1
|
Ramirez-Velez I, Namjoshi AA, Effiong UM, Peppas NA, Belardi B. Paracellular Delivery of Protein Drugs with Smart EnteroPatho Nanoparticles. ACS NANO 2024. [PMID: 39096293 DOI: 10.1021/acsnano.4c02116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/05/2024]
Abstract
A general platform for the safe and effective oral delivery of biologics would revolutionize the administration of protein-based drugs, improving access for patients and lowering the financial burden on the health-care industry. Because of their dimensions and physiochemical properties, nanomaterials stand as promising vehicles for navigating the complex and challenging environment in the gastrointestinal (GI) tract. Recent developments have led to materials that protect protein drugs from degradation and enable controlled release in the small intestine, the site of absorption for most proteins. Yet, once present in the small intestine, the protein must transit through the secreted mucus and epithelial cells of the intestinal mucosa into systemic circulation, a process that remains a bottleneck for nanomaterial-based delivery. One attractive pathway through the intestinal mucosa is the paracellular route, which avoids cell trafficking and other degradative processes in the interior of cells. Direct flux between cells is regulated by epithelial tight junctions (TJs) that seal the paracellular space and prevent protein flux. Here, we describe a smart nanoparticle system that directly and transiently disrupts TJs for improved protein delivery, an unrealized goal to-date. We take inspiration from enteropathogenic bacteria that adhere to intestinal epithelia and secrete inhibitors that block TJ interactions in the local environment. To mimic these natural mechanisms, we engineer nanoparticles (EnteroPatho NPs) that attach to the epithelial glycocalyx and release TJ modulators in response to the intestinal pH. We show that EnteroPatho NPs lead to TJ disruption and paracellular protein delivery, giving rise to a general platform for oral delivery.
Collapse
Affiliation(s)
- Isabela Ramirez-Velez
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Aditya A Namjoshi
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Unyime M Effiong
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Nicholas A Peppas
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
- Institute for Biomaterials, Drug Delivery and Regenerative Medicine, The University of Texas at Austin, Austin, Texas 78712, United States
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, Texas 78712, United States
- Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Brian Belardi
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
2
|
Przybylla R, Krohn M, Sellin ML, Frank M, Oswald S, Linnebacher M. Novel In Vitro Models for Cell Differentiation and Drug Transport Studies of the Human Intestine. Cells 2023; 12:2371. [PMID: 37830585 PMCID: PMC10572004 DOI: 10.3390/cells12192371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/14/2023] Open
Abstract
The most common in vitro model for absorption, distribution, metabolism, and excretion (ADME) purposes is currently the Caco-2 cell line. However, clear differences in gene and protein expression towards the small intestine and an, at best, fair prediction accuracy of intestinal drug absorption restrict the usefulness of a model for intestinal epithelial cells. To overcome these limitations, we evaluated a panel of low-passaged patient-derived colorectal cancer cell lines of the HROC collection concerning similarities to small intestinal epithelial cells and their potential to predict intestinal drug absorption. After initial screening of a larger panel, ten cell lines with confluent outgrowth and long-lasting barrier-forming potential were further characterized in close detail. Tight junctional complexes and microvilli structures were detected in all lines, anda higher degree of differentiation was observed in 5/10 cell lines. All lines expressed multiple transporter molecules, with the expression levels in three lines being close to those of small intestinal epithelial cells. Compared with the Caco-2 model, three HROC lines demonstrated both higher similarity to jejunal epithelial tissue cells and higher regulatory potential of relevant drug transporters. In summary, these lines would be better-suited human small intestinal epithelium models for basic and translational research, especially for ADME studies.
