1
|
Michielon E, Boninsegna M, Waaijman T, Fassini D, Spiekstra SW, Cramer J, Gaudriault P, Kodolányi J, de Gruijl TD, Homs-Corbera A, Gibbs S. Environmentally Controlled Microfluidic System Enabling Immune Cell Flow and Activation in an Endothelialised Skin-On-Chip. Adv Healthc Mater 2024; 13:e2400750. [PMID: 39370595 PMCID: PMC11582514 DOI: 10.1002/adhm.202400750] [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: 02/27/2024] [Revised: 09/17/2024] [Indexed: 10/08/2024]
Abstract
Integration of reconstructed human skin (RhS) into organ-on-chip (OoC) platforms addresses current limitations imposed by static culturing. This innovation, however, is not without challenges. Microfluidic devices, while powerful, often encounter usability, robustness, and gas bubble issues that hinder large-scale high-throughput setups. This study aims to develop a novel re-usable multi-well microfluidic adaptor (MMA) with the objective to provide a flexible tool for biologists implementing complex 3D biological models (e.g., skin) while enabling simultaneous user control over temperature, medium flow, oxigen (O2), nitrogen (N2), and carbon dioxide (CO2) without the need for an incubator. The presented MMA device is designed to be compatible with standard, commercially available 6-well multi-well plates (6MWPs) and 12-well transwells. This MMA-6MWP setup is employed to generate a skin-on-chip (SoC). RhS viability is maintained under flow for three days and their morphology closely resembles that of native human skin. A proof-of-concept study demonstrates the system's potential in toxicology applications by combining endothelialised RhS with flowing immune cells. This dynamic setting activates the monocyte-like MUTZ-3 cells (CD83 and CD86 upregulation) upon topical exposure of RhS to a sensitizer, revealing the MMA-6MWP's unique capabilities compared to static culturing, where such activation is absent.
Collapse
Affiliation(s)
- Elisabetta Michielon
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Amsterdam institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, Amsterdam UMC, Vrije Universiteit, Amsterdam, 1081 HV, The Netherlands
| | - Matteo Boninsegna
- Cherry Biotech SAS, 14 Rue De La Beaune, Bâtiment A, 2ème Étage, Montreuil, 93100, France
- Department of Physics, Bielefeld University, Universitätsstr 25, 33615, Bielefeld, Germany
| | - Taco Waaijman
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Amsterdam institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
| | - Dario Fassini
- Cherry Biotech SAS, 14 Rue De La Beaune, Bâtiment A, 2ème Étage, Montreuil, 93100, France
| | - Sander W Spiekstra
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Amsterdam institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
| | - Jeremy Cramer
- Cherry Biotech SAS, 14 Rue De La Beaune, Bâtiment A, 2ème Étage, Montreuil, 93100, France
| | - Pierre Gaudriault
- Cherry Biotech SAS, 14 Rue De La Beaune, Bâtiment A, 2ème Étage, Montreuil, 93100, France
| | - János Kodolányi
- Department of Dental Material Science, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, 1081 LA, The Netherlands
| | - Tanja D de Gruijl
- Amsterdam institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, Amsterdam UMC, Vrije Universiteit, Amsterdam, 1081 HV, The Netherlands
- Department of Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
| | - Antoni Homs-Corbera
- Cherry Biotech SAS, 14 Rue De La Beaune, Bâtiment A, 2ème Étage, Montreuil, 93100, France
| | - Susan Gibbs
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Amsterdam institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, 1081 LA, The Netherlands
| |
Collapse
|
2
|
Weitzberg E, Ingelman-Sundberg M, Lundberg JO, Engberg G, Schulte G, Lauschke VM. The 75-Year Anniversary of the Department of Physiology and Pharmacology at Karolinska Institutet-Examples of Recent Accomplishments and Future Perspectives. Pharmacol Rev 2024; 76:1089-1101. [PMID: 39414365 DOI: 10.1124/pharmrev.124.001433] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 09/09/2024] [Indexed: 10/18/2024] Open
Abstract
Karolinska Institutet is a medical university encompassing 21 departments distributed across three departmental or campus groups. Pharmacological research has a long and successful tradition at the institute with a multitude of seminal findings in the areas of neuronal control of vasodilatation, cardiovascular pharmacology, neuropsychopharmacology, receptor pharmacology, and pharmacogenomics that resulted in, among many other recognitions, two Nobel prizes in Physiology and Medicine, one in 1970 to Ulf von Euler for his discovery of the processes involved in storage, release, and inactivation of neurotransmitters and the other in 1982 to Sune Bergström and Bengt Samuelsson for their work on prostaglandins and the discovery of leukotrienes. Pharmacology at Karolinska Institutet has over the last decade been ranked globally among the top 10 according to the QS World University Ranking. With the Department of Physiology and Pharmacology now celebrating its 75-year anniversary, we wanted to take this as an opportunity to showcase recent research achievements and how they paved the way for current activities at the department. We emphasize examples from preclinical and clinical research where the dpartment's integrative environment and robust infrastructure have successfully facilitated the translation of findings into clinical applications and patient benefits. The close collaboration between preclinical scientists and clinical researchers across various disciplines, along with a strong network of partnerships within the department and beyond, positions us to continue leading world-class pharmacological research at the Department of Physiology and Pharmacology for decades to come. SIGNIFICANCE STATEMENT: Pharmacological research at Karolinska Institutet has a long and successful history. Given the 75-year anniversary of the Department of Physiology and Pharmacology, this perspective provides an overview of recent departmental achievements and future trajectories. For these developments, interdisciplinary and intersectoral collaborations and a clear focus on result translation are key elements to continue its legacy of world-leading pharmacological research.
Collapse
Affiliation(s)
- Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (E.W., M.I.-S., J.O.L., G.E., G.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.)
| | - Magnus Ingelman-Sundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (E.W., M.I.-S., J.O.L., G.E., G.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.)
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (E.W., M.I.-S., J.O.L., G.E., G.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.)
| | - Göran Engberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (E.W., M.I.-S., J.O.L., G.E., G.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.)
| | - Gunnar Schulte
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (E.W., M.I.-S., J.O.L., G.E., G.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 (E.W., M.I.-S., J.O.L., G.E., G.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
|
3
|
Du X, Jia H, Chang Y, Zhao Y, Song J. Progress of organoid platform in cardiovascular research. Bioact Mater 2024; 40:88-103. [PMID: 38962658 PMCID: PMC11220467 DOI: 10.1016/j.bioactmat.2024.05.043] [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: 03/04/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 07/05/2024] Open
Abstract
Cardiovascular disease is a significant cause of death in humans. Various models are necessary for the study of cardiovascular diseases, but once cellular and animal models have some defects, such as insufficient fidelity. As a new technology, organoid has certain advantages and has been used in many applications in the study of cardiovascular diseases. This article aims to summarize the application of organoid platforms in cardiovascular diseases, including organoid construction schemes, modeling, and application of cardiovascular organoids. Advances in cardiovascular organoid research have provided many models for different cardiovascular diseases in a variety of areas, including myocardium, blood vessels, and valves. Physiological and pathological models of different diseases, drug research models, and methods for evaluating and promoting the maturation of different kinds of organ tissues are provided for various cardiovascular diseases, including cardiomyopathy, myocardial infarction, and atherosclerosis. This article provides a comprehensive overview of the latest research progress in cardiovascular organ tissues, including construction protocols for cardiovascular organoid tissues and their evaluation system, different types of disease models, and applications of cardiovascular organoid models in various studies. The problems and possible solutions in organoid development are summarized.
Collapse
Affiliation(s)
- Xingchao Du
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Hao Jia
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Yuan Chang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Yiqi Zhao
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| |
Collapse
|
4
|
Fritschen A, Lindner N, Scholpp S, Richthof P, Dietz J, Linke P, Guttenberg Z, Blaeser A. High-Scale 3D-Bioprinting Platform for the Automated Production of Vascularized Organs-on-a-Chip. Adv Healthc Mater 2024; 13:e2304028. [PMID: 38511587 PMCID: PMC11469029 DOI: 10.1002/adhm.202304028] [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: 11/16/2023] [Revised: 03/18/2024] [Indexed: 03/22/2024]
Abstract
3D bioprinting possesses the potential to revolutionize contemporary methodologies for fabricating tissue models employed in pharmaceutical research and experimental investigations. This is enhanced by combining bioprinting with advanced organs-on-a-chip (OOCs), which includes a complex arrangement of multiple cell types representing organ-specific cells, connective tissue, and vasculature. However, both OOCs and bioprinting so far demand a high degree of manual intervention, thereby impeding efficiency and inhibiting scalability to meet technological requirements. Through the combination of drop-on-demand bioprinting with robotic handling of microfluidic chips, a print procedure is achieved that is proficient in managing three distinct tissue models on a chip within only a minute, as well as capable of consecutively processing numerous OOCs without manual intervention. This process rests upon the development of a post-printing sealable microfluidic chip, that is compatible with different types of 3D-bioprinters and easily connected to a perfusion system. The capabilities of the automized bioprint process are showcased through the creation of a multicellular and vascularized liver carcinoma model on the chip. The process achieves full vascularization and stable microvascular network formation over 14 days of culture time, with pronounced spheroidal cell growth and albumin secretion of HepG2 serving as a representative cell model.
Collapse
Affiliation(s)
- Anna Fritschen
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Nils Lindner
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Sebastian Scholpp
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Philipp Richthof
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Jonas Dietz
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | | | | | - Andreas Blaeser
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
- Centre for Synthetic BiologyTechnical University of Darmstadt64289DarmstadtGermany
| |
Collapse
|
5
|
Ino K, Wachi M, Utagawa Y, Konno A, Takinoue M, Abe H, Shiku H. Scanning electrochemical microscopy for determining oxygen consumption rates of cells in hydrogel fibers fabricated using an extrusion 3D bioprinter. Anal Chim Acta 2024; 1304:342539. [PMID: 38637037 DOI: 10.1016/j.aca.2024.342539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/04/2024] [Accepted: 03/25/2024] [Indexed: 04/20/2024]
Abstract
Three-dimensional (3D)-cultured cells have attracted the attention of researchers in tissue engineering- and drug screening-related fields. Among them, 3D cellular fibers have attracted significant attention because they can be stacked to prepare more complex tissues and organs. Cellular fibers are widely fabricated using extrusion 3D bioprinters. For these applications, it is necessary to evaluate cellular activities, such as the oxygen consumption rate (OCR), which is one of the major metabolic activities. We previously reported the use of scanning electrochemical microscopy (SECM) to evaluate the OCRs of cell spheroids. However, the SECM approach has not yet been applied to hydrogel fibers prepared using the bioprinters. To the best of our knowledge, this is the first study to evaluate the OCR of cellular fibers printed by extrusion 3D bioprinters. First, the diffusion theory was discussed to address this issue. Next, diffusion models were simulated to compare realistic models with this theory. Finally, the OCRs of MCF-7 cells in the printed hydrogel fibers were evaluated as a proof of concept. Our proposed approach could potentially be used to evaluate the OCRs of tissue-engineered fibers for organ transplantation and drug screening using in-vitro models.
