1
|
Damigos S, Caliskan A, Wajant G, Giddins S, Moldovan A, Kuhn S, Putz E, Dandekar T, Rudel T, Westermann AJ, Zdzieblo D. A Multicellular In Vitro Model of the Human Intestine with Immunocompetent Features Highlights Host-Pathogen Interactions During Early Salmonella Typhimurium Infection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2411233. [PMID: 39807570 DOI: 10.1002/advs.202411233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/24/2024] [Indexed: 01/16/2025]
Abstract
Studying the molecular basis of intestinal infections caused by enteric pathogens at the tissue level is challenging, because most human intestinal infection models have limitations, and results obtained from animals may not reflect the human situation. Infections with Salmonella enterica serovar Typhimurium (STm) have different outcomes between organisms. 3D tissue modeling of primary human material provides alternatives to animal experimentation, but epithelial co-culture with immune cells remains difficult. Macrophages, for instance, contribute to the immunocompetence of native tissue, yet their incorporation into human epithelial tissue models is challenging. A 3D immunocompetent tissue model of the human small intestine based on decellularized submucosa enriched with monocyte-derived macrophages (MDM) is established. The multicellular model recapitulated in vivo-like cellular diversity, especially the induction of GP2 positive microfold (M) cells. Infection studies with STm reveal that the pathogen physically interacts with these M-like cells. MDMs show trans-epithelial migration and phagocytosed STm within the model and the levels of inflammatory cytokines are induced upon STm infection. Infected epithelial cells are shed into the supernatant, potentially reflecting an intracellular reservoir of invasion-primed STm. Together, the 3D model of the human intestinal epithelium bears potential as an alternative to animals to identify human-specific processes underlying enteric bacterial infections.
Collapse
Affiliation(s)
- Spyridon Damigos
- Department for Functional Materials in Medicine and Dentistry, University Hospital Würzburg, Würzburg, Germany
| | - Aylin Caliskan
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Gisela Wajant
- Department for Functional Materials in Medicine and Dentistry, University Hospital Würzburg, Würzburg, Germany
| | - Sara Giddins
- Department for Functional Materials in Medicine and Dentistry, University Hospital Würzburg, Würzburg, Germany
| | - Adriana Moldovan
- Department of Microbiology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Sabine Kuhn
- Institute of Clinical Transfusion Medicine and Hemotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Evelyn Putz
- Institute of Clinical Transfusion Medicine and Hemotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Thomas Dandekar
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Thomas Rudel
- Department of Microbiology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Alexander J Westermann
- Department of Microbiology, Biocenter, University of Würzburg, Würzburg, Germany
- Helmholtz-Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Daniela Zdzieblo
- Department for Functional Materials in Medicine and Dentistry, University Hospital Würzburg, Würzburg, Germany
- Translational Center for Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research (ISC), 97070, Würzburg, Germany
| |
Collapse
|
2
|
Masete KV, Günzel D, Schulzke JD, Epple HJ, Hering NA. Matrix-free human 2D organoids recapitulate duodenal barrier and transport properties. BMC Biol 2025; 23:2. [PMID: 39757172 DOI: 10.1186/s12915-024-02105-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Traditionally, transformed cell line monolayers have been the standard model for studying epithelial barrier and transport function. Recently, intestinal organoids were proposed as superior in recapitulating the intestine. Typically, 3D organoids are digested and seeded as monolayers on gelatinous matrix pre-coated surfaces for anchorage. As this coat could potentially act as a diffusion barrier, we aimed to generate robust human duodenum-derived organoid monolayers that do not need a gelatinous matrix for anchorage to improve upon existing models to study epithelial transport and barrier function. RESULTS We characterized these monolayers phenotypically regarding polarization, tight junction formation and cellular composition, and functionally regarding uptake of nutrients, ion transport and cytokine-induced barrier dysfunction. The organoid monolayers recapitulated the duodenum phenotypically as well as functionally regarding glucose and short-chain fatty acid uptake. Tumour necrosis factor-alpha induced paracellular transport of 4-kDa Dextran and transcytosis of 44-kDa horseradish peroxidase. Notably, forskolin-stimulated chloride secretion was consistently lower when organoid monolayers were seeded on a layer of basement membrane extract (BME). CONCLUSIONS BME-free organoid monolayers represent an improved model for studying transcytotic, paracellular but especially transcellular transport. As BME is extracted from mice, our model furthers efforts to make organoid culture more animal-free.
Collapse
Affiliation(s)
- Kopano Valerie Masete
- Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Dorothee Günzel
- Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Jörg-Dieter Schulzke
- Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Hans-Jörg Epple
- Department of Gastroenterology, Rheumatology and Infectious Diseases, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
- Antibiotic Stewardship Team, Medical Directorate, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Nina A Hering
- Department of General and Visceral Surgery, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Hindenburgdamm 30, Berlin, 12203, Germany.
| |
Collapse
|
3
|
Zhu Z, Cheng Y, Liu X, Ding W, Liu J, Ling Z, Wu L. Advances in the Development and Application of Human Organoids: Techniques, Applications, and Future Perspectives. Cell Transplant 2025; 34:9636897241303271. [PMID: 39874083 PMCID: PMC11775963 DOI: 10.1177/09636897241303271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 10/10/2024] [Accepted: 11/11/2024] [Indexed: 01/30/2025] Open
Abstract
Organoids are three-dimensional (3D) cell cultures derived from human pluripotent stem cells or adult stem cells that recapitulate the cellular heterogeneity, structure, and function of human organs. These microstructures are invaluable for biomedical research due to their ability to closely mimic the complexity of native tissues while retaining human genetic material. This fidelity to native organ systems positions organoids as a powerful tool for advancing our understanding of human biology and for enhancing preclinical drug testing. Recent advancements have led to the successful development of a variety of organoid types, reflecting a broad range of human organs and tissues. This progress has expanded their application across several domains, including regenerative medicine, where organoids offer potential for tissue replacement and repair; disease modeling, which allows for the study of disease mechanisms and progression in a controlled environment; drug discovery and evaluation, where organoids provide a more accurate platform for testing drug efficacy and safety; and microecological research, where they contribute to understanding the interactions between microbes and host tissues. This review provides a comprehensive overview of the historical development of organoid technology, highlights the key achievements and ongoing challenges in the field, and discusses the current and emerging applications of organoids in both laboratory research and clinical practice.
Collapse
Affiliation(s)
- Zhangcheng Zhu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Yiwen Cheng
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xia Liu
- Department of Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wenwen Ding
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Jiaming Liu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Zongxin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lingbin Wu
- Department of Laboratory Medicine, Lishui Second People’s Hospital, Lishui, China
| |
Collapse
|
4
|
Song H, Hong Y, Lee H. Rapid automated production of tubular 3D intestine-on-a-chip with diverse cell types using coaxial bioprinting. LAB ON A CHIP 2024; 25:90-101. [PMID: 39648875 DOI: 10.1039/d4lc00731j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Despite considerable animal sacrifices and investments, drug development often falters in clinical trials due to species differences. To address this issue, specific in vitro models, such as organ-on-a-chip technology using human cells in microfluidic devices, are recognized as promising alternatives. Among the various organs, the human small intestine plays a pivotal role in drug development, particularly in the assessment of digestion and nutrient absorption. However, current intestine-on-a-chip devices struggle to accurately replicate the complex 3D tubular structures of the human small intestine, particularly when it comes to integrating a variety of cell types effectively. This limitation is primarily due to conventional fabrication methods, such as soft lithography and replica molding. In this research, we introduce a novel coaxial bioprinting method to construct 3D tubular structures that closely emulate the organization and functionality of the small intestine with multiple cell types. To ensure stable production of these small intestine-like tubular structures, we analyzed the rheological properties of bioinks to select the most suitable materials for coaxial bioprinting technology. Additionally, we conducted biological assessments to validate the gene expression patterns and functional attributes of the 3D intestine-on-a-chip. Our 3D intestine-on-a-chip, which faithfully replicates intestinal functions and organization, demonstrates clear superiority in both structure and biological function compared to the conventional 2D model. This innovative approach holds significant promise for a wide range of future applications.
Collapse
Affiliation(s)
- Heeju Song
- Department of Smart Health Science and Technology, Kangwon National University, Chuncheon, Republic of Korea.
| | - Yeonjin Hong
- Department of Smart Health Science and Technology, Kangwon National University, Chuncheon, Republic of Korea.
| | - Hyungseok Lee
- Department of Smart Health Science and Technology, Kangwon National University, Chuncheon, Republic of Korea.
- Department of Mechanical and Biomedical, Mechatronics Engineering, Kangwon National University, 1, Kangwondaehak-gil, Chuncheon-si, Gangwon-do, Republic of Korea
| |
Collapse
|
5
|
Königer L, Malkmus C, Mahdy D, Däullary T, Götz S, Schwarz T, Gensler M, Pallmann N, Cheufou D, Rosenwald A, Möllmann M, Groneberg D, Popp C, Groeber‐Becker F, Steinke M, Hansmann J. ReBiA-Robotic Enabled Biological Automation: 3D Epithelial Tissue Production. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406608. [PMID: 39324843 PMCID: PMC11615785 DOI: 10.1002/advs.202406608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/08/2024] [Indexed: 09/27/2024]
Abstract
The Food and Drug Administration's recent decision to eliminate mandatory animal testing for drug approval marks a significant shift to alternative methods. Similarly, the European Parliament is advocating for a faster transition, reflecting public preference for animal-free research practices. In vitro tissue models are increasingly recognized as valuable tools for regulatory assessments before clinical trials, in line with the 3R principles (Replace, Reduce, Refine). Despite their potential, barriers such as the need for standardization, availability, and cost hinder their widespread adoption. To address these challenges, the Robotic Enabled Biological Automation (ReBiA) system is developed. This system uses a dual-arm robot capable of standardizing laboratory processes within a closed automated environment, translating manual processes into automated ones. This reduces the need for process-specific developments, making in vitro tissue models more consistent and cost-effective. ReBiA's performance is demonstrated through producing human reconstructed epidermis, human airway epithelial models, and human intestinal organoids. Analyses confirm that these models match the morphology and protein expression of manually prepared and native tissues, with similar cell viability. These successes highlight ReBiA's potential to lower barriers to broader adoption of in vitro tissue models, supporting a shift toward more ethical and advanced research methods.
Collapse
Affiliation(s)
- Lukas Königer
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISC97070WürzburgGermany
| | - Christoph Malkmus
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISC97070WürzburgGermany
- Institute of Medical Engineering SchweinfurtTechnical University of Applied Sciences Würzburg‐Schweinfurt97421SchweinfurtGermany
| | - Dalia Mahdy
- Chair of Tissue Engineering and Regenerative MedicineUniversity Hospital Würzburg97070WürzburgGermany
| | - Thomas Däullary
- Chair of Tissue Engineering and Regenerative MedicineUniversity Hospital Würzburg97070WürzburgGermany
- Chair of Cellular ImmunotherapyUniversity Hospital Würzburg97080WürzburgGermany
| | - Susanna Götz
- Faculty of Design WürzburgTechnical University of Applied Sciences Würzburg‐Schweinfurt97070WürzburgGermany
| | - Thomas Schwarz
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISC97070WürzburgGermany
| | - Marius Gensler
- Chair of Tissue Engineering and Regenerative MedicineUniversity Hospital Würzburg97070WürzburgGermany
| | - Niklas Pallmann
- Chair of Tissue Engineering and Regenerative MedicineUniversity Hospital Würzburg97070WürzburgGermany
| | - Danjouma Cheufou
- Department of Thoracic SurgeryKlinikum Würzburg Mitte97070WürzburgGermany
| | | | - Marc Möllmann
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISC97070WürzburgGermany
| | - Dieter Groneberg
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISC97070WürzburgGermany
| | - Christina Popp
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISC97070WürzburgGermany
| | - Florian Groeber‐Becker
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISC97070WürzburgGermany
- Department of OphthalmologyUniversity Clinic Düsseldorf40225DüsseldorfGermany
| | - Maria Steinke
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISC97070WürzburgGermany
- Department of Oto‐Rhino‐LaryngologyPlasticAesthetic and Reconstructive Head and Neck SurgeryUniversity Hospital Würzburg97080WürzburgGermany
| | - Jan Hansmann
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISC97070WürzburgGermany
- Institute of Medical Engineering SchweinfurtTechnical University of Applied Sciences Würzburg‐Schweinfurt97421SchweinfurtGermany
| |
Collapse
|
6
|
Freer M, Cooper J, Goncalves K, Przyborski S. Bioengineering the Human Intestinal Mucosa and the Importance of Stromal Support for Pharmacological Evaluation In Vitro. Cells 2024; 13:1859. [PMID: 39594608 PMCID: PMC11592477 DOI: 10.3390/cells13221859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Drug discovery is associated with high levels of compound elimination in all stages of development. The current practices for the pharmacokinetic testing of intestinal absorption combine Transwell® inserts with the Caco-2 cell line and are associated with a wide range of limitations. The improvement of pharmacokinetic research relies on the development of more advanced in vitro intestinal constructs that better represent human native tissue and its response to drugs, providing greater predictive accuracy. Here, we present a humanized, bioengineered intestinal construct that recapitulates aspects of intestinal microanatomy. We present improved histotypic characteristics reminiscent of the human intestine, such as a reduction in transepithelial electrical resistance (TEER) and the formation of a robust basement membrane, which are contributed to in-part by a strong stromal foundation. We explore the link between stromal-epithelial crosstalk, paracrine communication, and the role of the keratinocyte growth factor (KGF) as a soluble mediator, underpinning the tissue-specific role of fibroblast subpopulations. Permeability studies adapted to a 96-well format allow for high throughput screening and demonstrate the role of the stromal compartment and tissue architecture on permeability and functionality, which is thought to be one of many factors responsible for unexpected drug outcomes using current approaches for pharmacokinetic testing.