Collapse
Affiliation(s)
- Randy Przybylla
- Molecular Oncology and Immunotherapy, Clinic of General Surgery, Rostock University Medical Centre, 18057 Rostock, Germany; (R.P.); (M.K.)
| | - Mathias Krohn
- Molecular Oncology and Immunotherapy, Clinic of General Surgery, Rostock University Medical Centre, 18057 Rostock, Germany; (R.P.); (M.K.)
| | - Marie-Luise Sellin
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopedics, Rostock University Medical Centre, 18057 Rostock, Germany;
| | - Marcus Frank
- Medical Biology and Electron Microscopy Centre, 18057 Rostock, Germany;
- Department Life, Light and Matter, University of Rostock, 18059 Rostock, Germany
| | - Stefan Oswald
- Institute of Pharmacology and Toxicology, Rostock University Medical Centre, 18057 Rostock, Germany;
| | - Michael Linnebacher
- Molecular Oncology and Immunotherapy, Clinic of General Surgery, Rostock University Medical Centre, 18057 Rostock, Germany; (R.P.); (M.K.)
| |
Collapse
|
3
|
Ohura K, Nakada Y, Imai T. Bioconversion and P-gp-Mediated Transport of Depot Fluphenazine Prodrugs after Intramuscular Injection. J Pharm Sci 2023; 112:1975-1984. [PMID: 37019360 DOI: 10.1016/j.xphs.2023.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/26/2023] [Accepted: 03/26/2023] [Indexed: 04/07/2023]
Abstract
Fluphenazine (FPZ) decanoate, an ester-type prodrug formulated as a long-acting injection (LAI), is used in the treatment of schizophrenia. FPZ enanthate was also developed as an LAI formulation, but is no longer in use clinically because of the short elimination half-life of FPZ, the parent drug, after intramuscular injection. In the present study, the hydrolysis of FPZ prodrugs was evaluated in human plasma and liver to clarify the reason for this difference in elimination half-lives. FPZ prodrugs were hydrolyzed in human plasma and liver microsomes. The rate of hydrolysis of FPZ enanthate in human plasma and liver microsomes was 15-fold and 6-fold, respectively, faster than that of FPZ decanoate. Butyrylcholinesterase (BChE) and human serum albumin (HSA) present in human plasma, and two carboxylesterase (CES) isozymes, hCE1 and hCE2, expressed in ubiquitous organs including liver, were mainly responsible for the hydrolysis of FPZ prodrugs. FPZ prodrugs may not be bioconverted in human skeletal muscle at the injection site because of lack of expression of BChE and CESs in muscle. Interestingly, although FPZ was a poor substrate for human P-glycoprotein, FPZ caproate was a good substrate. In conclusion, it is suggested that the shorter elimination half-life of FPZ following administration of FPZ enanthate compared with FPZ decanoate can be attributed to the more rapid hydrolysis of FPZ enanthate by BChE, HSA and CESs.
Collapse
Affiliation(s)
- Kayoko Ohura
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan; Headquarters for Admissions and Education, Kumamoto University, Kumamoto, Japan
| | | | - Teruko Imai
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan; Daiichi University of Pharmacy, Fukuoka, Japan.
| |
Collapse
|
4
|
Son YS, Son N, Yu WD, Baek A, Park YJ, Lee MS, Lee SJ, Kim DS, Son MY. Particulate matter 10 exposure affects intestinal functionality in both inflamed 2D intestinal epithelial cell and 3D intestinal organoid models. Front Immunol 2023; 14:1168064. [PMID: 37435069 PMCID: PMC10331606 DOI: 10.3389/fimmu.2023.1168064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/30/2023] [Indexed: 07/13/2023] Open
Abstract
Background A growing body of evidence suggests that particulate matter (PM10) enters the gastrointestinal (GI) tract directly, causing the GI epithelial cells to function less efficiently, leading to inflammation and an imbalance in the gut microbiome. PM10 may, however, act as an exacerbation factor in patients with inflamed intestinal epithelium, which is associated with inflammatory bowel disease. Objective The purpose of this study was to dissect the pathology mechanism of PM10 exposure in inflamed intestines. Methods In this study, we established chronically inflamed intestinal epithelium models utilizing two-dimensional (2D) human intestinal epithelial cells (hIECs) and 3D human intestinal organoids (hIOs), which mimic in vivo cellular diversity and function, in order to examine the deleterious effects of PM10 in human intestine-like in vitro models. Results Inflamed 2D hIECs and 3D hIOs exhibited pathological features, such as inflammation, decreased intestinal markers, and defective epithelial barrier function. In addition, we found that PM10 exposure induced a more severe disturbance of peptide uptake in inflamed 2D hIECs and 3D hIOs than in control cells. This was due to the fact that it interferes with calcium signaling, protein digestion, and absorption pathways. The findings demonstrate that PM10-induced epithelial alterations contribute to the exacerbation of inflammatory disorders caused by the intestine. Conclusions According to our findings, 2D hIEC and 3D hIO models could be powerful in vitro platforms for the evaluation of the causal relationship between PM exposure and abnormal human intestinal functions.
