciences, Department of Internal Medicine, Section of Gastroenterology, University of Manitoba, Winnipeg, MB, Canada.,University of Manitoba IBD Clinical and Research Centre, Winnipeg, MB, Canada
| | - Jean-Eric Ghia
- Immunology Department, University of Manitoba, Winnipeg, MB, Canada.,Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada.,Rady Faculty of Health Sciences, Department of Internal Medicine, Section of Gastroenterology, University of Manitoba, Winnipeg, MB, Canada.,University of Manitoba IBD Clinical and Research Centre, Winnipeg, MB, Canada
| |
Collapse
|
68
|
Berben P, Brouwers J, Augustijns P. Assessment of Passive Intestinal Permeability Using an Artificial Membrane Insert System. J Pharm Sci 2017; 107:250-256. [PMID: 28826878 DOI: 10.1016/j.xphs.2017.08.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/08/2017] [Accepted: 08/08/2017] [Indexed: 10/19/2022]
Abstract
Despite reasonable predictive power of current cell-based and cell-free absorption models for the assessment of intestinal drug permeability, high costs and lengthy preparation steps hamper their use. The use of a simple artificial membrane (without any lipids present) as intestinal barrier substitute would overcome these hurdles. In the present study, a set of 14 poorly water-soluble drugs, dissolved in 2 different media (fasted state simulated/human intestinal fluids [FaSSIF/FaHIF]), were applied to the donor compartment of an artificial membrane insert system (AMI-system) containing a regenerated cellulose membrane. Furthermore, to investigate the predictive capacity of the AMI-system as substitute for the well-established Caco-2 system to assess intestinal permeability, the same set of 14 drugs dissolved in FaHIF were applied to the donor compartment of a Caco-2 system. For 14 drugs, covering a broad range of physicochemical parameters, a reasonable correlation between both absorption systems was observed, characterized by a Pearson correlation coefficient r of 0.95 (FaHIF). Using the AMI-system, an excellent predictive capacity of FaSSIF as surrogate medium for FaHIF was demonstrated (r = 0.96). Based on the acquired data, the AMI-system appears to be a time- and cost-effective tool for the early-stage estimation of passive intestinal permeability for poorly water-soluble drugs.
Collapse
Affiliation(s)
- Philippe Berben
- Drug Delivery and Disposition, KU Leuven, Gasthuisberg O&N II, Herestraat 49 - Box 921, Leuven 3000, Belgium
| | - Joachim Brouwers
- Drug Delivery and Disposition, KU Leuven, Gasthuisberg O&N II, Herestraat 49 - Box 921, Leuven 3000, Belgium
| | - Patrick Augustijns
- Drug Delivery and Disposition, KU Leuven, Gasthuisberg O&N II, Herestraat 49 - Box 921, Leuven 3000, Belgium.
| |
Collapse
|
69
|
Blutt SE, Broughman JR, Zou W, Zeng XL, Karandikar UC, In J, Zachos NC, Kovbasnjuk O, Donowitz M, Estes MK. Gastrointestinal microphysiological systems. Exp Biol Med (Maywood) 2017; 242:1633-1642. [PMID: 28534432 DOI: 10.1177/1535370217710638] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Gastrointestinal diseases are a significant health care and economic burden. Prevention and treatment of these diseases have been limited by the available human biologic models. Microphysiological systems comprise organ-specific human cultures that recapitulate many structural, biological, and functional properties of the organ in smaller scale including aspects of flow, shear stress and chemical gradients. The development of intestinal microphysiological system platforms represents a critical component in improving our understanding, prevention, and treatment of gastrointestinal diseases. This minireview discusses: shortcomings of classical cell culture models of the gastrointestinal tract; human intestinal enteroids as a new model and their advantages compared to cell lines; why intestinal microphysiological systems are needed; potential functional uses of intestinal microphysiological systems in areas of drug development and modeling acute and chronic diseases; and current challenges in the development of intestinal microphysiological systems. Impact statement The development of a gastrointestinal MPS has the potential to facilitate the understanding of GI physiology. An ultimate goal is the integration of the intestinal MPS with other organ MPS. The development and characterization of nontransformed human intestinal cultures for use in MPS have progressed significantly since the inception of the MPS program in 2012, and these cultures are a key component of advancing MPS. Continued efforts are needed to optimize MPS to comprehensively and accurately recapitulate the complexity of the intestinal epithelium within intestinal tissue. These systems will need to include peristalsis, flow, and oxygen gradients, with incorporation of vascular, immune, and nerve cells. Regional cellular organization of crypt and villus areas will also be necessary to better model complete intestinal structure.
Collapse
Affiliation(s)
- Sarah E Blutt
- 1 Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - James R Broughman
- 1 Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Winnie Zou
- 1 Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xi-Lei Zeng
- 1 Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Umesh C Karandikar
- 1 Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Julie In
- 2 Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Nicholas C Zachos
- 2 Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Olga Kovbasnjuk
- 2 Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Mark Donowitz
- 2 Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Mary K Estes
- 1 Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA.,3 Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
70
|
Dalziel JE, Anderson RC, Bassett SA, Lloyd-West CM, Haggarty NW, Roy NC. Influence of Bovine Whey Protein Concentrate and Hydrolysate Preparation Methods on Motility in the Isolated Rat Distal Colon. Nutrients 2016; 8:nu8120809. [PMID: 27983629 PMCID: PMC5188464 DOI: 10.3390/nu8120809] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/30/2016] [Accepted: 12/08/2016] [Indexed: 01/19/2023] Open
Abstract
Whey protein concentrate (WPC) and hydrolysate (WPH) are protein ingredients used in sports, medical and pediatric formulations. Concentration and hydrolysis methods vary for whey sourced from cheese and casein co-products. The purpose of this research was to investigate the influence of whey processing methods on in vitro gastrointestinal (GI) health indicators for colonic motility, epithelial barrier integrity and immune modulation. WPCs from casein or cheese processing and WPH (11% or 19% degree of hydrolysis, DH) were compared for their effects on motility in a 1 cm section of isolated rat distal colon in an oxygenated tissue bath. Results showed that WPC decreased motility irrespective of whether it was a by-product of lactic acid or mineral acid casein production, or from cheese production. This indicated that regardless of the preparation methodology, the whey protein contained components that modulate aspects of motility within the distal colon. WPH (11% DH) increased contractile frequency by 27% in a delayed manner and WPH (19% DH) had an immediate effect on contractile properties, increasing tension by 65% and frequency by 131%. Increased motility was associated with increased hydrolysis that may be attributed to the abundance of bioactive peptides. Increased frequency of contractions by WPH (19% DH) was inhibited (by 44%) by naloxone, implicating a potential involvement of opioid receptors in modulation of motility. Trans-epithelial electrical resistance and cytokine expression assays revealed that the WPC proteins studied did not alter intestinal barrier integrity or elicit any discernible immune response.
Collapse
Affiliation(s)
- Julie E Dalziel
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch, Grasslands Research Centre, Palmerston North 4442, New Zealand.
| | - Rachel C Anderson
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch, Grasslands Research Centre, Palmerston North 4442, New Zealand.
| | - Shalome A Bassett
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch, Grasslands Research Centre, Palmerston North 4442, New Zealand.
| | - Catherine M Lloyd-West
- Bioinformatics and Statistics, AgResearch, Grasslands Research Centre, Palmerston North 4442, New Zealand.
| | - Neill W Haggarty
- Fonterra Co-operative Group, Palmerston North 4442, New Zealand.
| | - Nicole C Roy
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch, Grasslands Research Centre, Palmerston North 4442, New Zealand.
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand.
| |
Collapse
|
71
|
Anderson RC, Bassett SA, Haggarty NW, Gopal PK, Armstrong KM, Roy NC. Short communication: Early-lactation, but not mid-lactation, bovine lactoferrin preparation increases epithelial barrier integrity of Caco-2 cell layers. J Dairy Sci 2016; 100:886-891. [PMID: 27939537 DOI: 10.3168/jds.2016-11803] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 10/13/2016] [Indexed: 12/15/2022]
Abstract
Bovine lactoferrin is an important milk protein with many health-promoting properties, including improving intestinal barrier integrity. Dysfunction of this barrier, commonly referred to as "leaky gut," has been linked to inflammatory and autoimmune diseases. With some processing techniques, lactoferrin isolated from milk collected at the start of the milking season (early lactation) may have lower purity than that isolated from milk collected during the rest of the milking season (mid-lactation) and could result in differences in bioactivity based on the stage of lactation. We compared reversed-phase HPLC chromatographs of early-lactation and mid-lactation preparations and found that both had large chromatograph peaks at the time predicted for lactoferrin. The notable difference between the 2 chromatographs was a much larger peak in the early-lactation lactoferrin sample that was determined to be angiogenin. Angiogenin was first identified due to its ability to induce new blood vessel formation, but is now known to be involved in numerous physiological processes. Then, we compared the effects of early-lactation and mid-lactation lactoferrin preparations in 2 bioassays: trans-epithelial electrical resistance (TEER), a measure of intestinal barrier integrity, and peripheral blood mononuclear cell cytokine secretion, a measure of immune-stimulatory properties. We found that early-lactation lactoferrin increased TEER across Caco-2 cell layers compared with control from 10 to 48 h, mid-lactation lactoferrin did not alter TEER. We also found that early-lactation lactoferrin reduced the amount of IL-8 produced by peripheral blood mononuclear cells (compared with those treated with control medium) to a greater extent than mid-lactation lactoferrin. A pro-inflammatory chemokine, IL-8 is also known to decrease barrier function. These results suggest that the decrease in IL-8 production in the presence of early-lactation lactoferrin may be the mechanism by which it increases TEER. The anti-inflammatory effect of early-lactation lactoferrin may be related to the presence of angiogenin, which is known to suppress inflammatory responses. This work indicates that products rich in angiogenin may have intestinal health benefits, and further work to investigate this is warranted.