Collapse
Affiliation(s)
- Kosuke Ino
- Graduate School of Engineering, Tohoku University, 6-6-11-604, Aramaki-aza Aoba, Aoba-ku, Sendai, 980-8579, Japan.
| | - Mana Wachi
- School of Engineering, Tohoku University, 6-6-11-604, Aramaki-aza Aoba, Aoba-ku, Sendai, 980-8579, Japan
| | - Yoshinobu Utagawa
- Graduate School of Engineering, Tohoku University, 6-6-11-604, Aramaki-aza Aoba, Aoba-ku, Sendai, 980-8579, Japan
| | - An Konno
- Graduate School of Environmental Studies, Tohoku University, 6-6-11-604, Aramaki-aza Aoba, Aoba-ku, Sendai, 980-8579, Japan
| | - Masahiro Takinoue
- Department of Computer Science, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Hiroya Abe
- Graduate School of Engineering, Tohoku University, 6-6-11-604, Aramaki-aza Aoba, Aoba-ku, Sendai, 980-8579, Japan; Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Aramaki-aza Aoba 6-3, Aoba-ku, Sendai, 980-8578, Japan
| | - Hitoshi Shiku
- Graduate School of Engineering, Tohoku University, 6-6-11-604, Aramaki-aza Aoba, Aoba-ku, Sendai, 980-8579, Japan; Graduate School of Environmental Studies, Tohoku University, 6-6-11-604, Aramaki-aza Aoba, Aoba-ku, Sendai, 980-8579, Japan.
| |
Collapse
|
6
|
Koutsilieri S, Mickols E, Végvári Á, Lauschke VM. Proteomic workflows for deep phenotypic profiling of 3D organotypic liver models. Biotechnol J 2024; 19:e2300684. [PMID: 38509783 DOI: 10.1002/biot.202300684] [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: 12/03/2023] [Revised: 02/07/2024] [Accepted: 02/15/2024] [Indexed: 03/22/2024]
Abstract
Organotypic human tissue models constitute promising systems to facilitate drug discovery and development. They allow to maintain native cellular phenotypes and functions, which enables long-term pharmacokinetic and toxicity studies, as well as phenotypic screening. To trace relevant phenotypic changes back to specific targets or signaling pathways, comprehensive proteomic profiling is the gold-standard. A multitude of proteomic workflows have been applied on 3D tissue models to quantify their molecular phenotypes; however, their impact on analytical results and biological conclusions in this context has not been evaluated. The performance of twelve mass spectrometry-based global proteomic workflows that differed in the amount of cellular input, lysis protocols and quantification methods was compared for the analysis of primary human liver spheroids. Results differed majorly between protocols in the total number and subcellular compartment bias of identified proteins, which is particularly relevant for the reliable quantification of transporters and drug metabolizing enzymes. Using a model of metabolic dysfunction-associated steatotic liver disease, we furthermore show that critical disease pathways are robustly identified using a standardized high throughput-compatible workflow based on thermal lysis, even using only individual spheroids (1500 cells) as input. The results increase the applicability of proteomic profiling to phenotypic screens in organotypic microtissues and provide a scalable platform for deep phenotyping from limited biological material.
Collapse
Affiliation(s)
- Stefania Koutsilieri
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Evgeniya Mickols
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Ákos Végvári
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| |
Collapse
|
7
|
Monaghan MG, Borah R, Thomsen C, Browne S. Thou shall not heal: Overcoming the non-healing behaviour of diabetic foot ulcers by engineering the inflammatory microenvironment. Adv Drug Deliv Rev 2023; 203:115120. [PMID: 37884128 DOI: 10.1016/j.addr.2023.115120] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/01/2023] [Accepted: 10/23/2023] [Indexed: 10/28/2023]
Abstract
Diabetic foot ulcers (DFUs) are a devastating complication for diabetic patients that have debilitating effects and can ultimately lead to limb amputation. Healthy wounds progress through the phases of healing leading to tissue regeneration and restoration of the barrier function of the skin. In contrast, in diabetic patients dysregulation of these phases leads to chronic, non-healing wounds. In particular, unresolved inflammation in the DFU microenvironment has been identified as a key facet of chronic wounds in hyperglyceamic patients, as DFUs fail to progress beyond the inflammatory phase and towards resolution. Thus, control over and modulation of the inflammatory response is a promising therapeutic avenue for DFU treatment. This review discusses the current state-of-the art regarding control of the inflammatory response in the DFU microenvironment, with a specific focus on the development of biomaterials-based delivery strategies and their cargos to direct tissue regeneration in the DFU microenvironment.
Collapse
Affiliation(s)
- Michael G Monaghan
- Department of Mechanical, Manufacturing and Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland; Advanced Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons in Ireland, Dublin 2, Ireland; CÚRAM, Centre for Research in Medical Devices, National University of Ireland, H91 W2TY Galway, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
| | - Rajiv Borah
- Department of Mechanical, Manufacturing and Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland; Advanced Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons in Ireland, Dublin 2, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
| | - Charlotte Thomsen
- Department of Mechanical, Manufacturing and Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland; Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Shane Browne
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland, H91 W2TY Galway, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland; Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland.
| |
Collapse
|
8
|
Fan C, He J, Xu S, Yan J, Jin L, Dai J, Hu B. Advances in biomaterial-based cardiac organoids. BIOMATERIALS ADVANCES 2023; 153:213502. [PMID: 37352743 DOI: 10.1016/j.bioadv.2023.213502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/27/2023] [Accepted: 06/05/2023] [Indexed: 06/25/2023]
Abstract
Cardiovascular disease (CVD) is one of the important causes of death worldwide. The incidence and mortality rates are increasing annually with the intensification of social aging. The efficacy of drug therapy is limited in individuals suffering from severe heart failure due to the inability of myocardial cells to undergo regeneration and the challenging nature of cardiac tissue repair following injury. Consequently, surgical transplantation stands as the most efficient approach for treatment. Nevertheless, the shortage of donors and the considerable number of heart failure patients worldwide, estimated at 26 million, results in an alarming treatment deficit, with only around 5000 heart transplants feasible annually. The existing major alternatives, such as mechanical or xenogeneic hearts, have significant flaws, such as high cost and rejection, and are challenging to implement for large-scale, long-term use. An organoid is a three-dimensional (3D) cell tissue that mimics the characteristics of an organ. The critical application has been rated in annual biotechnology by authoritative journals, such as Science and Cell. Related industries have achieved rapid growth in recent years. Based on this technology, cardiac organoids are expected to pave the way for viable heart repair and treatment and play an essential role in pathological research, drug screening, and other areas. This review centers on the examination of biomaterials employed in cardiac repair, strategies employed for the reconstruction of cardiac structure and function, clinical investigations pertaining to cardiac repair, and the prospective applications of cardiac organoids. From basic research to clinical practice, the current status, latest progress, challenges, and prospects of biomaterial-based cardiac repair are summarized and discussed, providing a reference for future exploration and development of cardiac regeneration strategies.
Collapse
Affiliation(s)
- Caixia Fan
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing 312000, Zhejiang, China.
| | - Jiaxiong He
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing 312000, Zhejiang, China.
| | - Sijia Xu
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing 312000, Zhejiang, China
| | - Junyan Yan
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing 312000, Zhejiang, China.
| | - Lifang Jin
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing 312000, Zhejiang, China.
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China.
| | - Baowei Hu
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing 312000, Zhejiang, China.
| |
Collapse
|
9
|
Parafati M, Giza S, Shenoy TS, Mojica-Santiago JA, Hopf M, Malany LK, Platt D, Moore I, Jacobs ZA, Kuehl P, Rexroat J, Barnett G, Schmidt CE, McLamb WT, Clements T, Coen PM, Malany S. Human skeletal muscle tissue chip autonomous payload reveals changes in fiber type and metabolic gene expression due to spaceflight. NPJ Microgravity 2023; 9:77. [PMID: 37714852 PMCID: PMC10504373 DOI: 10.1038/s41526-023-00322-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 08/16/2023] [Indexed: 09/17/2023] Open
Abstract
Microphysiological systems provide the opportunity to model accelerated changes at the human tissue level in the extreme space environment. Spaceflight-induced muscle atrophy experienced by astronauts shares similar physiological changes to muscle wasting in older adults, known as sarcopenia. These shared attributes provide a rationale for investigating molecular changes in muscle cells exposed to spaceflight that may mimic the underlying pathophysiology of sarcopenia. We report the results from three-dimensional myobundles derived from muscle biopsies from young and older adults, integrated into an autonomous CubeLab™, and flown to the International Space Station (ISS) aboard SpaceX CRS-21 as part of the NIH/NASA funded Tissue Chips in Space program. Global transcriptomic RNA-Seq analyses comparing the myobundles in space and on the ground revealed downregulation of shared transcripts related to myoblast proliferation and muscle differentiation. The analyses also revealed downregulated differentially expressed gene pathways related to muscle metabolism unique to myobundles derived from the older cohort exposed to the space environment compared to ground controls. Gene classes related to inflammatory pathways were downregulated in flight samples cultured from the younger cohort compared to ground controls. Our muscle tissue chip platform provides an approach to studying the cell autonomous effects of spaceflight on muscle cell biology that may not be appreciated on the whole organ or organism level and sets the stage for continued data collection from muscle tissue chip experimentation in microgravity. We also report on the challenges and opportunities for conducting autonomous tissue-on-chip CubeLabTM payloads on the ISS.
Collapse
Affiliation(s)
- Maddalena Parafati
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Shelby Giza
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Tushar S Shenoy
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Jorge A Mojica-Santiago
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, 32610, USA
| | - Meghan Hopf
- Translational Research Institute, AdventHealth, Orlando, FL, 32804, USA
| | | | - Don Platt
- Micro Aerospace Solutions, INC, Melbourne, FL, 32935, USA
| | | | | | - Paul Kuehl
- Space Tango, LLC, Lexington, KY, 40505, USA
| | | | | | - Christine E Schmidt
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, 32610, USA
| | | | | | - Paul M Coen
- Translational Research Institute, AdventHealth, Orlando, FL, 32804, USA
| | - Siobhan Malany
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
10
|
Zandi Shafagh R, Youhanna S, Keulen J, Shen JX, Taebnia N, Preiss LC, Klein K, Büttner FA, Bergqvist M, van der Wijngaart W, Lauschke VM. Bioengineered Pancreas-Liver Crosstalk in a Microfluidic Coculture Chip Identifies Human Metabolic Response Signatures in Prediabetic Hyperglycemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203368. [PMID: 36285680 PMCID: PMC9731722 DOI: 10.1002/advs.202203368] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/05/2022] [Indexed: 05/19/2023]
Abstract
Aberrant glucose homeostasis is the most common metabolic disturbance affecting one in ten adults worldwide. Prediabetic hyperglycemia due to dysfunctional interactions between different human tissues, including pancreas and liver, constitutes the largest risk factor for the development of type 2 diabetes. However, this early stage of metabolic disease has received relatively little attention. Microphysiological tissue models that emulate tissue crosstalk offer emerging opportunities to study metabolic interactions. Here, a novel modular multitissue organ-on-a-chip device is presented that allows for integrated and reciprocal communication between different 3D primary human tissue cultures. Precisely controlled heterologous perfusion of each tissue chamber is achieved through a microfluidic single "synthetic heart" pneumatic actuation unit connected to multiple tissue chambers via specific configuration of microchannel resistances. On-chip coculture experiments of organotypic primary human liver spheroids and intact primary human islets demonstrate insulin secretion and hepatic insulin response dynamics at physiological timescales upon glucose challenge. Integration of transcriptomic analyses with promoter motif activity data of 503 transcription factors reveals tissue-specific interacting molecular networks that underlie β-cell stress in prediabetic hyperglycemia. Interestingly, liver and islet cultures show surprising counter-regulation of transcriptional programs, emphasizing the power of microphysiological coculture to elucidate the systems biology of metabolic crosstalk.