Collapse
Affiliation(s)
- Matthew Freer
- Department of Biosciences, Durham University, Durham DH1 3LE, UK;
| | - Jim Cooper
- European Collection of Authenticated Cell Cultures, Salisbury SP4 0JG, UK;
| | - Kirsty Goncalves
- Department of Biosciences, Durham University, Durham DH1 3LE, UK;
| | - Stefan Przyborski
- Department of Biosciences, Durham University, Durham DH1 3LE, UK;
- Reprocell Europe Ltd., Glasgow G20 0XA, UK
| |
Collapse
|
7
|
Jung N, Schreiner J, Baur F, Vogel-Kindgen S, Windbergs M. Predicting nanocarrier permeation across the human intestine in vitro: model matters. Biomater Sci 2024; 12:5775-5788. [PMID: 39402906 DOI: 10.1039/d4bm01092b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2024]
Abstract
For clinical translation of oral nanocarriers, simulation of the intestinal microenvironment during in vitro testing is crucial to evaluate interactions with the intestinal mucosa. However, studies are often conducted using simplistic cell culture models, overlooking key physiological factors, and potentially leading to an overestimation of nanocarrier permeation. In this study, we systematically investigate different tissue models of the human intestine under static cultivation and dynamic flow conditions and analyze the impact of altered tissue characteristics on nanocarrier permeation. Our results reveal that the selection of cell types as well as the respective culture condition have a notable impact on the physiological characteristics of the resulting tissues. Tissue layer thickness, mucus secretion, and barrier impairment, all increase with increasing amounts of goblet cells and the application of dynamic flow conditions. Permeation studies with poly(lactic-co-glycolic acid) (PLGA) nanocarriers with and without polyethylene glycol (PEG) coating elucidate that the amount of mucus present in the respective model is the limiting factor for the permeation of PLGA nanocarriers, while tissue topography presents the key factor influencing PEG-PLGA nanocarrier permeation. Furthermore, both nanocarriers exhibit diametrically opposite permeation kinetics compared to soluble compounds. In summary, these findings reveal the critical role of the implemented test systems on permeation assessment and emphasize that, in the context of preclinical nanocarrier testing, the choice of in vitro model matters.
Collapse
Affiliation(s)
- Nathalie Jung
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany.
| | - Jonas Schreiner
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany.
| | - Florentin Baur
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany.
| | - Sarah Vogel-Kindgen
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany.
| | - Maike Windbergs
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
8
|
Teo AJT, Ng SK, Khoo K, Wong SH, Li KHH. Microfluidic Gastrointestinal Cell Culture Technologies-Improvements in the Past Decade. BIOSENSORS 2024; 14:449. [PMID: 39329824 PMCID: PMC11429516 DOI: 10.3390/bios14090449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024]
Abstract
Gastrointestinal cell culture technology has evolved in the past decade with the integration of microfluidic technologies, bringing advantages with greater selectivity and cost effectiveness. Herein, these technologies are sorted into three categories, namely the cell-culture insert devices, conventional microfluidic devices, and 3D-printed microfluidic devices. Each category is discussed in brief with improvements also discussed here. Introduction of different companies and applications derived from each are also provided to encourage uptake. Subsequently, future perspectives of integrating microfluidics with trending topics like stool-derived in vitro communities and gut-immune-tumor axis investigations are discussed. Insights on modular microfluidics and its implications on gastrointestinal cell cultures are also discussed here. Future perspectives on point-of-care (POC) applications in relations to gastrointestinal microfluidic devices are also discussed here. In conclusion, this review presents an introduction of each microfluidic platform with an insight into the greater contribution of microfluidics in gastrointestinal cell cultures.
Collapse
Affiliation(s)
- Adrian J. T. Teo
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Singapore; (A.J.T.T.); (K.K.)
| | - Siu-Kin Ng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (S.-K.N.); (S.H.W.)
| | - Kaydeson Khoo
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Singapore; (A.J.T.T.); (K.K.)
| | - Sunny Hei Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (S.-K.N.); (S.H.W.)
- Department of Gastroenterology and Hepatology, Tan Tock Seng Hospital, Singapore 308433, Singapore
| | - King Ho Holden Li
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Singapore; (A.J.T.T.); (K.K.)
| |
Collapse
|
9
|
Almalla A, Alzain N, Elomaa L, Richter F, Scholz J, Lindner M, Siegmund B, Weinhart M. Hydrogel-Integrated Millifluidic Systems: Advancing the Fabrication of Mucus-Producing Human Intestinal Models. Cells 2024; 13:1080. [PMID: 38994934 PMCID: PMC11240340 DOI: 10.3390/cells13131080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 07/13/2024] Open
Abstract
The luminal surface of the intestinal epithelium is protected by a vital mucus layer, which is essential for lubrication, hydration, and fostering symbiotic bacterial relationships. Replicating and studying this complex mucus structure in vitro presents considerable challenges. To address this, we developed a hydrogel-integrated millifluidic tissue chamber capable of applying precise apical shear stress to intestinal models cultured on flat or 3D structured hydrogel scaffolds with adjustable stiffness. The chamber is designed to accommodate nine hydrogel scaffolds, 3D-printed as flat disks with a storage modulus matching the physiological range of intestinal tissue stiffness (~3.7 kPa) from bioactive decellularized and methacrylated small intestinal submucosa (dSIS-MA). Computational fluid dynamics simulations were conducted to confirm a laminar flow profile for both flat and 3D villi-comprising scaffolds in the physiologically relevant regime. The system was initially validated with HT29-MTX seeded hydrogel scaffolds, demonstrating accelerated differentiation, increased mucus production, and enhanced 3D organization under shear stress. These characteristic intestinal tissue features are essential for advanced in vitro models as they critically contribute to a functional barrier. Subsequently, the chamber was challenged with human intestinal stem cells (ISCs) from the terminal ileum. Our findings indicate that biomimicking hydrogel scaffolds, in combination with physiological shear stress, promote multi-lineage differentiation, as evidenced by a gene and protein expression analysis of basic markers and the 3D structural organization of ISCs in the absence of chemical differentiation triggers. The quantitative analysis of the alkaline phosphatase (ALP) activity and secreted mucus demonstrates the functional differentiation of the cells into enterocyte and goblet cell lineages. The millifluidic system, which has been developed and optimized for performance and cost efficiency, enables the creation and modulation of advanced intestinal models under biomimicking conditions, including tunable matrix stiffness and varying fluid shear stresses. Moreover, the readily accessible and scalable mucus-producing cellular tissue models permit comprehensive mucus analysis and the investigation of pathogen interactions and penetration, thereby offering the potential to advance our understanding of intestinal mucus in health and disease.
Collapse
Affiliation(s)
- Ahed Almalla
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany (N.A.); (L.E.); (F.R.); (J.S.); (M.L.)
| | - Nadra Alzain
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany (N.A.); (L.E.); (F.R.); (J.S.); (M.L.)
- Department of Gastroenterology, Infectious Diseases and Rheumatology (Including Nutrition Medicine), Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany;
| | - Laura Elomaa
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany (N.A.); (L.E.); (F.R.); (J.S.); (M.L.)
| | - Fiona Richter
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany (N.A.); (L.E.); (F.R.); (J.S.); (M.L.)
| | - Johanna Scholz
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany (N.A.); (L.E.); (F.R.); (J.S.); (M.L.)
| | - Marcus Lindner
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany (N.A.); (L.E.); (F.R.); (J.S.); (M.L.)
| | - Britta Siegmund
- Department of Gastroenterology, Infectious Diseases and Rheumatology (Including Nutrition Medicine), Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany;
| | - Marie Weinhart
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany (N.A.); (L.E.); (F.R.); (J.S.); (M.L.)
- Institute of Physical Chemistry and Electrochemistry, Leibniz Universität Hannover, Callinstr. 3A, 30167 Hannover, Germany
| |
Collapse
|
10
|
Ingelman-Sundberg M, Lauschke VM. Individualized Pharmacotherapy Utilizing Genetic Biomarkers and Novel In Vitro Systems As Predictive Tools for Optimal Drug Development and Treatment. Drug Metab Dispos 2024; 52:467-475. [PMID: 38575185 DOI: 10.1124/dmd.123.001302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/15/2024] [Accepted: 03/12/2024] [Indexed: 04/06/2024] Open
Abstract
In the area of drug development and clinical pharmacotherapy, a profound understanding of the pharmacokinetics and potential adverse reactions associated with the drug under investigation is paramount. Essential to this endeavor is a comprehensive understanding about interindividual variations in absorption, distribution, metabolism, and excretion (ADME) genetics and the predictive capabilities of in vitro systems, shedding light on metabolite formation and the risk of adverse drug reactions (ADRs). Both the domains of pharmacogenomics and the advancement of in vitro systems are experiencing rapid expansion. Here we present an update on these burgeoning fields, providing an overview of their current status and illuminating potential future directions. SIGNIFICANCE STATEMENT: There is very rapid development in the area of pharmacogenomics and in vitro systems for predicting drug pharmacokinetics and risk for adverse drug reactions. We provide an update of the current status of pharmacogenomics and developed in vitro systems on these aspects aimed to achieve a better personalized pharmacotherapy.
Collapse
Affiliation(s)
- Magnus Ingelman-Sundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (M.I.-S., V.M.L.); Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.); and University of Tübingen, Tübingen, Germany (V.M.L.)
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (M.I.-S., V.M.L.); Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.); and University of Tübingen, Tübingen, Germany (V.M.L.)
| |
Collapse
|
11
|
Liu T, Gu J, Fu C, Su L. Three-Dimensional Scaffolds for Intestinal Cell Culture: Fabrication, Utilization, and Prospects. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:158-175. [PMID: 37646409 DOI: 10.1089/ten.teb.2023.0124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
The intestine is a visceral organ that integrates absorption, metabolism, and immunity, which is vulnerable to external stimulus. Researchers in the fields such as food science, immunology, and pharmacology have committed to developing appropriate in vitro intestinal cell models to study the intestinal absorption and metabolism mechanisms of various nutrients and drugs, or pathogenesis of intestinal diseases. In the past three decades, the intestinal cell models have undergone a significant transformation from conventional two-dimensional cultures to three-dimensional (3D) systems, and the achievements of 3D cell culture have been greatly contributed by the fabrication of different scaffolds. In this review, we first introduce the developing trend of existing intestinal models. Then, four types of scaffolds, including Transwell, hydrogel, tubular scaffolds, and intestine-on-a-chip, are discussed for their 3D structure, composition, advantages, and limitations in the establishment of intestinal cell models. Excitingly, some of the in vitro intestinal cell models based on these scaffolds could successfully mimic the 3D structure, microenvironment, mechanical peristalsis, fluid system, signaling gradients, or other important aspects of the original human intestine. Furthermore, we discuss the potential applications of the intestinal cell models in drug screening, disease modeling, and even regenerative repair of intestinal tissues. This review presents an overview of state-of-the-art scaffold-based cell models within the context of intestines, and highlights their major advances and applications contributing to a better knowledge of intestinal diseases. Impact statement The intestine tract is crucial in the absorption and metabolism of nutrients and drugs, as well as immune responses against external pathogens or antigens in a complex microenvironment. The appropriate experimental cell model in vitro is needed for in-depth studies of intestines, due to the limitation of animal models in dynamic control and real-time assessment of key intestinal physiological and pathological processes, as well as the "R" principles in laboratory animal experiments. Three-dimensional (3D) scaffold-based cell cultivation has become a developing tendency because of the superior cell proliferation and differentiation and more physiologically relevant environment supported by the customized 3D scaffolds. In this review, we summarize four types of up-to-date 3D cell culture scaffolds fabricated by various materials and techniques for a better recapitulation of some essential physiological and functional characteristics of original intestines compared to conventional cell models. These emerging 3D intestinal models have shown promising results in not only evaluating the pharmacokinetic characteristics, security, and effectiveness of drugs, but also studying the pathological mechanisms of intestinal diseases at cellular and molecular levels. Importantly, the weakness of the representative 3D models for intestines is also discussed.
Collapse
Affiliation(s)
- Tiange Liu
- Department of Food Science and Technology, National University of Singapore (Suzhou) Research Institute, Suzhou, China
| | - Jia Gu
- Department of Food Science and Technology, National University of Singapore (Suzhou) Research Institute, Suzhou, China
| | - Caili Fu
- Department of Food Science and Technology, National University of Singapore (Suzhou) Research Institute, Suzhou, China
| | - Lingshan Su
- Department of Food Science and Technology, National University of Singapore (Suzhou) Research Institute, Suzhou, China
- Department of Food Science and Technology, National University of Singapore, Singapore, Singapore
| |
Collapse
|
12
|
Wang JB, Wu J, Zhang J, Guan LA, Feng HB, Zhu KY, Zhang Y, Zhao WJ, Peng Q, Meng B, Yang S, Sun H, Cheng YD, Zhang L. Bibliometric and visualized analysis of hydrogels in organoids research. Regen Ther 2024; 25:395-404. [PMID: 38435088 PMCID: PMC10905953 DOI: 10.1016/j.reth.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 03/05/2024] Open
Abstract
Over the past decades, there has been ongoing effort to develop complex biomimetic tissue engineering strategies for in vitro cultivation and maintenance of organoids. The defined hydrogels can create organoid models for various organs by changing their properties and various active molecules. An increasing number of researches has been done on the application of hydrogels in organoids, and a large number of articles have been published on the topic. Although there have been existing reviews describing the application of hydrogels in the field of organoids, there is still a lack of comprehensive studies summarizing and analyzing the overall research trends in this field. The citation can be used as an indicator of the scientific influence of an article in its field. This study aims to evaluate the application of hydrogels in organoids through bibliometric analysis, and to predict the hotspots and developing trends in this field.