Collapse
Affiliation(s)
- Rachel C Anderson
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch, Private Bag 11008, Palmerston North, 4442, New Zealand; Riddet Centre of Research Excellence, Massey University, Private Bag 11222, Palmerston North, 4442, New Zealand.
| | - Shalome A Bassett
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch, Private Bag 11008, Palmerston North, 4442, New Zealand
| | - Neill W Haggarty
- Fonterra Research and Development Centre, Private Bag 11029, Palmerston North, 4442, New Zealand
| | - Pramod K Gopal
- Fonterra Research and Development Centre, Private Bag 11029, Palmerston North, 4442, New Zealand
| | - Kelly M Armstrong
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch, Private Bag 11008, Palmerston North, 4442, New Zealand
| | - Nicole C Roy
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch, Private Bag 11008, Palmerston North, 4442, New Zealand; Riddet Centre of Research Excellence, Massey University, Private Bag 11222, Palmerston North, 4442, New Zealand
| |
Collapse
|
72
|
Cei D, Costa J, Gori G, Frediani G, Domenici C, Carpi F, Ahluwalia A. A bioreactor with an electro-responsive elastomeric membrane for mimicking intestinal peristalsis. BIOINSPIRATION & BIOMIMETICS 2016; 12:016001. [PMID: 27918289 DOI: 10.1088/1748-3190/12/1/016001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
This study describes an actuated bioreactor which mimics the pulsatile contractile motion of the intestinal barrier using electro-responsive elastomers as smart materials that undergo deformation upon electrical stimulation. The device consists of an annular dielectric elastomer actuator working as a radial artificial muscle able to rhythmically contract and relax a central cell culture well. The bioreactor maintained up to 4 h of actuation at a frequency of 0.15 Hz and a strain of 8%-10%, to those of the cyclic contraction and relaxation of the small intestine. In vitro tests demonstrated that the device was biocompatible and cell-adhesive for Caco-2 cells, which formed a confluent monolayer following 21 days of culture in the central well. In addition, cellular adhesion and cohesion were maintained after 4 h of continuous cyclic strain. These preliminary results encourage further investigations on the use of dielectric elastomer actuation as a versatile technology that might overcome the limitations of commercially available pneumatic driving systems to obtain bioreactors that can cyclically deform cell cultures in a biomimetic fashion.
Collapse
Affiliation(s)
- Daniele Cei
- Research Center 'E.Piaggio' and Department of Information Engineering, University of Pisa, Largo L Lazzarino, I-56126 Pisa, Italy
| | | | | | | | | | | | | |
Collapse
|
73
|
Samy W, Elnoby A, El-Gowelli HM, Elgindy N. Hybrid polymeric matrices for oral modified release of Desvenlafaxine succinate tablets. Saudi Pharm J 2016; 25:676-687. [PMID: 28725139 PMCID: PMC5506642 DOI: 10.1016/j.jsps.2016.10.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 10/07/2016] [Indexed: 11/29/2022] Open
Abstract
Purpose: Desvenlafaxine succinate (DSV) is a water soluble anti-depressant drug, which is rapidly absorbed after oral administration exaggerating its side effects. The current work aimed to prepare controllable release DSV matrix to reduce DSV side effects related to its initial burst. Methods: Fifteen DSV matrix formulations were prepared using different polymers, polymer/drug ratios and matrix excipients and characterized using Differential Scanning Calorimetry (DSC), infrared (IR) spectroscopy, water uptake and in vitro DSV release. The release kinetics were calculated to determine the drug release mechanism. Ex-vivo DSV absorption via rat intestinal mucosal cells and the calculation of the apparent permeability coefficient (Papp) were performed using everted sac technique. Results: Maltodextrin was the best matrix excipient (F7 and F10) showing acceptable decrease in the initial burst compared to the innovator. The addition of negatively charged polymers sodium carboxy methyl cellulose (SCMC) or sodium alginate resulted in an interaction that was proved by DSC and IR findings. This interaction slowed DSV release. F10 showed an excellent absorption of more than 80% of DSV after 4 h and the highest similarity factor with the innovator (84.7). Conclusion: A controllable release pattern of DSV was achieved using Methocel, Maltodextrin and SCMC. The obtained results could be used as a platform to control the release of cationic water soluble drugs that suffer from side effects associated with their initial burst after oral administration.
Collapse
Affiliation(s)
- Wael Samy
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Ayman Elnoby
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Hanan M El-Gowelli
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Nazik Elgindy
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
74
|
Dubray O, Jannin V, Demarne F, Pellequer Y, Lamprecht A, Béduneau A. In-vitro investigation regarding the effects of Gelucire ® 44/14 and Labrasol® ALF on the secretory intestinal transport of P-gp substrates. Int J Pharm 2016; 515:293-299. [PMID: 27720954 DOI: 10.1016/j.ijpharm.2016.10.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 09/29/2016] [Accepted: 10/05/2016] [Indexed: 10/20/2022]
Abstract
In this present study, the secretory transport of P-gp substrates, rhodamine 123 and digoxin, was evaluated using a Caco-2/HT29-MTX co-culture characterized by an efflux mechanism and a paracellular permeability closer to the human intestinal barrier compared to the Caco-2 monolayer gold standard. The influence of simulated intestinal fluids termed FeSSIF and FaSSIF on the intestinal absorption was also assessed in comparison with a conventional saline buffer. Labrasol® ALF and Gelucire® 44/14 in saline buffer significantly decreased to 83% and 62%, the P-gp-mediated transport of rhodamine 123 across the co-culture, respectively. The effects of Gelucire® 44/14 were much more exacerbated with the Caco-2 monolayer model with a reduced permeability to 34% but they were partially reversed in the co-culture with FeSSIF. The modulation by the lipid excipients of digoxin secretory transport across the Caco-2 monolayer and the co-culture was reduced compared with the rhodamine 123. This work also emphasizes the numerous parameters that have to be considered for predicting accurately the effects of potential P-gp inhibitors including the in-vitro model, the incubation media and the intrinsic properties of P-gp substrates.
Collapse
Affiliation(s)
- Océane Dubray
- FDE EA4267, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France
| | - Vincent Jannin
- Gattefossé SAS, 36 chemin de Genas, 69804, Saint-Priest cedex, France
| | - Fréderic Demarne
- Gattefossé SAS, 36 chemin de Genas, 69804, Saint-Priest cedex, France
| | - Yann Pellequer
- FDE EA4267, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France
| | - Alf Lamprecht
- FDE EA4267, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France; Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Germany
| | - Arnaud Béduneau
- FDE EA4267, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France.
| |
Collapse
|
75
|
Kodama N, Iwao T, Katano T, Ohta K, Yuasa H, Matsunaga T. Characteristic Analysis of Intestinal Transport in Enterocyte-Like Cells Differentiated from Human Induced Pluripotent Stem Cells. Drug Metab Dispos 2016; 44:0. [PMID: 27417181 DOI: 10.1124/dmd.116.069336] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 07/13/2016] [Indexed: 12/25/2022] Open
Abstract
We previously demonstrated that differentiated enterocytes from human induced pluripotent stem (iPS) cells exhibited drug-metabolizing activities and cytochrome P450 CYP3A4 inducibility. The aim of this study was to apply human iPS cell-derived enterocytes in pharmacokinetic studies by investigating the characteristics of drug transport into enterocyte-like cells. Human iPS cells cultured on feeder cells were differentiated into endodermal cells using activin A. These endodermal-like cells were then differentiated into intestinal stem cells by fibroblast growth factor 2. Finally, epidermal growth factor and small-molecule compounds induced the maturation of the intestinal stem cell-like cells. After differentiation, we performed transepithelial electrical resistance (TEER) measurements, immunofluorescence staining, and transport studies. TEER values increased in a time-dependent manner and reached approximately 100 Ω × cm(2) Efflux transport of Hoechst 33342, a substrate of breast cancer resistance protein (BCRP), was observed and inhibited by the BCRP inhibitor Ko143. The uptake of peptide transporter 1 substrate glycylsarcosine was also confirmed and suppressed when the temperature was lowered to 4°C. Using immunofluorescence staining, villin and Na(+)-K(+) ATPase were expressed. These results suggest that human iPS cell-derived enterocytes had loose tight junctions, polarity, as well as uptake and efflux transport functions. In addition, the rank order of apparent membrane permeability coefficient (Papp) values of these test compounds across the enterocyte-like cell membrane corresponded to the fraction absorbance (Fa) values. Therefore, differentiated enterocytes from human iPS cells may provide a useful comprehensive evaluation model of drug transport and metabolism in the small intestine.