Collapse
Affiliation(s)
- Ye Seul Son
- Department of Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Naeun Son
- Department of Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Bio-Molecular Science, Korea Research Institute of Bioscience and Biotechnology (KRIBB) School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Won Dong Yu
- Department of Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Bio-Molecular Science, Korea Research Institute of Bioscience and Biotechnology (KRIBB) School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Aruem Baek
- Department of Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Young-Jun Park
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Moo-Seung Lee
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Seon-Jin Lee
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Dae-Soo Kim
- Digital Biotech Innovation Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Mi-Young Son
- Department of Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Bio-Molecular Science, Korea Research Institute of Bioscience and Biotechnology (KRIBB) School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
5
|
Protective effects of Acetobacter ghanensis against gliadin toxicity in intestinal epithelial cells with immunoregulatory and gluten-digestive properties. Eur J Nutr 2023; 62:605-614. [PMID: 36175797 DOI: 10.1007/s00394-022-03015-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 09/22/2022] [Indexed: 11/04/2022]
Abstract
PURPOSE The aim of this study was to establish whether Acetobacter ghanensis, the probiotic characteristics of which were evaluated previously, attenuates gliadin-induced toxicity in intestinal epithelial cells with gluten-digestive and immunoregulatory properties. METHODS A co-culture model of human intestinal epithelial cell (Caco-2) monolayers on top of peripheral blood mononuclear cells (PBMCs) obtained from patients with celiac disease (CD) was established. The gluten-digestive properties of A. ghanensis were determined by checking bacterial growth in a medium containing gluten as the main nitrogen source. The mRNA levels of genes encoding TJ-associated proteins were measured by quantitative real-time PCR (qRT-PCR). The concentrations of IL-6 and TNFα were determined by enzyme-linked immunosorbent assay (ELISA). RESULTS We found that PT-gliadin disrupted intestinal barrier integrity by modulating the expression of TJ-associated genes encoding zonulin (increased by ~ 60%), zonula occludens-1 (ZO-1) (decreased by ~ 22%), and occludin (decreased by ~ 28%) in Caco-2 cells. Furthermore, PT-gliadin treatment in Caco-2 cells was associated with increased concentrations of IL-6 (~ 1.6-fold) and TNFα (~ twofold) from PBMCs. These modulatory effects of PT-gliadin, however, were suppressed when Caco-2 cells were subjected to A. ghanensis in the presence of PT-gliadin. As a factor underlying these protective effects, we showed that A. ghanensis could digest gluten peptides. CONCLUSIONS To our knowledge, the current study is the first to demonstrate that A. ghanensis improves intestinal barrier functions by attenuating the modulatory effects of PT-gliadin with immunoregulatory and gluten-digestive properties.