Collapse
Affiliation(s)
- Reza Zandi Shafagh
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17711, Sweden
- Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, 10044, Sweden
| | - Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17711, Sweden
| | - Jibbe Keulen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17711, Sweden
- Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, 10044, Sweden
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376, Stuttgart, Germany
- University of Tuebingen, 72074, Tuebingen, Germany
| | - Joanne X Shen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17711, Sweden
| | - Nayere Taebnia
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17711, Sweden
| | - Lena C Preiss
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17711, Sweden
- Department of Drug Metabolism and Pharmacokinetics (DMPK), The Healthcare Business of Merck KGaA, 64293, Darmstadt, Germany
| | - Kathrin Klein
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376, Stuttgart, Germany
- University of Tuebingen, 72074, Tuebingen, Germany
| | - Florian A Büttner
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376, Stuttgart, Germany
- University of Tuebingen, 72074, Tuebingen, Germany
| | - Mikael Bergqvist
- Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, 10044, Sweden
| | | | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17711, Sweden
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376, Stuttgart, Germany
- University of Tuebingen, 72074, Tuebingen, Germany
| |
Collapse
|
11
|
Middleton AM, Reynolds J, Cable S, Baltazar MT, Li H, Bevan S, Carmichael PL, Dent MP, Hatherell S, Houghton J, Kukic P, Liddell M, Malcomber S, Nicol B, Park B, Patel H, Scott S, Sparham C, Walker P, White A. Are Non-animal Systemic Safety Assessments Protective? A Toolbox and Workflow. Toxicol Sci 2022; 189:124-147. [PMID: 35822611 PMCID: PMC9412174 DOI: 10.1093/toxsci/kfac068] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
An important question in toxicological risk assessment is whether non-animal new approach methodologies (NAMs) can be used to make safety decisions that are protective of human health, without being overly conservative. In this work, we propose a core NAM toolbox and workflow for conducting systemic safety assessments for adult consumers. We also present an approach for evaluating how protective and useful the toolbox and workflow are by benchmarking against historical safety decisions. The toolbox includes physiologically based kinetic (PBK) models to estimate systemic Cmax levels in humans, and 3 bioactivity platforms, comprising high-throughput transcriptomics, a cell stress panel, and in vitro pharmacological profiling, from which points of departure are estimated. A Bayesian model was developed to quantify the uncertainty in the Cmax estimates depending on how the PBK models were parameterized. The feasibility of the evaluation approach was tested using 24 exposure scenarios from 10 chemicals, some of which would be considered high risk from a consumer goods perspective (eg, drugs that are systemically bioactive) and some low risk (eg, existing food or cosmetic ingredients). Using novel protectiveness and utility metrics, it was shown that up to 69% (9/13) of the low risk scenarios could be identified as such using the toolbox, whilst being protective against all (5/5) the high-risk ones. The results demonstrated how robust safety decisions could be made without using animal data. This work will enable a full evaluation to assess how protective and useful the toolbox and workflow are across a broader range of chemical-exposure scenarios.
Collapse
Affiliation(s)
| | - Joe Reynolds
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Sophie Cable
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | | | - Hequn Li
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | | | - Paul L Carmichael
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Matthew Philip Dent
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Sarah Hatherell
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Jade Houghton
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Predrag Kukic
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Mark Liddell
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Sophie Malcomber
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Beate Nicol
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | | | - Hiral Patel
- Charles River Laboratories, Cambridgeshire, CB10 1XL, UK
| | - Sharon Scott
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Chris Sparham
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Paul Walker
- Cyprotex Discovery Ltd, Cheshire SK10 4TG, UK
| | - Andrew White
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| |
Collapse
|
12
|
Rusyn I, Sakolish C, Kato Y, Stephan C, Vergara L, Hewitt P, Bhaskaran V, Davis M, Hardwick RN, Ferguson SS, Stanko JP, Bajaj P, Adkins K, Sipes NS, Hunter ES, Baltazar MT, Carmichael PL, Sadh K, Becker RA. Microphysiological Systems Evaluation: Experience of TEX-VAL Tissue Chip Testing Consortium. Toxicol Sci 2022; 188:143-152. [PMID: 35689632 PMCID: PMC9333404 DOI: 10.1093/toxsci/kfac061] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Much has been written and said about the promise and excitement of microphysiological systems, miniature devices that aim to recreate aspects of human physiology on a chip. The rapid explosion of the offerings and persistent publicity placed high expectations on both product manufacturers and regulatory agencies to adopt the data. Inevitably, discussions of where this technology fits in chemical testing paradigms are ongoing. Some end-users became early adopters, whereas others have taken a more cautious approach because of the high cost and uncertainties of their utility. Here, we detail the experience of a public-private collaboration established for testing of diverse microphysiological systems. Collectively, we present a number of considerations on practical aspects of using microphysiological systems in the context of their applications in decision-making. Specifically, future end-users need to be prepared for extensive on-site optimization and have access to a wide range of imaging and other equipment. We reason that cells, related reagents, and the technical skills of the research staff, not the devices themselves, are the most critical determinants of success. Extrapolation from concentration-response effects in microphysiological systems to human blood or oral exposures, difficulties with replicating the whole organ, and long-term functionality remain as critical challenges. Overall, we conclude that it is unlikely that a rodent- or human-equivalent model is achievable through a finite number of microphysiological systems in the near future; therefore, building consensus and promoting the gradual incorporation of these models into tiered approaches for safety assessment and decision-making is the sensible path to wide adoption.
Collapse
Affiliation(s)
- Ivan Rusyn
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, USA
| | - Courtney Sakolish
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, USA
| | - Yuki Kato
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, USA
| | - Clifford Stephan
- Institute of Biosciences and Technology, Texas A&M University, Houston, Texas 77030, USA
| | - Leoncio Vergara
- Institute of Biosciences and Technology, Texas A&M University, Houston, Texas 77030, USA
| | - Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, Darmstadt, Germany
| | - Vasanthi Bhaskaran
- Discovery Toxicology, Bristol Myers Squibb, Princeton, New Jersey 08543, USA
| | - Myrtle Davis
- Discovery Toxicology, Bristol Myers Squibb, Princeton, New Jersey 08543, USA
| | - Rhiannon N Hardwick
- Discovery Toxicology, Bristol Myers Squibb, San Diego, California 92130, USA
| | - Stephen S Ferguson
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, USA
| | - Jason P Stanko
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, USA
| | - Piyush Bajaj
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Framingham, Massachusetts 01701, USA
| | - Karissa Adkins
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Framingham, Massachusetts 01701, USA
| | - Nisha S Sipes
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, 27711, USA
| | - E Sidney Hunter
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, 27711, USA
| | - Maria T Baltazar
- Unilever Safety and Environmental Assurance Centre, Bedfordshire, Sharnbrook MK44 1LQ, UK
| | - Paul L Carmichael
- Unilever Safety and Environmental Assurance Centre, Bedfordshire, Sharnbrook MK44 1LQ, UK
| | - Kritika Sadh
- Unilever Safety and Environmental Assurance Centre, Bedfordshire, Sharnbrook MK44 1LQ, UK
| | - Richard A Becker
- American Chemistry Council, Washington, District of Columbia 20002, USA
| |
Collapse
|
13
|
Current insights into the bone marrow niche: From biology in vivo to bioengineering ex vivo. Biomaterials 2022; 286:121568. [DOI: 10.1016/j.biomaterials.2022.121568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 04/26/2022] [Accepted: 05/03/2022] [Indexed: 11/21/2022]
|
14
|
Fritschen A, Bell AK, Königstein I, Stühn L, Stark RW, Blaeser A. Investigation and comparison of resin materials in transparent DLP-printing for application in cell culture and organs-on-a-chip. Biomater Sci 2022; 10:1981-1994. [PMID: 35262097 DOI: 10.1039/d1bm01794b] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Organs-on-a-Chip (OOCs) have recently led to major discoveries and a better understanding of 3D cell organization, cell-cell interactions and tissue response to drugs and biological cues. However, their complexity and variability are still limited by the available fabrication technology. Transparent, cytocompatible and high-resolution 3D-printing could overcome these limitations, offering a flexible and low-cost alternative to soft lithography. Many advances have been made in stereolithography printing regarding resin formulation and the general printing process, but a systematic analysis of the printing process steps, employed resins and post-treatment procedures with a strong focus on the requirements in OOCs is missing. To fill this gap, this work provides an in-depth analysis of three different resin systems in comparison to polystyrene (PS) and poly(dimethylsiloxane) (PDMS), which can be considered the gold-standards in cell culture and microfluidics. The resins were characterized with respect to transparency, cytocompatibility and print resolution. These properties are not only governed by the resin composition, but additionally by the post-treatment procedure. The investigation of the mechanical (elastic modulus ∼2.2 GPa) and wetting properties (∼60° native / 20° plasma treated) showed a behavior very similar to PS. In addition, the absorbance of small molecules was two orders of magnitude lower in the applied resins (diffusion constant ∼0.01 μm2 s-1) than for PDMS (2.5 μm2 s-1), demonstrating the intrinsic suitability of these materials for OOCs. Raman spectroscopy and UV/VIS spectrophotometry revealed that post-treatment increased monomer conversion up to 2 times and removed photo initiator residues, leading to an increased transparency of up to 50% and up to 10-times higher cell viability. High magnification fluorescence imaging of HUVECs and L929 cells cultivated on printed dishes shows the high optical qualities of prints fabricated by the Digital Light Processing (DLP) printer. Finally, components of microfluidic chips such as high-aspect ratio pillars and holes with a diameter of 50 μm were printed. Concluding, the suitability of DLP-printing for OOCs was demonstrated by filling a printed chip with a cell-hydrogel mixture using a microvalve bioprinter, followed by the successful cultivation under perfusion. Our results highlight that DLP-printing has matured into a robust fabrication technology ready for application in extensive and versatile OOC research.
Collapse
Affiliation(s)
- Anna Fritschen
- Technical University of Darmstadt, Department of Mechanical Engineering, BioMedical Printing Technology, Magdalenenstr. 2, 64289 Darmstadt, Germany.
| | - Alena K Bell
- Technical University of Darmstadt, Institute of Materials Science, Physics of Surfaces, Alarich-Weiss-Str. 16, 64287 Darmstadt, Germany
| | - Inga Königstein
- Technical University of Darmstadt, Department of Mechanical Engineering, BioMedical Printing Technology, Magdalenenstr. 2, 64289 Darmstadt, Germany.
| | - Lukas Stühn
- Technical University of Darmstadt, Institute of Materials Science, Physics of Surfaces, Alarich-Weiss-Str. 16, 64287 Darmstadt, Germany
| | - Robert W Stark
- Technical University of Darmstadt, Institute of Materials Science, Physics of Surfaces, Alarich-Weiss-Str. 16, 64287 Darmstadt, Germany
| | - Andreas Blaeser
- Technical University of Darmstadt, Department of Mechanical Engineering, BioMedical Printing Technology, Magdalenenstr. 2, 64289 Darmstadt, Germany. .,Technical University of Darmstadt, Centre for Synthetic Biology, Schnittspahnstr. 10, 64287 Darmstadt, Germany
| |
Collapse
|
15
|
Hargrove-Grimes P, Low LA, Tagle DA. Microphysiological Systems: Stakeholder Challenges to Adoption in Drug Development. Cells Tissues Organs 2022; 211:269-281. [PMID: 34380142 PMCID: PMC8831652 DOI: 10.1159/000517422] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/14/2021] [Indexed: 01/03/2023] Open
Abstract
Microphysiological systems (MPS) or tissue chips/organs-on-chips are novel in vitro models that emulate human physiology at the most basic functional level. In this review, we discuss various hurdles to widespread adoption of MPS technology focusing on issues from multiple stakeholder sectors, e.g., academic MPS developers, commercial suppliers of platforms, the pharmaceutical and biotechnology industries, and regulatory organizations. Broad adoption of MPS technology has thus far been limited by a gap in translation between platform developers, end-users, regulatory agencies, and the pharmaceutical industry. In this brief review, we offer a perspective on the existing barriers and how end-users may help surmount these obstacles to achieve broader adoption of MPS technology.
Collapse
Affiliation(s)
- Passley Hargrove-Grimes
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Lucie A. Low
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Danilo A. Tagle
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
16
|
Kemas AM, Lauschke VM. Measuring Glucose Consumption and Gluconeogenesis in 3D Human Tissue Cultures with Nanoliter Input Volumes. Methods Mol Biol 2022; 2445:337-349. [PMID: 34973002 DOI: 10.1007/978-1-0716-2071-7_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Organotypic and microphysiological culture of primary human tissues and cancers has emerged as a powerful set of technologies that allow to faithfully mimic cellular metabolism and functions ex vivo. The predominant 3D culture methods include spheroids and microfluidic chips. These cultures use low cell numbers and culture volumes, which, however, poses important limitations for the available amounts of sample for downstream analyses. Here, we describe a detailed method for the measurement of glucose consumption dynamics in organotypic culture using a bienzymatic colorimetric assay that accurately quantifies glucose levels using nanoliter input volumes. As an example we utilize spheroids consisting of primary human hepatocytes. The assay has been carefully optimized and benchmarked and is compatible with both longitudinal and high-throughput screening in both static and perfused conditions. The method is straightforward and only requires a microplate reader capable of running absorbance kinetic measurements.