Collapse
Affiliation(s)
- Nao Kodama
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (N.K., T.I., T.M.), Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (T.K., K.O., H.Y.)
| | - Takahiro Iwao
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (N.K., T.I., T.M.), Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (T.K., K.O., H.Y.)
| | - Takahiro Katano
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (N.K., T.I., T.M.), Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (T.K., K.O., H.Y.)
| | - Kinya Ohta
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (N.K., T.I., T.M.), Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (T.K., K.O., H.Y.)
| | - Hiroaki Yuasa
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (N.K., T.I., T.M.), Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (T.K., K.O., H.Y.)
| | - Tamihide Matsunaga
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (N.K., T.I., T.M.), Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (T.K., K.O., H.Y.)
| |
Collapse
|
76
|
Joubert R, Steyn JD, Heystek HJ, Steenekamp JH, Du Preez JL, Hamman JH. In vitro oral drug permeation models: the importance of taking physiological and physico-chemical factors into consideration. Expert Opin Drug Deliv 2016; 14:179-187. [PMID: 27397695 DOI: 10.1080/17425247.2016.1211639] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION The assessment of intestinal membrane permeability properties of new chemical entities is a crucial step in the drug discovery and development process and a variety of in vitro models, methods and techniques are available to estimate the extent of oral drug absorption in humans. However, variations in certain physiological and physico-chemical factors are often not reflected in the results and the complex dynamic interplay between these factors is sometimes oversimplified with in vitro models. Areas covered: In vitro models to evaluate drug pharmacokinetics are briefly outlined, while both physiological and physico-chemical factors that may have an influence on these techniques are critically reviewed. The shortcomings identified for some of the in vitro techniques are discussed in conjunction with novel ways to improve and thereby overcome some challenges. Expert opinion: Although conventional in vitro methods and theories are used as basic guidelines to predict drug absorption, critical evaluations have identified some shortcomings. Advancements in technology have made it possible to investigate and understand the role of physiological and physico-chemical factors in drug delivery more clearly, which can be used to improve and refine the techniques to more closely mimic the in vivo environment.
Collapse
Affiliation(s)
- Ruan Joubert
- a Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences , North-West University , Potchefstroom , South Africa
| | - Johan Dewald Steyn
- a Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences , North-West University , Potchefstroom , South Africa
| | - Hendrik Jacobus Heystek
- a Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences , North-West University , Potchefstroom , South Africa
| | - Jan Harm Steenekamp
- a Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences , North-West University , Potchefstroom , South Africa
| | - Jan Lourens Du Preez
- a Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences , North-West University , Potchefstroom , South Africa
| | - Josias Hendrik Hamman
- a Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences , North-West University , Potchefstroom , South Africa
| |
Collapse
|
77
|
Peters SA, Jones CR, Ungell AL, Hatley OJD. Predicting Drug Extraction in the Human Gut Wall: Assessing Contributions from Drug Metabolizing Enzymes and Transporter Proteins using Preclinical Models. Clin Pharmacokinet 2016; 55:673-96. [PMID: 26895020 PMCID: PMC4875961 DOI: 10.1007/s40262-015-0351-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Intestinal metabolism can limit oral bioavailability of drugs and increase the risk of drug interactions. It is therefore important to be able to predict and quantify it in drug discovery and early development. In recent years, a plethora of models-in vivo, in situ and in vitro-have been discussed in the literature. The primary objective of this review is to summarize the current knowledge in the quantitative prediction of gut-wall metabolism. As well as discussing the successes of current models for intestinal metabolism, the challenges in the establishment of good preclinical models are highlighted, including species differences in the isoforms; regional abundances and activities of drug metabolizing enzymes; the interplay of enzyme-transporter proteins; and lack of knowledge on enzyme abundances and availability of empirical scaling factors. Due to its broad specificity and high abundance in the intestine, CYP3A is the enzyme that is frequently implicated in human gut metabolism and is therefore the major focus of this review. A strategy to assess the impact of gut wall metabolism on oral bioavailability during drug discovery and early development phases is presented. Current gaps in the mechanistic understanding and the prediction of gut metabolism are highlighted, with suggestions on how they can be overcome in the future.
Collapse
Affiliation(s)
- Sheila Annie Peters
- Translational Quantitative Pharmacology, BioPharma, R&D Global Early Development, Merck KGaA, Frankfurter Str. 250, F130/005, 64293, Darmstadt, Germany.
| | | | - Anna-Lena Ungell
- Investigative ADME, Non-Clinical Development, UCB New Medicines, BioPharma SPRL, Braine l'Alleud, Belgium
| | - Oliver J D Hatley
- Simcyp Limited (A Certara Company), Blades Enterprise Centre, Sheffield, UK
| |
Collapse
|
78
|
Oat β-glucan depresses SGLT1- and GLUT2-mediated glucose transport in intestinal epithelial cells (IEC-6). Nutr Res 2016; 36:541-52. [DOI: 10.1016/j.nutres.2016.02.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 02/10/2016] [Accepted: 02/16/2016] [Indexed: 01/28/2023]
|
79
|
Rtibi K, Selmi S, Jabri MA, Mamadou G, Limas-Nzouzi N, Sebai H, El-Benna J, Marzouki L, Eto B, Amri M. Effects of aqueous extracts from Ceratonia siliqua L. pods on small intestinal motility in rats and jejunal permeability in mice. RSC Adv 2016. [DOI: 10.1039/c6ra03457h] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The current study was performed to assess the effects of carob pod aqueous extracts (CPAE, pulp, seeds or mixture) on gastrointestinal transit (GIT) and intestinal epithelium permeability.
Collapse
Affiliation(s)
- Kaïs Rtibi
- Laboratory of Neurophysiology and Functional Pathology
- Department of Biological Sciences
- Faculty of Sciences of Tunis
- Tunis 2092
- Tunisia
| | - Slimen Selmi
- Laboratory Functional Physiology and Bio-resources Valorisation
- Higher Institute of Biotechnology of Beja
- University of Jendouba
- 9000 Beja
- Tunisia
| | - Mohamed-Amine Jabri
- Laboratory Functional Physiology and Bio-resources Valorisation
- Higher Institute of Biotechnology of Beja
- University of Jendouba
- 9000 Beja
- Tunisia
| | | | | | - Hichem Sebai
- Laboratory Functional Physiology and Bio-resources Valorisation
- Higher Institute of Biotechnology of Beja
- University of Jendouba
- 9000 Beja
- Tunisia
| | - Jamel El-Benna
- INSERM U1149 Biomedical Research Centre
- Faculty of Medicine X. Bichat
- 75018 Paris
- France
| | - Lamjed Marzouki
- Laboratory of Neurophysiology and Functional Pathology
- Department of Biological Sciences
- Faculty of Sciences of Tunis
- Tunis 2092
- Tunisia
| | - Bruno Eto
- TransCell-Lab
- Faculty of Medicine X. Bichat
- 75018 Paris
- France
| | - Mohamed Amri
- Laboratory of Neurophysiology and Functional Pathology
- Department of Biological Sciences
- Faculty of Sciences of Tunis
- Tunis 2092
- Tunisia
| |
Collapse
|
80
|
Obringer C, Manwaring J, Goebel C, Hewitt NJ, Rothe H. Suitability of the in vitro Caco-2 assay to predict the oral absorption of aromatic amine hair dyes. Toxicol In Vitro 2015; 32:1-7. [PMID: 26578466 DOI: 10.1016/j.tiv.2015.11.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 10/20/2015] [Accepted: 11/11/2015] [Indexed: 11/26/2022]
Abstract
Oral absorption is a key element for safety assessments of cosmetic ingredients, including hair dye molecules. Reliable in vitro methods are needed since the European Union has banned the use of animals for the testing of cosmetic ingredients. Caco-2 cells were used to measure the intestinal permeability characteristics (Papp) of 14 aromatic amine hair dye molecules with varying chemical structures, and the data were compared with historical in vivo oral absorption rat data. The majority of the hair dyes exhibited Papp values that indicated good in vivo absorption. The moderate to high oral absorption findings, i.e. ≥60%, were confirmed in in vivo rat studies. Moreover, the compound with a very low Papp value (APB: 3-((9,10-dihydro-9,10-dioxo-4-(methylamino)-1-anthracenyl)amino)-N,N-dimethyl-N-propyl-1-propanaminium) was poorly absorbed in vivo as well (5% of the dose). This data set suggests that the Caco-2 cell model is a reliable in vitro tool for the determination of the intestinal absorption of aromatic amines with diverse chemical structures. When used in combination with other in vitro assays for metabolism and skin penetration, the Caco-2 model can contribute to the prediction and mechanistic interpretation of the absorption, metabolism and elimination properties of cosmetic ingredients without the use of animals.
