1
|
Addario G, Moroni L, Mota C. Kidney Fibrosis In Vitro and In Vivo Models: Path Toward Physiologically Relevant Humanized Models. Adv Healthc Mater 2025; 14:e2403230. [PMID: 39906010 PMCID: PMC11973949 DOI: 10.1002/adhm.202403230] [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: 08/27/2024] [Revised: 01/14/2025] [Indexed: 02/06/2025]
Abstract
Chronic kidney disease (CKD) affects over 10% of the global population and is a leading cause of mortality. Kidney fibrosis, a key endpoint of CKD, disrupts nephron tubule anatomy and filtration function, and disease pathomechanisms are not fully understood. Kidney fibrosis is currently investigated with in vivo models, that gradually support the identification of possible mechanisms of fibrosis, but with limited translational research, as they do not fully recapitulate human kidney physiology, metabolism, and molecular pathways. In vitro 2D cell culture models are currently used, as a starting point in disease modeling and pharmacology, however, they lack the 3D kidney architecture complexity and functions. The failure of several therapies and drugs in clinical trials highlights the urgent need for advanced 3D in vitro models. This review discusses the urinary system's anatomy, associated diseases, and diagnostic methods, including biomarker analysis and tissue biopsy. It evaluates 2D and in vivo models, highlighting their limitations. The review explores the state-of-the-art 3D-humanized in vitro models, such as 3D cell aggregates, on-chip models, biofabrication techniques, and hybrid models, which aim to mimic kidney morphogenesis and functions. These advanced models hold promise for translating new therapies and drugs for kidney fibrosis into clinics.
Collapse
Affiliation(s)
- Gabriele Addario
- Department of Complex Tissue RegenerationMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityER Maastricht6229The Netherlands
| | - Lorenzo Moroni
- Department of Complex Tissue RegenerationMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityER Maastricht6229The Netherlands
| | - Carlos Mota
- Department of Complex Tissue RegenerationMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityER Maastricht6229The Netherlands
| |
Collapse
|
2
|
Zhang H, Zhao L, Brodský J, Migliaccio L, Gablech I, Neužil P, You M. Proteomics-on-a-Chip - Microfluidics meets proteomics. Biosens Bioelectron 2025; 273:117122. [PMID: 39813764 DOI: 10.1016/j.bios.2024.117122] [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: 12/09/2024] [Accepted: 12/30/2024] [Indexed: 01/18/2025]
Abstract
Proteomics provides an understanding of biological systems by enabling the detailed study of protein expression profiles, which is crucial for early disease diagnosis. Microfluidic-based proteomics enhances this field by integrating complex proteome analysis into compact and efficient systems. This review focuses on developing microfluidic chip structures for proteomics, covering on-chip sample pretreatment, protein extraction, purification, and identification in recent years. Furthermore, our work aims to inspire researchers to select proper methodologies in designing novel, efficient assays for proteomics applications by analyzing trends and innovations in this field.
Collapse
Affiliation(s)
- Haoqing Zhang
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, PR China; TFX Group-Xi'an Jiaotong University Institute of Life Health, Xi'an 710049, PR China
| | - Lei Zhao
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Jan Brodský
- Department of Microelectronics, Faculty of Electrical Engineering and Communication, Brno University of Technology, Technická 3058/10, 616 00, Brno, Czech Republic
| | - Ludovico Migliaccio
- Department of Microelectronics, Faculty of Electrical Engineering and Communication, Brno University of Technology, Technická 3058/10, 616 00, Brno, Czech Republic
| | - Imrich Gablech
- Department of Microelectronics, Faculty of Electrical Engineering and Communication, Brno University of Technology, Technická 3058/10, 616 00, Brno, Czech Republic
| | - Pavel Neužil
- Ministry of Education Key Laboratory of Micro and Nano Systems for Aerospace, School of Mechanical Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi, 710072, PR China.
| | - Minli You
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, PR China; TFX Group-Xi'an Jiaotong University Institute of Life Health, Xi'an 710049, PR China.
| |
Collapse
|
3
|
Ceccotti E, Semnani A, Bussolati B, Bruno S. Human kidney organoids for modeling the development of different diseases. Curr Top Dev Biol 2025; 163:364-393. [PMID: 40254349 DOI: 10.1016/bs.ctdb.2024.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
The increasing incidence of kidney diseases has highlighted the need for in vitro experimental models to mimic disease development and to test new therapeutic approaches. Traditional two-dimensional in vitro experimental models are not fully able to recapitulate renal diseases. Instead, kidney organoids represent three-dimensional models that better mimic the human organ from both structural and functional points of view. Human pluripotent stem cells (PSCs), both embryonic and induced, are ideal sources for generating renal organoids. These organoids contain all renal cell types and the protocols to differentiate PSCs into renal organoids consist of three different stages that recapitulate embryonic development: mesodermal induction, nephron progenitor formation, and nephron differentiation. Recently it has been establish a renal organoid model where collecting ducts are also present. In this case, the presence of ureteric bud progenitor cells is essential. Renal organoids are particularly useful for studying genetic diseases, by introducing the specific mutations in PSCs by genome editing or generating organoids from patient-derived PSCs. Moreover, renal organoids represent promising models in toxicology studies and testing new therapeutic approaches. Renal organoids can be established also from adult stem cells. This type of organoid, named tubuloid, is composed only of epithelial cells and recapitulates the tissue repair process. The tubuloids can be generated from adult stem or progenitor cells, obtained from renal biopsies or urine, and are promising in vitro models for studying tubular functions, diseases, and regeneration. Tubuloids can be derived from patients and permit the study of genetic diseases, performing personalized drug screening and modeling renal pathologies.
Collapse
Affiliation(s)
- Elena Ceccotti
- Department of Medical Sciences, University of Torino, Corso Dogliotti, Torino, Italy
| | - Armina Semnani
- Department of Medical Sciences, University of Torino, Corso Dogliotti, Torino, Italy
| | - Benedetta Bussolati
- Department of Medical Sciences, University of Torino, Corso Dogliotti, Torino, Italy; Molecular Biotechnology Center "Guido Tarone", Via Nizza, Torino, Italy
| | - Stefania Bruno
- Department of Medical Sciences, University of Torino, Corso Dogliotti, Torino, Italy.
| |
Collapse
|
4
|
Nie J, Lou S, Pollet AMAO, van Vegchel M, Bouten CVC, den Toonder JMJ. A Cell Pre-Wrapping Seeding Technique for Hydrogel-Based Tubular Organ-On-A-Chip. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400970. [PMID: 38872259 PMCID: PMC11321624 DOI: 10.1002/advs.202400970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/28/2024] [Indexed: 06/15/2024]
Abstract
Organ-on-a-chip (OOC) models based on microfluidic technology are increasingly used to obtain mechanistic insight into (patho)physiological processes in humans, and they hold great promise for application in drug development and regenerative medicine. Despite significant progress in OOC development, several limitations of conventional microfluidic devices pose challenges. First, most microfluidic systems have rectangular cross sections and flat walls, and therefore tubular/ curved structures, like blood vessels and nephrons, are not well represented. Second, polymers used as base materials for microfluidic devices are much stiffer than in vivo extracellular matrix (ECM). Finally, in current cell seeding methods, challenges exist regarding precise control over cell seeding location, unreachable spaces due to flow resistances, and restricted dimensions/geometries. To address these limitations, an alternative cell seeding technique and a corresponding workflow is introduced to create circular cross-sectioned tubular OOC models by pre-wrapping cells around sacrificial fiber templates. As a proof of concept, a perfusable renal proximal tubule-on-a-chip is demonstrated with a diameter as small as 50 µm, cellular tubular structures with branches and curvature, and a preliminary vascular-renal tubule interaction model. The cell pre-wrapping seeding technique promises to enable the construction of diverse physiological/pathological models, providing tubular OOC systems for mechanistic investigations and drug development.