Collapse
Affiliation(s)
- Aurino M Kemas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.
- University of Tuebingen, Tuebingen, Germany.
| |
Collapse
|
17
|
Nashimoto Y, Abe M, Fujii R, Taira N, Ida H, Takahashi Y, Ino K, Ramon‐Azcon J, Shiku H. Topography and Permeability Analyses of Vasculature-on-a-Chip Using Scanning Probe Microscopies. Adv Healthc Mater 2021; 10:e2101186. [PMID: 34409770 DOI: 10.1002/adhm.202101186] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/22/2021] [Indexed: 11/08/2022]
Abstract
Microphysiological systems (MPS) or organs-on-chips (OoC) can emulate the physiological functions of organs in vitro and are effective tools for determining human drug responses in preclinical studies. However, the analysis of MPS has relied heavily on optical tools, resulting in difficulties in real-time and high spatial resolution imaging of the target cell functions. In this study, the role of scanning probe microscopy (SPM) as an analytical tool for MPS is evaluated. An access hole is made in a typical MPS system with stacked microchannels to insert SPM probes into the system. For the first study, a simple vascular model composed of only endothelial cells is prepared for SPM analysis. Changes in permeability and local chemical flux are quantitatively evaluated during the construction of the vascular system. The morphological changes in the endothelial cells after flow stimulation are imaged at the single-cell level for topographical analysis. Finally, the possibility of adapting the permeability and topographical analysis using SPM for the intestinal vascular system is further evaluated. It is believed that this study will pave the way for an in situ permeability assay and structural analysis of MPS using SPM.
Collapse
Affiliation(s)
- Yuji Nashimoto
- Frontier Research Institute for Interdisciplinary Sciences (FRIS) Tohoku University Miyagi 980‐8578 Japan
- Graduate School of Engineering Tohoku University Miyagi 980‐8579 Japan
- Graduate School of Environmental Studies Tohoku University Miyagi 980‐8579 Japan
| | - Minori Abe
- Graduate School of Environmental Studies Tohoku University Miyagi 980‐8579 Japan
| | - Ryota Fujii
- Graduate School of Environmental Studies Tohoku University Miyagi 980‐8579 Japan
| | - Noriko Taira
- Graduate School of Environmental Studies Tohoku University Miyagi 980‐8579 Japan
| | - Hiroki Ida
- Frontier Research Institute for Interdisciplinary Sciences (FRIS) Tohoku University Miyagi 980‐8578 Japan
- Graduate School of Environmental Studies Tohoku University Miyagi 980‐8579 Japan
- WPI‐Advanced Institute for Materials Research Tohoku University Miyagi 980‐8577 Japan
- Precursory Research for Embryonic Science and Technology (PRESTO) Science and Technology Agency (JST) Saitama 332‐0012 Japan
| | - Yasufumi Takahashi
- Precursory Research for Embryonic Science and Technology (PRESTO) Science and Technology Agency (JST) Saitama 332‐0012 Japan
- WPI‐Nano Life Science Institute Kanazawa University Ishikawa 920‐1192 Japan
| | - Kosuke Ino
- Graduate School of Engineering Tohoku University Miyagi 980‐8579 Japan
| | - Javier Ramon‐Azcon
- Institute for Bioengineering of Catalonia (IBEC) The Barcelona Institute of Science and Technology Barcelona 08028 Spain
- Institució Catalana de Reserca I Estudis Avançats (ICREA) Passeig de Lluís Companys, 23 Barcelona E08010 Spain
| | - Hitoshi Shiku
- Graduate School of Engineering Tohoku University Miyagi 980‐8579 Japan
- Graduate School of Environmental Studies Tohoku University Miyagi 980‐8579 Japan
| |
Collapse
|
18
|
Utoh R, Enomoto S, Yamada M, Yamanaka K, Yajima Y, Furusawa K, Seki M. Polyanion-induced, microfluidic engineering of fragmented collagen microfibers for reconstituting extracellular environments of 3D hepatocyte culture. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 129:112417. [PMID: 34579926 DOI: 10.1016/j.msec.2021.112417] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 12/11/2022]
Abstract
Artificial biological scaffolds made of extracellular matrix (ECM) components, such as type I collagen, provide ideal physicochemical cues to various cell culture platforms. However, it remains a challenge to fabricate micrometer-sized ECM materials with precisely controlled morphologies that could reconstitute the 3-dimensional (3D) microenvironments surrounding cells. In the present study, we proposed a unique process to fabricate fragmented collagen microfibers using a microfluidic laminar-flow system. The continuous flow of an acidic collagen solution was neutralized to generate solid fibers, which were subsequently fragmented by applying a gentle shear stress in a polyanion-containing phosphate buffer. The morphology of the fiber fragment was controllable in a wide range by changing the type and/or concentration of the polyanion and by tuning the applied shear stress. The biological benefits of the fragmented fibers were investigated through the formation of multicellular spheroids composed of primary rat hepatocytes and microfibers on non-cell-adhesive micro-vessels. The microfibers enhanced the survival and functions of the hepatocytes and reproduced proper cell polarity, because the fibers facilitated the formation of cell-cell and cell-matrix interactions while modulating the close packing of cells. These results clearly indicated that the microengineered fragmented collagen fibers have great potential to reconstitute extracellular microenvironments for hepatocytes in 3D culture, which will be of significant benefit for cell-based drug testing and bottom-up tissue engineering.
Collapse
Affiliation(s)
- Rie Utoh
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Sakiko Enomoto
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Masumi Yamada
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan.
| | - Keigo Yamanaka
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Yuya Yajima
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Kazuya Furusawa
- Department of Applied Chemistry and Food Science, Faculty of Environmental and Information Sciences, Fukui University of Technology, 3-6-1 Gakuen, Fukui 910-8505, Japan
| | - Minoru Seki
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| |
Collapse
|
19
|
Sano E, Deguchi S, Matsuoka N, Tsuda M, Wang M, Kosugi K, Mori C, Yagi K, Wada A, Yamasaki S, Kawai T, Yodogawa M, Mizuguchi H, Nakazawa N, Yamashita F, Torisawa YS, Takayama K. Generation of Tetrafluoroethylene-Propylene Elastomer-Based Microfluidic Devices for Drug Toxicity and Metabolism Studies. ACS OMEGA 2021; 6:24859-24865. [PMID: 34604667 PMCID: PMC8482466 DOI: 10.1021/acsomega.1c03719] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Indexed: 06/13/2023]
Abstract
Polydimethylsiloxane (PDMS) is widely used to fabricate microfluidic organs-on-chips. Using these devices (PDMS-based devices), the mechanical microenvironment of living tissues, such as pulmonary respiration and intestinal peristalsis, can be reproduced in vitro. However, the use of PDMS-based devices in drug discovery research is limited because of their extensive absorption of drugs. In this study, we investigated the feasibility of the tetrafluoroethylene-propylene (FEPM) elastomer to fabricate a hepatocyte-on-a-chip (FEPM-based hepatocyte chip) with lower drug absorption. The FEPM-based hepatocyte chip expressed drug-metabolizing enzymes, drug-conjugating enzymes, and drug transporters. Also, it could produce human albumin. Although the metabolites of midazolam and bufuralol were hardly detected in the PDMS-based hepatocyte chip, they were detected abundantly in the FEPM-based hepatocyte chip. Finally, coumarin-induced hepatocyte cytotoxicity was less severe in the PDMS-based hepatocyte chip than in the FEPM-based hepatocyte chip, reflecting the different drug absorptions of the two chips. In conclusion, the FEPM-based hepatocyte chip could be a useful tool in drug discovery research, including drug metabolism and toxicity studies.
Collapse
Affiliation(s)
- Emi Sano
- Center
for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
- Department
of Micro Engineering, Kyoto University, Kyoto 615-8540, Japan
| | - Sayaka Deguchi
- Center
for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
- Laboratory
of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical
Sciences, Osaka University, Osaka 565-0871, Japan
| | | | - Masahiro Tsuda
- Department
of Applied Pharmaceutics and Pharmacokinetics, Graduate School of
Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Mengyang Wang
- Department
of Applied Pharmaceutics and Pharmacokinetics, Graduate School of
Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Kaori Kosugi
- Center
for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
- Department
of Micro Engineering, Kyoto University, Kyoto 615-8540, Japan
| | - Chihiro Mori
- Department
of Micro Engineering, Kyoto University, Kyoto 615-8540, Japan
| | | | | | | | | | | | - Hiroyuki Mizuguchi
- Laboratory
of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical
Sciences, Osaka University, Osaka 565-0871, Japan
| | | | - Fumiyoshi Yamashita
- Department
of Applied Pharmaceutics and Pharmacokinetics, Graduate School of
Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
- Department
of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Yu-suke Torisawa
- Department
of Micro Engineering, Kyoto University, Kyoto 615-8540, Japan
| | - Kazuo Takayama
- Center
for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
20
|
Loskill P, Hardwick RN, Roth A. Challenging the pipeline. Stem Cell Reports 2021; 16:2033-2037. [PMID: 34525380 PMCID: PMC8452595 DOI: 10.1016/j.stemcr.2021.08.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 12/24/2022] Open
Abstract
This commentary presents a thought experiment seeking to answer the key question: "If you were to put aside all the traditional drug discovery processes and start a new drug discovery program that places the highest priority on human and disease-relevant models throughout the entire process, how could it be done?"
Collapse
Affiliation(s)
- Peter Loskill
- Department of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany; NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany; 3R-Center for In vitro Models and Alternatives to Animal Testing, Eberhard Karls University Tübingen, Tübingen, Germany.
| | - Rhiannon N Hardwick
- Translational Safety Sciences, Theravance Biopharma US, Inc., South San Francisco, CA, USA
| | - Adrian Roth
- Personalized Healthcare Safety Interface, Product Development Safety, Roche Innovation Centre Basel, Basel, Switzerland
| |
Collapse
|
21
|
Ching T, Toh YC, Hashimoto M, Zhang YS. Bridging the academia-to-industry gap: organ-on-a-chip platforms for safety and toxicology assessment. Trends Pharmacol Sci 2021; 42:715-728. [PMID: 34187693 PMCID: PMC8364498 DOI: 10.1016/j.tips.2021.05.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/04/2021] [Accepted: 05/27/2021] [Indexed: 12/14/2022]
Abstract
Some organ-on-a-chip (OoC) systems for drug evaluation show better predictive capabilities than planar, static cell cultures and animal models. One of the ongoing initiatives led by OoC developers is to bridge the academia-to-industry gap in the hope of gaining wider adoption by end-users - academic biological researchers and industry. We discuss several recommendations that can help to drive the adoption of OoC systems by the market. We first review some key challenges faced by OoC developers before highlighting current advances in OoC platforms. We then offer recommendations for OoC developers to promote the uptake of OoC systems by the industry.
Collapse
Affiliation(s)
- Terry Ching
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Pillar of Engineering Product Development, Singapore University of Technology and Design, Singapore 487373; Digital Manufacturing and Design Centre, Singapore University of Technology and Design, Singapore 4873724; Department of Biomedical Engineering, National University of Singapore, Singapore 117583
| | - Yi-Chin Toh
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD 4000, Australia; Centre for Biomedical Technologies, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia.
| | - Michinao Hashimoto
- Pillar of Engineering Product Development, Singapore University of Technology and Design, Singapore 487373; Digital Manufacturing and Design Centre, Singapore University of Technology and Design, Singapore 4873724.