Collapse
Affiliation(s)
- Jia-bo Wang
- Department of Orthopedics, Clinical Medical College of Yangzhou University, Yangzhou 225001, China
- Huai'an 82 Hospital, Huai'an 223001, China
| | - Jie Wu
- Huai'an 82 Hospital, Huai'an 223001, China
| | - Jian Zhang
- Huai'an 82 Hospital, Huai'an 223001, China
| | - Li-an Guan
- Huai'an 82 Hospital, Huai'an 223001, China
| | | | - Ke-yan Zhu
- The Fifth People's Hospital of Huai'an, Huai'an 223001, China
| | - Yu Zhang
- Department of Orthopedics, Clinical Medical College of Yangzhou University, Yangzhou 225001, China
| | - Wen-jie Zhao
- Graduate School of Dalian Medical University, Dalian 116000, China
| | - Qing Peng
- Department of Orthopedics, Clinical Medical College of Yangzhou University, Yangzhou 225001, China
| | - Bo Meng
- Graduate School of Dalian Medical University, Dalian 116000, China
| | - Sheng Yang
- Graduate School of Dalian Medical University, Dalian 116000, China
| | - Hua Sun
- Department of Orthopedics, Clinical Medical College of Yangzhou University, Yangzhou 225001, China
| | | | - Liang Zhang
- Department of Orthopedics, Clinical Medical College of Yangzhou University, Yangzhou 225001, China
| |
Collapse
|
13
|
Fu A, Mao S, Kasai N, Zhu H, Zeng H. Dynamic tissue model in vitro and its application for assessment of microplastics-induced toxicity to air-blood barrier (ABB). Biosens Bioelectron 2024; 246:115858. [PMID: 38039733 DOI: 10.1016/j.bios.2023.115858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/07/2023] [Accepted: 11/15/2023] [Indexed: 12/03/2023]
Abstract
The replication of the hominine physiological environment was identified as an effectual strategy to develop the physiological model in vitro to perform the intuitionistic assessment of toxicity of contaminations. Herein, we proposed a dynamic interface strategy that accurately mimicked the blood flow and shear stress in human capillaries to subtly evaluate the physiological damages. To proof the concept, the dynamic air-blood barrier (ABB) model in vitro was developed by the dynamic interface strategy and was utilized to assess the toxicity of polyethylene terephthalate microplastics (PET-MPs). The developed dynamic ABB model was compared with the static ABB model developed by the conventional Transwell® system and the animal model, then the performance of the dynamic ABB model in evaluation of the PET-MPs induced pulmonary damage via replicating the hominine ABB. The experimental data revealed that the developed dynamic ABB model in vitro effectively mimicked the physiological structure and barrier functions of human ABB, in which more sophisticated physiological microenvironment enabled the distinguishment of the toxicities of PET-MPs in different sizes and different concentrations comparing with the static ABB model constructed on Transwell® systems. Furthermore, the consistent physiological and biochemical characters adopted dynamic ABB model could be achieved in a quick manner referring with that of the mouse model in the evaluation of the microplastics-induced pulmonary damage. The proposed dynamic interface strategy supplied a general approach to develop the hominine physiological environment in vitro and exhibited a potential to develop the ABB model in vitro to evaluate the hazards of inhaled airborne pollutants.
Collapse
Affiliation(s)
- Anchen Fu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, China
| | - Sifeng Mao
- Department of Applied Chemistry, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, Minamiohsawa, Hachioji, Tokyo, 192-0397, Japan.
| | - Nahoko Kasai
- Department of Applied Chemistry, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, Minamiohsawa, Hachioji, Tokyo, 192-0397, Japan
| | - Haiyan Zhu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, China
| | - Hulie Zeng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, China.
| |
Collapse
|
14
|
Leal F, Zeiringer S, Jeitler R, Costa PF, Roblegg E. A comprehensive overview of advanced dynamic in vitro intestinal and hepatic cell culture models. Tissue Barriers 2024; 12:2163820. [PMID: 36680530 PMCID: PMC10832944 DOI: 10.1080/21688370.2022.2163820] [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: 08/23/2022] [Accepted: 12/22/2022] [Indexed: 01/22/2023] Open
Abstract
Orally administered drugs pass through the gastrointestinal tract before being absorbed in the small intestine and metabolised in the liver. To test the efficacy and toxicity of drugs, animal models are often employed; however, they are not suitable for investigating drug-tissue interactions and making reliable predictions, since the human organism differs drastically from animals in terms of absorption, distribution, metabolism and excretion of substances. Likewise, simple static in vitro cell culture systems currently used in preclinical drug screening often do not resemble the native characteristics of biological barriers. Dynamic models, on the other hand, provide in vivo-like cell phenotypes and functionalities that offer great potential for safety and efficacy prediction. Herein, current microfluidic in vitro intestinal and hepatic models are reviewed, namely single- and multi-tissue micro-bioreactors, which are associated with different methods of cell cultivation, i.e., scaffold-based versus scaffold-free.
Collapse
Affiliation(s)
- Filipa Leal
- BIOFABICS, Rua Alfredo Allen 455, 4200-135 Porto, Portugal
| | - Scarlett Zeiringer
- Department of Pharmaceutical Technology and Biopharmacy, University of Graz, Institute of Pharmaceutical Sciences, Universitaetsplatz 1, Graz, Austria
| | - Ramona Jeitler
- Department of Pharmaceutical Technology and Biopharmacy, University of Graz, Institute of Pharmaceutical Sciences, Universitaetsplatz 1, Graz, Austria
| | - Pedro F. Costa
- BIOFABICS, Rua Alfredo Allen 455, 4200-135 Porto, Portugal
| | - Eva Roblegg
- Department of Pharmaceutical Technology and Biopharmacy, University of Graz, Institute of Pharmaceutical Sciences, Universitaetsplatz 1, Graz, Austria
| |
Collapse
|
15
|
Fey C, Truschel T, Nehlsen K, Damigos S, Horstmann J, Stradal T, May T, Metzger M, Zdzieblo D. Enhancing pre-clinical research with simplified intestinal cell line models. J Tissue Eng 2024; 15:20417314241228949. [PMID: 38449469 PMCID: PMC10916479 DOI: 10.1177/20417314241228949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 01/12/2024] [Indexed: 03/08/2024] Open
Abstract
Two-dimensional culture remains widely employed to determine the bioavailability of orally delivered drugs. To gain more knowledge about drug uptake mechanisms and risk assessment for the patient after oral drug admission, intestinal in vitro models demonstrating a closer similarity to the in vivo situation are needed. In particular, Caco-2 cell-based Transwell® models show advantages as they are reproducible, cost-efficient, and standardized. However, cellular complexity is impaired and cell function is strongly modified as important transporters in the apical membrane are missing. To overcome these limitations, primary organoid-based human small intestinal tissue models were developed recently but the application of these cultures in pre-clinical research still represents an enormous challenge, as culture setup is complex as well as time- and cost-intensive. To overcome these hurdles, we demonstrate the establishment of primary organoid-derived intestinal cell lines by immortalization. Besides exhibiting cellular diversity of the organoid, these immortalized cell lines enable a standardized and more cost-efficient culture. Further, our cell line-based Transwell®-like models display an organ-specific epithelial barrier integrity, ultrastructural features and representative transport functions. Altogether, our novel model systems are cost-efficient with close similarity to the in vivo situation, therefore favoring their use in bioavailability studies in the context of pre-clinical screenings.
Collapse
Affiliation(s)
- Christina Fey
- Translational Center for Regenerative Therapies (TLZ-RT) Würzburg, Branch of the Fraunhofer Institute for Silicate Research (ISC), Würzburg, Germany
| | | | | | - Spyridon Damigos
- Department of Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, Würzburg, Germany
| | - Julia Horstmann
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | | | - Marco Metzger
- Translational Center for Regenerative Therapies (TLZ-RT) Würzburg, Branch of the Fraunhofer Institute for Silicate Research (ISC), Würzburg, Germany
- Department of Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, Würzburg, Germany
| | - Daniela Zdzieblo
- Translational Center for Regenerative Therapies (TLZ-RT) Würzburg, Branch of the Fraunhofer Institute for Silicate Research (ISC), Würzburg, Germany
- Department of Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, Würzburg, Germany
- Project Center for Stem Cell Process Engineering (PZ-SPT), Branch of the Fraunhofer Institute for Silicate Research (ISC), Würzburg, Germany
| |
Collapse
|
16
|
Hevia A, Ruas-Madiedo P, Faria MA, Petit V, Alves B, Alvito P, Arranz E, Bastiaan-Net S, Corredig M, Dijk W, Dupont D, Giblin L, Graf BA, Kondrashina A, Ramos H, Ruiz L, Santos-Hernández M, Soriano-Romaní L, Tomás-Cobos L, Vivanco-Maroto SM, Recio I, Miralles B. A Shared Perspective on in Vitro and in Vivo Models to Assay Intestinal Transepithelial Transport of Food Compounds. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:19265-19276. [PMID: 38035628 PMCID: PMC10723066 DOI: 10.1021/acs.jafc.3c05479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/29/2023] [Accepted: 11/07/2023] [Indexed: 12/02/2023]
Abstract
Assessing nutrient bioavailability is complex, as the process involves multiple digestion steps, several cellular environments, and regulatory-metabolic mechanisms. Several in vitro models of different physiological relevance are used to study nutrient absorption, providing significant challenges in data evaluation. However, such in vitro models are needed for mechanistic studies as well as to screen for biological functionality of the food structures designed. This collaborative work aims to put into perspective the wide-range of models to assay the permeability of food compounds considering the particular nature of the different molecules, and, where possible, in vivo data are provided for comparison.
Collapse
Affiliation(s)
- Arancha Hevia
- Dairy
Research Institute of Asturias (IPLA-CSIC), Paseo Río Linares, sn. Villaviciosa 33300, Asturias. Spain
| | - Patricia Ruas-Madiedo
- Dairy
Research Institute of Asturias (IPLA-CSIC), Paseo Río Linares, sn. Villaviciosa 33300, Asturias. Spain
| | - Miguel Angelo Faria
- LAQV/REQUIMTE,
Laboratório de Bromatologia e Hidrologia, Departamento de Ciências
Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Valérie Petit
- Nestlé
Research, Société des Produits
Nestlé SA, 1000 Lausanne, Switzerland
| | - Bruna Alves
- Faculty
of Sciences, University of Lisboa, Campo Grande, 1749-016 Lisboa Portugal
| | - Paula Alvito
- Food
and Nutrition Department, National Institute
of Health Dr. Ricardo Jorge, 1649-016 Lisbon, Portugal
- CESAM - Centre
for Environmental and Marine Studies, University
of Aveiro, 3810-193 Aveiro, Portugal
| | - Elena Arranz
- Department
of Nutrition and Food Science, Faculty of Pharmacy, Complutense University of Madrid (UCM), E-28040 Madrid, Spain
| | - Shanna Bastiaan-Net
- Wageningen
Food & Biobased Research, Wageningen
University & Research, 6708 WG Wageningen, The Netherlands
| | - Milena Corredig
- Department
of Food Science, Aarhus University, Agro Food Park 48, 8200 Aarhus, Denmark
| | | | - Didier Dupont
- INRAE Agrocampus Ouest, STLO, F-35042 Rennes, France
| | - Linda Giblin
- Teagasc Food Research Centre, Moorepark, Fermoy, P61
C996 County Cork, Ireland
| | - Brigitte Anna Graf
- Department
of Health Professions, Faculty of Health and Education, Manchester Metropolitan University, M15 6BH Manchester, U.K.
| | - Alina Kondrashina
- H&H
Group, H&H Research, Global Research
and Technology Centre, P61
K202 Cork, County Cork, Ireland
| | - Helena Ramos
- LAQV/REQUIMTE,
Laboratório de Bromatologia e Hidrologia, Departamento de Ciências
Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Lorena Ruiz
- Dairy
Research Institute of Asturias (IPLA-CSIC), Paseo Río Linares, sn. Villaviciosa 33300, Asturias. Spain
| | - Marta Santos-Hernández
- Wellcome
Trust - MRC Institute of Metabolic Science, Metabolic Research laboratories, Addenbrooke’s Hospital, Hills Road, CB2 0QQ Cambridge, U.K.
| | - Laura Soriano-Romaní
- AINIA
in Vitro Preclinical Studies Area, Parque
Tecnológico de Valencia. c/Benjamín Franklin, 5-11, E46980 Paterna, Spain
| | - Lidia Tomás-Cobos
- AINIA
in Vitro Preclinical Studies Area, Parque
Tecnológico de Valencia. c/Benjamín Franklin, 5-11, E46980 Paterna, Spain
| | | | - Isidra Recio
- Institute of Food
Science Research CIAL (CSIC-UAM), C/Nicolás Cabrera 9, 28049 Madrid, Spain
| | - Beatriz Miralles
- Institute of Food
Science Research CIAL (CSIC-UAM), C/Nicolás Cabrera 9, 28049 Madrid, Spain
| |
Collapse
|
17
|
Wright CW, Li N, Shaffer L, Hill A, Boyer N, Alves SE, Venkataraman S, Biswas K, Lieberman LA, Mohammadi S. Establishment of a 96-well transwell system using primary human gut organoids to capture multiple quantitative pathway readouts. Sci Rep 2023; 13:16357. [PMID: 37773535 PMCID: PMC10541891 DOI: 10.1038/s41598-023-43656-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 09/26/2023] [Indexed: 10/01/2023] Open
Abstract
Disruptions in the gut epithelial barrier can lead to the development of chronic indications such as inflammatory bowel disease (IBD). Historically, barrier function has been assessed in cancer cell lines, which do not contain all human intestinal cell types, leading to poor translatability. To bridge this gap, we adapted human primary gut organoids grown as monolayers to quantify transcription factor phosphorylation, gene expression, cytokine production, and barrier function. In this work we describe and characterize a novel 96-well human gut organoid-derived monolayer system that enables quantitative assessment of candidate therapeutics. Normal human intestine differentiation patterns and barrier function were characterized and confirmed to recapitulate key aspects of in vivo biology. Next, cellular response to TNF-α (a central driver of IBD) was determined using a diverse cadre of quantitative readouts. We showed that TNF-α pathway antagonists rescued damage caused by TNF-α in a dose-dependent manner, indicating that this system is suitable for quantitative assessment of barrier modulating factors. Taken together, we have established a robust primary cell-based 96-well system capable of interrogating questions around mucosal response. This system is well suited to provide pivotal functional data to support translational target and drug discovery efforts.
Collapse
Affiliation(s)
- Charles W Wright
- Discovery Immunology, Merck & Co., Inc., Cambridge, MA, 02141, USA
| | - Naomi Li
- Quantitative Biosciences, Merck & Co., Inc., Boston, MA, 02115, USA
| | - Lynsey Shaffer
- Quantitative Biosciences, Merck & Co., Inc., Boston, MA, 02115, USA
- Moderna, Inc., Cambridge, MA, USA
| | - Armetta Hill
- Quantitative Biosciences, Merck & Co., Inc., Boston, MA, 02115, USA
| | - Nicolas Boyer
- Discovery Chemistry, Merck & Co., Inc., Boston, MA, 02115, USA
| | - Stephen E Alves
- Discovery Immunology, Merck & Co., Inc., Cambridge, MA, 02141, USA
| | | | - Kaustav Biswas
- Discovery Chemistry, Merck & Co., Inc., Boston, MA, 02115, USA
| | | | - Sina Mohammadi
- Discovery Immunology, Merck & Co., Inc., Cambridge, MA, 02141, USA.
| |
Collapse
|
18
|
Koziolek M, Augustijns P, Berger C, Cristofoletti R, Dahlgren D, Keemink J, Matsson P, McCartney F, Metzger M, Mezler M, Niessen J, Polli JE, Vertzoni M, Weitschies W, Dressman J. Challenges in Permeability Assessment for Oral Drug Product Development. Pharmaceutics 2023; 15:2397. [PMID: 37896157 PMCID: PMC10609725 DOI: 10.3390/pharmaceutics15102397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/12/2023] [Accepted: 09/19/2023] [Indexed: 10/29/2023] Open
Abstract
Drug permeation across the intestinal epithelium is a prerequisite for successful oral drug delivery. The increased interest in oral administration of peptides, as well as poorly soluble and poorly permeable compounds such as drugs for targeted protein degradation, have made permeability a key parameter in oral drug product development. This review describes the various in vitro, in silico and in vivo methodologies that are applied to determine drug permeability in the human gastrointestinal tract and identifies how they are applied in the different stages of drug development. The various methods used to predict, estimate or measure permeability values, ranging from in silico and in vitro methods all the way to studies in animals and humans, are discussed with regard to their advantages, limitations and applications. A special focus is put on novel techniques such as computational approaches, gut-on-chip models and human tissue-based models, where significant progress has been made in the last few years. In addition, the impact of permeability estimations on PK predictions in PBPK modeling, the degree to which excipients can affect drug permeability in clinical studies and the requirements for colonic drug absorption are addressed.