Collapse
Affiliation(s)
| | | | | | | | - Helga Rothe
- Procter & Gamble Service GmbH, Schwalbach, Germany
| |
Collapse
|
81
|
Ozawa T, Takayama K, Okamoto R, Negoro R, Sakurai F, Tachibana M, Kawabata K, Mizuguchi H. Generation of enterocyte-like cells from human induced pluripotent stem cells for drug absorption and metabolism studies in human small intestine. Sci Rep 2015; 5:16479. [PMID: 26559489 PMCID: PMC4642303 DOI: 10.1038/srep16479] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 10/14/2015] [Indexed: 12/14/2022] Open
Abstract
Enterocytes play an important role in drug absorption and metabolism. However, a widely used enterocyte model, Caco-2 cell, has difficulty in evaluating both drug absorption and metabolism because the expression levels of some drug absorption and metabolism-related genes in these cells differ largely from those of human enterocytes. Therefore, we decided to generate the enterocyte-like cells from human induced pluripotent stem (iPS) cells (hiPS-ELCs), which are applicable to drug absorption and metabolism studies. The efficiency of enterocyte differentiation from human iPS cells was significantly improved by using EGF, SB431542, and Wnt3A, and extending the differentiation period. The gene expression levels of cytochrome P450 3A4 (CYP3A4) and peptide transporter 1 in the hiPS-ELCs were higher than those in Caco-2 cells. In addition, CYP3A4 expression in the hiPS-ELCs was induced by treatment with 1, 25-dihydroxyvitamin D3 or rifampicin, which are known to induce CYP3A4 expression, indicating that the hiPS-ELCs have CYP3A4 induction potency. Moreover, the transendothelial electrical resistance (TEER) value of the hiPS-ELC monolayer was approximately 240 Ω*cm2, suggesting that the hiPS-ELC monolayer could form a barrier. In conclusion, we succeeded in establishing an enterocyte model from human iPS cells which have potential to be applied for drug absorption and metabolism studies.
Collapse
Affiliation(s)
- Tatsuya Ozawa
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan.,Laboratory of Hepatocyte Regulation, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Kazuo Takayama
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan.,Laboratory of Hepatocyte Regulation, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan.,iPS Cell-based Research Project on Hepatic Toxicity and Metabolism, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Ryota Okamoto
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan.,Laboratory of Hepatocyte Regulation, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Ryosuke Negoro
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Fuminori Sakurai
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan.,Laboratory of Regulatory Sciences for Oligonucleotide Therapeutics, Clinical Drug Development Project, Graduate School of Pharmaceutical Sciences, Osaka University Osaka 565-0871, Japan
| | - Masashi Tachibana
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Kenji Kawabata
- Laboratory of Stem Cell Regulation, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan.,Laboratory of Biomedical Innovation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan.,Laboratory of Hepatocyte Regulation, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan.,iPS Cell-based Research Project on Hepatic Toxicity and Metabolism, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan.,Global Center for Medical Engineering and Informatics, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
82
|
Cui HM, Zhang QY, Wang JL, Chen JL, Zhang YL, Tong XL. Poor permeability and absorption affect the activity of four alkaloids from Coptis. Mol Med Rep 2015; 12:7160-8. [PMID: 26352530 DOI: 10.3892/mmr.2015.4288] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 08/04/2015] [Indexed: 11/06/2022] Open
Abstract
Coptidis rhizoma (Coptis) and its alkaloids exert various pharmacological functions in cells and tissues; however, the oral absorption of these alkaloids requires further elucidation. The present study aimed to examine the mechanism underlying the poor absorption of alkaloids, including berberine (BER), coptisine (COP), palmatine (PAL) and jatrorrhizine (JAT). An ultra‑performance liquid chromatography (UPLC) method was validated for the determination of BER, COP, PAL and JAT in the above experimental medium. In addition, the apparent oil‑water partition coefficient (Po/w); apparent permeability coefficient (Papp), determined using a parallel artificial membrane permeability assay (PAMPA) plate; membrane retention coefficient (R %); and effect of P‑glycoprotein (P‑gp) inhibitor on the Papp of the four alkaloids were investigated. The intestinal absorption rate constant (Ka) and absorption percentage (A %) of the four alkaloids were also determined. The results of the present study demonstrated that the Po/w of the four alkaloids in 0.1 mol·l‑1 HCl medium was significantly higher (P<0.01), compared with those of the alkaloids in phosphate buffer (pH 7.4). The Papp of BER was 1.0‑1.2x10‑6 cm·s‑1, determined using a PAMPA plate, and the Papp of BER, COP, PAL and JAT decreased sequentially. The concentrations of the four alkaloids on the apical‑to‑basolateral (AP‑BL) surface and the basolateral‑to‑apical (BL‑AP) surface increased in a linear manner, with increasing concentrations between 10 and 100 µmol. In addition, the transportation of BER on the BL‑AP surface was significantly faster (P<0.01), compared with that on the AP‑BL surface and, following the addition of verpamil (a P‑gp inhibitor), the Papp (AP‑BL) of the four alkaloids increased, whereas the Papp (BL‑AP) was significantly decreased (P<0.01). The rat intestinal perfusion experiment demonstrated that the four alkaloids were poorly absorbed; however, the Ka of BER was significantly higher, compared with the three other alkaloids. Furthermore, the A % and Ka provided evidence that the absorption of BER was increased in the jejunum, compared with in the ileum. In conclusion, the four alkaloids from Coptis appeared to be poorly absorbed, determined using a shake flask, pre‑coated PAMPA plates, a Caco‑2 cell monolayer model and intestinal perfusion; however, absorption was higher in the jejunum than in the ileum. Among the four alkaloids, the permeability of BER was markedly higher than the others, and P‑gp efflux had a significant effect on the absorption of those alkaloids.
Collapse
Affiliation(s)
- Han-Ming Cui
- Department of Chinese Traditional Medicine Research and Development Center, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| | - Qiu-Yan Zhang
- Department of Chinese Traditional Medicine Research and Development Center, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| | - Jia-Long Wang
- Department of Chinese Traditional Medicine Research and Development Center, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| | - Jian-Long Chen
- Department of Chinese Traditional Medicine Research and Development Center, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| | - Yu-Ling Zhang
- Department of Chinese Traditional Medicine Research and Development Center, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| | - Xiao-Lin Tong
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| |
Collapse
|
83
|
Araújo F, Pereira C, Costa J, Barrias C, Granja PL, Sarmento B. In vitroM-like cells genesis through a tissue-engineered triple-culture intestinal model. J Biomed Mater Res B Appl Biomater 2015; 104:782-8. [DOI: 10.1002/jbm.b.33508] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Revised: 07/18/2015] [Accepted: 08/14/2015] [Indexed: 01/22/2023]
Affiliation(s)
- Francisca Araújo
- I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto; Portugal
- INEB-Instituto de Engenharia Biomédica, University of Porto; Rua do Campo Alegre, 823 Porto 4150-180 Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, University of Porto; Rua de Jorge Viterbo Ferreira Porto 4050-313 Portugal
| | - Carla Pereira
- I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto; Portugal
- INEB-Instituto de Engenharia Biomédica, University of Porto; Rua do Campo Alegre, 823 Porto 4150-180 Portugal
- FEUP-Faculdade de Engenharia, University of Porto; Rua Dr. Roberto Frias Porto 4200-465 Portugal
| | - Joana Costa
- I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto; Portugal
- INEB-Instituto de Engenharia Biomédica, University of Porto; Rua do Campo Alegre, 823 Porto 4150-180 Portugal
- FEUP-Faculdade de Engenharia, University of Porto; Rua Dr. Roberto Frias Porto 4200-465 Portugal
| | - Cristina Barrias
- I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto; Portugal
- INEB-Instituto de Engenharia Biomédica, University of Porto; Rua do Campo Alegre, 823 Porto 4150-180 Portugal
| | - Pedro L. Granja
- I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto; Portugal
- INEB-Instituto de Engenharia Biomédica, University of Porto; Rua do Campo Alegre, 823 Porto 4150-180 Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, University of Porto; Rua de Jorge Viterbo Ferreira Porto 4050-313 Portugal
- FEUP-Faculdade de Engenharia, University of Porto; Rua Dr. Roberto Frias Porto 4200-465 Portugal
| | - Bruno Sarmento
- I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto; Portugal
- INEB-Instituto de Engenharia Biomédica, University of Porto; Rua do Campo Alegre, 823 Porto 4150-180 Portugal
- CESPU-Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Instituto Universitário de Ciências da Saúde; Rua Central de Grandra, 1317 Gandra 4585-116 Portugal
| |
Collapse
|
84
|
Pan F, Han L, Zhang Y, Yu Y, Liu J. Optimization of Caco-2 and HT29 co-culture in vitro cell models for permeability studies. Int J Food Sci Nutr 2015; 66:680-5. [PMID: 26299896 DOI: 10.3109/09637486.2015.1077792] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The purpose of this study was to investigate the appropriate proportion of Caco-2 and HT29 co-culture in vitro cell models for permeability studies. The results showed that the transepithelial electrical resistance values of 9:1 and 1:0 groups (263 ± 3.61 and 300 ± 7.55) after 21-day culture were >250 Ω cm(2), which were suitable for further experiments. The confocal laser microscopy showed that the group of 9:1 (Caco-2:HT29) had the highest integrity, whereas the group of 0:1 (Caco-2:HT29) exhibited the lowest. The staining study confirmed that mucus was successfully produced by HT29 cells, and it was also produced in co-cultures with Caco-2 cells model, but the Caco-2 monocultures did not have any blue staining, which made us affirm that mucus is only produced in the presence of HT29 cells. The real-time PCR results showed that the total highest expression level of ALPi and MUC5AC was the ratio of 9:1 (Caco-2:HT29) and lowest is 1:1 (Caco-2:HT29). So we concluded that 9:1 (Caco-2:HT29) is the optimal Caco-2 to HT29 ratio in the in vitro model co-culture for permeability studies.