Collapse
Affiliation(s)
- Jing Nie
- Microsystems Research SectionDepartment of Mechanical EngineeringEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
- Institute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyEindhoven5600 MBThe Netherlands
- Soft Tissue Engineering & Mechanobiology Research SectionDepartment of Biomedical EngineeringEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Sha Lou
- Microsystems Research SectionDepartment of Mechanical EngineeringEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
- Institute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyEindhoven5600 MBThe Netherlands
- Soft Tissue Engineering & Mechanobiology Research SectionDepartment of Biomedical EngineeringEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Andreas M. A. O. Pollet
- Microsystems Research SectionDepartment of Mechanical EngineeringEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
- Institute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Manon van Vegchel
- Microsystems Research SectionDepartment of Mechanical EngineeringEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
- Soft Tissue Engineering & Mechanobiology Research SectionDepartment of Biomedical EngineeringEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Carlijn V. C. Bouten
- Institute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyEindhoven5600 MBThe Netherlands
- Soft Tissue Engineering & Mechanobiology Research SectionDepartment of Biomedical EngineeringEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Jaap M. J. den Toonder
- Microsystems Research SectionDepartment of Mechanical EngineeringEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
- Institute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| |
Collapse
|
5
|
Bourg S, Rakotozandriny K, Lucas IT, Letavernier E, Bonhomme C, Babonneau F, Abou-Hassan A. Confining calcium oxalate crystal growth in a carbonated apatite-coated microfluidic channel to better understand the role of Randall's plaque in kidney stone formation. LAB ON A CHIP 2024; 24:2017-2024. [PMID: 38407354 DOI: 10.1039/d3lc01050c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Effective prevention of recurrent kidney stone disease requires the understanding of the mechanisms of its formation. Numerous in vivo observations have demonstrated that a large number of pathological calcium oxalate kidney stones develop on an apatitic calcium phosphate deposit, known as Randall's plaque. In an attempt to understand the role of the inorganic hydroxyapatite phase in the formation and habits of calcium oxalates, we confined their growth under dynamic physicochemical and flow conditions in a reversible microfluidic channel coated with hydroxyapatite. Using multi-scale characterization techniques including scanning electron and Raman microscopy, we showed the successful formation of carbonated hydroxyapatite as found in Randall's plaque. This was possible due to a new two-step flow seed-mediated growth strategy which allowed us to coat the channel with carbonated hydroxyapatite. Precipitation of calcium oxalates under laminar flow from supersaturated solutions of oxalate and calcium ions showed that the formation of crystals is a substrate and time dependent complex process where diffusion of oxalate ions to the surface of carbonated hydroxyapatite and the solubility of the latter are among the most important steps for the formation of calcium oxalate crystals. Indeed when an oxalate solution was flushed for 24 h, dissolution of the apatite layer and formation of calcium carbonate calcite crystals occurred which seems to promote calcium oxalate crystal formation. Such a growth route has never been observed in vivo in the context of kidney stones. Under our experimental conditions, our results do not show any direct promoting role of carbonated hydroxyapatite in the formation of calcium oxalate crystals, consolidating therefore the important role that macromolecules can play in the process of nucleation and growth of calcium oxalate crystals on Randall's plaque.
Collapse
Affiliation(s)
- Samantha Bourg
- Laboratoire Physicochimie des Electrolytes et Nanosystèmes Interfaciaux (PHENIX), CNRS, Sorbonne Université, UMR 8234, Campus Jussieu, 4 place Jussieu, F-75005 Paris, France.
| | - Karol Rakotozandriny
- Laboratoire Physicochimie des Electrolytes et Nanosystèmes Interfaciaux (PHENIX), CNRS, Sorbonne Université, UMR 8234, Campus Jussieu, 4 place Jussieu, F-75005 Paris, France.
- Laboratoire de Chimie de la Matière Condensée de Paris (LCMCP), CNRS, Sorbonne Université, UMR 7574, Campus Jussieu, 4 place Jussieu, F-75005 Paris, France
| | - Ivan T Lucas
- Laboratoire Interfaces et Systèmes Electrochimiques (LISE), CNRS, Sorbonne Université, UMR 8235, Campus Jussieu, 4 place Jussieu, F-75005 Paris, France
| | - Emmanuel Letavernier
- AP-HP, Hôpital Tenon, Explorations Fonctionnelles Multidisciplinaires et Laboratoire des Lithiases, F-75020 Paris, France
| | - Christian Bonhomme
- Laboratoire de Chimie de la Matière Condensée de Paris (LCMCP), CNRS, Sorbonne Université, UMR 7574, Campus Jussieu, 4 place Jussieu, F-75005 Paris, France
| | - Florence Babonneau
- Laboratoire de Chimie de la Matière Condensée de Paris (LCMCP), CNRS, Sorbonne Université, UMR 7574, Campus Jussieu, 4 place Jussieu, F-75005 Paris, France
| | - Ali Abou-Hassan
- Laboratoire Physicochimie des Electrolytes et Nanosystèmes Interfaciaux (PHENIX), CNRS, Sorbonne Université, UMR 8234, Campus Jussieu, 4 place Jussieu, F-75005 Paris, France.
- Institut Universitaire de France (IUF), 75231 Paris Cedex 05, France
| |
Collapse
|
6
|
Guimaraes APP, Calori IR, Stilhano RS, Tedesco AC. Renal proximal tubule-on-a-chip in PDMS: fabrication, functionalization, and RPTEC:HUVEC co-culture evaluation. Biofabrication 2024; 16:025024. [PMID: 38408383 DOI: 10.1088/1758-5090/ad2d2f] [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: 11/14/2023] [Accepted: 02/26/2024] [Indexed: 02/28/2024]
Abstract
'On-a-chip' technology advances the development of physiologically relevant organ-mimicking architecture by integrating human cells into three-dimensional microfluidic devices. This method also establishes discrete functional units, faciliting focused research on specific organ components. In this study, we detail the development and assessment of a convoluted renal proximal tubule-on-a-chip (PT-on-a-chip). This platform involves co-culturing Renal Proximal Tubule Epithelial Cells (RPTEC) and Human Umbilical Vein Endothelial Cells (HUVEC) within a polydimethylsiloxane microfluidic device, crafted through a combination of 3D printing and molding techniques. Our PT-on-a-chip significantly reduced high glucose level, exhibited albumin uptake, and simulated tubulopathy induced by amphotericin B. Remarkably, the RPTEC:HUVEC co-culture exhibited efficient cell adhesion within 30 min on microchannels functionalized with plasma, 3-aminopropyltriethoxysilane, and type-I collagen. This approach significantly reduced the required incubation time for medium perfusion. In comparison, alternative methods such as plasma and plasma plus polyvinyl alcohol were only effective in promoting cell attachment to flat surfaces. The PT-on-a-chip holds great promise as a valuable tool for assessing the nephrotoxic potential of new drug candidates, enhancing our understanding of drug interactions with co-cultured renal cells, and reducing the need for animal experimentation, promoting the safe and ethical development of new pharmaceuticals.