Collapse
Affiliation(s)
- Mirko Koziolek
- NCE Drug Product Development, Development Sciences, AbbVie Deutschland GmbH & Co. KG, 67061 Ludwigshafen, Germany
| | - Patrick Augustijns
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Constantin Berger
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, 97070 Würzburg, Germany;
| | - Rodrigo Cristofoletti
- Department of Pharmaceutics, University of Florida, 6550 Sanger Road, Orlando, FL 32827, USA
| | - David Dahlgren
- Department of Pharmaceutical Biosciences, Uppsala University, 75124 Uppsala, Sweden (J.N.)
| | - Janneke Keemink
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, 4070 Basel, Switzerland;
| | - Pär Matsson
- Department of Pharmacology and SciLifeLab Gothenburg, University of Gothenburg, 40530 Gothenburg, Sweden;
| | - Fiona McCartney
- School of Veterinary Medicine, University College Dublin, D04 V1W8 Dublin, Ireland;
| | - Marco Metzger
- Translational Center for Regenerative Therapies (TLZ-RT) Würzburg, Branch of the Fraunhofer Institute for Silicate Research (ISC), 97082 Würzburg, Germany
| | - Mario Mezler
- Quantitative, Translational & ADME Sciences, AbbVie Deutschland GmbH & Co. KG, 67061 Ludwigshafen, Germany;
| | - Janis Niessen
- Department of Pharmaceutical Biosciences, Uppsala University, 75124 Uppsala, Sweden (J.N.)
| | - James E. Polli
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD 21021, USA;
| | - Maria Vertzoni
- Department of Pharmacy, National and Kapodistrian University of Athens, 157 84 Zografou, Greece;
| | - Werner Weitschies
- Institute of Pharmacy, University of Greifswald, 17489 Greifswald, Germany
| | - Jennifer Dressman
- Fraunhofer Institute of Translational Medicine and Pharmacology, 60596 Frankfurt, Germany
| |
Collapse
|
19
|
Mehta M, Polli JE, Seo P, Bhoopathy S, Berginc K, Kristan K, Cook J, Dressman JB, Mandula H, Munshi U, Shanker R, Volpe DA, Gordon J, Veerasingham S, Welink J, Almeida S, Gonzalez P, Painter D, Tsang YC, Vaidyanathan J, Velagapudi R. Drug Permeability - Best Practices for Biopharmaceutics Classification System (BCS)-Based Biowaivers: A workshop Summary Report. J Pharm Sci 2023; 112:1749-1762. [PMID: 37142122 DOI: 10.1016/j.xphs.2023.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 04/28/2023] [Accepted: 04/28/2023] [Indexed: 05/06/2023]
Abstract
The workshop "Drug Permeability - Best Practices for Biopharmaceutics Classification System (BCS) Based Biowaivers" was held virtually on December 6, 2021, organized by the University of Maryland Center of Excellence in Regulatory Science and Innovation (M-CERSI), and the Food and Drug Administration (FDA). The workshop focused on the industrial, academic, and regulatory experiences in generating and evaluating permeability data, with the aim to further facilitate implementation of the BCS and efficient development of high-quality drug products globally. As the first international permeability workshop since the BCS based biowaivers was finalized as the ICH M9 guideline, the workshop included lectures, panel discussions, and breakout sessions. Lecture and panel discussion topics covered case studies at IND, NDA, and ANDA stages, typical deficiencies relating to permeability assessment supporting BCS biowaiver, types of evidence that are available to demonstrate high permeability, method suitability of a permeability assay, impact of excipients, importance of global acceptance of permeability methods, opportunities to expand the use of biowaivers (e.g. non-Caco-2 cell lines, totality-of-evidence approach to demonstrate high permeability) and future of permeability testing. Breakout sessions focused on 1) in vitro and in silico intestinal permeability methods; 2) potential excipient effects on permeability and; 3) use of label and literature data to designate permeability class.
Collapse
Affiliation(s)
- M Mehta
- US Food & Drug Administration, Silver Spring, MD, USA.
| | - J E Polli
- University of Maryland, Baltimore, MD, USA
| | - P Seo
- US Food & Drug Administration, Silver Spring, MD, USA
| | | | | | | | - J Cook
- Pfizer Inc, Groton, CT, USA
| | - J B Dressman
- Fraunhofer Institute of Translational Medicine and Pharmacology, Frankfurt, Germany
| | - H Mandula
- US Food & Drug Administration, Silver Spring, MD, USA
| | - U Munshi
- US Food & Drug Administration, Silver Spring, MD, USA
| | | | - D A Volpe
- US Food & Drug Administration, Silver Spring, MD, USA
| | - J Gordon
- World Health Organization, Geneva, Switzerland
| | | | - J Welink
- European Medicines Agency, Amsterdam, the Netherlands
| | - S Almeida
- Medicines for Europe, Brussels, Belgium
| | - P Gonzalez
- Biopharmaceutical Evaluation Center, Santiago, Chile
| | | | | | | | | |
Collapse
|
20
|
Kort-Mascort J, Flores-Torres S, Peza-Chavez O, Jang JH, Pardo LA, Tran SD, Kinsella J. Decellularized ECM hydrogels: prior use considerations, applications, and opportunities in tissue engineering and biofabrication. Biomater Sci 2023; 11:400-431. [PMID: 36484344 DOI: 10.1039/d2bm01273a] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Tissue development, wound healing, pathogenesis, regeneration, and homeostasis rely upon coordinated and dynamic spatial and temporal remodeling of extracellular matrix (ECM) molecules. ECM reorganization and normal physiological tissue function, require the establishment and maintenance of biological, chemical, and mechanical feedback mechanisms directed by cell-matrix interactions. To replicate the physical and biological environment provided by the ECM in vivo, methods have been developed to decellularize and solubilize tissues which yield organ and tissue-specific bioactive hydrogels. While these biomaterials retain several important traits of the native ECM, the decellularizing process, and subsequent sterilization, and solubilization result in fragmented, cleaved, or partially denatured macromolecules. The final product has decreased viscosity, moduli, and yield strength, when compared to the source tissue, limiting the compatibility of isolated decellularized ECM (dECM) hydrogels with fabrication methods such as extrusion bioprinting. This review describes the physical and bioactive characteristics of dECM hydrogels and their role as biomaterials for biofabrication. In this work, critical variables when selecting the appropriate tissue source and extraction methods are identified. Common manual and automated fabrication techniques compatible with dECM hydrogels are described and compared. Fabrication and post-manufacturing challenges presented by the dECM hydrogels decreased mechanical and structural stability are discussed as well as circumvention strategies. We further highlight and provide examples of the use of dECM hydrogels in tissue engineering and their role in fabricating complex in vitro 3D microenvironments.
Collapse
Affiliation(s)
| | | | - Omar Peza-Chavez
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada.
| | - Joyce H Jang
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada.
| | | | - Simon D Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Joseph Kinsella
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
21
|
Däullary T, Imdahl F, Dietrich O, Hepp L, Krammer T, Fey C, Neuhaus W, Metzger M, Vogel J, Westermann AJ, Saliba AE, Zdzieblo D. A primary cell-based in vitro model of the human small intestine reveals host olfactomedin 4 induction in response to Salmonella Typhimurium infection. Gut Microbes 2023; 15:2186109. [PMID: 36939013 PMCID: PMC10038062 DOI: 10.1080/19490976.2023.2186109] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/21/2023] Open
Abstract
Infection research largely relies on classical cell culture or mouse models. Despite having delivered invaluable insights into host-pathogen interactions, both have limitations in translating mechanistic principles to human pathologies. Alternatives can be derived from modern Tissue Engineering approaches, allowing the reconstruction of functional tissue models in vitro. Here, we combined a biological extracellular matrix with primary tissue-derived enteroids to establish an in vitro model of the human small intestinal epithelium exhibiting in vivo-like characteristics. Using the foodborne pathogen Salmonella enterica serovar Typhimurium, we demonstrated the applicability of our model to enteric infection research in the human context. Infection assays coupled to spatio-temporal readouts recapitulated the established key steps of epithelial infection by this pathogen in our model. Besides, we detected the upregulation of olfactomedin 4 in infected cells, a hitherto unrecognized aspect of the host response to Salmonella infection. Together, this primary human small intestinal tissue model fills the gap between simplistic cell culture and animal models of infection, and shall prove valuable in uncovering human-specific features of host-pathogen interplay.
Collapse
Affiliation(s)
- Thomas Däullary
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg (UKW), Würzburg, Germany
- Faculty of Biology, Biocenter, Chair of Microbiology, Julius-Maximilians-Universität Würzburg (JMU), Würzburg, Germany
| | - Fabian Imdahl
- Helmholtz-Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Oliver Dietrich
- Helmholtz-Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Laura Hepp
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg (UKW), Würzburg, Germany
| | - Tobias Krammer
- Helmholtz-Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Christina Fey
- Fraunhofer Institute for Silicate Research (ISC),Translational Center Regenerative Therapies (TLC-RT), Würzburg, Germany
| | - Winfried Neuhaus
- Austrian Institute of Technology (AIT), Vienna, Austria
- Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University (DPU), Krems, Austria
| | - Marco Metzger
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg (UKW), Würzburg, Germany
- Fraunhofer Institute for Silicate Research (ISC),Translational Center Regenerative Therapies (TLC-RT), Würzburg, Germany
- Fraunhofer Institute for Silicate Research, Project Center for Stem Cell Process Engineering, Würzburg, Germany
| | - Jörg Vogel
- Helmholtz-Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
- Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Alexander J Westermann
- Helmholtz-Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
- Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz-Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Daniela Zdzieblo
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg (UKW), Würzburg, Germany
- Fraunhofer Institute for Silicate Research (ISC),Translational Center Regenerative Therapies (TLC-RT), Würzburg, Germany
- Fraunhofer Institute for Silicate Research, Project Center for Stem Cell Process Engineering, Würzburg, Germany
| |
Collapse
|
22
|
Petrosyan A, Montali F, Peloso A, Citro A, Byers LN, La Pointe C, Suleiman M, Marchetti A, Mcneill EP, Speer AL, Ng WH, Ren X, Bussolati B, Perin L, Di Nardo P, Cardinale V, Duisit J, Monetti AR, Savino JR, Asthana A, Orlando G. Regenerative medicine technologies applied to transplant medicine. An update. Front Bioeng Biotechnol 2022; 10:1015628. [PMID: 36263358 PMCID: PMC9576214 DOI: 10.3389/fbioe.2022.1015628] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Regenerative medicine (RM) is changing how we think and practice transplant medicine. In regenerative medicine, the aim is to develop and employ methods to regenerate, restore or replace damaged/diseased tissues or organs. Regenerative medicine investigates using tools such as novel technologies or techniques, extracellular vesicles, cell-based therapies, and tissue-engineered constructs to design effective patient-specific treatments. This review illustrates current advancements in regenerative medicine that may pertain to transplant medicine. We highlight progress made and various tools designed and employed specifically for each tissue or organ, such as the kidney, heart, liver, lung, vasculature, gastrointestinal tract, and pancreas. By combing both fields of transplant and regenerative medicine, we can harbor a successful collaboration that would be beneficial and efficacious for the repair and design of de novo engineered whole organs for transplantations.
Collapse
Affiliation(s)
- Astgik Petrosyan
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Saban Research Institute, Division of Urology, Children’s Hospital Los Angeles, Los Angeles, CA, United States
| | - Filippo Montali
- Department of General Surgery, di Vaio Hospital, Fidenza, Italy
| | - Andrea Peloso
- Visceral Surgery Division, University Hospitals of Geneva, Geneva, Switzerland
| | - Antonio Citro
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Lori N. Byers
- Wake Forest School of Medicine, Winston Salem, NC, United States
| | | | - Mara Suleiman
- Wake Forest School of Medicine, Winston Salem, NC, United States
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Alice Marchetti
- Wake Forest School of Medicine, Winston Salem, NC, United States
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Eoin P. Mcneill
- Department of Pediatric Surgery, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, United States
| | - Allison L Speer
- Department of Pediatric Surgery, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, United States
| | - Wai Hoe Ng
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Xi Ren
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Laura Perin
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Saban Research Institute, Division of Urology, Children’s Hospital Los Angeles, Los Angeles, CA, United States
| | - Paolo Di Nardo
- Centro Interdipartimentale per la Medicina Rigenerativa (CIMER), Università Degli Studi di Roma Tor Vergata, Rome, Italy
| | - Vincenzo Cardinale
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Jerome Duisit
- Department of Plastic, Reconstructive and Aesthetic Surgery, CHU Rennes, University of Rennes I, Rennes, France
| | | | | | - Amish Asthana
- Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Giuseppe Orlando
- Wake Forest School of Medicine, Winston Salem, NC, United States
| |
Collapse
|
23
|
Schaller-Ammann R, Kreß S, Feiel J, Schwagerle G, Priedl J, Birngruber T, Kasper C, Egger D. Advanced Online Monitoring of In Vitro Human 3D Full-Thickness Skin Equivalents. Pharmaceutics 2022; 14:pharmaceutics14071436. [PMID: 35890329 PMCID: PMC9315769 DOI: 10.3390/pharmaceutics14071436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/27/2022] [Accepted: 06/30/2022] [Indexed: 12/17/2022] Open
Abstract
Skin equivalents and skin explants are widely used for dermal penetration studies in the pharmacological development of drugs. Environmental parameters, such as the incubation and culture conditions affect cellular responses and thus the relevance of the experimental outcome. However, available systems such as the Franz diffusion chamber, only measure in the receiving culture medium, rather than assessing the actual conditions for cells in the tissue. We developed a sampling design that combines open flow microperfusion (OFM) sampling technology for continuous concentration measurements directly in the tissue with microfluidic biosensors for online monitoring of culture parameters. We tested our design with real-time measurements of oxygen, glucose, lactate, and pH in full-thickness skin equivalent and skin explants. Furthermore, we compared dermal penetration for acyclovir, lidocaine, and diclofenac in skin equivalents and skin explants. We observed differences in oxygen, glucose, and drug concentrations in skin equivalents compared to the respective culture medium and to skin explants.