Collapse
Affiliation(s)
- Fengguang Pan
- a Laboratory of Nutrition and Functional Food , Jilin University , Changchun , PR China
| | - Lu Han
- a Laboratory of Nutrition and Functional Food , Jilin University , Changchun , PR China
| | - Yan Zhang
- a Laboratory of Nutrition and Functional Food , Jilin University , Changchun , PR China
| | - Yiding Yu
- a Laboratory of Nutrition and Functional Food , Jilin University , Changchun , PR China
| | - Jingbo Liu
- a Laboratory of Nutrition and Functional Food , Jilin University , Changchun , PR China
| |
Collapse
|
85
|
Meyer S, Raber G, Ebert F, Taleshi MS, Francesconi KA, Schwerdtle T. Arsenic-containing hydrocarbons and arsenic-containing fatty acids: Transfer across and presystemic metabolism in the Caco-2 intestinal barrier model. Mol Nutr Food Res 2015; 59:2044-56. [PMID: 26153761 DOI: 10.1002/mnfr.201500286] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 06/26/2015] [Accepted: 06/26/2015] [Indexed: 01/03/2023]
Abstract
SCOPE Arsenic-containing hydrocarbons (AsHCs) and arsenic-containing fatty acids (AsFAs) represent two classes of arsenolipids occurring naturally in marine food. Toxicological data are yet scarce and an assessment regarding the risk to human health has not been possible. Here, we investigated the transfer and presystemic metabolism of five arsenolipids in an intestinal barrier model. METHODS AND RESULTS Three AsHCs and two AsFAs were applied to the Caco-2 intestinal barrier model. Thereby, the short-chain AsHCs reached up to 50% permeability. Transport is likely to occur via passive diffusion. The AsFAs showed lower intestinal bioavailability, but respective permeabilities were still two to five times higher as compared to arsenobetaine or arsenosugars. Interestingly, AsFAs were effectively biotransformed while passing the in vitro intestinal barrier, whereas AsHCs were transported to the blood-facing compartment essentially unchanged. CONCLUSION AsFAs can be presystemically metabolised and the amount of transferred arsenic is lower than that for AsHCs. In contrast, AsHCs are likely to be highly intestinally bioavailable to humans. Since AsHCs exert strong toxicity in vitro and in vivo, toxicity studies with experimental animals as well as a human exposure assessment are needed to assess the risk to human health related to the presence of AsHCs in seafood.
Collapse
Affiliation(s)
- Sören Meyer
- Graduate School of Chemistry, University of Münster, Münster, Germany.,Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Georg Raber
- Institute of Chemistry-Analytical Chemistry, NAWI Graz, University of Graz, Graz, Austria
| | - Franziska Ebert
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Mojtaba S Taleshi
- Institute of Chemistry-Analytical Chemistry, NAWI Graz, University of Graz, Graz, Austria
| | - Kevin A Francesconi
- Institute of Chemistry-Analytical Chemistry, NAWI Graz, University of Graz, Graz, Austria
| | - Tanja Schwerdtle
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| |
Collapse
|
86
|
Sengupta R, Anderson RC, Altermann E, McNabb WC, Ganesh S, Armstrong KM, Moughan PJ, Roy NC. Lactobacillus fermentum AGR1487 cell surface structures and supernatant increase paracellular permeability through different pathways. Microbiologyopen 2015; 4:541-52. [PMID: 25943073 PMCID: PMC4554451 DOI: 10.1002/mbo3.260] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 03/19/2015] [Accepted: 03/23/2015] [Indexed: 12/21/2022] Open
Abstract
Lactobacillus fermentum is commonly found in food products, and some strains are known to have beneficial effects on human health. However, our previous research indicated that L. fermentum AGR1487 decreases in vitro intestinal barrier integrity. The hypothesis was that cell surface structures of AGR1487 are responsible for the observed in vitro effect. AGR1487 was compared to another human oral L. fermentum strain, AGR1485, which does not cause the same effect. The examination of phenotypic traits associated with the composition of cell surface structures showed that compared to AGR1485, AGR1487 had a smaller genome, utilized different sugars, and had greater tolerance to acid and bile. The effect of the two strains on intestinal barrier integrity was determined using two independent measures of paracellular permeability of the intestinal epithelial Caco-2 cell line. The transepithelial electrical resistance (TEER) assay specifically measures ion permeability, whereas the mannitol flux assay measures the passage of uncharged molecules. Both live and UV-inactivated AGR1487 decreased TEER across Caco-2 cells implicating the cell surfaces structures in the effect. However, only live AGR1487, and not UV-inactivated AGR1487, increased the rate of passage of mannitol, implying that a secreted component(s) is responsible for this effect. These differences in barrier integrity results are likely due to the TEER and mannitol flux assays measuring different characteristics of the epithelial barrier, and therefore imply that there are multiple mechanisms involved in the effect of AGR1487 on barrier integrity.
Collapse
Affiliation(s)
- Ranjita Sengupta
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch GrasslandsPalmerston North, 4442, New Zealand
- Riddet Institute, Massey UniversityPalmerston North, 4442, New Zealand
| | - Rachel C Anderson
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch GrasslandsPalmerston North, 4442, New Zealand
- Riddet Institute, Massey UniversityPalmerston North, 4442, New Zealand
| | - Eric Altermann
- Riddet Institute, Massey UniversityPalmerston North, 4442, New Zealand
- Rumen Microbiology Team, Animal Nutrition & Health Group, AgResearch GrasslandsPalmerston North, 4442, New Zealand
| | - Warren C McNabb
- Riddet Institute, Massey UniversityPalmerston North, 4442, New Zealand
- AgResearch GrasslandsPalmerston North, 4442, New Zealand
| | - Siva Ganesh
- Bioinformatics Mathematics & Statistics Team, AgResearch GrasslandsPalmerston North, 4442, New Zealand
| | - Kelly M Armstrong
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch GrasslandsPalmerston North, 4442, New Zealand
| | - Paul J Moughan
- Riddet Institute, Massey UniversityPalmerston North, 4442, New Zealand
| | - Nicole C Roy
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch GrasslandsPalmerston North, 4442, New Zealand
- Riddet Institute, Massey UniversityPalmerston North, 4442, New Zealand
- Gravida: National Centre for Growth and Development, The University of AucklandAuckland, New Zealand
| |
Collapse
|
87
|
Sallam MA, Marín Boscá MT. Optimization, ex vivo permeation, and stability study of lipid nanocarrier loaded gelatin capsules for treatment of intermittent claudication. Int J Nanomedicine 2015. [PMID: 26203244 PMCID: PMC4508069 DOI: 10.2147/ijn.s83123] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
In this study, an optimized nanodispersible oral dosage form (containing a lactate ester) was developed for cilostazol (CZL). CZL is a phosphodiesterase inhibitor used for intermittent claudication. We aimed to improve the dissolution rate and absorption of CZL giving it a better chance of oral bioavailability, and to evaluate its stability on storage. Suitable compositions of nanoemulsion preconcentrate formulations were screened via solubility and compatibility tests. Response surface methodology and a desirability approach were applied to optimize preconcentrates containing minimum amount of surfactant mixture, maximum amount of lipid, and possessing the smallest globule size, with the highest emulsification and dissolution rates and minimum risk of drug precipitation. As part of the optimization process, the main effect, interaction effects and quadratic effects of amounts of lipid, and surfactant/co-surfactant ratio on % transmittance, globule size, emulsification time, drug precipitation, and drug release were investigated. The optimized formulation consisting of 28.9% butyl lactate, 28.9% Capryol, 27.82% Solubilisant Gamma 2429, and 14.18% Transcutol possessing a globule size of 60 nm was mixed with Aerosil 200. This gave uniform free flowing granules, which were characterized for surface and powder properties. The self-nanoemulsifying granules (SNEGs) filled into hard gelatin capsules showed two- and threefold increase in CZL released compared with conventional tablet and pure drug, respectively. The amount of drug permeated using non-everted sac technique from the SNEGs was twofold higher than that permeated from the tablet suspension. The shelf life was 526 days at 25°C. Our study illustrated that the developed SNEGs, with bioenhancing ingredients, held great potential as a superior alternative to traditional oral formulations of CZL.