Collapse
Affiliation(s)
- Ana Paula Pereira Guimaraes
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering- Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, São Paulo, Ribeirão Preto 14040-901, Brazil
| | - Italo Rodrigo Calori
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering- Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, São Paulo, Ribeirão Preto 14040-901, Brazil
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Labs, Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, Oxford, MS 38677, United States of America
| | - Roberta Sessa Stilhano
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Sao Paulo, Brazil
| | - Antonio Claudio Tedesco
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering- Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, São Paulo, Ribeirão Preto 14040-901, Brazil
| |
Collapse
|
7
|
Aljabali AAA, Obeid MA, Mishra V, El-Tanani M, Tambuwala MM. Customizable Microfluidic Devices: Progress, Constraints, and Future Advances. Curr Drug Deliv 2024; 21:1285-1299. [PMID: 39034714 DOI: 10.2174/0115672018264064231017113813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/13/2023] [Accepted: 08/31/2023] [Indexed: 07/23/2024]
Abstract
The field of microfluidics encompasses the study of fluid behavior within micro-channels and the development of miniature systems featuring internal compartments or passageways tailored for fluid control and manipulation. Microfluidic devices capitalize on the unique chemical and physical properties exhibited by fluids at the microscopic scale. In contrast to their larger counterparts, microfluidic systems offer a multitude of advantages. Their implementation facilitates the investigation and utilization of reduced sample, solvent, and reagent volumes, thus yielding decreased operational expenses. Owing to their compact dimensions, these devices allow for the concurrent execution of multiple procedures, leading to expedited experimental timelines. Over the past two decades, microfluidics has undergone remarkable advancements, evolving into a multifaceted discipline. Subfields such as organ-on-a-chip and paper-based microfluidics have matured into distinct fields of study. Nonetheless, while scientific progress within the microfluidics realm has been notable, its translation into autonomous end-user applications remains a frontier to be fully explored. This paper sets forth the central objective of scrutinizing the present research paradigm, prevailing limitations, and potential prospects of customizable microfluidic devices. Our inquiry revolves around the latest strides achieved, prevailing constraints, and conceivable trajectories for adaptable microfluidic technologies. We meticulously delineate existing iterations of microfluidic systems, elucidate their operational principles, deliberate upon encountered limitations, and provide a visionary outlook toward the future trajectory of microfluidic advancements. In summation, this work endeavors to shed light on the current state of microfluidic systems, underscore their operative intricacies, address incumbent challenges, and unveil promising pathways that chart the course toward the next frontier of microfluidic innovation.
Collapse
Affiliation(s)
- Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid 21163, Jordan
| | - Mohammad A Obeid
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid 21163, Jordan
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Mohamed El-Tanani
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, England, UK
| |
Collapse
|
8
|
R N, Aggarwal A, Sravani AB, Mallya P, Lewis S. Organ-On-A-Chip: An Emerging Research Platform. Organogenesis 2023; 19:2278236. [PMID: 37965897 PMCID: PMC10653779 DOI: 10.1080/15476278.2023.2278236] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 10/27/2023] [Indexed: 11/16/2023] Open
Abstract
In drug development, conventional preclinical and clinical testing stages rely on cell cultures and animal experiments, but these methods may fall short of fully representing human biology. To overcome this limitation, the emergence of organ-on-a-chip (OOC) technology has sparked interest as a transformative approach in drug testing research. By closely replicating human organ responses to external signals, OOC devices hold immense potential in revolutionizing drug efficacy and safety predictions. This review focuses on the advancements, applications, and prospects of OOC devices in drug testing. Based on the latest advances in the field of OOC systems and their clinical applications, this review reflects the effectiveness of OOC devices in replacing human volunteers in certain clinical studies. This review underscores the critical role of OOC technology in transforming drug testing methodologies.
Collapse
Affiliation(s)
- Nithin R
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Ayushi Aggarwal
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Anne Boyina Sravani
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Pooja Mallya
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Shaila Lewis
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| |
Collapse
|
9
|
Syahruddin MH, Anggraeni R, Ana ID. A microfluidic organ-on-a-chip: into the next decade of bone tissue engineering applied in dentistry. Future Sci OA 2023; 9:FSO902. [PMID: 37753360 PMCID: PMC10518836 DOI: 10.2144/fsoa-2023-0061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 08/21/2023] [Indexed: 09/28/2023] Open
Abstract
A comprehensive understanding of the complex physiological and pathological processes associated with alveolar bones, their responses to different therapeutics strategies, and cell interactions with biomaterial becomes necessary in precisely treating patients with severe progressive periodontitis, as a bone-related issue in dentistry. However, existing monolayer cell culture or pre-clinical models have been unable to mimic the complex physiological, pathological and regeneration processes in the bone microenvironment in response to different therapeutic strategies. In this point, 'organ-on-a-chip' (OOAC) technology, specifically 'alveolar-bone-on-a-chip', is expected to resolve the problems by better imitating infection site microenvironment and microphysiology within the oral tissues. The OOAC technology is assessed in this study toward better approaches in disease modeling and better therapeutics strategy for bone tissue engineering applied in dentistry.
Collapse
Affiliation(s)
- Muhammad Hidayat Syahruddin
- Postgraduate Student, Dental Science Doctoral Study Program, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | - Rahmi Anggraeni
- Research Center for Preclinical & Clinical Medicine, National Research & Innovation Agency of the Republic of Indonesia, Cibinong Science Center, Bogor, 16915, Indonesia
- Research Collaboration Center for Biomedical Scaffolds, National Research & Innovation Agency (BRIN) – Universitas Gadjah Mada (UGM), Yogyakarta, 55281, Indonesia
| | - Ika Dewi Ana
- Department of Dental Biomedical Sciences, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
- Research Collaboration Center for Biomedical Scaffolds, National Research & Innovation Agency (BRIN) – Universitas Gadjah Mada (UGM), Yogyakarta, 55281, Indonesia
| |
Collapse
|
10
|
Kwon EJ, Hwang SH, Seo S, Park J, Park S, Kim S. Efficacy of Mesenchymal-Stromal-Cell-Derived Extracellular Vesicles in Ameliorating Cisplatin Nephrotoxicity, as Modeled Using Three-Dimensional, Gravity-Driven, Two-Layer Tubule-on-a-Chip (3D-MOTIVE Chip). Int J Mol Sci 2023; 24:15726. [PMID: 37958709 PMCID: PMC10647511 DOI: 10.3390/ijms242115726] [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: 09/18/2023] [Revised: 10/22/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Mesenchymal stromal cell (MSC)-derived extracellular vesicles (EVs) are known to have a therapeutic effect on nephrotoxicity. As animal models require significant time and resources to evaluate drug effects, there is a need for a new experimental technique that can accurately predict drug effects in humans. We evaluated the therapeutic effect of MSC-derived EVs in cisplatin nephrotoxicity using a three-dimensional, gravity-driven, two-layer tubule-on-a-chip (3D-MOTIVE chip). In the 3D-MOTIVE chip, 10 μM cisplatin decreased the number of attached cells compared to the vehicle. Conversely, annexin V and reactive oxygen species (ROS) were increased. Cell viability was increased 2.8-fold and 2.5-fold after treatment with EVs at 4 and 8 µg/mL, respectively, compared to the cisplatin-induced nephrotoxicity group. Cell attachment was increased 2.25-fold by treatment with 4 µg/mL EVs and 2.02-fold by 8 µg/mL EVs. Annexin V and ROS levels were decreased compared to those in the cisplatin-induced nephrotoxicity group. There were no significant differences in annexin V and ROS levels according to EV concentration. In sum, we created a cisplatin-induced nephrotoxicity model on a 3D-MOTIVE chip and found that MSC-derived EVs could restore cell viability. Thus, MSC-derived EVs may have the potential to ameliorate cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Eun-Jeong Kwon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea; (E.-J.K.); (S.-H.H.)
| | - Seong-Hye Hwang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea; (E.-J.K.); (S.-H.H.)
| | - Seungwan Seo
- Osong Medical Innovation Foundation, Cheongju-si 28161, Republic of Korea;
| | - Jaesung Park
- Department of School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea;
| | - Seokwoo Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea; (E.-J.K.); (S.-H.H.)
| | - Sejoong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea; (E.-J.K.); (S.-H.H.)