Collapse
Affiliation(s)
- Roland Schaller-Ammann
- Health—Institute for Biomedicine and Health Sciences, Joanneum Research Forschungsgesellschaft mbH, Neue Stiftingtalstrasse 2, 8010 Graz, Austria; (R.S.-A.); (J.F.); (G.S.); (J.P.)
| | - Sebastian Kreß
- Institute of Cell and Tissue Culture Technologies, Department of Biotechnology, University of Natural, Resources and Life Sciences, Vienna, Muthgasse 18, 1190 Vienna, Austria; (S.K.); (C.K.)
| | - Jürgen Feiel
- Health—Institute for Biomedicine and Health Sciences, Joanneum Research Forschungsgesellschaft mbH, Neue Stiftingtalstrasse 2, 8010 Graz, Austria; (R.S.-A.); (J.F.); (G.S.); (J.P.)
| | - Gerd Schwagerle
- Health—Institute for Biomedicine and Health Sciences, Joanneum Research Forschungsgesellschaft mbH, Neue Stiftingtalstrasse 2, 8010 Graz, Austria; (R.S.-A.); (J.F.); (G.S.); (J.P.)
| | - Joachim Priedl
- Health—Institute for Biomedicine and Health Sciences, Joanneum Research Forschungsgesellschaft mbH, Neue Stiftingtalstrasse 2, 8010 Graz, Austria; (R.S.-A.); (J.F.); (G.S.); (J.P.)
| | - Thomas Birngruber
- Health—Institute for Biomedicine and Health Sciences, Joanneum Research Forschungsgesellschaft mbH, Neue Stiftingtalstrasse 2, 8010 Graz, Austria; (R.S.-A.); (J.F.); (G.S.); (J.P.)
- Correspondence: (T.B.); (D.E.)
| | - Cornelia Kasper
- Institute of Cell and Tissue Culture Technologies, Department of Biotechnology, University of Natural, Resources and Life Sciences, Vienna, Muthgasse 18, 1190 Vienna, Austria; (S.K.); (C.K.)
| | - Dominik Egger
- Institute of Cell and Tissue Culture Technologies, Department of Biotechnology, University of Natural, Resources and Life Sciences, Vienna, Muthgasse 18, 1190 Vienna, Austria; (S.K.); (C.K.)
- Correspondence: (T.B.); (D.E.)
| |
Collapse
|
24
|
Tullie L, Jones BC, De Coppi P, Li VSW. Building gut from scratch - progress and update of intestinal tissue engineering. Nat Rev Gastroenterol Hepatol 2022; 19:417-431. [PMID: 35241800 DOI: 10.1038/s41575-022-00586-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/31/2022] [Indexed: 12/18/2022]
Abstract
Short bowel syndrome (SBS), a condition defined by insufficient absorptive intestinal epithelium, is a rare disease, with an estimated prevalence up to 0.4 in 10,000 people. However, it has substantial morbidity and mortality for affected patients. The mainstay of treatment in SBS is supportive, in the form of intravenous parenteral nutrition, with the aim of achieving intestinal autonomy. The lack of a definitive curative therapy has led to attempts to harness innate developmental and regenerative mechanisms to engineer neo-intestine as an alternative approach to addressing this unmet clinical need. Exciting advances have been made in the field of intestinal tissue engineering (ITE) over the past decade, making a review in this field timely. In this Review, we discuss the latest advances in the components required to engineer intestinal grafts and summarize the progress of ITE. We also explore some key factors to consider and challenges to overcome when transitioning tissue-engineered intestine towards clinical translation, and provide the future outlook of ITE in therapeutic applications and beyond.
Collapse
Affiliation(s)
- Lucinda Tullie
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK.,Stem Cell and Regenerative Medicine Section, DBC, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Brendan C Jones
- Stem Cell and Regenerative Medicine Section, DBC, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Paolo De Coppi
- Stem Cell and Regenerative Medicine Section, DBC, Great Ormond Street Institute of Child Health, University College London, London, UK. .,Specialist Neonatal and Paediatric Surgery Unit, Great Ormond Street Hospital, London, UK.
| | - Vivian S W Li
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
25
|
Cecen B, Bal-Ozturk A, Yasayan G, Alarcin E, Kocak P, Tutar R, Kozaci LD, Shin SR, Miri AK. Selection of natural biomaterials for micro-tissue and organ-on-chip models. J Biomed Mater Res A 2022; 110:1147-1165. [PMID: 35102687 PMCID: PMC10700148 DOI: 10.1002/jbm.a.37353] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/14/2022]
Abstract
The desired organ in micro-tissue models of organ-on-a-chip (OoC) devices dictates the optimum biomaterials, divided into natural and synthetic biomaterials. They can resemble biological tissues' biological functions and architectures by constructing bioactivity of macromolecules, cells, nanoparticles, and other biological agents. The inclusion of such components in OoCs allows them having biological processes, such as basic biorecognition, enzymatic cleavage, and regulated drug release. In this report, we review natural-based biomaterials that are used in OoCs and their main characteristics. We address the preparation, modification, and characterization methods of natural-based biomaterials and summarize recent reports on their applications in the design and fabrication of micro-tissue models. This article will help bioengineers select the proper biomaterials based on developing new technologies to meet clinical expectations and improve patient outcomes fusing disease modeling.
Collapse
Affiliation(s)
- Berivan Cecen
- Department of Mechanical Engineering, Rowan University, Glassboro, New Jersey, USA
| | - Ayca Bal-Ozturk
- Department of Analytical Chemistry, Faculty of Pharmacy, Istinye University, Istanbul, Turkey
- Department of Stem Cell and Tissue Engineering, Institute of Health Sciences, Istinye University, Istanbul, Turkey
| | - Gokcen Yasayan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Marmara University, Istanbul, Turkey
| | - Emine Alarcin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Marmara University, Istanbul, Turkey
| | - Polen Kocak
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey
| | - Rumeysa Tutar
- Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Leyla Didem Kozaci
- Faculty of Medicine, Department of Medical Biochemistry, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, Massachusetts, USA
| | - Amir K. Miri
- Department of Mechanical Engineering, Rowan University, Glassboro, New Jersey, USA
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| |
Collapse
|
26
|
Taelman J, Diaz M, Guiu J. Human Intestinal Organoids: Promise and Challenge. Front Cell Dev Biol 2022; 10:854740. [PMID: 35359445 PMCID: PMC8962662 DOI: 10.3389/fcell.2022.854740] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/24/2022] [Indexed: 12/12/2022] Open
Abstract
The study of human intestinal biology in healthy and diseased conditions has always been challenging. Primary obstacles have included limited tissue accessibility, inadequate in vitro maintenance and ethical constrains. The development of three-dimensional organoid cultures has transformed this entirely. Intestinal organoids are self-organized three-dimensional structures that partially recapitulate the identity, cell heterogeneity and cell behaviour of the original tissue in vitro. This includes the capacity of stem cells to self-renew, as well as to differentiate towards major intestinal lineages. Therefore, over the past decade, the use of human organoid cultures has been instrumental to model human intestinal development, homeostasis, disease, and regeneration. Intestinal organoids can be derived from pluripotent stem cells (PSC) or from adult somatic intestinal stem cells (ISC). Both types of organoid sources harbour their respective strengths and weaknesses. In this mini review, we describe the applications of human intestinal organoids, discussing the differences, advantages, and disadvantages of PSC-derived and ISC-derived organoids.
Collapse
Affiliation(s)
- Jasin Taelman
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d’Investigació Biomèdica de Bellvitge–IDIBELL, L’Hospitalet de Llobregat, Spain
- Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Spain
| | - Mònica Diaz
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d’Investigació Biomèdica de Bellvitge–IDIBELL, L’Hospitalet de Llobregat, Spain
- Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Spain
| | - Jordi Guiu
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d’Investigació Biomèdica de Bellvitge–IDIBELL, L’Hospitalet de Llobregat, Spain
- Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Spain
- *Correspondence: Jordi Guiu,
| |
Collapse
|
27
|
Weigel T, Malkmus C, Weigel V, Wußmann M, Berger C, Brennecke J, Groeber-Becker F, Hansmann J. Fully Synthetic 3D Fibrous Scaffolds for Stromal Tissues-Replacement of Animal-Derived Scaffold Materials Demonstrated by Multilayered Skin. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2106780. [PMID: 34933407 DOI: 10.1002/adma.202106780] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 12/14/2021] [Indexed: 06/14/2023]
Abstract
The extracellular matrix (ECM) of soft tissues in vivo has remarkable biological and structural properties. Thereby, the ECM provides mechanical stability while it still can be rearranged via cellular remodeling during tissue maturation or healing processes. However, modern synthetic alternatives fail to provide these key features among basic properties. Synthetic matrices are usually completely degraded or are inert regarding cellular remodeling. Based on a refined electrospinning process, a method is developed to generate synthetic scaffolds with highly porous fibrous structures and enhanced fiber-to-fiber distances. Since this approach allows for cell migration, matrix remodeling, and ECM synthesis, the scaffold provides an ideal platform for the generation of soft tissue equivalents. Using this matrix, an electrospun-based multilayered skin equivalent composed of a stratified epidermis, a dermal compartment, and a subcutis is able to be generated without the use of animal matrix components. The extension of classical dense electrospun scaffolds with high porosities and motile fibers generates a fully synthetic and defined alternative to collagen-gel-based tissue models and is a promising system for the construction of tissue equivalents as in vitro models or in vivo implants.
Collapse
Affiliation(s)
- Tobias Weigel
- Translational Center for Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research (ISC), 97082, Würzburg, Germany
- Department for Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, 97070, Würzburg, Germany
| | - Christoph Malkmus
- Department for Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, 97070, Würzburg, Germany
| | - Verena Weigel
- Translational Center for Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research (ISC), 97082, Würzburg, Germany
- Department for Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, 97070, Würzburg, Germany
| | - Maximiliane Wußmann
- Translational Center for Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research (ISC), 97082, Würzburg, Germany
| | - Constantin Berger
- Department for Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, 97070, Würzburg, Germany
| | - Julian Brennecke
- Department for Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, 97070, Würzburg, Germany
| | - Florian Groeber-Becker
- Translational Center for Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research (ISC), 97082, Würzburg, Germany
- Department for Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, 97070, Würzburg, Germany
| | - Jan Hansmann
- Department for Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, 97070, Würzburg, Germany
- Faculty of Electrical Engineering, University of Applied Sciences Würzburg-Schweinfurt, 97421, Schweinfurt, Germany
| |
Collapse
|
28
|
Lipreri MV, Baldini N, Graziani G, Avnet S. Perfused Platforms to Mimic Bone Microenvironment at the Macro/Milli/Microscale: Pros and Cons. Front Cell Dev Biol 2022; 9:760667. [PMID: 35047495 PMCID: PMC8762164 DOI: 10.3389/fcell.2021.760667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 11/30/2021] [Indexed: 11/26/2022] Open
Abstract
As life expectancy increases, the population experiences progressive ageing. Ageing, in turn, is connected to an increase in bone-related diseases (i.e., osteoporosis and increased risk of fractures). Hence, the search for new approaches to study the occurrence of bone-related diseases and to develop new drugs for their prevention and treatment becomes more pressing. However, to date, a reliable in vitro model that can fully recapitulate the characteristics of bone tissue, either in physiological or altered conditions, is not available. Indeed, current methods for modelling normal and pathological bone are poor predictors of treatment outcomes in humans, as they fail to mimic the in vivo cellular microenvironment and tissue complexity. Bone, in fact, is a dynamic network including differently specialized cells and the extracellular matrix, constantly subjected to external and internal stimuli. To this regard, perfused vascularized models are a novel field of investigation that can offer a new technological approach to overcome the limitations of traditional cell culture methods. It allows the combination of perfusion, mechanical and biochemical stimuli, biological cues, biomaterials (mimicking the extracellular matrix of bone), and multiple cell types. This review will discuss macro, milli, and microscale perfused devices designed to model bone structure and microenvironment, focusing on the role of perfusion and encompassing different degrees of complexity. These devices are a very first, though promising, step for the development of 3D in vitro platforms for preclinical screening of novel anabolic or anti-catabolic therapeutic approaches to improve bone health.
Collapse
Affiliation(s)
| | - Nicola Baldini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,Biomedical Science and Technologies Lab, IRCSS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Gabriela Graziani
- Laboratory for NanoBiotechnology (NaBi), IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Sofia Avnet
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
29
|
Youhanna S, Kemas AM, Preiss L, Zhou Y, Shen JX, Cakal SD, Paqualini FS, Goparaju SK, Shafagh RZ, Lind JU, Sellgren CM, Lauschke VM. Organotypic and Microphysiological Human Tissue Models for Drug Discovery and Development-Current State-of-the-Art and Future Perspectives. Pharmacol Rev 2022; 74:141-206. [PMID: 35017176 DOI: 10.1124/pharmrev.120.000238] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 10/12/2021] [Indexed: 12/11/2022] Open
Abstract
The number of successful drug development projects has been stagnant for decades despite major breakthroughs in chemistry, molecular biology, and genetics. Unreliable target identification and poor translatability of preclinical models have been identified as major causes of failure. To improve predictions of clinical efficacy and safety, interest has shifted to three-dimensional culture methods in which human cells can retain many physiologically and functionally relevant phenotypes for extended periods of time. Here, we review the state of the art of available organotypic culture techniques and critically review emerging models of human tissues with key importance for pharmacokinetics, pharmacodynamics, and toxicity. In addition, developments in bioprinting and microfluidic multiorgan cultures to emulate systemic drug disposition are summarized. We close by highlighting important trends regarding the fabrication of organotypic culture platforms and the choice of platform material to limit drug absorption and polymer leaching while supporting the phenotypic maintenance of cultured cells and allowing for scalable device fabrication. We conclude that organotypic and microphysiological human tissue models constitute promising systems to promote drug discovery and development by facilitating drug target identification and improving the preclinical evaluation of drug toxicity and pharmacokinetics. There is, however, a critical need for further validation, benchmarking, and consolidation efforts ideally conducted in intersectoral multicenter settings to accelerate acceptance of these novel models as reliable tools for translational pharmacology and toxicology. SIGNIFICANCE STATEMENT: Organotypic and microphysiological culture of human cells has emerged as a promising tool for preclinical drug discovery and development that might be able to narrow the translation gap. This review discusses recent technological and methodological advancements and the use of these systems for hit discovery and the evaluation of toxicity, clearance, and absorption of lead compounds.