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA.
| |
Collapse
|
22
|
Kemas AM, Youhanna S, Zandi Shafagh R, Lauschke VM. Insulin-dependent glucose consumption dynamics in 3D primary human liver cultures measured by a sensitive and specific glucose sensor with nanoliter input volume. FASEB J 2021; 35:e21305. [PMID: 33566368 DOI: 10.1096/fj.202001989rr] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/06/2020] [Accepted: 12/09/2020] [Indexed: 12/27/2022]
Abstract
The liver plays a central role in glucose homeostasis and hepatic insulin resistance constitutes a key feature of type 2 diabetes. However, platforms that accurately mimic human hepatic glucose disposition and allow for rapid and scalable quantification of glucose consumption dynamics are lacking. Here, we developed and optimized a colorimetric glucose assay based on the glucose oxidase-peroxidase system and demonstrate that the system can monitor glucose consumption in 3D primary human liver cell cultures over multiple days. The system was highly sensitive (limit of detection of 3.5 µM) and exceptionally accurate (R2 = 0.999) while requiring only nanoliter input volumes (250 nL), enabling longitudinal profiling of individual liver microtissues. By utilizing a novel polymer, off-stoichiometric thiol-ene (OSTE), and click-chemistry based on thiol-Michael additions, we furthermore show that the assay can be covalently bound to custom-build chips, facilitating the integration of the sensor into microfluidic devices. Using this system, we find that glucose uptake of our 3D human liver cultures closely resembles human hepatic glucose uptake in vivo as measured by euglycemic-hyperinsulinemic clamp. By comparing isogenic insulin-resistant and insulin-sensitive liver cultures we furthermore show that insulin and extracellular glucose levels account for 55% and 45% of hepatic glucose consumption, respectively. In conclusion, the presented data show that the integration of accurate and scalable nanoliter glucose sensors with physiologically relevant organotypic human liver models enables longitudinal profiling of hepatic glucose consumption dynamics that will facilitate studies into the biology and pathobiology of glycemic control, as well as antidiabetic drug screening.
Collapse
Affiliation(s)
- Aurino M Kemas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Reza Zandi Shafagh
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Department of Micro and Nanosystem, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
23
|
Kinstlinger IS, Calderon GA, Royse MK, Means AK, Grigoryan B, Miller JS. Perfusion and endothelialization of engineered tissues with patterned vascular networks. Nat Protoc 2021; 16:3089-3113. [PMID: 34031610 DOI: 10.1038/s41596-021-00533-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 03/04/2021] [Indexed: 02/04/2023]
Abstract
As engineered tissues progress toward therapeutically relevant length scales and cell densities, it is critical to deliver oxygen and nutrients throughout the tissue volume via perfusion through vascular networks. Furthermore, seeding of endothelial cells within these networks can recapitulate the barrier function and vascular physiology of native blood vessels. In this protocol, we describe how to fabricate and assemble customizable open-source tissue perfusion chambers and catheterize tissue constructs inside them. Human endothelial cells are seeded along the lumenal surfaces of the tissue constructs, which are subsequently connected to fluid pumping equipment. The protocol is agnostic with respect to biofabrication methodology as well as cell and material composition, and thus can enable a wide variety of experimental designs. It takes ~14 h over the course of 3 d to prepare perfusion chambers and begin a perfusion experiment. We envision that this protocol will facilitate the adoption and standardization of perfusion tissue culture methods across the fields of biomaterials and tissue engineering.
Collapse
Affiliation(s)
| | | | - Madison K Royse
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - A Kristen Means
- Department of Bioengineering, Rice University, Houston, TX, USA
| | | | - Jordan S Miller
- Department of Bioengineering, Rice University, Houston, TX, USA.
| |
Collapse
|
24
|
CRISPR: The Multidrug Resistance Endgame? Mol Biotechnol 2021; 63:676-685. [PMID: 34021472 DOI: 10.1007/s12033-021-00340-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/15/2021] [Indexed: 12/25/2022]
Abstract
The flexibility of microbes to undergo or adapt to the changes in their physiology and genotypical traits has enabled the microbes acquiring resistance to latest or recently discovered drugs which have consequently led to the menace of multidrug resistance (MDR). There is a surge in the discovery of novel antibiotics to counter the rising MDR phenomena, and in such a quest, for investigating an efficient alternative mechanism or compound to combat MDR, Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) has piqued the interests of the researchers across the globe. CRISPR-Cas9 technology is a genome-editing tool with successful widespread applications in cell lines, plants, animals, and even in human clinical trials, and it is seriously being considered as a potential candidate for countering MDR. This review encompasses the broad scope of CRISPR-Cas9 along with its various variations, underlying principles, mechanisms, as well as applications. Furthermore, the implications of recent advancements in various disciplines are highlighted to enhance the applicability of this technique. Consequently, its research gaps and challenges are also identified so that they can be addressed in the possible future thereby further expanding the lore of CRISPR-Cas9 technique.
Collapse
|
25
|
Pin C, Collins T, Gibbs M, Kimko H. Systems Modeling to Quantify Safety Risks in Early Drug Development: Using Bifurcation Analysis and Agent-Based Modeling as Examples. AAPS JOURNAL 2021; 23:77. [PMID: 34018069 PMCID: PMC8137611 DOI: 10.1208/s12248-021-00580-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 03/09/2021] [Indexed: 11/30/2022]
Abstract
Quantitative Systems Toxicology (QST) models, recapitulating pharmacokinetics and mechanism of action together with the organic response at multiple levels of biological organization, can provide predictions on the magnitude of injury and recovery dynamics to support study design and decision-making during drug development. Here, we highlight the application of QST models to predict toxicities of cancer treatments, such as cytopenia(s) and gastrointestinal adverse effects, where narrow therapeutic indexes need to be actively managed. The importance of bifurcation analysis is demonstrated in QST models of hematologic toxicity to understand how different regions of the parameter space generate different behaviors following cancer treatment, which results in asymptotically stable predictions, yet highly irregular for specific schedules, or oscillating predictions of blood cell levels. In addition, an agent-based model of the intestinal crypt was used to simulate how the spatial location of the injury within the crypt affects the villus disruption severity. We discuss the value of QST modeling approaches to support drug development and how they align with technological advances impacting trial design including patient selection, dose/regimen selection, and ultimately patient safety.
Collapse
Affiliation(s)
- Carmen Pin
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge Science Park, Milton Road, Cambridge, UK
| | - Teresa Collins
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge Science Park, Milton Road, Cambridge, UK
| | - Megan Gibbs
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Holly Kimko
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gaithersburg, Maryland, USA.
| |
Collapse
|
26
|
Ingelman-Sundberg M, Lauschke VM. 3D human liver spheroids for translational pharmacology and toxicology. Basic Clin Pharmacol Toxicol 2021; 130 Suppl 1:5-15. [PMID: 33872466 DOI: 10.1111/bcpt.13587] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 12/14/2022]
Abstract
Drug development is a failure-prone endeavour, and more than 85% of drugs fail during clinical development, showcasing that current preclinical systems for compound selection are clearly inadequate. Liver toxicity remains a major reason for safety failures. Furthermore, all efforts to develop pharmacological therapies for a variety of chronic liver diseases, such as non-alcoholic steatohepatitis (NASH) and fibrosis, remain unsuccessful. Considering the time and expense of clinical trials, as well as the substantial burden on patients, new strategies are thus of paramount importance to increase clinical success rates. To this end, human liver spheroids are becoming increasingly utilized as they allow to preserve patient-specific phenotypes and functions for multiple weeks in culture. We here review the recent application of such systems for i) predictive and mechanistic analyses of drug hepatotoxicity, ii) the evaluation of hepatic disposition and metabolite formation of low clearance drugs and iii) the development of drugs for metabolic and infectious liver diseases, including NASH, fibrosis, malaria and viral hepatitis. We envision that with increasing dissemination, liver spheroids might become the new gold standard for such applications in translational pharmacology and toxicology.
Collapse
Affiliation(s)
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
27
|
Gough A, Soto-Gutierrez A, Vernetti L, Ebrahimkhani MR, Stern AM, Taylor DL. Human biomimetic liver microphysiology systems in drug development and precision medicine. Nat Rev Gastroenterol Hepatol 2021; 18:252-268. [PMID: 33335282 PMCID: PMC9106093 DOI: 10.1038/s41575-020-00386-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/02/2020] [Indexed: 02/07/2023]
Abstract
Microphysiology systems (MPS), also called organs-on-chips and tissue chips, are miniaturized functional units of organs constructed with multiple cell types under a variety of physical and biochemical environmental cues that complement animal models as part of a new paradigm of drug discovery and development. Biomimetic human liver MPS have evolved from simpler 2D cell models, spheroids and organoids to address the increasing need to understand patient-specific mechanisms of complex and rare diseases, the response to therapeutic treatments, and the absorption, distribution, metabolism, excretion and toxicity of potential therapeutics. The parallel development and application of transdisciplinary technologies, including microfluidic devices, bioprinting, engineered matrix materials, defined physiological and pathophysiological media, patient-derived primary cells, and pluripotent stem cells as well as synthetic biology to engineer cell genes and functions, have created the potential to produce patient-specific, biomimetic MPS for detailed mechanistic studies. It is projected that success in the development and maturation of patient-derived MPS with known genotypes and fully matured adult phenotypes will lead to advanced applications in precision medicine. In this Review, we examine human biomimetic liver MPS that are designed to recapitulate the liver acinus structure and functions to enhance our knowledge of the mechanisms of disease progression and of the absorption, distribution, metabolism, excretion and toxicity of therapeutic candidates and drugs as well as to evaluate their mechanisms of action and their application in precision medicine and preclinical trials.
Collapse
Affiliation(s)
- Albert Gough
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alejandro Soto-Gutierrez
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lawrence Vernetti
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mo R Ebrahimkhani
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andrew M Stern
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - D Lansing Taylor
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA.
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
28
|
Cromwell EF, Leung M, Hammer M, Thai A, Rajendra R, Sirenko O. Disease Modeling with 3D Cell-Based Assays Using a Novel Flowchip System and High-Content Imaging. SLAS Technol 2021; 26:237-248. [PMID: 33783259 DOI: 10.1177/24726303211000688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
There is an increasing interest in using three-dimensional (3D) cell structures for modeling tumors, organs, and tissue to accelerate translational research. We describe here a novel automated organoid assay system (the Pu·MA System) combined with microfluidic-based flowchips that can facilitate 3D cell-based assays. The flowchip is composed of sample wells, which contain organoids, connected to additional multiple wells that can hold various assay reagents. Organoids are positioned in a protected chamber in sample wells, and fluids are exchanged from side reservoirs using pressure-driven flow. Media exchange, sample staining, wash steps, and other processes can be performed without disruption to or loss of 3D sample. The bottom of the sample chamber is thin, optically clear plastic compatible with high-content imaging (HCI). The whole system can be kept in an incubator, allowing long-term cellular assays to be performed. We present two examples of use of the system for biological research. In the first example, cytotoxicity effects of anticancer drugs were evaluated on HeLa and HepG2 spheroids using HCI and vascular endothelial growth factor expression. In the second application, the flowchip system was used for the functional evaluation of Ca2+ oscillations in neurospheroids. Neurospheres were incubated with neuroactive compounds, and neuronal activity was assessed using Ca2+-sensitive dyes and fast kinetic fluorescence imaging. This novel assay system using microfluidics enables automation of 3D cell-based cultures that mimic in vivo conditions, performs multidosing protocols and multiple media exchanges, provides gentle handling of spheroids and organoids, and allows a wide range of assay detection modalities.
Collapse
|
29
|
Sung JH. Multi-organ-on-a-chip for pharmacokinetics and toxicokinetic study of drugs. Expert Opin Drug Metab Toxicol 2021; 17:969-986. [PMID: 33764248 DOI: 10.1080/17425255.2021.1908996] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Accurate prediction of pharmacokinetic (PK) and toxicokinetics (TK) of drugs is imperative for successful development of new pharmaceutics. Although conventional in vitro methods for predicting the PK and TK of drugs are well established, limitations still exist and more advanced chip-based in vitro platforms combined with mathematical models can help researchers overcome the limitations. Areas covered: We will review recent progress in the development of multi-organ-on-a-chip platforms for predicting PK and TK of drugs, as well as mathematical approaches that can be combined with these platforms for experiment design, data analysis and in vitro-in vivo extrapolation (IVIVE) for application to humans. Expert opinion: Although there remain some challenges to be addressed, the remarkable progress in the area of multi-organ-on-a-chip in recent years indicate that we will see tangible outcomes that can be utilized in the pharmaceutical industry in near future.