Collapse
|
6
|
Lopez-Escalera S, Wellejus A. Evaluation of Caco-2 and human intestinal epithelial cells as in vitro models of colonic and small intestinal integrity. Biochem Biophys Rep 2022; 31:101314. [PMID: 35873654 PMCID: PMC9304606 DOI: 10.1016/j.bbrep.2022.101314] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/12/2022] [Accepted: 07/12/2022] [Indexed: 11/29/2022] Open
Abstract
Although the colonic cell line Caco-2 is widely used as a model of the small intestinal barrier function, it has limitations such as overestimated transepithelial electrical resistance (TEER) compared to in vivo conditions. Therefore, we investigated Human Intestinal Epithelial Cells (HIECs) as an alternative in vitro model. We explored whether cell seeding number of HIEC-6, and the number of incubation days for HIEC and Caco-2 cells had an impact on TEER, and tight junction expression was examined for both cell lines via immunofluorescence in the presence and absence of probiotic bacteria. We observed no significant difference in TEER readings for either cell lines when cultured for different days. Further, the HIEC TEER readings did not change with increased seeding number and were not significantly different from a control with no cells. HIECs expressed Claudin-1 and Zonula Occludens-1 but not Occludin. Caco-2 co-culture with probiotic bacteria demonstrated a significant increase in TEER, particularly for the lactobacillus strains, whereas HIEC TEER did not respond to bacterial co-incubation. Our study shows that although HIECs express certain TJ proteins, a significant TEER was not observed, likely due to the embryonic origin of the cells, which limits the application of this cell line as a suitable model for small intestinal barrier function. Human embryonic intestinal epithelial cells cannot form a significant barrier. Contrary to Caco-2 cells HIECs do not express the tight junction protein occludin. Probiotic bacteria induce a tight barrier in Caco-2 cells but not in HIECs.
Collapse
Affiliation(s)
- Silvia Lopez-Escalera
- Human Health Research, Scientific Affairs, Chr. Hansen A/S, Bøge Alle 10-12, DK-2970, Hørsholm, Denmark
- Friedrich-Schiller Universität Jena, Fakultät für Biowissenschaften, Bachstraβe 18K, 07743, Jena, Germany
| | - Anja Wellejus
- Human Health Research, Scientific Affairs, Chr. Hansen A/S, Bøge Alle 10-12, DK-2970, Hørsholm, Denmark
- Corresponding author.
| |
Collapse
|
7
|
A Novel Curcumin-Mycophenolic Acid Conjugate Inhibited Hyperproliferation of Tumor Necrosis Factor-Alpha-Induced Human Keratinocyte Cells. Pharmaceutics 2021; 13:pharmaceutics13070956. [PMID: 34201974 PMCID: PMC8308932 DOI: 10.3390/pharmaceutics13070956] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/20/2021] [Accepted: 06/22/2021] [Indexed: 12/21/2022] Open
Abstract
Curcumin (CUR) has been used as adjuvant therapy for therapeutic application in the treatment of psoriasis through several mechanisms of action. Due to the poor oral bioavailability of CUR, several approaches have been developed to overcome the limitations of CUR, including the prodrug strategy. In this study, CUR was esterified with mycophenolic acid (MPA) as a novel conjugate prodrug. The MPA-CUR conjugate was structurally elucidated using FT-IR, 1H-NMR, 13C-NMR, and MS techniques. Bioavailable fractions (BFs) across Caco-2 cells of CUR, MPA, and MPA-CUR were collected for further biological activity evaluation representing an in vitro cellular transport model for oral administration. The antipsoriatic effect of the BFs was determined using antiproliferation and anti-inflammation assays against hyperproliferation of tumor necrosis factor-alpha (TNF-α)-induced human keratinocytes (HaCaT). The BF of MPA-CUR provided better antiproliferation than that of CUR (p < 0.001). The enhanced hyperproliferation suppression of the BF of MPA-CUR resulted from the reduction of several inflammatory cytokines, including IL-6, IL-8, and IL-1β. The molecular mechanisms of anti-inflammatory activity were mediated by an attenuated signaling cascade of MAPKs protein, i.e., p38, ERK, and JNK. Our results present evidence for the MPA-CUR conjugate as a promising therapeutic agent for treating psoriasis by antiproliferative and anti-inflammatory actions.