Collapse
Affiliation(s)
- Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Aurino M Kemas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Lena Preiss
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Joanne X Shen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Selgin D Cakal
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Francesco S Paqualini
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Sravan K Goparaju
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Reza Zandi Shafagh
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Johan Ulrik Lind
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| |
Collapse
|
30
|
Hashimoto Y, Michiba K, Maeda K, Kusuhara H. Quantitative prediction of pharmacokinetic properties of drugs in humans: Recent advance in in vitro models to predict the impact of efflux transporters in the small intestine and blood-brain barrier. J Pharmacol Sci 2021; 148:142-151. [PMID: 34924119 DOI: 10.1016/j.jphs.2021.10.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/17/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022] Open
Abstract
Efflux transport systems are essential to suppress the absorption of xenobiotics from the intestinal lumen and protect the critical tissues at the blood-tissue barriers, such as the blood-brain barrier. The function of drug efflux transport is dominated by various transporters. Accumulated clinical evidences have revealed that genetic variations of the transporters, together with coadministered drugs, affect the expression and/or function of transporters and subsequently the pharmacokinetics of substrate drugs. Thus, in the preclinical stage of drug development, quantitative prediction of the impact of efflux transporters as well as that of uptake transporters and metabolic enzymes on the pharmacokinetics of drugs in humans has been performed using various in vitro experimental tools. Various kinds of human-derived cell systems can be applied to the precise prediction of drug transport in humans. Mathematical modeling consisting of each intrinsic metabolic or transport process enables us to understand the disposition of drugs both at the organ level and at the level of the whole body by integrating a variety of experimental results into model parameters. This review focuses on the role of efflux transporters in the intestinal absorption and brain distribution of drugs, in addition to recent advances in predictive tools and methodologies.
Collapse
Affiliation(s)
- Yoshiki Hashimoto
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kazuyoshi Michiba
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kazuya Maeda
- Laboratory of Pharmaceutics, Kitasato University School of Pharmacy, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
31
|
Aguilar C, Alves da Silva M, Saraiva M, Neyazi M, Olsson IAS, Bartfeld S. Organoids as host models for infection biology - a review of methods. Exp Mol Med 2021; 53:1471-1482. [PMID: 34663936 PMCID: PMC8521091 DOI: 10.1038/s12276-021-00629-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/26/2021] [Accepted: 02/24/2021] [Indexed: 01/10/2023] Open
Abstract
Infectious diseases are a major threat worldwide. With the alarming rise of antimicrobial resistance and emergence of new potential pathogens, a better understanding of the infection process is urgently needed. Over the last century, the development of in vitro and in vivo models has led to remarkable contributions to the current knowledge in the field of infection biology. However, applying recent advances in organoid culture technology to research infectious diseases is now taking the field to a higher level of complexity. Here, we describe the current methods available for the study of infectious diseases using organoid cultures.
Collapse
Affiliation(s)
- Carmen Aguilar
- grid.8379.50000 0001 1958 8658Research Centre for Infectious Diseases, Institute for Molecular Infection Biology, Julius Maximilians Universität Wuerzburg, Wuerzburg, Germany
| | - Marta Alves da Silva
- grid.5808.50000 0001 1503 7226i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226IBMC- Instituto de Biologia Celular e Molecular, Universidade do Porto, Porto, Portugal
| | - Margarida Saraiva
- grid.5808.50000 0001 1503 7226i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226IBMC- Instituto de Biologia Celular e Molecular, Universidade do Porto, Porto, Portugal
| | - Mastura Neyazi
- grid.8379.50000 0001 1958 8658Research Centre for Infectious Diseases, Institute for Molecular Infection Biology, Julius Maximilians Universität Wuerzburg, Wuerzburg, Germany
| | - I. Anna S. Olsson
- grid.5808.50000 0001 1503 7226i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226IBMC- Instituto de Biologia Celular e Molecular, Universidade do Porto, Porto, Portugal
| | - Sina Bartfeld
- grid.8379.50000 0001 1958 8658Research Centre for Infectious Diseases, Institute for Molecular Infection Biology, Julius Maximilians Universität Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
32
|
Jelodari S, Sadroddiny E. Decellularization of Small Intestinal Submucosa. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1345:71-84. [PMID: 34582015 DOI: 10.1007/978-3-030-82735-9_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Small intestinal submucosa (SIS) is the most studied extracellular matrix (ECM) for repair and regeneration of different organs and tissues. Promising results of SIS-ECM as a vascular graft, led scientists to examine its applicability for repairing other tissues. Overall results indicated that SIS grafts induce tissue regeneration and remodeling to almost native condition. Investigating immunomodulatory effects of SIS is another interesting field of research. SIS can be utilized in different forms for multiple clinical and experimental studies. The aim of this chapter is to investigate the decellularization process of SIS and its common clinical application.
Collapse
Affiliation(s)
- Sahar Jelodari
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Esmaeil Sadroddiny
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
33
|
Saleh J, Mercier B, Xi W. Bioengineering methods for organoid systems. Biol Cell 2021; 113:475-491. [PMID: 34580889 DOI: 10.1111/boc.202000119] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 08/05/2021] [Accepted: 09/16/2021] [Indexed: 12/23/2022]
Abstract
Organoids have been widely used in fundamental, biomimetic, and therapeutic studies. These multicellular systems form via cell-autonomous self-organization where a cohort of stem cells undergoes in vivo-like proliferation, differentiation, and morphogenesis. They also recapitulate a series of physiological cell organization, complexity and functions that are untouchable by conventional bio-model systems using immortal cell lines. However, the development of organoids is often not easily controlled and their shape and size are yet fully physiological. Recent research has demonstrated that multiple bioengineering tools could be harnessed to control important internal and external cues that dictate stem cell behavior and stem-cell based organoid development. In this review, we introduce the current development of organoid systems and their potentials, as well as their limitations that impede their further utility in research and clinical fields. In comparison to conventional autonomous organoid system, we then review bioengineering approaches that offer improved control over organoid growth and development. We focus on the genetic editing tools that allow the program of build-in responses and phenotypes for organoid systems with enhanced physiological relevance. We also highlight the advances in bioengineering methods to modify cellular external milieus to generate desirable cell composition, 3D micro-architectures, and complex microfluidic systems. We conclude that the emerging biomimetic methods that employ multidisciplinary approaches could prevail in the future development of organoid systems.
Collapse
Affiliation(s)
- Jad Saleh
- Université de Paris, CNRS, Institut Jacques Monod, Paris, France
| | - Barbara Mercier
- Université de Paris, CNRS, Institut Jacques Monod, Paris, France
| | - Wang Xi
- Université de Paris, CNRS, Institut Jacques Monod, Paris, France
| |
Collapse
|
34
|
Chong HB, Youn J, Shin W, Kim HJ, Kim DS. Multiplex recreation of human intestinal morphogenesis on a multi-well insert platform by basolateral convective flow. LAB ON A CHIP 2021; 21:3316-3327. [PMID: 34323906 DOI: 10.1039/d1lc00404b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Here, we report a multiplex culture system that enables simultaneous recreation of multiple replications of the three-dimensional (3D) microarchitecture of the human intestinal epithelium in vitro. The "basolateral convective flow-generating multi-well insert platform (BASIN)" contains 24 nano-porous inserts and an open basolateral chamber applying controllable convective flow in the basolateral compartment that recreates a biomimetic morphogen gradient using a conventional orbital shaker. The mechanistic approach by which the removal of morphogen inhibitors in the basolateral medium can induce intestinal morphogenesis was applied to manipulate the basolateral convective flow in space and time. In a multiplex BASIN, we successfully regenerated a 3D villi-like intestinal microstructure using the Caco-2 human intestinal epithelium that presents high barrier function with minimal insert-to-insert variations. The enhanced cytodifferentiation and proliferation of the 3D epithelial layers formed in the BASIN were visualized with markers of absorptive (villin) and proliferative cells (Ki67). The paracellular transport and efflux profiles of the microengineered 3D epithelial layers in the BASIN confirmed its reproducibility, robustness, and scalability for multiplex biochemical or pharmaceutical studies. Finally, the BASIN was used to investigate the effects of dextran sodium sulfate on the intestinal epithelial barrier and morphology to validate its practical applicability for investigating the effects of external chemicals on the intestinal epithelium and constructing a leaky-gut model. We envision that the BASIN may provide an improved multiplex, scalable, and physiological intestinal epithelial model that is readily accessible to researchers in both basic and applied sciences.
Collapse
Affiliation(s)
- Hyeon Beom Chong
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, South Korea.
| | | | | | | | | |
Collapse
|
35
|
Routier A, Blaizot A, Agossa K, Dubar M. What do we know about the mechanisms of action of probiotics on factors involved in the pathogenesis of periodontitis? A scoping review of in vitro studies. Arch Oral Biol 2021; 129:105196. [PMID: 34153538 DOI: 10.1016/j.archoralbio.2021.105196] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 06/03/2021] [Accepted: 06/07/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Probiotics are increasingly used in oral prevention and treatment conditions, but little is known about their abilities. The aim of this review is to clarify, summarize and disseminate current knowledge about the mode of action of in vitro probiotics on factors involved in the pathogenesis of periodontitis. METHOD 2495 articles were identified in three databases (Medline, Web of Science, SpringerLink) and 26 studies included in this scoping review. RESULTS Twenty-three probiotic species were identified, the majority of which were Lactobacilli or Bifidobacteria. Lactobacillus rhamnosus (30.8 %) and Lactobacillus reuteri (42.3 %) were found to be the two predominantly studied probiotic species and three main mechanisms of action of probiotics could be classified as: (i) modulation of the immuno-inflammatory response, (ii) direct actions of probiotics on periodontopathogens by adhesion or nutritive competitions and/or the secretion of antimicrobial molecules and (iii) indirect actions through environmental modifications. A combination of several probiotic strains seems to be beneficial via synergistic action amplifying the functions of each strain used. However, heterogeneity of the methodologies and probiotic species included in studies leads us to consider the following avenues for future research: (i) implementation of standardized periodontal models as close as possible to in vivo periodontal conditions to identify the functions of each strain for appropriate medication, (ii) updating data about interactions within oral biofilms to identify new candidates and to predict then analyze their behavior within these biofilms. CONCLUSION Probiotics may have their place in the response to inter-individual variability in periodontitis, provided that the choice of the probiotic strain or combination of them will be personalized and optimal for each patient.
Collapse
Affiliation(s)
- Arthur Routier
- School of Dentistry, Lille University Hospital, Lille, France.
| | - Alessandra Blaizot
- Department of Public Health, Faculty of Dental Surgery, Lille University Hospital, Lille, France.
| | - Kevimy Agossa
- Department of Periodontology, Faculty of Dental Surgery, Lille University Hospital, Lille, France; University of Lille, Inserm, Lille University Hospital, U1008, F-59000 Lille, France.
| | - Marie Dubar
- Department of Periodontology, Faculty of Dental Surgery, Lille University Hospital, Lille, France; University of Lille, Inserm, Lille University Hospital, UMR-S 1172, F-59000 Lille, France.
| |
Collapse
|
36
|
Zietek T, Boomgaarden WAD, Rath E. Drug Screening, Oral Bioavailability and Regulatory Aspects: A Need for Human Organoids. Pharmaceutics 2021; 13:1280. [PMID: 34452240 PMCID: PMC8399541 DOI: 10.3390/pharmaceutics13081280] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/12/2021] [Accepted: 08/14/2021] [Indexed: 12/31/2022] Open
Abstract
The intestinal epithelium critically contributes to oral bioavailability of drugs by constituting an important site for drug absorption and metabolism. In particular, intestinal epithelial cells (IEC) actively serve as gatekeepers of drug and nutrient availability. IECs' transport processes and metabolism are interrelated to the whole-body metabolic state and represent potential points of origin as well as therapeutic targets for a variety of diseases. Human intestinal organoids represent a superior model of the intestinal epithelium, overcoming limitations of currently used in vitro models. Caco-2 cells or rodent explant models face drawbacks such as their cancer and non-human origin, respectively, but are commonly used to study intestinal nutrient absorption, enterocyte metabolism and oral drug bioavailability, despite poorly correlative data. In contrast, intestinal organoids allow investigating distinct aspects of bioavailability including spatial resolution of transport, inter-individual differences and high-throughput screenings. As several countries have already developed strategic roadmaps to phase out animal experiments for regulatory purposes, intestinal organoid culture and organ-on-a-chip technology in combination with in silico approaches are roads to go in the preclinical and regulatory setup and will aid implementing the 3Rs (reduction, refinement and replacement) principle in basic science.