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
11
|
da Silva RGL, Blasimme A. Organ chip research in Europe: players, initiatives, and policies. Front Bioeng Biotechnol 2023; 11:1237561. [PMID: 37731764 PMCID: PMC10507620 DOI: 10.3389/fbioe.2023.1237561] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/17/2023] [Indexed: 09/22/2023] Open
Abstract
Background: Organ chips are microfabricated devices containing living engineered organ substructures in a controlled microenvironment. Research on organ chips has increased considerably over the past two decades. Aim: This paper offers an overview of the emerging knowledge ecosystem of organ chip research in Europe. Method: This study is based on queries and analyses undertaken through the bibliometric software Dimensions.ai. Results: Organ chip research has been rapidly growing in Europe in recent years, supported by robust academic science consortia, public-private initiatives, dedicated funding, and science policy instruments. Our data shows that previous investment in basic and fundamental research in centers of excellence in bioengineering science and technology are relevant to future investment in organ chips. Moreover, organ chip research in Europe is characterized by collaborative infrastructures to promote convergence of scientific, technical, and clinical capabilities. Conclusion: According to our study, the knowledge ecosystem of organ chip research in Europe has been growing sustainably. This growth is due to relevant institutional diversity, public-private initiatives, and ongoing research collaborations supported by robust funding schemes.
Collapse
|
12
|
Gao AY, Haak AJ, Bakri SJ. In vitro laboratory models of proliferative vitreoretinopathy. Surv Ophthalmol 2023; 68:861-874. [PMID: 37209723 DOI: 10.1016/j.survophthal.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 05/13/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
Proliferative vitreoretinopathy (PVR), the most common cause of recurrent retinal detachment, is characterized by the formation and contraction of fibrotic membranes on the surface of the retina. There are no Food and Drug Administration (FDA)-approved drugs to prevent or treat PVR. Therefore, it is necessary to develop accurate in vitro models of the disease that will enable researchers to screen drug candidates and prioritize the most promising candidates for clinical studies. We provide a summary of recent in vitro PVR models, as well as avenues for model improvement. Several in vitro PVR models were identified, including various types of cell cultures. Additionally, novel techniques that have not been used to model PVR were identified, including organoids, hydrogels, and organ-on-a-chip models. Novel ideas for improving in vitro PVR models are highlighted. Researchers may consult this review to help design in vitro models of PVR, which will aid in the development of therapies to treat the disease.
Collapse
Affiliation(s)
- Ashley Y Gao
- Mayo Clinic, Department of Ophthalmology, Rochester, Minnesota, USA
| | - Andrew J Haak
- Mayo Clinic, Department of Physiology and Biomedical Engineering, Rochester, Minnesota, USA
| | - Sophie J Bakri
- Mayo Clinic, Department of Ophthalmology, Rochester, Minnesota, USA.
| |
Collapse
|
13
|
Namazian Jam N, Gottlöber F, Hempel M, Dzekhtsiarova Y, Behrens S, Sonntag F, Sradnick J, Hugo C, Schmieder F. Microphysiological Conditions Do Not Affect MDR1-Mediated Transport of Rhodamine 123 above an Artificial Proximal Tubule. Biomedicines 2023; 11:2045. [PMID: 37509683 PMCID: PMC10376999 DOI: 10.3390/biomedicines11072045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/15/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Despite disadvantages, such as high cost and their poor predictive value, animal experiments are still the state of the art for pharmaceutical substance testing. One reason for this problem is the inability of standard cell culture methods to emulate the physiological environment necessary to recapitulate in vivo processes. Microphysiological systems offer the opportunity to close this gap. In this study, we utilize a previously employed microphysiological system to examine the impact of pressure and flow on the transportation of substances mediated by multidrug resistance protein 1 (MDR1) across an artificial cell-based tubular barrier. By using a miniaturized fluorescence measurement device, we could continuously track the MDR1-mediated transport of rhodamine 123 above the artificial barrier over 48 h. We proved that applying pressure and flow affects both active and passive transport of rhodamine 123. Using experimental results and curve fittings, the kinetics of MDR1-mediated transport as well as passive transport were investigated; thus, a kinetic model that explains this transport above an artificial tubular barrier was identified. This kinetic model demonstrates that the simple Michaelis-Menten model is not an appropriate model to explain the MDR1-mediated transport; instead, Hill kinetics, with Hill slope of n = 2, is a better fit. The kinetic values, Km, Vmax, and apparent permeability (Papp), obtained in this study are comparable with other in vivo and in vitro studies. Finally, the presented proximal tubule-on-a-chip can be used for pharmaceutical substance testing and to investigate pharmacokinetics of the renal transporter MDR1.
Collapse
Affiliation(s)
- Negin Namazian Jam
- Fraunhofer Institute for Material and Beam Technology IWS, 01277 Dresden, Germany
| | - Felix Gottlöber
- Fraunhofer Institute for Material and Beam Technology IWS, 01277 Dresden, Germany
| | - Melanie Hempel
- Fraunhofer Institute for Material and Beam Technology IWS, 01277 Dresden, Germany
| | - Yuliya Dzekhtsiarova
- Fraunhofer Institute for Material and Beam Technology IWS, 01277 Dresden, Germany
| | - Stephan Behrens
- Fraunhofer Institute for Material and Beam Technology IWS, 01277 Dresden, Germany
| | - Frank Sonntag
- Fraunhofer Institute for Material and Beam Technology IWS, 01277 Dresden, Germany
| | - Jan Sradnick
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, 01307 Dresden, Germany
| | - Christian Hugo
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, 01307 Dresden, Germany
| | - Florian Schmieder
- Fraunhofer Institute for Material and Beam Technology IWS, 01277 Dresden, Germany
| |
Collapse
|
14
|
Hou C, Gu Y, Yuan W, Zhang W, Xiu X, Lin J, Gao Y, Liu P, Chen X, Song L. Application of microfluidic chips in the simulation of the urinary system microenvironment. Mater Today Bio 2023; 19:100553. [PMID: 36747584 PMCID: PMC9898763 DOI: 10.1016/j.mtbio.2023.100553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/01/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023] Open
Abstract
The urinary system, comprising the kidneys, ureters, bladder, and urethra, has a unique mechanical and fluid microenvironment, which is essential to the urinary system growth and development. Microfluidic models, based on micromachining and tissue engineering technology, can integrate pathophysiological characteristics, maintain cell-cell and cell-extracellular matrix interactions, and accurately simulate the vital characteristics of human tissue microenvironments. Additionally, these models facilitate improved visualization and integration and meet the requirements of the laminar flow environment of the urinary system. However, several challenges continue to impede the development of a tissue microenvironment with controllable conditions closely resemble physiological conditions. In this review, we describe the biochemical and physical microenvironment of the urinary system and explore the feasibility of microfluidic technology in simulating the urinary microenvironment and pathophysiological characteristics in vitro. Moreover, we summarize the current research progress on adapting microfluidic chips for constructing the urinary microenvironment. Finally, we discuss the current challenges and suggest directions for future development and application of microfluidic technology in constructing the urinary microenvironment in vitro.
Collapse
Affiliation(s)
- Changhao Hou
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| | - Yubo Gu
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| | - Wei Yuan
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| | - Wukai Zhang
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xianjie Xiu
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| | - Jiahao Lin
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| | - Yue Gao
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Peichuan Liu
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiang Chen
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lujie Song
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| |
Collapse
|
15
|
Biosensor integrated tissue chips and their applications on Earth and in space. Biosens Bioelectron 2023; 222:114820. [PMID: 36527831 PMCID: PMC10143284 DOI: 10.1016/j.bios.2022.114820] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 09/24/2022] [Accepted: 10/13/2022] [Indexed: 12/27/2022]
Abstract
The development of space exploration technologies has positively impacted everyday life on Earth in terms of communication, environmental, social, and economic perspectives. The human body constantly fluctuates during spaceflight, even for a short-term mission. Unfortunately, technology is evolving faster than humans' ability to adapt, and many therapeutics entering clinical trials fail even after being subjected to vigorous in vivo testing due to toxicity and lack of efficacy. Therefore, tissue chips (also mentioned as organ-on-a-chip) with biosensors are being developed to compensate for the lack of relevant models to help improve the drug development process. There has been a push to monitor cell and tissue functions, based on their biological signals and utilize the integration of biosensors into tissue chips in space to monitor and assess cell microenvironment in real-time. With the collaboration between the Center for the Advancement of Science in Space (CASIS), the National Aeronautics and Space Administration (NASA) and other partners, they are providing the opportunities to study the effects of microgravity environment has on the human body. Institutions such as the National Institute of Health (NIH) and National Science Foundation (NSF) are partnering with CASIS and NASA to utilize tissue chips onboard the International Space Station (ISS). This article reviews the endless benefits of space technology, the development of integrated biosensors in tissue chips and their applications to better understand human biology, physiology, and diseases in space and on Earth, followed by future perspectives of tissue chip applications on Earth and in space.