Collapse
|
8
|
Ohta Y, Kazuki K, Abe S, Oshimura M, Kobayashi K, Kazuki Y. Development of Caco-2 cells expressing four CYPs via a mammalian artificial chromosome. BMC Biotechnol 2020; 20:44. [PMID: 32819341 PMCID: PMC7441628 DOI: 10.1186/s12896-020-00637-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 08/10/2020] [Indexed: 12/30/2022] Open
Abstract
Background Oral administration is the most common way to deliver drugs to the systemic circulation or target organs. Orally administered drugs are absorbed in the intestine and metabolized in the intestine and liver. In the early stages of drug development, it is important to predict first-pass metabolism accurately to select candidate drugs with high bioavailability. The Caco-2 cell line derived from colorectal cancer is widely used as an intestinal model to assess drug membrane permeability. However, because the expression of major drug-metabolizing enzymes, such as cytochrome P450 (CYP), is extremely low in Caco-2 cells, it is difficult to predict intestinal metabolism, which is a significant factor in predicting oral drug bioavailability. Previously, we constructed a mouse artificial chromosome vector carrying the CYP (CYP2C9, CYP2C19, CYP2D6, and CYP3A4) and P450 oxidoreductase (POR) (4CYPs-MAC) genes and increased CYP expression and metabolic activity in HepG2 cells via transfer of this vector. Results In the current study, to improve the Caco-2 cell assay model by taking metabolism into account, we attempted to increase CYP expression by transferring the 4CYPs-MAC into Caco-2 cells. The Caco-2 cells carrying the 4CYPs-MAC showed higher CYP mRNA expression and activity. In addition, high metabolic activity, availability for permeation test, and the potential to assess drug–drug interactions were confirmed. Conclusions The established Caco-2 cells with the 4CYPs-MAC are expected to enable more accurate prediction of the absorption and metabolism in the human intestine than parental Caco-2 cells. The mammalian artificial chromosome vector system would provide useful models for drug development.
Collapse
Affiliation(s)
- Yumi Ohta
- Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori, 683-8503, Japan
| | - Kanako Kazuki
- Chromosome Engineering Research Center (CERC), Tottori University, 86 Nishi-cho, Yonago, Tottori, 683-8503, Japan
| | - Satoshi Abe
- Trans Chromosomics, Inc, 86 Nishi-cho, Yonago, Tottori, 683-8503, Japan
| | - Mitsuo Oshimura
- Trans Chromosomics, Inc, 86 Nishi-cho, Yonago, Tottori, 683-8503, Japan
| | - Kaoru Kobayashi
- Laboratory of Biopharmaceutics, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Yasuhiro Kazuki
- Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori, 683-8503, Japan. .,Chromosome Engineering Research Center (CERC), Tottori University, 86 Nishi-cho, Yonago, Tottori, 683-8503, Japan.
| |
Collapse
|
9
|
Hori T, Kurosawa O, Ishihara K, Mizuta T, Iwata H. Three-Dimensional Cell Sheet Construction Method with a Polyester Micromesh Sheet. Tissue Eng Part C Methods 2020; 26:170-179. [PMID: 32186996 DOI: 10.1089/ten.tec.2019.0330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cell sheet engineering has become important in a variety of fields, including regenerative medicine and transplantation. Our research group previously developed micromesh cultures that enable cells to form a cell sheet on a microstructured mesh sheet. Here, we present a more usable micromesh culture and devices that make it possible, aiming for widespread use. The devices are mainly constituted of a polyester micromesh sheet and three-dimensional (3D)-printed simple frames that fix the mesh sheet on it. Cells such as fibroblast Tig-1-20 cells, hepatoma HepG2 cells, or mesenchymal stem cells (MSC) were easily seeded on the polyester mesh sheet in the device and cultured for 16 days, which was followed by the formation of a 100-400-μm-thick cell sheet. The cell sheet was very robust, easy to handle, and could be readily removed from the device for subsequent analysis. Optical coherence tomography revealed the structure of the cell sheet as having the mesh sheet layer in the center of the cell sheet. Confocal microscopy demonstrated that Tig-1-20 cells in the cell sheet were aligned according to the shape of the mesh apertures, indicating that cell orientation can be controlled with this micromesh culture. As for another application, the device was used to construct a multilayered cell sheet that consists of three different types of cells. Furthermore, for mass production, the device frames were made using polyoxymethylene (POM) instead of 3D printing materials. Using the POM devices, a large MSC sheet for 10 cm dishes was successfully produced 7 days after cell seeding. This micromesh culture may become one of the useful cell sheet construction methods in future for medical and research fields. Impact statement Currently, cell sheets are constructed, for example, on a temperature-responsive polymer-coated dish or a porous membrane. These cell sheets are widely used but are not completely suitable in terms of robustness, ease of handling, cost, ease of microscopic cell observation, or nutrient supply. We previously reported that the micromesh culture can provide a three-dimensional (3D) cell sheet that has advantages for cell observation and nutrient supply. In this study, the micromesh culture was enhanced with a polyester micromesh sheet and a series of devices of polyoxymethylene, helping us to produce a robust, cost-effective, easily layered, and easy-to-use 3D cell sheet.