Collapse
Affiliation(s)
- Tamara Zietek
- Doctors against Animal Experiments, 51143 Köln, Germany
| | | | - Eva Rath
- Chair of Nutrition and Immunology, Technische Universität München, 85354 Freising, Germany
| |
Collapse
|
37
|
Signore MA, De Pascali C, Giampetruzzi L, Siciliano PA, Francioso L. Gut-on-Chip microphysiological systems: Latest advances in the integration of sensing strategies and adoption of mature detection mechanisms. SENSING AND BIO-SENSING RESEARCH 2021. [DOI: 10.1016/j.sbsr.2021.100443] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
38
|
Kasendra M, Troutt M, Broda T, Bacon WC, Wang TC, Niland JC, Helmrath MA. Intestinal organoids: roadmap to the clinic. Am J Physiol Gastrointest Liver Physiol 2021; 321:G1-G10. [PMID: 33950707 PMCID: PMC8321798 DOI: 10.1152/ajpgi.00425.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 01/31/2023]
Abstract
Recent advances in intestinal organoid research, along with encouraging preclinical proof-of-concept studies, have revealed significant therapeutic potential for induced pluripotent stem cell (iPSC)-derived organoids in the healing and replacement of severely injured or diseased bowel (Finkbeiner et al. Biol Open 4: 1462-1472, 2015; Kitano et al. Nat Commun 8: 765, 2017; Cruz-Acuna et al. Nat Cell Biol 19: 1326-1335, 2017). To fully realize the tremendous promise of stem cell organoid-based therapies, careful planning aligned with significant resources and efforts must be devoted demonstrating their safety and efficacy to meet critical regulatory requirements. Early recognition of the inherent preclinical and clinical obstacles that occur with the novel use of pluripotent stem cell-derived products will accelerate their bench-to-bedside translation (Neofytou et al. J Clin Invest 125: 2551-2557, 2015; O'Brien et al. Stem Cell Res Ther 6: 146, 2015; Ouseph et al. Cytotherapy 17: 339-343, 2015). To overcome many of these hurdles, a close and effective collaboration is needed between experts from various disciplines, including basic and clinical research, product development and manufacturing, quality assurance and control, and regulatory affairs. Therefore, the purpose of this article is to outline the critical areas and challenges that must be addressed when transitioning laboratory-based discovery, through an investigational new drug (IND) application to first-in-human clinical trial, and to encourage investigators to consider the required regulatory steps from the earliest stage of the translational process. The ultimate goal is to provide readers with a draft roadmap that they could use while navigating this exciting cell therapy space.
Collapse
Affiliation(s)
- Magdalena Kasendra
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Misty Troutt
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Taylor Broda
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - W Clark Bacon
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Columbia University Medical Center, New York City, New York
| | - Joyce C Niland
- Department of Diabetes and Cancer Discovery Science, City of Hope, Duarte, California
| | - Michael A Helmrath
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
39
|
Younes M, Aggett P, Aguilar F, Crebelli R, Dusemund B, Filipič M, Frutos MJ, Galtier P, Gundert‐Remy U, Kuhnle GG, Lambré C, Leblanc J, Lillegaard IT, Moldeus P, Mortensen A, Oskarsson A, Stankovic I, Waalkens‐Berendsen I, Woutersen RA, Wright M, Di Domenico A, Fairweather‐Tait S, McArdle HJ, Smeraldi C, Gott D. Guidance on safety evaluation of sources of nutrients and bioavailability of nutrient from the sources (Revision 1). EFSA J 2021; 19:e06552. [PMID: 33815621 PMCID: PMC8002907 DOI: 10.2903/j.efsa.2021.6552] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
[Table: see text] This guidance describes the scientific data required to allow an evaluation of the safety of new substances that are proposed for use as sources of nutrients in food supplements, foods for the general population or foods for specific groups and an assessment of the bioavailability of the nutrient from the proposed source. This guidance describes the scientific data required to allow an evaluation of the safety of the source within the established framework for risk assessment of food additives and novel food ingredients and the bioavailability of the nutrient from this source. This document is arranged in five main sections: one on technical data aimed at characterising the proposed source and at identifying potential hazards resulting from its manufacture and stability in food; one on existing authorisations and evaluation, providing an overview of previous assessments on the proposed source and their conclusions; one on proposed uses and exposure assessment section, allowing an estimate of the dietary exposure to the source and the nutrient based on the proposed uses and use levels; one on toxicological data, describing approaches which can be used to identify (in conjunction with data on manufacture and composition) and to characterise hazards of the source and any relevant breakdown products; the final section on bioavailability focuses on determining the extent to which the nutrient from the proposed source is available for use by the body in comparison with one or more forms of the same nutrient that are already permitted for use on the positive lists. This guidance was adopted by the Panel on Food Additives and Nutrient Sources added to Food (ANS Panel) on 16 May 2018. Upon request from EFSA, the present guidance has been revised to inform applicants of new provisions set out in Regulation (EC) No 178/2002, as amended by Regulation (EU) 2019/1381 on the transparency and sustainability of the EU risk assessment in the food chain.
Collapse
|
40
|
Heydarian M, Schweinlin M, Schwarz T, Rawal R, Walles H, Metzger M, Rudel T, Kozjak-Pavlovic V. Triple co-culture and perfusion bioreactor for studying the interaction between Neisseria gonorrhoeae and neutrophils: A novel 3D tissue model for bacterial infection and immunity. J Tissue Eng 2021; 12:2041731420988802. [PMID: 33796248 PMCID: PMC7970704 DOI: 10.1177/2041731420988802] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/31/2020] [Indexed: 01/13/2023] Open
Abstract
Gonorrhea, a sexually transmitted disease caused by the bacteria Neisseria gonorrhoeae, is characterized by a large number of neutrophils recruited to the site of infection. Therefore, proper modeling of the N. gonorrhoeae interaction with neutrophils is very important for investigating and understanding the mechanisms that gonococci use to evade the immune response. We have used a combination of a unique human 3D tissue model together with a dynamic culture system to study neutrophil transmigration to the site of N. gonorrhoeae infection. The triple co-culture model consisted of epithelial cells (T84 human colorectal carcinoma cells), human primary dermal fibroblasts, and human umbilical vein endothelial cells on a biological scaffold (SIS). After the infection of the tissue model with N. gonorrhoeae, we introduced primary human neutrophils to the endothelial side of the model using a perfusion-based bioreactor system. By this approach, we were able to demonstrate the activation and transmigration of neutrophils across the 3D tissue model and their recruitment to the site of infection. In summary, the triple co-culture model supplemented by neutrophils represents a promising tool for investigating N. gonorrhoeae and other bacterial infections and interactions with the innate immunity cells under conditions closely resembling the native tissue environment.
Collapse
Affiliation(s)
| | - Matthias Schweinlin
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Thomas Schwarz
- Translational Centre Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research (ISC), Würzburg, Bayern, Germany
| | - Ravisha Rawal
- Biocenter, Chair of Microbiology, University of Würzburg, Würzburg, Germany
| | - Heike Walles
- Research Center "Dynamic Systems: Systems Engineering" (CDS), Otto von-Guericke-University, Magdeburg, Sachsen-Anhalt, Germany
| | - Marco Metzger
- Translational Centre Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research (ISC), Würzburg, Bayern, Germany
| | - Thomas Rudel
- Biocenter, Chair of Microbiology, University of Würzburg, Würzburg, Germany
| | | |
Collapse
|
41
|
Kessie DK, Lodes N, Oberwinkler H, Goldman WE, Walles T, Steinke M, Gross R. Activity of Tracheal Cytotoxin of Bordetella pertussis in a Human Tracheobronchial 3D Tissue Model. Front Cell Infect Microbiol 2021; 10:614994. [PMID: 33585281 PMCID: PMC7873972 DOI: 10.3389/fcimb.2020.614994] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/02/2020] [Indexed: 11/13/2022] Open
Abstract
Bordetella pertussis is a highly contagious pathogen which causes whooping cough in humans. A major pathophysiology of infection is the extrusion of ciliated cells and subsequent disruption of the respiratory mucosa. Tracheal cytotoxin (TCT) is the only virulence factor produced by B. pertussis that has been able to recapitulate this pathology in animal models. This pathophysiology is well characterized in a hamster tracheal model, but human data are lacking due to scarcity of donor material. We assessed the impact of TCT and lipopolysaccharide (LPS) on the functional integrity of the human airway mucosa by using in vitro airway mucosa models developed by co-culturing human tracheobronchial epithelial cells and human tracheobronchial fibroblasts on porcine small intestinal submucosa scaffold under airlift conditions. TCT and LPS either alone and in combination induced blebbing and necrosis of the ciliated epithelia. TCT and LPS induced loss of ciliated epithelial cells and hyper-mucus production which interfered with mucociliary clearance. In addition, the toxins had a disruptive effect on the tight junction organization, significantly reduced transepithelial electrical resistance and increased FITC-Dextran permeability after toxin incubation. In summary, the results indicate that TCT collaborates with LPS to induce the disruption of the human airway mucosa as reported for the hamster tracheal model.
Collapse
Affiliation(s)
- David K. Kessie
- Biocentre, Chair of Microbiology, University of Würzburg, Würzburg, Germany
| | - Nina Lodes
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Heike Oberwinkler
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - William E. Goldman
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Thorsten Walles
- Department of Thoracic Surgery, University of Medicine Magdeburg, Magdeburg, Germany
| | - Maria Steinke
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Roy Gross
- Biocentre, Chair of Microbiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
42
|
Meir M, Salm J, Fey C, Schweinlin M, Kollmann C, Kannapin F, Germer CT, Waschke J, Beck C, Burkard N, Metzger M, Schlegel N. Enteroids Generated from Patients with Severe Inflammation in Crohn's Disease Maintain Alterations of Junctional Proteins. J Crohns Colitis 2020; 14:1473-1487. [PMID: 32342109 DOI: 10.1093/ecco-jcc/jjaa085] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The mechanisms underlying loss of intestinal epithelial barrier [IEB] function in Crohn's disease [CD] are poorly understood. We tested whether human enteroids generated from isolated intestinal crypts of CD patients serve as an appropriate in vitro model to analyse changes of IEB proteins observed in patients' specimens. METHODS Gut samples from CD patients and healthy individuals who underwent surgery were collected. Enteroids were generated from intestinal crypts and analyses of junctional proteins in comparison to full wall samples were performed. RESULTS Histopathology confirmed the presence of CD and the extent of inflammation in intestinal full wall sections. As revealed by immunostaining and Western blot analysis, profound changes in expression patterns of tight junction, adherens junction and desmosomal proteins were observed in full wall specimens when CD was present. Unexpectedly, when enteroids were generated from specimens of CD patients with severe inflammation, alterations of most tight junction proteins and the majority of changes in desmosomal proteins but not E-cadherin were maintained under culture conditions. Importantly, these changes were maintained without any additional stimulation of cytokines. Interestingly, qRT-PCR demonstrated that mRNA levels of junctional proteins were not different when enteroids from CD patients were compared to enteroids from healthy controls. CONCLUSIONS These data indicate that enteroids generated from patients with severe inflammation in CD maintain some characteristics of intestinal barrier protein changes on a post-transcriptional level. The enteroid in vitro model represents an appropriate tool to gain further cellular and molecular insights into the pathogenesis of barrier dysfunction in CD.
Collapse
Affiliation(s)
- Michael Meir
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Jonas Salm
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Christina Fey
- Chair for Tissue Engineering and Regenerative Medicine, Wuerzburg, Germany
| | | | - Catherine Kollmann
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Felix Kannapin
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Christoph-Thomas Germer
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Jens Waschke
- Institute of Anatomy and Cell Biology, Ludwig-Maximilians-University, Munich, Germany
| | | | - Natalie Burkard
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Marco Metzger
- Chair for Tissue Engineering and Regenerative Medicine, Wuerzburg, Germany.,Fraunhofer Institute for Silicate Research ISC, Translational Centre for Regenerative Therapies TLC-RT, Wuerzburg, Germany
| | - Nicolas Schlegel
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
43
|
Meran L, Massie I, Campinoti S, Weston AE, Gaifulina R, Tullie L, Faull P, Orford M, Kucharska A, Baulies A, Novellasdemunt L, Angelis N, Hirst E, König J, Tedeschi AM, Pellegata AF, Eli S, Snijders AP, Collinson L, Thapar N, Thomas GMH, Eaton S, Bonfanti P, De Coppi P, Li VSW. Engineering transplantable jejunal mucosal grafts using patient-derived organoids from children with intestinal failure. Nat Med 2020; 26:1593-1601. [PMID: 32895569 PMCID: PMC7116539 DOI: 10.1038/s41591-020-1024-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 07/17/2020] [Indexed: 11/08/2022]
Abstract
Intestinal failure, following extensive anatomical or functional loss of small intestine, has debilitating long-term consequences for children1. The priority of patient care is to increase the length of functional intestine, particularly the jejunum, to promote nutritional independence2. Here we construct autologous jejunal mucosal grafts using biomaterials from pediatric patients and show that patient-derived organoids can be expanded efficiently in vitro. In parallel, we generate decellularized human intestinal matrix with intact nanotopography, which forms biological scaffolds. Proteomic and Raman spectroscopy analyses reveal highly analogous biochemical profiles of human small intestine and colon scaffolds, indicating that they can be used interchangeably as platforms for intestinal engineering. Indeed, seeding of jejunal organoids onto either type of scaffold reliably reconstructs grafts that exhibit several aspects of physiological jejunal function and that survive to form luminal structures after transplantation into the kidney capsule or subcutaneous pockets of mice for up to 2 weeks. Our findings provide proof-of-concept data for engineering patient-specific jejunal grafts for children with intestinal failure, ultimately aiding in the restoration of nutritional autonomy.
Collapse
Affiliation(s)
- Laween Meran
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK
- Stem Cell and Regenerative Medicine Section, DBC, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Isobel Massie
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK
| | - Sara Campinoti
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK
- Stem Cell and Regenerative Medicine Section, DBC, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Anne E Weston
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK
| | - Riana Gaifulina
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Lucinda Tullie
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK
- Stem Cell and Regenerative Medicine Section, DBC, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Peter Faull
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK
| | - Michael Orford
- Stem Cell and Regenerative Medicine Section, DBC, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Anna Kucharska
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK
| | - Anna Baulies
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK
| | - Laura Novellasdemunt
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK
| | - Nikolaos Angelis
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK
| | - Elizabeth Hirst
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK
| | - Julia König
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK
| | - Alfonso Maria Tedeschi
- Stem Cell and Regenerative Medicine Section, DBC, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Alessandro Filippo Pellegata
- Stem Cell and Regenerative Medicine Section, DBC, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Susanna Eli
- Stem Cell and Regenerative Medicine Section, DBC, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Ambrosius P Snijders
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK
| | - Lucy Collinson
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK
| | - Nikhil Thapar
- Stem Cell and Regenerative Medicine Section, DBC, Great Ormond Street Institute of Child Health, University College London, London, UK
- Gastroenterology, Hepatology and Liver Transplant, Queensland Children's Hospital, Brisbane, Queensland, Australia
| | - Geraint M H Thomas
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Simon Eaton
- Stem Cell and Regenerative Medicine Section, DBC, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Paola Bonfanti
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK
- Stem Cell and Regenerative Medicine Section, DBC, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Paolo De Coppi
- Stem Cell and Regenerative Medicine Section, DBC, Great Ormond Street Institute of Child Health, University College London, London, UK.