Collapse
Affiliation(s)
- Marwa Ahmed Sallam
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - María Teresa Marín Boscá
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Granada University, Granada, Spain
| |
Collapse
|
88
|
Graves RA, Ledet GA, Glotser EY, Mitchner DM, Bostanian LA, Mandal TK. Formulation and evaluation of biodegradable nanoparticles for the oral delivery of fenretinide. Eur J Pharm Sci 2015; 76:1-9. [PMID: 25933716 DOI: 10.1016/j.ejps.2015.04.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/27/2015] [Accepted: 04/27/2015] [Indexed: 12/31/2022]
Abstract
Fenretinide is an anticancer drug with low water solubility and poor bioavailability. The goal of this study was to develop biodegradable polymeric nanoparticles of fenretinide with the intent of increasing its apparent aqueous solubility and intestinal permeability. Three biodegradable polymers were investigated for this purpose: two different poly lactide-co-glycolide (PLGA) polymers, one acid terminated and one ester terminated, and one poly lactide-co-glycolide/polyethylene glycol (PLGA/PEG) diblock copolymer. Nanoparticles were obtained by using an emulsification solvent evaporation technique. The formulations were characterized by differential scanning calorimetry (DSC), scanning electron microscopy (SEM), and particle size analysis. Dissolution studies and Caco-2 cell permeation studies were also carried out for all formulations. Ultra high performance liquid chromatography coupled with mass spectrometry (UPLC/MS) and ultraviolet detection was used for the quantitative determination of fenretinide. Drug loading and the type of polymer affected the nanoparticles' physical properties, drug release rate, and cell permeability. While the acid terminated PLGA nanoparticles performed the best in drug release, the ester terminated PLGA nanoparticles performed the best in the Caco-2 cell permeability assays. The PLGA/PEG copolymer nanoparticles performed better than the formulations with ester terminated PLGA in terms of drug release but had the poorest performance in terms of cell permeation. All three categories of formulations performed better than the drug alone in both drug release and cell permeation studies.
Collapse
Affiliation(s)
- Richard A Graves
- College of Pharmacy, Division of Basic Pharmaceutical Sciences, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, LA 70125-1098, United States
| | - Grace A Ledet
- College of Pharmacy, Division of Basic Pharmaceutical Sciences, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, LA 70125-1098, United States
| | - Elena Y Glotser
- College of Pharmacy, Division of Basic Pharmaceutical Sciences, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, LA 70125-1098, United States
| | - Demaurian M Mitchner
- College of Pharmacy, Division of Basic Pharmaceutical Sciences, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, LA 70125-1098, United States
| | - Levon A Bostanian
- College of Pharmacy, Division of Basic Pharmaceutical Sciences, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, LA 70125-1098, United States.
| | - Tarun K Mandal
- College of Pharmacy, Division of Basic Pharmaceutical Sciences, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, LA 70125-1098, United States
| |
Collapse
|
89
|
Pereira C, Araújo F, Barrias CC, Granja PL, Sarmento B. Dissecting stromal-epithelial interactions in a 3D in vitro cellularized intestinal model for permeability studies. Biomaterials 2015; 56:36-45. [PMID: 25934277 DOI: 10.1016/j.biomaterials.2015.03.054] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 03/28/2015] [Indexed: 12/20/2022]
Abstract
Absorption evaluation plays an increasingly important role at the early stage of drug discovery due to its potential to scan the ADME (absorption, distribution, metabolism and excretion) properties of new drug candidates. Therefore, a new three-dimensional (3D) in vitro model replicating the intestinal functioning is herein proposed aiming to dissect the stromal-epithelial interactions and evaluate the permeation of a model drug, insulin. Inspired on the intestinal mucosal architecture, the present model comprises intestinal myofibroblasts (CCD18-Co cells) embedded in Matrigel, onto which epithelial enterocytes (Caco-2 cells) and mucus-producing cells (HT29-MTX cells) were seeded. CCD18-Co myofibroblasts showed to have a central role in the remodeling of the surrounding matrix confirmed by the production of fibronectin. Subsequently, this matrix revealed to be essential to the maintenance of the model architecture by supporting the overlying epithelial cells. In terms of functionality, this model allowed the efficient prediction of insulin permeability in which the presence of mucus, the less tight character between Caco-2 and HT29-MTX epithelial cells and the 3D assembly were critical factors. Concluding, this model constitutes a robust tool in the drug development field with potential to bridge the traditional 2D cell culture models and in vivo animal models.
Collapse
Affiliation(s)
- Carla Pereira
- I3S - Instituto de Investigação e Inovação em Saúde and INEB - Instituto de Engenharia Biomédica, University of Porto, Rua do Campo Alegre, 823, 4150-180, Porto, Portugal; FEUP - Faculdade de Engenharia, University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal
| | - Francisca Araújo
- I3S - Instituto de Investigação e Inovação em Saúde and INEB - Instituto de Engenharia Biomédica, University of Porto, Rua do Campo Alegre, 823, 4150-180, Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Rua de Jorge Viterbo Ferreira, 4050-313, Porto, Portugal
| | - Cristina C Barrias
- I3S - Instituto de Investigação e Inovação em Saúde and INEB - Instituto de Engenharia Biomédica, University of Porto, Rua do Campo Alegre, 823, 4150-180, Porto, Portugal
| | - Pedro L Granja
- I3S - Instituto de Investigação e Inovação em Saúde and INEB - Instituto de Engenharia Biomédica, University of Porto, Rua do Campo Alegre, 823, 4150-180, Porto, Portugal; FEUP - Faculdade de Engenharia, University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Rua de Jorge Viterbo Ferreira, 4050-313, Porto, Portugal
| | - Bruno Sarmento
- I3S - Instituto de Investigação e Inovação em Saúde and INEB - Instituto de Engenharia Biomédica, University of Porto, Rua do Campo Alegre, 823, 4150-180, Porto, Portugal; CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Instituto Superior de Ciências da Saúde-Norte, Rua Central de Grandra, 1317, 4585-116, Gandra, Portugal.
| |
Collapse
|
90
|
Maschmeyer I, Hasenberg T, Jaenicke A, Lindner M, Lorenz AK, Zech J, Garbe LA, Sonntag F, Hayden P, Ayehunie S, Lauster R, Marx U, Materne EM. Chip-based human liver-intestine and liver-skin co-cultures--A first step toward systemic repeated dose substance testing in vitro. Eur J Pharm Biopharm 2015; 95:77-87. [PMID: 25857839 DOI: 10.1016/j.ejpb.2015.03.002] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 02/13/2015] [Accepted: 03/03/2015] [Indexed: 01/10/2023]
Abstract
Systemic repeated dose safety assessment and systemic efficacy evaluation of substances are currently carried out on laboratory animals and in humans due to the lack of predictive alternatives. Relevant international regulations, such as OECD and ICH guidelines, demand long-term testing and oral, dermal, inhalation, and systemic exposure routes for such evaluations. So-called "human-on-a-chip" concepts are aiming to replace respective animals and humans in substance evaluation with miniaturized functional human organisms. The major technical hurdle toward success in this field is the life-like combination of human barrier organ models, such as intestine, lung or skin, with parenchymal organ equivalents, such as liver, at the smallest biologically acceptable scale. Here, we report on a reproducible homeostatic long-term co-culture of human liver equivalents with either a reconstructed human intestinal barrier model or a human skin biopsy applying a microphysiological system. We used a multi-organ chip (MOC) platform, which provides pulsatile fluid flow within physiological ranges at low media-to-tissue ratios. The MOC supports submerse cultivation of an intact intestinal barrier model and an air-liquid interface for the skin model during their co-culture with the liver equivalents respectively at (1)/100.000 the scale of their human counterparts in vivo. To increase the degree of organismal emulation, microfluidic channels of the liver-skin co-culture could be successfully covered with human endothelial cells, thus mimicking human vasculature, for the first time. Finally, exposure routes emulating oral and systemic administration in humans have been qualified by applying a repeated dose administration of a model substance - troglitazone - to the chip-based co-cultures.
Collapse
Affiliation(s)
- Ilka Maschmeyer
- TissUse GmbH, Markgrafenstrasse 18, 15528 Spreenhagen, Germany; Technische Universität Berlin, Institute of Biotechnology, Department Medical Biotechnology, Gustav-Meyer-Allee 25, 13355 Berlin, Germany.
| | - Tobias Hasenberg
- Technische Universität Berlin, Institute of Biotechnology, Department Medical Biotechnology, Gustav-Meyer-Allee 25, 13355 Berlin, Germany.
| | - Annika Jaenicke
- Technische Universität Berlin, Institute of Biotechnology, Department Medical Biotechnology, Gustav-Meyer-Allee 25, 13355 Berlin, Germany.
| | - Marcus Lindner
- Technische Universität Berlin, Institute of Biotechnology, Department Medical Biotechnology, Gustav-Meyer-Allee 25, 13355 Berlin, Germany.
| | - Alexandra Katharina Lorenz
- Technische Universität Berlin, Institute of Biotechnology, Department Medical Biotechnology, Gustav-Meyer-Allee 25, 13355 Berlin, Germany.
| | - Julie Zech
- Technische Universität Berlin, Institute of Biotechnology, Department Bioanalytics, Seestrasse 13, 13353 Berlin, Germany; Research and Teaching Institute for Brewing in Berlin (VLB), Research Institute for Special Analyses (FIS), Seestrasse 13, 13353 Berlin, Germany.