Collapse
Affiliation(s)
- Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul, sejong, Republic of Korea
| |
Collapse
|
30
|
Ashford MB, England RM, Akhtar N. Highway to Success—Developing Advanced Polymer Therapeutics. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000285] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Marianne B. Ashford
- Advanced Drug Delivery Pharmaceutical Sciences, R&D, AstraZeneca Macclesfield SK10 2NA UK
| | - Richard M. England
- Advanced Drug Delivery Pharmaceutical Sciences, R&D, AstraZeneca Macclesfield SK10 2NA UK
| | - Nadim Akhtar
- New Modalities & Parenteral Development Pharmaceutical Technology & Development, Operations, AstraZeneca Macclesfield SK10 2NA UK
| |
Collapse
|
31
|
Yamanaka M, Wen X, Imamura S, Sakai R, Terada S, Kamei KI. Cyclo olefin polymer-based solvent-free mass-productive microphysiological systems. Biomed Mater 2021; 16. [PMID: 33588402 DOI: 10.1088/1748-605x/abe660] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 02/15/2021] [Indexed: 12/26/2022]
Abstract
A microphysiological system (MPS) holds great promise for drug screening and toxicological testing as an alternative to animal models. However, this platform faces several challenges in terms of the materials used (e.g., polydimethylsiloxane; PDMS). For instance, absorption of drug candidates and fluorescent dyes into PDMS, as well as the effect elicited by materials on cultured cells, can cause inaccurate or misleading results in cell assays. The use of PDMS also poses challenges for mass production and long-term storage of fabricated MPSs. Hence, to circumvent these issues, herein we describe the development of a cyclo olefin polymer (COP)-based MPS using photobonding processes and vacuum ultraviolet (VUV), designated as COP-VUV-MPS. COP is an amorphous polymer with chemical/physical stability, high purity and optical clarity. Due to the thermostability and high modulus of COP, the metal molding processes was applied for mass production of MPSs without deformation of microstructures and with quick fabrication cycle time (approx. 10 min/cycle). Moreover, VUV photobonding process with an excimer light at a 172-nm wavelength allowed assembling COP materials without the use of additional solvents and tapes, which might cause cell damages. In comparison with the conventional MPS made of PDMS (PDMS-MPS), COP-VUV-MPS showed improved chemical resistance without causing molecule absorption. Moreover, COP-VUV-MPS maintained the stemness of environmentally sensitive human-induced pluripotent stem cells without causing undesired cellular phenotypes or gene expression. These results suggest that COP-VUV-MPS may be broadly applicable for the advancement of MPS and applications in drug development, as well as in vitro toxicological testing.
Collapse
Affiliation(s)
- Makoto Yamanaka
- Ushio Inc, 1-6-5 Marunouchi, Chiyoda-ku, Chiyoda-ku, Tokyo, 100-8150, JAPAN
| | - Xiaopeng Wen
- Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto, 606-8501, JAPAN
| | - Satoshi Imamura
- Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto, 606-8501, JAPAN
| | - Risako Sakai
- Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto, 606-8501, JAPAN
| | - Shiho Terada
- Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto, 606-8501, JAPAN
| | - Ken-Ichiro Kamei
- Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto, 606-8501, JAPAN
| |
Collapse
|
32
|
Pointon A, Maher J, Davis M, Baker T, Cichocki J, Ramsden D, Hale C, Kolaja KL, Levesque P, Sura R, Stresser DM, Gintant G. Cardiovascular microphysiological systems (CVMPS) for safety studies - a pharma perspective. LAB ON A CHIP 2021; 21:458-472. [PMID: 33471007 DOI: 10.1039/d0lc01040e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The integrative responses of the cardiovascular (CV) system are essential for maintaining blood flow to provide oxygenation, nutrients, and waste removal for the entire body. Progress has been made in independently developing simple in vitro models of two primary components of the CV system, namely the heart (using induced pluripotent stem-cell derived cardiomyocytes) and the vasculature (using endothelial cells and smooth muscle cells). These two in vitro biomimics are often described as immature and simplistic, and typically lack the structural complexity of native tissues. Despite these limitations, they have proven useful for specific "fit for purpose" applications, including early safety screening. More complex in vitro models offer the tantalizing prospect of greater refinement in risk assessments. To this end, efforts to physically link cardiac and vascular components to mimic a true CV microphysiological system (CVMPS) are ongoing, with the goal of providing a more holistic and integrated CV response model. The challenges of building and implementing CVMPS in future pharmacological safety studies are many, and include a) the need for more complex (and hence mature) cell types and tissues, b) the need for more realistic vasculature (within and across co-modeled tissues), and c) the need to meaningfully couple these two components to allow for integrated CV responses. Initial success will likely come with simple, bioengineered tissue models coupled with fluidics intended to mirror a vascular component. While the development of more complex integrated CVMPS models that are capable of differentiating safe compounds and providing mechanistic evaluations of CV liabilities may be feasible, adoption by pharma will ultimately hinge on model efficiency, experimental reproducibility, and added value above current strategies.
Collapse
Affiliation(s)
- Amy Pointon
- Functional Mechanistic Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Jonathan Maher
- Translational Safety Sciences, Theravance Biopharma, South San Francisco, CA 94080, USA
| | - Myrtle Davis
- Discovery Toxicology, Bristol-Myers Squibb Company, 3553 Lawrenceville Rd Princeton, NJ 08540, USA
| | - Thomas Baker
- Eli Lilly, Lilly Corporate Center, Indianapolis IN 46285, USA
| | | | - Diane Ramsden
- Takeda Pharmaceuticals, 35 Landsdowne St., Cambridge, MA 02139, UK
| | - Christopher Hale
- Amgen Research, 1120 Veterans Blvd., S. San Francisco, 94080, USA
| | - Kyle L Kolaja
- Investigative Toxicology and Cell Therapy, Bristol-Myers Squibb Company, 556 Morris Avenue, Summit NJ 07042, USA
| | - Paul Levesque
- Discovery Toxicology, Bristol-Myers Squibb Company, 3553 Lawrenceville Rd Princeton, NJ 08540, USA
| | | | - David M Stresser
- Drug Metabolism, Pharmacokinetics and Translational Modeling, AbbVie, 1 Waukegan Rd, N Chicago, IL 60064, USA
| | - Gary Gintant
- Integrative Pharmacology, Integrated Science and Technology, AbbVie, 1 Waukegan Rd, N Chicago, IL 60064, USA.
| |
Collapse
|
33
|
Tadevosyan K, Iglesias-García O, Mazo MM, Prósper F, Raya A. Engineering and Assessing Cardiac Tissue Complexity. Int J Mol Sci 2021; 22:ijms22031479. [PMID: 33540699 PMCID: PMC7867236 DOI: 10.3390/ijms22031479] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 01/28/2021] [Accepted: 01/28/2021] [Indexed: 01/14/2023] Open
Abstract
Cardiac tissue engineering is very much in a current focus of regenerative medicine research as it represents a promising strategy for cardiac disease modelling, cardiotoxicity testing and cardiovascular repair. Advances in this field over the last two decades have enabled the generation of human engineered cardiac tissue constructs with progressively increased functional capabilities. However, reproducing tissue-like properties is still a pending issue, as constructs generated to date remain immature relative to native adult heart. Moreover, there is a high degree of heterogeneity in the methodologies used to assess the functionality and cardiac maturation state of engineered cardiac tissue constructs, which further complicates the comparison of constructs generated in different ways. Here, we present an overview of the general approaches developed to generate functional cardiac tissues, discussing the different cell sources, biomaterials, and types of engineering strategies utilized to date. Moreover, we discuss the main functional assays used to evaluate the cardiac maturation state of the constructs, both at the cellular and the tissue levels. We trust that researchers interested in developing engineered cardiac tissue constructs will find the information reviewed here useful. Furthermore, we believe that providing a unified framework for comparison will further the development of human engineered cardiac tissue constructs displaying the specific properties best suited for each particular application.
Collapse
Affiliation(s)
- Karine Tadevosyan
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL) and Program for Clinical Translation of Regenerative Medicine in Catalonia (P-CMRC), 08908 L’Hospitalet del Llobregat, Spain;
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Olalla Iglesias-García
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL) and Program for Clinical Translation of Regenerative Medicine in Catalonia (P-CMRC), 08908 L’Hospitalet del Llobregat, Spain;
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, 31008 Pamplona, Spain; (M.M.M.); (F.P.)
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Correspondence: (O.I.-G.); (A.R.)
| | - Manuel M. Mazo
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, 31008 Pamplona, Spain; (M.M.M.); (F.P.)
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Hematology and Cell Therapy Area, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Felipe Prósper
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, 31008 Pamplona, Spain; (M.M.M.); (F.P.)
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Hematology and Cell Therapy Area, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- Center for Networked Biomedical Research on Cancer (CIBERONC), 28029 Madrid, Spain
| | - Angel Raya
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL) and Program for Clinical Translation of Regenerative Medicine in Catalonia (P-CMRC), 08908 L’Hospitalet del Llobregat, Spain;
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
- Correspondence: (O.I.-G.); (A.R.)
| |
Collapse
|
34
|
Hodgkinson T, Tsimbouri PM, Llopis-Hernandez V, Campsie P, Scurr D, Childs PG, Phillips D, Donnelly S, Wells JA, O'Brien FJ, Salmeron-Sanchez M, Burgess K, Alexander M, Vassalli M, Oreffo ROC, Reid S, France DJ, Dalby MJ. The use of nanovibration to discover specific and potent bioactive metabolites that stimulate osteogenic differentiation in mesenchymal stem cells. SCIENCE ADVANCES 2021; 7:7/9/eabb7921. [PMID: 33637520 PMCID: PMC7909882 DOI: 10.1126/sciadv.abb7921] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 01/12/2021] [Indexed: 05/02/2023]
Abstract
Bioactive metabolites have wide-ranging biological activities and are a potential source of future research and therapeutic tools. Here, we use nanovibrational stimulation to induce osteogenic differentiation of mesenchymal stem cells, in the absence of off-target, nonosteogenic differentiation. We show that this differentiation method, which does not rely on the addition of exogenous growth factors to culture media, provides an artifact-free approach to identifying bioactive metabolites that specifically and potently induce osteogenesis. We first identify a highly specific metabolite, cholesterol sulfate, an endogenous steroid. Next, a screen of other small molecules with a similar steroid scaffold identified fludrocortisone acetate with both specific and highly potent osteogenic-inducing activity. Further, we implicate cytoskeletal contractility as a measure of osteogenic potency and cell stiffness as a measure of specificity. These findings demonstrate that physical principles can be used to identify bioactive metabolites and then enable optimization of metabolite potency can be optimized by examining structure-function relationships.