- Specialist Neonatal and Paediatric Service, Great Ormond Street Hospital, London, UK.
| | - Vivian S W Li
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
44
|
Zietek T, Giesbertz P, Ewers M, Reichart F, Weinmüller M, Urbauer E, Haller D, Demir IE, Ceyhan GO, Kessler H, Rath E. Organoids to Study Intestinal Nutrient Transport, Drug Uptake and Metabolism - Update to the Human Model and Expansion of Applications. Front Bioeng Biotechnol 2020; 8:577656. [PMID: 33015026 PMCID: PMC7516017 DOI: 10.3389/fbioe.2020.577656] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/19/2020] [Indexed: 12/22/2022] Open
Abstract
Intestinal transport and sensing processes and their interconnection to metabolism are relevant to pathologies such as malabsorption syndromes, inflammatory diseases, obesity and type 2 diabetes. Constituting a highly selective barrier, intestinal epithelial cells absorb, metabolize, and release nutrients into the circulation, hence serving as gatekeeper of nutrient availability and metabolic health for the whole organism. Next to nutrient transport and sensing functions, intestinal transporters including peptide transporter 1 (PEPT1) are involved in the absorption of drugs and prodrugs, including certain inhibitors of angiotensin-converting enzyme, protease inhibitors, antivirals, and peptidomimetics like β-lactam antibiotics. Here, we verify the applicability of 3D organoids for in vitro investigation of intestinal biochemical processes related to transport and metabolism of nutrients and drugs. Establishing a variety of methodologies including illustration of transporter-mediated nutrient and drug uptake and metabolomics approaches, we highlight intestinal organoids as robust and reliable tool in this field of research. Currently used in vitro models to study intestinal nutrient absorption, drug transport and enterocyte metabolism, such as Caco-2 cells or rodent explant models are of limited value due to their cancer and non-human origin, respectively. Particularly species differences result in poorly correlative data and findings obtained in these models cannot be extrapolated reliably to humans, as indicated by high failure rates in drug development pipelines. In contrast, human intestinal organoids represent a superior model of the intestinal epithelium and might help to implement the 3Rs (Reduction, Refinement and Replacement) principle in basic science as well as the preclinical and regulatory setup.
Collapse
Affiliation(s)
- Tamara Zietek
- Chair of Nutritional Physiology, Technische Universität München, Munich, Germany
| | - Pieter Giesbertz
- Chair of Nutritional Physiology, Technische Universität München, Munich, Germany
| | - Maren Ewers
- Pediatric Nutritional Medicine, Klinikum Rechts der Isar, Else Kröner-Fresenius-Zentrum für Ernährungsmedizin, Technische Universität München, Munich, Germany
| | - Florian Reichart
- Institute for Advanced Study, Department of Chemistry and Center for Integrated Protein Science (CIPSM), Technische Universität München, Garching, Germany
| | - Michael Weinmüller
- Institute for Advanced Study, Department of Chemistry and Center for Integrated Protein Science (CIPSM), Technische Universität München, Garching, Germany
| | - Elisabeth Urbauer
- Chair of Nutrition and Immunology, Technische Universität München, Munich, Germany
| | - Dirk Haller
- Chair of Nutrition and Immunology, Technische Universität München, Munich, Germany.,ZIEL Institute for Food and Health, Technische Universität München, Munich, Germany
| | - Ihsan Ekin Demir
- Department of Surgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany.,Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey.,German Cancer Consortium (DKTK), Munich, Germany.,CRC 1321 Modeling and Targeting Pancreatic Cancer, Klinikum rechts der Isar, School of Medicine, Technische Universität München, Munich, Germany
| | - Güralp O Ceyhan
- Department of Surgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany.,Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Horst Kessler
- Institute for Advanced Study, Department of Chemistry and Center for Integrated Protein Science (CIPSM), Technische Universität München, Garching, Germany
| | - Eva Rath
- Chair of Nutrition and Immunology, Technische Universität München, Munich, Germany
| |
Collapse
|
45
|
Darling NJ, Mobbs CL, González-Hau AL, Freer M, Przyborski S. Bioengineering Novel in vitro Co-culture Models That Represent the Human Intestinal Mucosa With Improved Caco-2 Structure and Barrier Function. Front Bioeng Biotechnol 2020; 8:992. [PMID: 32984279 PMCID: PMC7487342 DOI: 10.3389/fbioe.2020.00992] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 07/29/2020] [Indexed: 12/17/2022] Open
Abstract
The Caco-2 monolayer is the most widely used in vitro model of the human intestinal mucosa to study absorption. However, models lack communication from other cells present in the native intestine, such as signals from fibroblasts in the lamina propria. In this study, we have investigated the effects of fibroblasts upon the Caco-2 epithelium through two mechanisms: indirect signaling from fibroblasts and direct contact with fibroblasts. Culture of Caco-2 cells with paracrine signals from fibroblasts, through the use of conditioned media, did not induce a significant change in epithelial cell morphology or function. To examine the effects of direct contact between the epithelium and fibroblasts, we developed novel, humanized three-dimensional (3D) co-culture models whereby Caco-2 cells are grown on the surface of a subepithelial-like tissue construct containing intestinal or dermal fibroblasts. In our models, we observed endogenous extracellular matrix production from the fibroblasts that provides support to the above epithelium. The Caco-2 epithelium displayed morphological changes in 3D co-culture including enhanced polarization and the formation of a basement membrane-like attachment to the underlying stromal compartment. An important structural alteration was the significantly straightened lateral membrane that closely mimics the structure of the in vivo intestinal mucosa. This enhanced lateral membrane phenotype, in correlation with an reduction in TEER to levels more similar to the human intestine, is thought to be responsible for the increased paracellular permeability observed in 3D co-cultures. Our results demonstrate that direct contact between epithelial and mesenchymal cells results in an enhanced epithelial barrier. The in vitro models described herein have the potential to be used for studying intestinal epithelial-fibroblast interactions and could provide more accurate tools for drug permeability studies.
Collapse
Affiliation(s)
- Nicole J Darling
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - Claire L Mobbs
- Department of Biosciences, Durham University, Durham, United Kingdom.,Reprocell Europe Ltd, Sedgefield, United Kingdom
| | | | - Matthew Freer
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - Stefan Przyborski
- Department of Biosciences, Durham University, Durham, United Kingdom.,Reprocell Europe Ltd, Sedgefield, United Kingdom
| |
Collapse
|
46
|
Costa J, Almonti V, Cacopardo L, Poli D, Rapposelli S, Ahluwalia A. Investigating Curcumin/Intestinal Epithelium Interaction in a Millifluidic Bioreactor. Bioengineering (Basel) 2020; 7:bioengineering7030100. [PMID: 32858899 PMCID: PMC7552770 DOI: 10.3390/bioengineering7030100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/14/2020] [Accepted: 08/18/2020] [Indexed: 01/25/2023] Open
Abstract
Multidrug resistance is still an obstacle for chemotherapeutic treatments. One of the proteins involved in this phenomenon is the P-glycoprotein, P-gp, which is known to be responsible for the efflux of therapeutic substances from the cell cytoplasm. To date, the identification of a drug that can efficiently inhibit P-gp activity remains a challenge, nevertheless some studies have identified natural compounds suitable for that purpose. Amongst them, curcumin has shown an inhibitory effect on the protein in in vitro studies using Caco-2 cells. To understand if flow can modulate the influence of curcumin on the protein's activity, we studied the uptake of a P-gp substrate under static and dynamic conditions. Caco-2 cells were cultured in bioreactors and in Transwells and the basolateral transport of rhodamine-123 was assessed in the two systems as a function of the P-gp activity. Experiments were performed with and without pre-treatment of the cells with an extract of curcumin or an arylmethyloxy-phenyl derivative to evaluate the inhibitory effect of the natural substance with respect to a synthetic compound. The results indicated that the P-gp activity of the cells cultured in the bioreactors was intrinsically lower, and that the effect of both natural and synthetic inhibitors was up modulated by the presence of flow. Our study underlies the fact that the use of more sophisticated and physiologically relevant in vitro models can bring new insights on the therapeutic effects of natural substances such as curcumin.
Collapse
Affiliation(s)
- Joana Costa
- Research Center “E. Piaggio”, University of Pisa, 56122 Pisa, Italy; (L.C.); (D.P.); (A.A.)
- Correspondence:
| | - Vanessa Almonti
- LARF-DIMES, Department of Experimental Medicine, University of Genoa, 16126 Genoa, Italy;
- Centro 3R (Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research), 56122 Pisa, Italy;
| | - Ludovica Cacopardo
- Research Center “E. Piaggio”, University of Pisa, 56122 Pisa, Italy; (L.C.); (D.P.); (A.A.)
| | - Daniele Poli
- Research Center “E. Piaggio”, University of Pisa, 56122 Pisa, Italy; (L.C.); (D.P.); (A.A.)
| | - Simona Rapposelli
- Centro 3R (Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research), 56122 Pisa, Italy;
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | - Arti Ahluwalia
- Research Center “E. Piaggio”, University of Pisa, 56122 Pisa, Italy; (L.C.); (D.P.); (A.A.)
- Centro 3R (Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research), 56122 Pisa, Italy;
| |
Collapse
|
47
|
Youhanna S, Lauschke VM. The Past, Present and Future of Intestinal In Vitro Cell Systems for Drug Absorption Studies. J Pharm Sci 2020; 110:50-65. [PMID: 32628951 DOI: 10.1016/j.xphs.2020.07.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/02/2020] [Accepted: 07/02/2020] [Indexed: 12/23/2022]
Abstract
The intestinal epithelium acts as a selective barrier for the absorption of water, nutrients and orally administered drugs. To evaluate the gastrointestinal permeability of a candidate molecule, scientists and drug developers have a multitude of cell culture models at their disposal. Static transwell cultures constitute the most extensively characterized intestinal in vitro system and can accurately categorize molecules into low, intermediate and high permeability compounds. However, they lack key aspects of intestinal physiology, including the cellular complexity of the intestinal epithelium, flow, mechanical strain, or interactions with intestinal mucus and microbes. To emulate these features, a variety of different culture paradigms, including microfluidic chips, organoids and intestinal slice cultures have been developed. Here, we provide an updated overview of intestinal in vitro cell culture systems and critically review their suitability for drug absorption studies. The available data show that these advanced culture models offer impressive possibilities for emulating intestinal complexity. However, there is a paucity of systematic absorption studies and benchmarking data and it remains unclear whether the increase in model complexity and costs translates into improved drug permeability predictions. In the absence of such data, conventional static transwell cultures remain the current gold-standard paradigm for drug absorption studies.
Collapse
Affiliation(s)
- Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden.
| |
Collapse
|
48
|
Corrò C, Novellasdemunt L, Li VSW. A brief history of organoids. Am J Physiol Cell Physiol 2020; 319:C151-C165. [PMID: 32459504 PMCID: PMC7468890 DOI: 10.1152/ajpcell.00120.2020] [Citation(s) in RCA: 224] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/12/2020] [Accepted: 05/26/2020] [Indexed: 12/22/2022]
Abstract
In vitro cell cultures are crucial research tools for modeling human development and diseases. Although the conventional monolayer cell cultures have been widely used in the past, the lack of tissue architecture and complexity of such model fails to inform the true biological processes in vivo. Recent advances in the organoid technology have revolutionized the in vitro culture tools for biomedical research by creating powerful three-dimensional (3D) models to recapitulate the cellular heterogeneity, structure, and functions of the primary tissues. Such organoid technology enables researchers to recreate human organs and diseases in a dish and thus holds great promises for many translational applications such as regenerative medicine, drug discovery, and precision medicine. In this review, we provide an overview of the organoid history and development. We discuss the strengths and limitations of organoids as well as their potential applications in the laboratory and the clinic.
Collapse
Affiliation(s)
- Claudia Corrò
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London United Kingdom
| | - Laura Novellasdemunt
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London United Kingdom
| | - Vivian S W Li
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London United Kingdom
| |
Collapse
|
49
|
Fey C, Betz J, Rosenbaum C, Kralisch D, Vielreicher M, Friedrich O, Metzger M, Zdzieblo D. Bacterial nanocellulose as novel carrier for intestinal epithelial cells in drug delivery studies. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 109:110613. [DOI: 10.1016/j.msec.2019.110613] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 12/21/2019] [Accepted: 12/26/2019] [Indexed: 01/14/2023]
|
50
|
Berger C, Bjørlykke Y, Hahn L, Mühlemann M, Kress S, Walles H, Luxenhofer R, Ræder H, Metzger M, Zdzieblo D. Matrix decoded - A pancreatic extracellular matrix with organ specific cues guiding human iPSC differentiation. Biomaterials 2020; 244:119766. [PMID: 32199284 DOI: 10.1016/j.biomaterials.2020.119766] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/29/2019] [Accepted: 01/04/2020] [Indexed: 12/19/2022]
Abstract
The extracellular matrix represents a dynamic microenvironment regulating essential cell functions in vivo. Tissue engineering approaches aim to recreate the native niche in vitro using biological scaffolds generated by organ decellularization. So far, the organ specific origin of such scaffolds was less considered and potential consequences for in vitro cell culture remain largely elusive. Here, we show that organ specific cues of biological scaffolds affect cellular behavior. In detail, we report on the generation of a well-preserved pancreatic bioscaffold and introduce a scoring system allowing standardized inter-study quality assessment. Using multiple analysis tools for in-depth-characterization of the biological scaffold, we reveal unique compositional, physico-structural, and biophysical properties. Finally, we prove the functional relevance of the biological origin by demonstrating a regulatory effect of the matrix on multi-lineage differentiation of human induced pluripotent stem cells emphasizing the significance of matrix specificity for cellular behavior in artificial microenvironments.
Collapse
Affiliation(s)
- Constantin Berger
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Yngvild Bjørlykke
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Lukas Hahn
- Functional Polymer Materials, Department of Chemistry and Pharmacy and Bavarian Polymer Institute, Würzburg University, Würzburg, Germany
| | - Markus Mühlemann
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Sebastian Kress
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Heike Walles
- Translational Center Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research ISC, Würzburg, Germany; Otto-von Guericke University, Core Facility Tissue Engineering, Magdeburg, Germany
| | - Robert Luxenhofer
- Functional Polymer Materials, Department of Chemistry and Pharmacy and Bavarian Polymer Institute, Würzburg University, Würzburg, Germany
| | - Helge Ræder
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Marco Metzger
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany; Translational Center Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research ISC, Würzburg, Germany
| | - Daniela Zdzieblo
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany; Translational Center Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research ISC, Würzburg, Germany.
| |
Collapse
|