Collapse
|
16
|
Su M, Ruan L, Dong X, Tian S, Lang W, Wu M, Chen Y, Lv Q, Lei L. Current state of knowledge on intelligent-response biological and other macromolecular hydrogels in biomedical engineering: A review. Int J Biol Macromol 2023; 227:472-492. [PMID: 36549612 DOI: 10.1016/j.ijbiomac.2022.12.148] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/07/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Because intelligent hydrogels have good biocompatibility, a rapid response, and good degradability as well as a stimulus response mode that is rich, hydrophilic, and similar to the softness and elasticity of living tissue, they have received widespread attention and are widely used in biomedical engineering. In this article, we conduct a systematic review of the use of smart hydrogels in biomedical engineering. First, we introduce the properties and applications of hydrogels and compare the similarities and differences between traditional hydrogels and smart hydrogels. Secondly, we summarize the intelligent hydrogel types, the mechanisms of action used by different hydrogels, and the materials for preparing different types of hydrogels, such as the materials for the preparation of temperature-responsive hydrogels, which mainly include gelatin, carrageenan, agarose, amylose, etc.; summarize the morphologies of different hydrogels, such as films, fibers and microspheres; and summarize the application of smart hydrogels in biomedical engineering, such as for the delivery of proteins, antibiotics, deoxyribonucleic acid, etc. Finally, we summarize the shortcomings of current research and present future prospects for smart hydrogels. The purpose of this paper is to provide researchers engaged in related fields with a systematic review of the application of intelligent hydrogels in biomedical engineering. We hope that they will get some inspiration from this work to provide new directions for the development of related fields.
Collapse
Affiliation(s)
- Mengrong Su
- College of Biology & Pharmacy, Yulin Normal University, Yulin 537000, China
| | - Lian Ruan
- College of Biology & Pharmacy, Yulin Normal University, Yulin 537000, China
| | - Xiaoyu Dong
- Institute of Medicine Nursing, Hubei University of Medicine, Shiyan 442000, China
| | - Shujing Tian
- College of Biology & Pharmacy, Yulin Normal University, Yulin 537000, China
| | - Wen Lang
- College of Biology & Pharmacy, Yulin Normal University, Yulin 537000, China
| | - Minhui Wu
- College of Biology & Pharmacy, Yulin Normal University, Yulin 537000, China
| | - Yujie Chen
- College of Biology & Pharmacy, Yulin Normal University, Yulin 537000, China
| | - Qizhuang Lv
- College of Biology & Pharmacy, Yulin Normal University, Yulin 537000, China; Guangxi Key Laboratory of Agricultural Resources Chemistry and Biotechnology, Yulin 537000, China.
| | - Lanjie Lei
- Jiangxi Provincial Key Lab of System Biomedicine, Jiujiang University, Jiujiang 332000, China.
| |
Collapse
|
17
|
Zommiti M, Connil N, Tahrioui A, Groboillot A, Barbey C, Konto-Ghiorghi Y, Lesouhaitier O, Chevalier S, Feuilloley MGJ. Organs-on-Chips Platforms Are Everywhere: A Zoom on Biomedical Investigation. Bioengineering (Basel) 2022; 9:646. [PMID: 36354557 PMCID: PMC9687856 DOI: 10.3390/bioengineering9110646] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/13/2022] [Accepted: 10/27/2022] [Indexed: 08/28/2023] Open
Abstract
Over the decades, conventional in vitro culture systems and animal models have been used to study physiology, nutrient or drug metabolisms including mechanical and physiopathological aspects. However, there is an urgent need for Integrated Testing Strategies (ITS) and more sophisticated platforms and devices to approach the real complexity of human physiology and provide reliable extrapolations for clinical investigations and personalized medicine. Organ-on-a-chip (OOC), also known as a microphysiological system, is a state-of-the-art microfluidic cell culture technology that sums up cells or tissue-to-tissue interfaces, fluid flows, mechanical cues, and organ-level physiology, and it has been developed to fill the gap between in vitro experimental models and human pathophysiology. The wide range of OOC platforms involves the miniaturization of cell culture systems and enables a variety of novel experimental techniques. These range from modeling the independent effects of biophysical forces on cells to screening novel drugs in multi-organ microphysiological systems, all within microscale devices. As in living biosystems, the development of vascular structure is the salient feature common to almost all organ-on-a-chip platforms. Herein, we provide a snapshot of this fast-evolving sophisticated technology. We will review cutting-edge developments and advances in the OOC realm, discussing current applications in the biomedical field with a detailed description of how this technology has enabled the reconstruction of complex multi-scale and multifunctional matrices and platforms (at the cellular and tissular levels) leading to an acute understanding of the physiopathological features of human ailments and infections in vitro.
Collapse
Affiliation(s)
- Mohamed Zommiti
- Research Unit Bacterial Communication and Anti-infectious Strategies (CBSA, UR4312), University of Rouen Normandie, 27000 Evreux, France
| | | | | | | | | | | | | | | | - Marc G. J. Feuilloley
- Research Unit Bacterial Communication and Anti-infectious Strategies (CBSA, UR4312), University of Rouen Normandie, 27000 Evreux, France
| |
Collapse
|
18
|
Hajiabbas M, D'Agostino C, Simińska-Stanny J, Tran SD, Shavandi A, Delporte C. Bioengineering in salivary gland regeneration. J Biomed Sci 2022; 29:35. [PMID: 35668440 PMCID: PMC9172163 DOI: 10.1186/s12929-022-00819-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/26/2022] [Indexed: 11/16/2022] Open
Abstract
Salivary gland (SG) dysfunction impairs the life quality of many patients, such as patients with radiation therapy for head and neck cancer and patients with Sjögren’s syndrome. Multiple SG engineering strategies have been considered for SG regeneration, repair, or whole organ replacement. An in-depth understanding of the development and differentiation of epithelial stem and progenitor cells niche during SG branching morphogenesis and signaling pathways involved in cell–cell communication constitute a prerequisite to the development of suitable bioengineering solutions. This review summarizes the essential bioengineering features to be considered to fabricate an engineered functional SG model using various cell types, biomaterials, active agents, and matrix fabrication methods. Furthermore, recent innovative and promising approaches to engineering SG models are described. Finally, this review discusses the different challenges and future perspectives in SG bioengineering.