Collapse
Affiliation(s)
- Takeshi Hori
- Compass to Healthy Life Research Complex Program, RIKEN, Kobe, Japan
| | - Osamu Kurosawa
- Compass to Healthy Life Research Complex Program, RIKEN, Kobe, Japan
| | | | | | - Hiroo Iwata
- Compass to Healthy Life Research Complex Program, RIKEN, Kobe, Japan
| |
Collapse
|
10
|
Abstract
The first-pass hydrolysis of oral ester-type prodrugs in the liver and intestine is mediated mainly by hCE1 and hCE2 of the respective predominant carboxylesterase (CES) isozymes. In order to provide high blood concentrations of the parent drugs, it is preferable that prodrugs are absorbed as an intact ester in the intestine, then rapidly converted to active parent drugs by hCE1 in the liver. In the present study, we designed a prodrug of fexofenadine (FXD) as a model parent drug that is resistant to hCE2 but hydrolyzed by hCE1, utilizing the differences in catalytic characteristics of hCE1 and hCE2. In order to precisely predict the intestinal absorption of an FXD prodrug candidate, we developed a novel high-throughput system by modifying Caco-2 cells. Further, we evaluated species differences and aging effects in the intestinal and hepatic hydrolysis of prodrugs to improve the estimation of in vivo first-pass hydrolysis of ester-type prodrugs. Consequently, it was possible to design a hepatotropic prodrug utilizing the differences in tissue distribution and substrate specificity of CESs. In addition, we successfully established three useful in vitro systems for predicting the intestinal absorption of hCE1 substrate using Caco-2 cells. However, some factors involved in estimating the bioavailability of prodrugs in human, such as changes in recognition of drug transporters by esterification, and species differences of the first-pass hydrolysis, should be comprehensively considered in prodrug development.
Collapse
Affiliation(s)
- Kayoko Ohura
- Priority Organization for Innovation and Excellence, Kumamoto University
| |
Collapse
|
11
|
Gibiansky L, Ravva P, Parrott NJ, Bhardwaj R, Zwanziger E, Grimsey P, Clinch B, Sturm S. Mechanistic Population Pharmacokinetic Model of Oseltamivir and Oseltamivir Carboxylate Accounting for Physiological Changes to Predict Exposures in Neonates and Infants. Clin Pharmacol Ther 2020; 108:126-135. [PMID: 31957010 PMCID: PMC7325316 DOI: 10.1002/cpt.1791] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/04/2020] [Indexed: 11/12/2022]
Abstract
A mechanistic population‐pharmacokinetic model was developed to predict oseltamivir exposures in neonates and infants accounting for physiological changes during the first 2 years of life. The model included data from 13 studies, comprising 436 subjects with normal renal function (317 pediatric subjects (≥ 38 weeks postmenstrual age (PMA), ≥ 13 days old) and 119 adult subjects < 40 years). Concentration–time profiles of oseltamivir and its active metabolite, oseltamivir carboxylate (OC), were characterized by a four‐compartment model, with absorption described by three additional compartments. Renal maturational changes were implemented by description of OC clearance with allometric function of weight and Hill function of PMA. Clearance of OC increased with weight up to 43 kg (allometric coefficient 0.75). Half the adult OC clearance was reached at a PMA of 45.6 weeks (95% confidence interval (CI) 41.6–49.6) with a Hill coefficient of 2.35 (95% CI 1.67–3.04). The model supports the European Union/United States‐approved 3 mg/kg twice‐daily oseltamivir dose for infants < 1 year (PMA ≥ 38 weeks) and allows prediction of exposures in preterm neonates.