| | - Leif-Alexander Garbe
- Technische Universität Berlin, Institute of Biotechnology, Department Bioanalytics, Seestrasse 13, 13353 Berlin, Germany.
| | - Frank Sonntag
- Fraunhofer Institute for Material and Beam Technology, Winterbergstr. 28, 01277 Dresden, Germany.
| | | | | | - Roland Lauster
- Technische Universität Berlin, Institute of Biotechnology, Department Medical Biotechnology, Gustav-Meyer-Allee 25, 13355 Berlin, Germany.
| | - Uwe Marx
- TissUse GmbH, Markgrafenstrasse 18, 15528 Spreenhagen, Germany; Technische Universität Berlin, Institute of Biotechnology, Department Medical Biotechnology, Gustav-Meyer-Allee 25, 13355 Berlin, Germany.
| | - Eva-Maria Materne
- Technische Universität Berlin, Institute of Biotechnology, Department Medical Biotechnology, Gustav-Meyer-Allee 25, 13355 Berlin, Germany.
| |
Collapse
|
91
|
Ferretti V, Dalpiaz A, Bertolasi V, Ferraro L, Beggiato S, Spizzo F, Spisni E, Pavan B. Indomethacin co-crystals and their parent mixtures: does the intestinal barrier recognize them differently? Mol Pharm 2015; 12:1501-11. [PMID: 25794305 DOI: 10.1021/mp500826y] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Co-crystals are crystalline complexes of two or more molecules bound together in crystal lattices through noncovalent interactions. The solubility and dissolution properties of co-crystals can allow to increase the bioavailability of poorly water-soluble active pharmaceutical ingredients (APIs). It is currently believed that the co-crystallization strategy should not induce changes on the pharmacological profile of the APIs, even if it is not yet clear whether a co-crystal would be defined as a physical mixture or as a new chemical entity. In order to clarify these aspects, we chose indomethacin as guest poorly aqueous soluble molecule and compared its properties with those of its co-crystals obtained with 2-hydroxy-4-methylpyridine (co-crystal 1), 2-methoxy-5-nitroaniline (co-crystal 2), and saccharine (co-crystal 3). In particular, we performed a systematic comparison among indomethacin, its co-crystals, and their parent physical mixtures by evaluating via HPLC analysis the API dissolution profile, its ability to permeate across intestinal cell monolayers (NCM460), and its oral bioavailability in rat. The indomethacin dissolution profile was not altered by the presence of co-crystallizing agents as physical mixtures, whereas significant changes were observed by the dissolution of the co-crystals. Furthermore, there was a qualitative concordance between the API dissolution patterns and the relative oral bioavailabilities in rats. Co-crystal 1 induced a drastic decrease of the transepithelial electrical resistance (TEER) value of NCM460 cell monolayers, whereas its parent mixture did not evidence any effect. The saccharin-indomethacin mixture induced a drastic decrease of the TEER value of monolayers, whereas its parent co-crystal 3 did not induce any effects on their integrity, being anyway able to increase the permeation of indomethacin. Taken together, these results demonstrate for the first time different effects induced by co-crystals and their parent physical mixtures on a biologic system, findings that could raise serious concerns about the use of co-crystal strategy to improve API bioavailability without performing appropriate investigations.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Enzo Spisni
- ∥Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | | |
Collapse
|
92
|
Zeller P, Bricks T, Vidal G, Jacques S, Anton PM, Leclerc E. Multiparametric temporal analysis of the Caco-2/TC7 demonstrated functional and differentiated monolayers as early as 14 days of culture. Eur J Pharm Sci 2015; 72:1-11. [PMID: 25725134 DOI: 10.1016/j.ejps.2015.02.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 01/20/2015] [Accepted: 02/14/2015] [Indexed: 11/16/2022]
Abstract
Reducing the differentiation period for obtaining an in vitro intestinal barrier model is required to reduce the duration and cost for drug screening assays. In this frame, the Caco-2/TC7 subclone differentiation state was investigated from day 0 (D0) to day 32 (D32). As such, the expression of 45 genes (including cell junction, cell polarization, cell functionality, drug transport and metabolism genes) was followed throughout the 32 days. In parallel, the monolayer polarization and the formation of the cellular junctions were characterized by the immuno-staining of occludin, claudin-1 and actin proteins. The cell monolayer permeability was analyzed via transepithelial electric resistance measurements and paracellular transport of Lucifer Yellow. The P-gp efflux efficiency was assessed by rhodamine 123 transport. Alkaline phosphate activity was quantified to assess the cell differentiation. Three stages of differentiation were observed using the clustering of principal component analysis of the RTqPCR data and the overall assays. From D0 to D10, cells were in a proliferation stage and under-differentiated; from D14 to D21 a stable differentiation stage was reached; from D25 to D32 the epithelium seemed to enter into a post-differentiated stage. This study demonstrates that Caco-2/TC7 cells are functional and ready for use in drug screening permeability assays from 14 days in culture when compared with conventional 21 days for Caco-2 cells. In addition, this study provides a refined set of data allowing temporal and multi scale investigations, due to the intracellular kinetics and mRNA levels that can be correlated with membrane protein kinetics and functional extracellular activities. Therefore, shorter time in culture combined with a better knowledge of the cells during the time in culture will in turn help to improve the quality and cost of Caco-2/TC7 assays for drug development.
Collapse
Affiliation(s)
- Perrine Zeller
- CNRS UMR 7338, Laboratoire de Biomécanique et Bio ingénierie, Université de Technologie de Compiègne, France
| | - Thibault Bricks
- CNRS UMR 7338, Laboratoire de Biomécanique et Bio ingénierie, Université de Technologie de Compiègne, France
| | - Guillaume Vidal
- CNRS UMR 7338, Laboratoire de Biomécanique et Bio ingénierie, Université de Technologie de Compiègne, France
| | - Sébastien Jacques
- INSERM U1016, Plate-forme génomique, institut Cochin, 22 rue Méchain, 75014 Paris, France
| | - Pauline M Anton
- EGEAL, Institut Polytechnique Lasalle Beauvais, Beauvais, France
| | - Eric Leclerc
- CNRS UMR 7338, Laboratoire de Biomécanique et Bio ingénierie, Université de Technologie de Compiègne, France.
| |
Collapse
|
93
|
Srinivasan B, Kolli AR, Esch MB, Abaci HE, Shuler ML, Hickman JJ. TEER measurement techniques for in vitro barrier model systems. ACTA ACUST UNITED AC 2015; 20:107-26. [PMID: 25586998 DOI: 10.1177/2211068214561025] [Citation(s) in RCA: 1417] [Impact Index Per Article: 141.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Transepithelial/transendothelial electrical resistance (TEER) is a widely accepted quantitative technique to measure the integrity of tight junction dynamics in cell culture models of endothelial and epithelial monolayers. TEER values are strong indicators of the integrity of the cellular barriers before they are evaluated for transport of drugs or chemicals. TEER measurements can be performed in real time without cell damage and generally are based on measuring ohmic resistance or measuring impedance across a wide spectrum of frequencies. The measurements for various cell types have been reported with commercially available measurement systems and also with custom-built microfluidic implementations. Some of the barrier models that have been widely characterized using TEER include the blood-brain barrier (BBB), gastrointestinal (GI) tract, and pulmonary models. Variations in these values can arise due to factors such as temperature, medium formulation, and passage number of cells. The aim of this article is to review the different TEER measurement techniques and analyze their strengths and weaknesses, determine the significance of TEER in drug toxicity studies, examine the various in vitro models and microfluidic organs-on-chips implementations using TEER measurements in some widely studied barrier models (BBB, GI tract, and pulmonary), and discuss the various factors that can affect TEER measurements.
Collapse
Affiliation(s)
- Balaji Srinivasan
- NanoScience Technology Center, University of Central Florida, Orlando, FL, USA
| | - Aditya Reddy Kolli
- NanoScience Technology Center, University of Central Florida, Orlando, FL, USA
| | | | | | | | - James J Hickman
- NanoScience Technology Center, University of Central Florida, Orlando, FL, USA Biomolecular Science Center, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
94
|
Newby D, Freitas AA, Ghafourian T. Decision trees to characterise the roles of permeability and solubility on the prediction of oral absorption. Eur J Med Chem 2014; 90:751-65. [PMID: 25528330 DOI: 10.1016/j.ejmech.2014.12.006] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 12/02/2014] [Accepted: 12/03/2014] [Indexed: 01/11/2023]
Abstract
Oral absorption of compounds depends on many physiological, physiochemical and formulation factors. Two important properties that govern oral absorption are in vitro permeability and solubility, which are commonly used as indicators of human intestinal absorption. Despite this, the nature and exact characteristics of the relationship between these parameters are not well understood. In this study a large dataset of human intestinal absorption was collated along with in vitro permeability, aqueous solubility, melting point, and maximum dose for the same compounds. The dataset allowed a permeability threshold to be established objectively to predict high or low intestinal absorption. Using this permeability threshold, classification decision trees incorporating a solubility-related parameter such as experimental or predicted solubility, or the melting point based absorption potential (MPbAP), along with structural molecular descriptors were developed and validated to predict oral absorption class. The decision trees were able to determine the individual roles of permeability and solubility in oral absorption process. Poorly permeable compounds with high solubility show low intestinal absorption, whereas poorly water soluble compounds with high or low permeability may have high intestinal absorption provided that they have certain molecular characteristics such as a small polar surface or specific topology.