Collapse
Affiliation(s)
- Maryam Hajiabbas
- Laboratory of Pathophysiological and Nutritional Biochemistry, Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Blg G/E CP 611, B-1070, Brussels, Belgium
| | - Claudia D'Agostino
- Laboratory of Pathophysiological and Nutritional Biochemistry, Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Blg G/E CP 611, B-1070, Brussels, Belgium
| | - Julia Simińska-Stanny
- Department of Process Engineering and Technology of Polymer and Carbon Materials, Faculty of Chemistry, Wroclaw University of Science and Technology, Norwida 4/6, 50-373, Wroclaw, Poland.,3BIO-BioMatter, École Polytechnique de Bruxelles, Université Libre de Bruxelles, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| | - Simon D Tran
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, H3A 0C7, Canada
| | - Amin Shavandi
- 3BIO-BioMatter, École Polytechnique de Bruxelles, Université Libre de Bruxelles, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| | - Christine Delporte
- Laboratory of Pathophysiological and Nutritional Biochemistry, Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Blg G/E CP 611, B-1070, Brussels, Belgium.
| |
Collapse
|
19
|
Koyilot MC, Natarajan P, Hunt CR, Sivarajkumar S, Roy R, Joglekar S, Pandita S, Tong CW, Marakkar S, Subramanian L, Yadav SS, Cherian AV, Pandita TK, Shameer K, Yadav KK. Breakthroughs and Applications of Organ-on-a-Chip Technology. Cells 2022; 11:cells11111828. [PMID: 35681523 PMCID: PMC9180073 DOI: 10.3390/cells11111828] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/28/2022] [Accepted: 05/30/2022] [Indexed: 12/10/2022] Open
Abstract
Organ-on-a-chip (OOAC) is an emerging technology based on microfluid platforms and in vitro cell culture that has a promising future in the healthcare industry. The numerous advantages of OOAC over conventional systems make it highly popular. The chip is an innovative combination of novel technologies, including lab-on-a-chip, microfluidics, biomaterials, and tissue engineering. This paper begins by analyzing the need for the development of OOAC followed by a brief introduction to the technology. Later sections discuss and review the various types of OOACs and the fabrication materials used. The implementation of artificial intelligence in the system makes it more advanced, thereby helping to provide a more accurate diagnosis as well as convenient data management. We introduce selected OOAC projects, including applications to organ/disease modelling, pharmacology, personalized medicine, and dentistry. Finally, we point out certain challenges that need to be surmounted in order to further develop and upgrade the current systems.
Collapse
Affiliation(s)
- Mufeeda C. Koyilot
- Molecular Robotics, Cochin 682033, India; (M.C.K.); (P.N.); (S.S.); (R.R.); (S.J.); (S.M.); (A.V.C.)
| | - Priyadarshini Natarajan
- Molecular Robotics, Cochin 682033, India; (M.C.K.); (P.N.); (S.S.); (R.R.); (S.J.); (S.M.); (A.V.C.)
| | - Clayton R. Hunt
- Houston Methodist Research Institute, Houston, TX 77030, USA;
| | - Sonish Sivarajkumar
- Molecular Robotics, Cochin 682033, India; (M.C.K.); (P.N.); (S.S.); (R.R.); (S.J.); (S.M.); (A.V.C.)
| | - Romy Roy
- Molecular Robotics, Cochin 682033, India; (M.C.K.); (P.N.); (S.S.); (R.R.); (S.J.); (S.M.); (A.V.C.)
| | - Shreeram Joglekar
- Molecular Robotics, Cochin 682033, India; (M.C.K.); (P.N.); (S.S.); (R.R.); (S.J.); (S.M.); (A.V.C.)
| | - Shruti Pandita
- Mays Cancer Center, University of Texas Health Sciences Center at San Antonio, San Antonio, TX 78229, USA;
| | - Carl W. Tong
- School of Engineering Medicine, Texas A&M University, Houston, TX 77030, USA;
| | - Shamsudheen Marakkar
- Molecular Robotics, Cochin 682033, India; (M.C.K.); (P.N.); (S.S.); (R.R.); (S.J.); (S.M.); (A.V.C.)
| | | | - Shalini S. Yadav
- Department of Immunology, UT MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Anoop V. Cherian
- Molecular Robotics, Cochin 682033, India; (M.C.K.); (P.N.); (S.S.); (R.R.); (S.J.); (S.M.); (A.V.C.)
| | - Tej K. Pandita
- Houston Methodist Research Institute, Houston, TX 77030, USA;
- Center for Genomic and Precision Medicine, Institute of Biosciences and Technology, Department of Translational Medical Sciences, Texas A&M University, Houston, TX 77030, USA
- Correspondence: (T.K.P.); (K.S.); (K.K.Y.)
| | - Khader Shameer
- School of Public Health, Faculty of Medicine, Imperial College London, South Kensington, London SW7 2AZ, UK
- Correspondence: (T.K.P.); (K.S.); (K.K.Y.)
| | - Kamlesh K. Yadav
- School of Engineering Medicine, Texas A&M University, Houston, TX 77030, USA;
- Center for Genomic and Precision Medicine, Institute of Biosciences and Technology, Department of Translational Medical Sciences, Texas A&M University, Houston, TX 77030, USA
- Correspondence: (T.K.P.); (K.S.); (K.K.Y.)
| |
Collapse
|
20
|
Galateanu B, Hudita A, Biru EI, Iovu H, Zaharia C, Simsensohn E, Costache M, Petca RC, Jinga V. Applications of Polymers for Organ-on-Chip Technology in Urology. Polymers (Basel) 2022; 14:1668. [PMID: 35566836 PMCID: PMC9105302 DOI: 10.3390/polym14091668] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/05/2022] [Accepted: 04/18/2022] [Indexed: 02/07/2023] Open
Abstract
Organ-on-chips (OOCs) are microfluidic devices used for creating physiological organ biomimetic systems. OOC technology brings numerous advantages in the current landscape of preclinical models, capable of recapitulating the multicellular assemblage, tissue-tissue interaction, and replicating numerous human pathologies. Moreover, in cancer research, OOCs emulate the 3D hierarchical complexity of in vivo tumors and mimic the tumor microenvironment, being a practical cost-efficient solution for tumor-growth investigation and anticancer drug screening. OOCs are compact and easy-to-use microphysiological functional units that recapitulate the native function and the mechanical strain that the cells experience in the human bodies, allowing the development of a wide range of applications such as disease modeling or even the development of diagnostic devices. In this context, the current work aims to review the scientific literature in the field of microfluidic devices designed for urology applications in terms of OOC fabrication (principles of manufacture and materials used), development of kidney-on-chip models for drug-toxicity screening and kidney tumors modeling, bladder-on-chip models for urinary tract infections and bladder cancer modeling and prostate-on-chip models for prostate cancer modeling.
Collapse
Affiliation(s)
- Bianca Galateanu
- Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei Street, 050095 Bucharest, Romania; (B.G.); (M.C.)
| | - Ariana Hudita
- Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei Street, 050095 Bucharest, Romania; (B.G.); (M.C.)
| | - Elena Iuliana Biru
- Advanced Polymer Materials Group, Department of Bioresources and Polymer Science, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (H.I.); (C.Z.)
| | - Horia Iovu
- Advanced Polymer Materials Group, Department of Bioresources and Polymer Science, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (H.I.); (C.Z.)