Collapse
Affiliation(s)
- Tom Hodgkinson
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin D2, Ireland
| | - P Monica Tsimbouri
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Virginia Llopis-Hernandez
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Paul Campsie
- SUPA Department of Biomedical Engineering, University of Strathclyde, Glasgow G1 1QE, UK
| | - David Scurr
- School of Pharmacy, The University of Nottingham, Nottingham NG7 2RD, UK
| | - Peter G Childs
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow G12 8LT, UK
| | - David Phillips
- School of Chemistry, College of Science and Engineering, University of Glasgow, Glasgow G12 8QQ, UK
| | - Sam Donnelly
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Julia A Wells
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, UK
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin D2, Ireland
| | - Manuel Salmeron-Sanchez
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow G12 8LT, UK
| | - Karl Burgess
- Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, University of Glasgow, Switchback Rd., Bearsden, Glasgow G61 1BD, UK
| | - Morgan Alexander
- School of Pharmacy, The University of Nottingham, Nottingham NG7 2RD, UK
| | - Massimo Vassalli
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow G12 8LT, UK
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, UK
| | - Stuart Reid
- SUPA Department of Biomedical Engineering, University of Strathclyde, Glasgow G1 1QE, UK
| | - David J France
- School of Chemistry, College of Science and Engineering, University of Glasgow, Glasgow G12 8QQ, UK
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK.
| |
Collapse
|
35
|
Rusyn I, Roth A. Editorial overview of the special issue on application of tissue chips in toxicology. Toxicology 2021; 450:152687. [PMID: 33484733 DOI: 10.1016/j.tox.2021.152687] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 11/29/2022]
Affiliation(s)
- Ivan Rusyn
- Department of Veterinary Integrative Biosciences, College of veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA.
| | - Adrian Roth
- Product Development Safety, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
36
|
Dufau J, Shen JX, Couchet M, De Castro Barbosa T, Mejhert N, Massier L, Griseti E, Mouisel E, Amri EZ, Lauschke VM, Rydén M, Langin D. In vitro and ex vivo models of adipocytes. Am J Physiol Cell Physiol 2021; 320:C822-C841. [PMID: 33439778 DOI: 10.1152/ajpcell.00519.2020] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adipocytes are specialized cells with pleiotropic roles in physiology and pathology. Several types of fat cells with distinct metabolic properties coexist in various anatomically defined fat depots in mammals. White, beige, and brown adipocytes differ in their handling of lipids and thermogenic capacity, promoting differences in size and morphology. Moreover, adipocytes release lipids and proteins with paracrine and endocrine functions. The intrinsic properties of adipocytes pose specific challenges in culture. Mature adipocytes float in suspension culture due to high triacylglycerol content and are fragile. Moreover, a fully differentiated state, notably acquirement of the unilocular lipid droplet of white adipocyte, has so far not been reached in two-dimensional culture. Cultures of mouse and human-differentiated preadipocyte cell lines and primary cells have been established to mimic white, beige, and brown adipocytes. Here, we survey various models of differentiated preadipocyte cells and primary mature adipocyte survival describing main characteristics, culture conditions, advantages, and limitations. An important development is the advent of three-dimensional culture, notably of adipose spheroids that recapitulate in vivo adipocyte function and morphology in fat depots. Challenges for the future include isolation and culture of adipose-derived stem cells from different anatomic location in animal models and humans differing in sex, age, fat mass, and pathophysiological conditions. Further understanding of fat cell physiology and dysfunction will be achieved through genetic manipulation, notably CRISPR-mediated gene editing. Capturing adipocyte heterogeneity at the single-cell level within a single fat depot will be key to understanding diversities in cardiometabolic parameters among lean and obese individuals.
Collapse
Affiliation(s)
- Jérémy Dufau
- Inserm, Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR1297, Toulouse, France.,Faculté de Médecine, I2MC, UMR1297, Université de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Joanne X Shen
- Karolinska Institutet, Department of Physiology and Pharmacology, Stockholm, Sweden
| | - Morgane Couchet
- Karolinska Institutet, Department of Medicine (H7), Stockholm, Sweden
| | | | - Niklas Mejhert
- Karolinska Institutet, Department of Medicine (H7), Stockholm, Sweden
| | - Lucas Massier
- Karolinska Institutet, Department of Medicine (H7), Stockholm, Sweden
| | - Elena Griseti
- Inserm, Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR1297, Toulouse, France.,Faculté de Médecine, I2MC, UMR1297, Université de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Etienne Mouisel
- Inserm, Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR1297, Toulouse, France.,Faculté de Médecine, I2MC, UMR1297, Université de Toulouse, Université Paul Sabatier, Toulouse, France
| | | | - Volker M Lauschke
- Karolinska Institutet, Department of Physiology and Pharmacology, Stockholm, Sweden
| | - Mikael Rydén
- Karolinska Institutet, Department of Medicine (H7), Stockholm, Sweden
| | - Dominique Langin
- Inserm, Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR1297, Toulouse, France.,Faculté de Médecine, I2MC, UMR1297, Université de Toulouse, Université Paul Sabatier, Toulouse, France.,Toulouse University Hospitals, Department of Biochemistry, Toulouse, France
| |
Collapse
|
37
|
Abstract
Microphysiological systems (MPS), often referred to as "organ-on-chips," are microfluidic-based in vitro models that aim to recapitulate the dynamic chemical and mechanical microenvironment of living organs. MPS promise to bridge the gap between in vitro and in vivo models and ultimately improve the translation from preclinical animal studies to clinical trials. However, despite the explosion of interest in this area in recent years, and the obvious rewards for such models that could improve R&D efficiency and reduce drug attrition in the clinic, the pharmaceutical industry has been slow to fully adopt this technology. The ability to extract robust, quantitative information from MPS at scale is a key requirement if these models are to impact drug discovery and the subsequent drug development process. Microscopy imaging remains a core technology that enables the capture of information at the single-cell level and with subcellular resolution. Furthermore, such imaging techniques can be automated, increasing throughput and enabling compound screening. In this review, we discuss a range of imaging techniques that have been applied to MPS of varying focus, such as organoids and organ-chip-type models. We outline the opportunities these technologies can bring in terms of understanding mechanistic biology, but also how they could be used in higher-throughput screens, widening the scope of their impact in drug discovery. We discuss the associated challenges of imaging these complex models and the steps required to enable full exploitation. Finally, we discuss the requirements for MPS, if they are to be applied at a scale necessary to support drug discovery projects.
Collapse
Affiliation(s)
- Samantha Peel
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, United Kingdom
| | - Mark Jackman
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Cambridge, United Kingdom
| |
Collapse
|
38
|
Li J, Hua Y, Miyagawa S, Zhang J, Li L, Liu L, Sawa Y. hiPSC-Derived Cardiac Tissue for Disease Modeling and Drug Discovery. Int J Mol Sci 2020; 21:E8893. [PMID: 33255277 PMCID: PMC7727666 DOI: 10.3390/ijms21238893] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 12/20/2022] Open
Abstract
Relevant, predictive normal, or disease model systems are of vital importance for drug development. The difference between nonhuman models and humans could contribute to clinical trial failures despite ideal nonhuman results. As a potential substitute for animal models, human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs) provide a powerful tool for drug toxicity screening, modeling cardiovascular diseases, and drug discovery. Here, we review recent hiPSC-CM disease models and discuss the features of hiPSC-CMs, including subtype and maturation and the tissue engineering technologies for drug assessment. Updates from the international multisite collaborators/administrations for development of novel drug discovery paradigms are also summarized.
Collapse
Affiliation(s)
- Junjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
- Department of Cell Design for Tissue Construction, Faculty of Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ying Hua
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Jingbo Zhang
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Lingjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Li Liu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
- Department of Design for Tissue Regeneration, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| |
Collapse
|
39
|
Fritschen A, Blaeser A. Biosynthetic, biomimetic, and self-assembled vascularized Organ-on-a-Chip systems. Biomaterials 2020; 268:120556. [PMID: 33310539 DOI: 10.1016/j.biomaterials.2020.120556] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/15/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023]
Abstract
Organ-on-a-Chip (OOC) devices have seen major advances in the last years with respect to biological complexity, physiological composition and biomedical relevance. In this context, integration of vasculature has proven to be a crucial element for long-term culture of thick tissue samples as well as for realistic pharmacokinetic, toxicity and metabolic modelling. With the emergence of digital production technologies and the reinvention of existing tools, a multitude of design approaches for guided angio- and vasculogenesis is available today. The underlying production methods can be categorized into biosynthetic, biomimetic and self-assembled vasculature formation. The diversity and importance of production approaches, vascularization strategies as well as biomaterials and cell sourcing are illustrated in this work. A comprehensive technological review with a strong focus on the challenge of producing physiologically relevant vascular structures is given. Finally, the remaining obstacles and opportunities in the development of vascularized Organ-on-a-Chip platforms for advancing drug development and predictive disease modelling are noted.
Collapse
Affiliation(s)
- Anna Fritschen
- Institute for BioMedical Printing Technology, Technical University of Darmstadt, Germany.
| | - Andreas Blaeser
- Institute for BioMedical Printing Technology, Technical University of Darmstadt, Germany; Centre for Synthetic Biology, Technical University of Darmstadt, Germany.
| |
Collapse
|
40
|
Langley G. Lush Prize Conference 2018: The Times They are A-Changin. Altern Lab Anim 2020; 48:12S-16S. [PMID: 33106007 DOI: 10.1177/0261192920911340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
41
|
|
42
|
Bae M, Yi HG, Jang J, Cho DW. Microphysiological Systems for Neurodegenerative Diseases in Central Nervous System. MICROMACHINES 2020; 11:E855. [PMID: 32947879 PMCID: PMC7570039 DOI: 10.3390/mi11090855] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/13/2020] [Accepted: 09/14/2020] [Indexed: 12/19/2022]
Abstract
Neurodegenerative diseases are among the most severe problems in aging societies. Various conventional experimental models, including 2D and animal models, have been used to investigate the pathogenesis of (and therapeutic mechanisms for) neurodegenerative diseases. However, the physiological gap between humans and the current models remains a hurdle to determining the complexity of an irreversible dysfunction in a neurodegenerative disease. Therefore, preclinical research requires advanced experimental models, i.e., those more physiologically relevant to the native nervous system, to bridge the gap between preclinical stages and patients. The neural microphysiological system (neural MPS) has emerged as an approach to summarizing the anatomical, biochemical, and pathological physiology of the nervous system for investigation of neurodegenerative diseases. This review introduces the components (such as cells and materials) and fabrication methods for designing a neural MPS. Moreover, the review discusses future perspectives for improving the physiological relevance to native neural systems.
Collapse
Affiliation(s)
- Mihyeon Bae
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Chungam-ro, Nam-gu, Pohang 37673, Korea;
| | - Hee-Gyeong Yi
- Department of Rural and Biosystems Engineering, College of Agricultural Sciences, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Korea
| | - Jinah Jang
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Chungam-ro, Nam-gu, Pohang 37673, Korea;
- Department of Creative IT Engineering, Pohang University of Science and Technology (POSTECH), 77 Chungam-ro, Nam-gu, Pohang 37673, Korea
- Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Chungam-ro, Nam-gu, Pohang 37673, Korea;
- Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| |
Collapse
|
43
|
Deal HE, Brown AC, Daniele MA. Microphysiological systems for the modeling of wound healing and evaluation of pro-healing therapies. J Mater Chem B 2020; 8:7062-7075. [PMID: 32756718 PMCID: PMC7460719 DOI: 10.1039/d0tb00544d] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Wound healing is a multivariate process involving the coordinated response of numerous proteins and cell types. Accordingly, biomedical research has seen an increased adoption of the use of in vitro wound healing assays with complexity beyond that offered by traditional well-plate constructs. These microphysiological systems (MPS) seek to recapitulate one or more physiological features of the in vivo microenvironment, while retaining the analytical capacity of more reductionist assays. Design efforts to achieve relevant wound healing physiology include the use of dynamic perfusion over static culture, the incorporation of multiple cell types, the arrangement of cells in three dimensions, the addition of biomechanically and biochemically relevant hydrogels, and combinations thereof. This review provides a brief overview of the wound healing process and in vivo assays, and we critically review the current state of MPS and supporting technologies for modelling and studying wound healing. We distinguish between MPS that seek to inform a particular phase of wound healing, and constructs that have the potential to inform multiple phases of wound healing. This distinction is a product of whether analysis of a particular process is prioritized, or a particular physiology is prioritized, during design. Material selection is emphasized throughout, and relevant fabrication techniques discussed.