Collapse
Affiliation(s)
- Danielle Newby
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham, Kent ME4 4TB, UK
| | - Alex A Freitas
- School of Computing, University of Kent, Canterbury, Kent CT2 7NF, UK
| | - Taravat Ghafourian
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham, Kent ME4 4TB, UK; Drug Applied Research Centre and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
95
|
Vázquez M, Devesa V, Vélez D. Characterization of the intestinal absorption of inorganic mercury in Caco-2 cells. Toxicol In Vitro 2014; 29:93-102. [PMID: 25283090 DOI: 10.1016/j.tiv.2014.09.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 09/16/2014] [Accepted: 09/23/2014] [Indexed: 11/16/2022]
Abstract
The main form of mercury exposure in the general population is through food. Intestinal absorption is therefore a key step in the penetration of mercury into the systemic circulation, and should be considered when evaluating exposure risk. Many studies have investigated the transport of mercury species in different cell lines, though the mechanisms underlying their intestinal absorption are not clear. This study evaluates the accumulation and transport of Hg(II), one of the mercury species ingested in food, using Caco-2 cells as intestinal epithelium model with the purpose of clarifying the mechanisms involved in its absorption. Hg(II) shows moderate absorption, and its transport fundamentally takes place via a carrier-mediated transcellular mechanism. The experiments indicate the participation of an energy-dependent transport mechanism. In addition, H(+)- and Na(+)-dependent transport is also observed. These data, together with those obtained from inhibition studies using specific substrates or inhibitors of different transporter families, suggest the participation of divalent cation and amino acid transporters, and even some organic anion transporters, in Hg(II) intestinal transport. An important cellular accumulation of up to 51% is observed - a situation which in view of the toxic nature of this species could affect intestinal mucosal function. This study contributes new information on the mechanisms of transport of Hg(II) at intestinal level, and which may be responsible for penetration of this mercurial form into the systemic circulation.
Collapse
Affiliation(s)
- M Vázquez
- Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Avenida Agustín Escardino 7, 46980 Paterna, Valencia, Spain
| | - V Devesa
- Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Avenida Agustín Escardino 7, 46980 Paterna, Valencia, Spain
| | - D Vélez
- Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Avenida Agustín Escardino 7, 46980 Paterna, Valencia, Spain.
| |
Collapse
|
96
|
Cai Y, Xu C, Chen P, Hu J, Hu R, Huang M, Bi H. Development, validation, and application of a novel 7-day Caco-2 cell culture system. J Pharmacol Toxicol Methods 2014; 70:175-81. [DOI: 10.1016/j.vascn.2014.07.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/05/2014] [Accepted: 07/08/2014] [Indexed: 11/26/2022]
|
97
|
Júnior MADR, de Faria ACM, Velozo EDS, Dalla Costa T, de Andrade FP, de Castro WV. Determination of fexofenadine in Hank's balanced salt solution by high-performance liquid chromatography with ultraviolet detection: application to Caco-2 cell permeability studies. Biomed Chromatogr 2014; 29:537-44. [DOI: 10.1002/bmc.3310] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 06/26/2014] [Accepted: 07/22/2014] [Indexed: 01/18/2023]
|
98
|
Karim FT, Kalam A, Anwar R, Miah MM, Rahman MS, Islam SMA. Preparation and evaluation of SEDDS of simvastatin byin vivo, in vitroandex vivotechnique. Drug Dev Ind Pharm 2014; 41:1338-42. [DOI: 10.3109/03639045.2014.950271] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
99
|
Ilboudo S, Fouche E, Rizzati V, Toé AM, Gamet-Payrastre L, Guissou PI. In vitro impact of five pesticides alone or in combination on human intestinal cell line Caco-2. Toxicol Rep 2014; 1:474-489. [PMID: 28962261 PMCID: PMC5598529 DOI: 10.1016/j.toxrep.2014.07.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 07/01/2014] [Accepted: 07/14/2014] [Indexed: 01/24/2023] Open
Abstract
In Burkina Faso, as in most Sahelian countries, the failure to follow good agricultural practices coupled with poor soil and climate conditions in the locust control context lead to high environmental contaminations with pesticide residues. Thus, consumers being orally exposed to a combination of multiple pesticide residues through food and water intake, the digestive tract is a tissue susceptible to be directly exposed to these food contaminants. The aim of our work was to compare in vitro the impact of five desert locust control pesticides (Deltamethrin DTM, Fenitrothion FNT, Fipronil FPN, Lambda-cyalothrine LCT, and Teflubenzuron TBZ) alone and in combination on the human intestinal Caco-2 cells viability and function. Cells were exposed to 0.1–100 μM pesticides for 10 days alone or in mixture (MIX). Our results showed a cytotoxic effect of DTM, FNT, FPN, LCT, and TBZ alone or in combination in human intestinal Caco-2 cells. The most efficient were shown to be FPN and FNT impacting the cell layer integrity and/or barrier function, ALP activity, antioxidant enzyme activity, lipid peroxidation, Akt activation, and apoptosis. The presence of antioxidant reduced lipid peroxidation level and attenuated the pesticides-induced cell toxicity, suggesting that key mechanism of pesticides cytotoxicity may be linked to their pro-oxidative potential. A comparative analysis with the predicted cytotoxic effect of pesticides mixture using mathematical modeling shown that the combination of these pesticides led to synergistic effects rather than to a simple independent or dose addition effect.
Collapse
Affiliation(s)
- Sylvain Ilboudo
- INRA UMR 1331Toxalim (Research centre in food Toxicology), 180 Chemin de Tournefeuille, F-31027 Toulouse, France.,Institut de Recherche en Science de la Santé (IRSS/CNRST), 03, BP 7192, Ouagadougou, Burkina Faso.,Laboratoire de Toxicologie, Environnement et Santé; Ecole Doctorale de la Santé, Université de Ouagadougou, 03, BP 7021, Ouagadougou, Burkina Faso
| | - Edwin Fouche
- INRA UMR 1331Toxalim (Research centre in food Toxicology), 180 Chemin de Tournefeuille, F-31027 Toulouse, France
| | - Virginie Rizzati
- INRA UMR 1331Toxalim (Research centre in food Toxicology), 180 Chemin de Tournefeuille, F-31027 Toulouse, France
| | - Adama M Toé
- Institut de Recherche en Science de la Santé (IRSS/CNRST), 03, BP 7192, Ouagadougou, Burkina Faso
| | - Laurence Gamet-Payrastre
- INRA UMR 1331Toxalim (Research centre in food Toxicology), 180 Chemin de Tournefeuille, F-31027 Toulouse, France
| | - Pierre I Guissou
- Institut de Recherche en Science de la Santé (IRSS/CNRST), 03, BP 7192, Ouagadougou, Burkina Faso.,Laboratoire de Toxicologie, Environnement et Santé; Ecole Doctorale de la Santé, Université de Ouagadougou, 03, BP 7021, Ouagadougou, Burkina Faso
| |
Collapse
|
100
|
Eum SY, Jaraki D, Bertrand L, András IE, Toborek M. Disruption of epithelial barrier by quorum-sensing N-3-(oxododecanoyl)-homoserine lactone is mediated by matrix metalloproteinases. Am J Physiol Gastrointest Liver Physiol 2014; 306:G992-G1001. [PMID: 24742991 PMCID: PMC4042118 DOI: 10.1152/ajpgi.00016.2014] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The intestinal epithelium forms a selective barrier maintained by tight junctions (TJs) and separating the luminal environment from the submucosal tissues. N-acylhomoserine lactone (AHL) quorum-sensing molecules produced by gram-negative bacteria in the gut can influence homeostasis of the host intestinal epithelium. In the present study, we evaluated the regulatory mechanisms affecting the impact of two representative long- and short-chain AHLs, N-3-(oxododecanoyl)-homoserine lactone (C12-HSL) and N-butyryl homoserine lactone (C4-HSL), on barrier function of human intestinal epithelial Caco-2 cells. Treatment with C12-HSL, but not with C4-HSL, perturbed Caco-2 barrier function; the effect was associated with decreased levels of the TJ proteins occludin and tricellulin and their delocalization from the TJs. C12-HSL also induced matrix metalloprotease (MMP)-2 and MMP-3 activation via lipid raft- and protease-activated receptor (PAR)-dependent signaling. Pretreatment with lipid raft disruptors, PAR antagonists, or MMP inhibitors restored the C12-HSL-induced loss of the TJ proteins and increased permeability of Caco-2 cell monolayers. These results indicate that PAR/lipid raft-dependent MMP-2 and -3 activation followed by degradation of occludin and tricellulin are involved in C12-HSL-induced alterations of epithelial paracellular barrier functions.
Collapse
Affiliation(s)
- Sung Yong Eum
- 1Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Dima Jaraki
- 1Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Luc Bertrand
- 1Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Ibolya E. András
- 1Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida; and Jerzy Kukuczka Academy of Physical Education, Katowice, Poland
| |
Collapse
|