- Academy of Romanian Scientists, Ilfov Street, 50044 Bucharest, Romania
| | - Catalin Zaharia
- Advanced Polymer Materials Group, Department of Bioresources and Polymer Science, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (H.I.); (C.Z.)
| | - Eliza Simsensohn
- “Carol Davila” University of Medicine and Pharmacy Bucharest, 050474 Bucharest, Romania; (E.S.); (R.-C.P.); (V.J.)
| | - Marieta Costache
- Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei Street, 050095 Bucharest, Romania; (B.G.); (M.C.)
| | - Razvan-Cosmin Petca
- “Carol Davila” University of Medicine and Pharmacy Bucharest, 050474 Bucharest, Romania; (E.S.); (R.-C.P.); (V.J.)
| | - Viorel Jinga
- “Carol Davila” University of Medicine and Pharmacy Bucharest, 050474 Bucharest, Romania; (E.S.); (R.-C.P.); (V.J.)
| |
Collapse
|
21
|
Adaptation of the Kirkstall QV600 LLI Microfluidics System for the Study of Gastrointestinal Absorption by Mass Spectrometry Imaging and LC-MS/MS. Pharmaceutics 2022; 14:pharmaceutics14020364. [PMID: 35214096 PMCID: PMC8878338 DOI: 10.3390/pharmaceutics14020364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/26/2022] [Accepted: 02/02/2022] [Indexed: 02/04/2023] Open
Abstract
Absorption studies on oral drugs can be difficult due to the challenge of replicating the complex structure and environment of the GI tract. Drug absorption studies can be conducted using in vivo and ex vivo animal tissue or animal-free techniques. These studies typically involve the use of Caco-2 cells. However, Caco-2 cells do not incorporate all the cell types found in intestinal tissue and lack P450 metabolizing enzymes. The QV600 LLI system is a microfluidics system designed for use with cell culture. Here, it has been adapted to house appropriate sections of ex vivo porcine tissue to act as a system that models the duodenum section of the small intestine. A pH regulated solution of Atorvastatin was flowed over the apical layer of the GI tissue and a nutrient solution flowed over the basal layer of the tissue to maintain tissue viability. The tissue samples were snap-frozen, cryosectioned, and imaged using MALDI Mass Spectrometry Imaging (MSI). A proof-of-concept study on the effect of excipients on absorption was conducted. Different concentrations of the solubilizing agent were added to the donor circuit of the QV600 LLI. The amount of Atorvastatin in the acceptor circuit was determined to study the effect of the excipient on the amount of drug that had permeated through the tissue. Using these data, Papp, pig values were calculated and compared with the literature.
Collapse
|
22
|
From organ-on-chip to body-on-chip: The next generation of microfluidics platforms for in vitro drug efficacy and toxicity testing. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 187:41-91. [PMID: 35094781 DOI: 10.1016/bs.pmbts.2021.07.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The high failure rate in drug development is often attributed to the lack of accurate pre-clinical models that may lead to false discoveries and inconclusive data when the compounds are eventually tested in clinical phase. With the evolution of cell culture technologies, drug testing systems have widely improved, and today, with the emergence of microfluidics devices, drug screening seems to be at the dawn of an important revolution. An organ-on-chip allows the culture of living cells in continuously perfused microchambers to reproduce physiological functions of a particular tissue or organ. The advantages of such systems are not only their ability to recapitulate the complex biochemical interactions between different human cell types but also to incorporate physical forces, including shear stress and mechanical stretching or compression. To improve this model, and to reproduce the absorption, distribution, metabolism, and elimination process of an exogenous compound, organ-on-chips can even be linked fluidically to mimic physiological interactions between different organs, leading to the development of body-on-chips. Although these technologies are still at a young age and need to address a certain number of limitations, they already demonstrated their relevance to study the effect of drugs or toxins on organs, displaying a similar response to what is observed in vivo. The purpose of this review is to present the evolution from organ-on-chip to body-on-chip, examine their current use for drug testing and discuss their advantages and future challenges they will face in order to become an essential pillar of pharmaceutical research.
Collapse
|
23
|
Zhao P, Wang J, Chen C, Wang J, Liu G, Nandakumar K, Li Y, Wang L. Microfluidic Applications in Drug Development: Fabrication of Drug Carriers and Drug Toxicity Screening. MICROMACHINES 2022; 13:200. [PMID: 35208324 PMCID: PMC8877367 DOI: 10.3390/mi13020200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/23/2022] [Accepted: 01/23/2022] [Indexed: 01/09/2023]
Abstract
Microfluidic technology has been highly useful in nanovolume sample preparation, separation, synthesis, purification, detection and assay, which are advantageous in drug development. This review highlights the recent developments and trends in microfluidic applications in two areas of drug development. First, we focus on how microfluidics has been developed as a facile tool for the fabrication of drug carriers including microparticles and nanoparticles. Second, we discuss how microfluidic chips could be used as an independent platform or integrated with other technologies in drug toxicity screening. Challenges and future perspectives of microfluidic applications in drug development have also been provided considering the present technological limitations.
Collapse
Affiliation(s)
- Pei Zhao
- Energy Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; (P.Z.); (J.W.); (C.C.); (J.W.); (G.L.); (K.N.)
- School of Energy and Power Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Jianchun Wang
- Energy Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; (P.Z.); (J.W.); (C.C.); (J.W.); (G.L.); (K.N.)
- School of Energy and Power Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Chengmin Chen
- Energy Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; (P.Z.); (J.W.); (C.C.); (J.W.); (G.L.); (K.N.)
- School of Energy and Power Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Jianmei Wang
- Energy Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; (P.Z.); (J.W.); (C.C.); (J.W.); (G.L.); (K.N.)
- School of Energy and Power Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Guangxia Liu
- Energy Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; (P.Z.); (J.W.); (C.C.); (J.W.); (G.L.); (K.N.)
- School of Energy and Power Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Krishnaswamy Nandakumar
- Energy Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; (P.Z.); (J.W.); (C.C.); (J.W.); (G.L.); (K.N.)
- School of Energy and Power Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Cain Department of Chemical Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Yan Li
- Energy Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; (P.Z.); (J.W.); (C.C.); (J.W.); (G.L.); (K.N.)
- School of Energy and Power Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Liqiu Wang
- Department of Mechanical Engineering, Faculty of Engineering, The University of Hong Kong, Hong Kong 999077, China
| |
Collapse
|
24
|
Tajeddin A, Mustafaoglu N. Design and Fabrication of Organ-on-Chips: Promises and Challenges. MICROMACHINES 2021; 12:1443. [PMID: 34945293 PMCID: PMC8707724 DOI: 10.3390/mi12121443] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/14/2021] [Accepted: 11/21/2021] [Indexed: 02/07/2023]
Abstract
The advent of the miniaturization approach has influenced the research trends in almost all disciplines. Bioengineering is one of the fields benefiting from the new possibilities of microfabrication techniques, especially in cell and tissue culture, disease modeling, and drug discovery. The limitations of existing 2D cell culture techniques, the high time and cost requirements, and the considerable failure rates have led to the idea of 3D cell culture environments capable of providing physiologically relevant tissue functions in vitro. Organ-on-chips are microfluidic devices used in this context as a potential alternative to in vivo animal testing to reduce the cost and time required for drug evaluation. This emerging technology contributes significantly to the development of various research areas, including, but not limited to, tissue engineering and drug discovery. However, it also brings many challenges. Further development of the technology requires interdisciplinary studies as some problems are associated with the materials and their manufacturing techniques. Therefore, in this paper, organ-on-chip technologies are presented, focusing on the design and fabrication requirements. Then, state-of-the-art materials and microfabrication techniques are described in detail to show their advantages and also their limitations. A comparison and identification of gaps for current use and further studies are therefore the subject of the final discussion.
Collapse
Affiliation(s)
- Alireza Tajeddin
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla 34596, Istanbul, Turkey;
| | - Nur Mustafaoglu
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla 34596, Istanbul, Turkey;
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Tuzla 34596, Istanbul, Turkey
| |
Collapse
|
25
|
Pattanayak P, Singh SK, Gulati M, Vishwas S, Kapoor B, Chellappan DK, Anand K, Gupta G, Jha NK, Gupta PK, Prasher P, Dua K, Dureja H, Kumar D, Kumar V. Microfluidic chips: recent advances, critical strategies in design, applications and future perspectives. MICROFLUIDICS AND NANOFLUIDICS 2021; 25:99. [PMID: 34720789 PMCID: PMC8547131 DOI: 10.1007/s10404-021-02502-2] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/19/2021] [Indexed: 05/12/2023]
Abstract
Microfluidic chip technology is an emerging tool in the field of biomedical application. Microfluidic chip includes a set of groves or microchannels that are engraved on different materials (glass, silicon, or polymers such as polydimethylsiloxane or PDMS, polymethylmethacrylate or PMMA). The microchannels forming the microfluidic chip are interconnected with each other for desired results. This organization of microchannels trapped into the microfluidic chip is associated with the outside by inputs and outputs penetrating through the chip, as an interface between the macro- and miniature world. With the help of a pump and a chip, microfluidic chip helps to determine the behavioral change of the microfluids. Inside the chip, there are microfluidic channels that permit the processing of the fluid, for example, blending and physicochemical responses. Microfluidic chip has numerous points of interest including lesser time and reagent utilization and alongside this, it can execute numerous activities simultaneously. The miniatured size of the chip fastens the reaction as the surface area increases. It is utilized in different biomedical applications such as food safety sensing, peptide analysis, tissue engineering, medical diagnosis, DNA purification, PCR activity, pregnancy, and glucose estimation. In the present study, the design of various microfluidic chips has been discussed along with their biomedical applications.