Collapse
Affiliation(s)
- Halston E Deal
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA. and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA
| | - Ashley C Brown
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA. and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA
| | - Michael A Daniele
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA. and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA and Department of Electrical & Computer Engineering, North Carolina State University, 890 Oval Dr., Raleigh, NC 27695, USA
| |
Collapse
|
44
|
Filippo Buono M, von Boehmer L, Strang J, P. Hoerstrup S, Y. Emmert M, Nugraha B. Human Cardiac Organoids for Modeling Genetic Cardiomyopathy. Cells 2020; 9:cells9071733. [PMID: 32698471 PMCID: PMC7409052 DOI: 10.3390/cells9071733] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Genetic cardiomyopathies are characterized by changes in the function and structure of the myocardium. The development of a novel in vitro model could help to better emulate healthy and diseased human heart conditions and may improve the understanding of disease mechanisms. In this study, for the first time, we demonstrated the generation of cardiac organoids using a triculture approach of human induced pluripotent stem-cell-derived cardiomyocytes (hiPS-CMs)-from healthy subjects and cardiomyopathy patients-human cardiac microvascular endothelial cells (HCMECs) and human cardiac fibroblasts (HCFs). We assessed the organoids' suitability as a 3D cellular model for the representation of phenotypical features of healthy and cardiomyopathic hearts. We observed clear differences in structure and beating behavior between the organoid groups, depending on the type of hiPS-CMs (healthy versus cardiomyopathic) used. Organoids may thus prove a promising tool for the design and testing of patient-specific treatments as well as provide a platform for safer and more efficacious drug development.
Collapse
Affiliation(s)
- Michele Filippo Buono
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland; (M.F.B.); (L.v.B.); (S.P.H.)
| | - Lisa von Boehmer
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland; (M.F.B.); (L.v.B.); (S.P.H.)
| | - Jaan Strang
- Zurich University of Applied Sciences, 8820 Wädenswil, Switzerland;
| | - Simon P. Hoerstrup
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland; (M.F.B.); (L.v.B.); (S.P.H.)
- Wyss Translational Center Zurich, 8006 Zurich, Switzerland
| | - Maximilian Y. Emmert
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland; (M.F.B.); (L.v.B.); (S.P.H.)
- Wyss Translational Center Zurich, 8006 Zurich, Switzerland
- Department of Cardiovascular Surgery, University Hospital Zurich, 8091 Zurich, Switzerland
- Department of Cardiovascular Surgery, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
- Correspondence: (M.Y.E.); (B.N.); Tel.: +41-44-634-5610 (M.Y.E.); +41-635-8533 (B.N.)
| | - Bramasta Nugraha
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland; (M.F.B.); (L.v.B.); (S.P.H.)
- Correspondence: (M.Y.E.); (B.N.); Tel.: +41-44-634-5610 (M.Y.E.); +41-635-8533 (B.N.)
| |
Collapse
|
45
|
Nudischer R, Renggli K, Hierlemann A, Roth AB, Bertinetti-Lapatki C. Characterization of a long-term mouse primary liver 3D tissue model recapitulating innate-immune responses and drug-induced liver toxicity. PLoS One 2020; 15:e0235745. [PMID: 32645073 PMCID: PMC7347206 DOI: 10.1371/journal.pone.0235745] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 06/22/2020] [Indexed: 02/07/2023] Open
Abstract
Three-dimensional liver in vitro systems have recently attracted a lot of attention in drug development. These systems help to gain unprecedented insights into drug-induced liver injury (DILI), as they more closely reproduce liver biology, and as drug effects can be studied in isolated and controllable microenvironments. Many groups established human-based in vitro models but so far neglected the animal equivalent, although the availability of both models would be desirable. Animal in vitro models enable back- and forward translation of in vitro and in vivo findings, bridge the gap between rodent in vivo and human in vitro scenarios, and ultimately support the interpretation of data generated with preclinical species and humans. Since mice are often used in drug development and physiologically relevant in vitro systems are lacking, we established, for the first time, a mouse liver model that encompasses primary parenchymal and non-parenchymal cells with preserved viability and functionality over three weeks. Using our three-dimensional liver spheroids, we were able to predict the toxicity of known DILI compounds, demonstrated the interaction cascades between the different cell types and showed evidence of drug-induced steatosis and cholestasis. In summary, our mouse liver spheroids represent a valuable in vitro model that can be applied to study DILI findings, reported from mouse studies, and offers the potential to detect immune-mediated drug-induced liver toxicity.
Collapse
Affiliation(s)
- Ramona Nudischer
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
- * E-mail:
| | - Kasper Renggli
- Bioengineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Zurich, Switzerland
| | - Andreas Hierlemann
- Bioengineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Zurich, Switzerland
| | - Adrian B. Roth
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Cristina Bertinetti-Lapatki
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
46
|
An FDA/CDER perspective on nonclinical testing strategies: Classical toxicology approaches and new approach methodologies (NAMs). Regul Toxicol Pharmacol 2020; 114:104662. [DOI: 10.1016/j.yrtph.2020.104662] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/03/2020] [Accepted: 04/15/2020] [Indexed: 02/08/2023]
|
47
|
Baltazar MT, Cable S, Carmichael PL, Cubberley R, Cull T, Delagrange M, Dent MP, Hatherell S, Houghton J, Kukic P, Li H, Lee MY, Malcomber S, Middleton AM, Moxon TE, Nathanail AV, Nicol B, Pendlington R, Reynolds G, Reynolds J, White A, Westmoreland C. A Next-Generation Risk Assessment Case Study for Coumarin in Cosmetic Products. Toxicol Sci 2020; 176:236-252. [PMID: 32275751 PMCID: PMC7357171 DOI: 10.1093/toxsci/kfaa048] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Next-Generation Risk Assessment is defined as an exposure-led, hypothesis-driven risk assessment approach that integrates new approach methodologies (NAMs) to assure safety without the use of animal testing. These principles were applied to a hypothetical safety assessment of 0.1% coumarin in face cream and body lotion. For the purpose of evaluating the use of NAMs, existing animal and human data on coumarin were excluded. Internal concentrations (plasma Cmax) were estimated using a physiologically based kinetic model for dermally applied coumarin. Systemic toxicity was assessed using a battery of in vitro NAMs to identify points of departure (PoDs) for a variety of biological effects such as receptor-mediated and immunomodulatory effects (Eurofins SafetyScreen44 and BioMap Diversity 8 Panel, respectively), and general bioactivity (ToxCast data, an in vitro cell stress panel and high-throughput transcriptomics). In addition, in silico alerts for genotoxicity were followed up with the ToxTracker tool. The PoDs from the in vitro assays were plotted against the calculated in vivo exposure to calculate a margin of safety with associated uncertainty. The predicted Cmax values for face cream and body lotion were lower than all PoDs with margin of safety higher than 100. Furthermore, coumarin was not genotoxic, did not bind to any of the 44 receptors tested and did not show any immunomodulatory effects at consumer-relevant exposures. In conclusion, this case study demonstrated the value of integrating exposure science, computational modeling and in vitro bioactivity data, to reach a safety decision without animal data.
Collapse
Affiliation(s)
- Maria T Baltazar
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Sophie Cable
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Paul L Carmichael
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Richard Cubberley
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Tom Cull
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Mona Delagrange
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Matthew P Dent
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Sarah Hatherell
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Jade Houghton
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Predrag Kukic
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Hequn Li
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Mi-Young Lee
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Sophie Malcomber
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Alistair M Middleton
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Thomas E Moxon
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Alexis V Nathanail
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Beate Nicol
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Ruth Pendlington
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Georgia Reynolds
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Joe Reynolds
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Andrew White
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Carl Westmoreland
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| |
Collapse
|
48
|
Busquet F, Hartung T, Pallocca G, Rovida C, Leist M. Harnessing the power of novel animal-free test methods for the development of COVID-19 drugs and vaccines. Arch Toxicol 2020; 94:2263-2272. [PMID: 32447523 PMCID: PMC7245508 DOI: 10.1007/s00204-020-02787-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/18/2020] [Indexed: 01/13/2023]
Abstract
The COVID-19-inducing virus, SARS-CoV2, is likely to remain a threat to human health unless efficient drugs or vaccines become available. Given the extent of the current pandemic (people in over one hundred countries infected) and its disastrous effect on world economy (associated with limitations of human rights), speedy drug discovery is critical. In this situation, past investments into the development of new (animal-free) approach methods (NAM) for drug safety, efficacy, and quality evaluation can be leveraged. For this, we provide an overview of repurposing ideas to shortcut drug development times. Animal-based testing would be too lengthy, and it largely fails, when a pathogen is species-specific or if the desired drug is based on specific features of human biology. Fortunately, industry has already largely shifted to NAM, and some public funding programs have advanced the development of animal-free technologies. For instance, NAM can predict genotoxicity (a major aspect of carcinogenicity) within days, human antibodies targeting virus epitopes can be generated in molecular biology laboratories within weeks, and various human cell-based organoids are available to test virus infectivity and the biological processes controlling them. The European Medicines Agency (EMA) has formed an expert group to pave the way for the use of such approaches for accelerated drug development. This situation illustrates the importance of diversification in drug discovery strategies and clearly shows the shortcomings of an approach that invests 95% of resources into a single technology (animal experimentation) in the face of challenges that require alternative approaches.
Collapse
Affiliation(s)
- Francois Busquet
- CAAT-Europe at the University of Konstanz, 78457, Konstanz, Germany
- ALTERTOX, 1000, Brussels, Belgium
| | - Thomas Hartung
- CAAT-Europe at the University of Konstanz, 78457, Konstanz, Germany
- CAAT, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Giorgia Pallocca
- CAAT-Europe at the University of Konstanz, 78457, Konstanz, Germany
| | - Costanza Rovida
- CAAT-Europe at the University of Konstanz, 78457, Konstanz, Germany
| | - Marcel Leist
- CAAT-Europe at the University of Konstanz, 78457, Konstanz, Germany.
- In Vitro Toxicology and Biomedicine, Department Inaugurated By the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457, Konstanz, Germany.
| |
Collapse
|
49
|
Walker PA, Ryder S, Lavado A, Dilworth C, Riley RJ. The evolution of strategies to minimise the risk of human drug-induced liver injury (DILI) in drug discovery and development. Arch Toxicol 2020; 94:2559-2585. [PMID: 32372214 PMCID: PMC7395068 DOI: 10.1007/s00204-020-02763-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 04/22/2020] [Indexed: 12/15/2022]
Abstract
Early identification of toxicity associated with new chemical entities (NCEs) is critical in preventing late-stage drug development attrition. Liver injury remains a leading cause of drug failures in clinical trials and post-approval withdrawals reflecting the poor translation between traditional preclinical animal models and human clinical outcomes. For this reason, preclinical strategies have evolved over recent years to incorporate more sophisticated human in vitro cell-based models with multi-parametric endpoints. This review aims to highlight the evolution of the strategies adopted to improve human hepatotoxicity prediction in drug discovery and compares/contrasts these with recent activities in our lab. The key role of human exposure and hepatic drug uptake transporters (e.g. OATPs, OAT2) is also elaborated.
Collapse
Affiliation(s)
- Paul A Walker
- Cyprotex Discovery Ltd., No.24 Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, UK.
| | - Stephanie Ryder
- Cyprotex Discovery Ltd., No.24 Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, UK
| | - Andrea Lavado
- Cyprotex Discovery Ltd., No.24 Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, UK
| | - Clive Dilworth
- Cyprotex Discovery Ltd., No.24 Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, UK.,Alderley Park Accelerator, Alderley Park, Macclesfield, Cheshire, SK10 4TG, UK
| | - Robert J Riley
- Cyprotex Discovery Ltd., No.24 Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, UK
| |
Collapse
|
50
|
Collins TA, Rolf MG, Pointon A. Current and future approaches to nonclinical cardiovascular safety assessment. Drug Discov Today 2020; 25:1129-1134. [PMID: 32209428 DOI: 10.1016/j.drudis.2020.03.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/03/2020] [Accepted: 03/16/2020] [Indexed: 12/21/2022]
Abstract
Our goal is to accurately predict all types of cardiovascular events in patients utilising nonclinical cardiovascular safety data. In the past two decades, cardiovascular safety science has primarily focused on events associated with the electrocardiogram. Broadening out to other cardiovascular parameters, we share real-life case studies that highlight our progress towards improved and better-informed project progression based upon use of disease models, mechanism-based translation and structure-function relationships. To fulfil this goal, further advances in patient-relevant humanised models will be required to enable cardiovascular safety science to keep pace with the ever-changing landscape of novel therapeutic paradigms.
Collapse
Affiliation(s)
- Teresa A Collins
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Michael G Rolf
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Amy Pointon
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK.
| |
Collapse
|