Collapse
Affiliation(s)
| | - Patanjali Ravva
- Roche Innovation Center New York, Roche Pharmaceutical Research and Early Development, New York, New York, USA.,Pfizer Inc, Global Clinical Pharmacology, New York, New York, USA
| | - Neil J Parrott
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Rajinder Bhardwaj
- Integrated Drug Development, Certara Strategic Consulting, Parsippany, New Jersey, USA
| | - Elke Zwanziger
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Paul Grimsey
- Roche Innovation Center Welwyn, Roche Pharmaceutical Research and Early Development, Welwyn Garden City, UK
| | - Barry Clinch
- Roche Products Limited, Product Development, Welwyn Garden City, UK
| | - Stefan Sturm
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| |
Collapse
|
12
|
Ishizaki Y, Furihata T, Oyama Y, Ohura K, Imai T, Hosokawa M, Akita H, Chiba K. Development of a Caco-2 Cell Line Carrying the Human Intestine-Type CES Expression Profile as a Promising Tool for Ester-Containing Drug Permeability Studies. Biol Pharm Bull 2018; 41:697-706. [DOI: 10.1248/bpb.b17-00880] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yuma Ishizaki
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University
| | - Tomomi Furihata
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University
- Department of Pharmacology, Graduate School of Medicine, Chiba University
| | - Yusuke Oyama
- Department of Metabolism-Based Drug Design and Delivery, Graduate School of Pharmaceutical Science, Kumamoto University
| | - Kayoko Ohura
- Department of Metabolism-Based Drug Design and Delivery, Graduate School of Pharmaceutical Science, Kumamoto University
| | - Teruko Imai
- Department of Metabolism-Based Drug Design and Delivery, Graduate School of Pharmaceutical Science, Kumamoto University
| | - Masakiyo Hosokawa
- Laboratory of Drug Metabolism and Biopharmaceutics, Faculty of Pharmaceutical Sciences, Chiba Institute of Science
| | - Hidetaka Akita
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University
| | - Kan Chiba
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University
| |
Collapse
|
13
|
Inhibitory Influence of Panax notoginseng Saponins on Aspirin Hydrolysis in Human Intestinal Caco-2 Cells. Molecules 2018; 23:molecules23020455. [PMID: 29463025 PMCID: PMC6016969 DOI: 10.3390/molecules23020455] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 02/15/2018] [Accepted: 02/15/2018] [Indexed: 01/18/2023] Open
Abstract
Herb-drug interactions are important safety concerns in clinical practice. The interactions occur firstly in the intestinal absorption for orally administered drugs. Aspirin and Panax notoginseng saponins (PNS)-based drugs are often combined in China to prevent larger-artery atherosclerosis. Here, we aimed to characterize the aspirin transport across Caco-2 cell monolayers, a model of the intestinal absorption, and further to evaluate the influence of PNS on aspirin hydrolysis and the relating mechanisms. Transcellular transport of aspirin and the influence of PNS were explored using Caco-2 cell monolayers. The protein expression of human carboxylesterase 1 (hCE1) and hCE2 in Caco-2 cells after PNS treatment was analyzed by ELISA, and the mRNA level were determined by qRT-PCR. In the study, Caco-2 cells showed high level of hydrolase activity, and most aspirin was hydrolyzed inside the cells during the transport process. Interestingly, PNS were demonstrated to inhibit the esterase activities responsible for aspirin hydrolysis in Caco-2 cells. PNS could also decrease the protein expression of hCE1 and hCE2, whereas exhibited minor effect on the mRNA expression. These results indicated that oral administration of PNS-based drugs might inhibit the hydrolysis of aspirin during intestinal absorption thus promoting its bioavailability.
Collapse
|