Collapse
Affiliation(s)
- Prapti Pattanayak
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411 India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411 India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411 India
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411 India
| | - Bhupinder Kapoor
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411 India
| | - Dinesh Kumar Chellappan
- School of Pharmacy, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Krishnan Anand
- Department of Chemical Pathology, School of Pathology, Faculty of Health Sciences and National Health Laboratory Service, University of the Free State, Bloemfontein, South Africa
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, Uttar Pradesh 201310 India
| | - Piyush Kumar Gupta
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Plot no. 32-34, Knowledge Park III, Greater Noida, Uttar Pradesh 201310 India
| | - Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Energy Acres, Dehradun, 248007 India
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007 Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, Australia
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana 12401 India
| | - Deepak Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, 173229 India
| | - Vijay Kumar
- School of Bioengineering and Bioscience, Lovely Professional University, Phagwara, Punjab 144411 India
| |
Collapse
|
26
|
Hwang SH, Lee YM, Choi Y, Son HE, Ryu JY, Na KY, Chin HJ, Jeon NL, Kim S. Role of Human Primary Renal Fibroblast in TGF-β1-Mediated Fibrosis-Mimicking Devices. Int J Mol Sci 2021; 22:ijms221910758. [PMID: 34639099 PMCID: PMC8509581 DOI: 10.3390/ijms221910758] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 01/21/2023] Open
Abstract
Renal fibrosis is a progressive chronic kidney disease that ultimately leads to end-stage renal failure. Despite several approaches to combat renal fibrosis, an experimental model to evaluate currently available drugs is not ideal. We developed fibrosis-mimicking models using three-dimensional (3D) co-culture devices designed with three separate layers of tubule interstitium, namely, epithelial, fibroblastic, and endothelial layers. We introduced human renal proximal tubular epithelial cells (HK-2), human umbilical-vein endothelial cells, and patient-derived renal fibroblasts, and evaluated the effects of transforming growth factor-β (TGF-β) and TGF-β inhibitor treatment on this renal fibrosis model. The expression of the fibrosis marker alpha smooth muscle actin upon TGF-β1 treatment was augmented in monolayer-cultured HK-2 cells in a 3D disease model. In the vascular compartment of renal fibrosis models, the density of vessels was increased and decreased in the TGF-β-treated group and TGF-β-inhibitor treatment group, respectively. Multiplex ELISA using supernatants in the TGF-β-stimulating 3D models showed that pro-inflammatory cytokine and growth factor levels including interleukin-1 beta, tumor necrosis factor alpha, basic fibroblast growth factor, and TGF-β1, TGF-β2, and TGF-β3 were increased, which mimicked the fibrotic microenvironments of human kidneys. This study may enable the construction of a human renal fibrosis-mimicking device model beyond traditional culture experiments.
Collapse
Affiliation(s)
- Seong-Hye Hwang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si 13620, Korea; (S.-H.H.); (Y.-M.L.); (Y.C.); (H.E.S.); (J.Y.R.); (K.Y.N.); (H.J.C.)
| | - Yun-Mi Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si 13620, Korea; (S.-H.H.); (Y.-M.L.); (Y.C.); (H.E.S.); (J.Y.R.); (K.Y.N.); (H.J.C.)
| | - Yunyeong Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si 13620, Korea; (S.-H.H.); (Y.-M.L.); (Y.C.); (H.E.S.); (J.Y.R.); (K.Y.N.); (H.J.C.)
| | - Hyung Eun Son
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si 13620, Korea; (S.-H.H.); (Y.-M.L.); (Y.C.); (H.E.S.); (J.Y.R.); (K.Y.N.); (H.J.C.)
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Ji Young Ryu
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si 13620, Korea; (S.-H.H.); (Y.-M.L.); (Y.C.); (H.E.S.); (J.Y.R.); (K.Y.N.); (H.J.C.)
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Ki Young Na
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si 13620, Korea; (S.-H.H.); (Y.-M.L.); (Y.C.); (H.E.S.); (J.Y.R.); (K.Y.N.); (H.J.C.)
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Ho Jun Chin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si 13620, Korea; (S.-H.H.); (Y.-M.L.); (Y.C.); (H.E.S.); (J.Y.R.); (K.Y.N.); (H.J.C.)
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Noo Li Jeon
- Program for Bioengineering, School of Engineering, Seoul National University, Seoul 08826, Korea
- Correspondence: (N.L.J.); (S.K.); Tel.: +82-31-787-7051 (S.K.)
| | - Sejoong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si 13620, Korea; (S.-H.H.); (Y.-M.L.); (Y.C.); (H.E.S.); (J.Y.R.); (K.Y.N.); (H.J.C.)
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
- Correspondence: (N.L.J.); (S.K.); Tel.: +82-31-787-7051 (S.K.)
| |
Collapse
|
27
|
Bellucci L, Montini G, Collino F, Bussolati B. Mesenchymal Stromal Cell-Derived Extracellular Vesicles Pass through the Filtration Barrier and Protect Podocytes in a 3D Glomerular Model under Continuous Perfusion. Tissue Eng Regen Med 2021; 18:549-560. [PMID: 34313970 PMCID: PMC8325748 DOI: 10.1007/s13770-021-00374-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 06/30/2021] [Accepted: 07/07/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Dynamic cultures, characterized by continuous fluid reperfusion, elicit physiological responses from cultured cells. Mesenchymal stem cell-derived EVs (MSC-EVs) has been proposed as a novel approach in treating several renal diseases, including acute glomerular damage, by using traditional two-dimensional cell cultures and in vivo models. We here aimed to use a fluidic three-dimensional (3D) glomerular model to study the EV dynamics within the glomerular structure under perfusion. METHODS To this end, we set up a 3D glomerular model culturing human glomerular endothelial cells and podocytes inside a bioreactor on the opposite sides of a porous membrane coated with type IV collagen. The bioreactor was connected to a circuit that allowed fluid passage at the rate of 80 µl/min. To mimic glomerular damage, the system was subjected to doxorubicin administration in the presence of therapeutic MSC-EVs. RESULTS The integrity of the glomerular basal membrane in the 3D glomerulus was assessed by a permeability assay, demonstrating that the co-culture could limit the passage of albumin through the filtration barrier. In dynamic conditions, serum EVs engineered with cel-miR-39 passed through the glomerular barrier and transferred the exogenous microRNA to podocyte cell lines. Doxorubicin treatment increased podocyte apoptosis, whereas MSC-EV within the endothelial circuit protected podocytes from damage, decreasing cell death and albumin permeability. CONCLUSION Using an innovative millifluidic model, able to mimic the human glomerular barrier, we were able to trace the EV passage and therapeutic effect in dynamic conditions.
Collapse
Affiliation(s)
- Linda Bellucci
- Department of Molecular Biotechnology and Health Sciences, University of Torino, via Nizza 52, 10126, Turin, Italy
- Laboratory of Translational Research in Paediatric Nephro-Urology, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Giovanni Montini
- Laboratory of Translational Research in Paediatric Nephro-Urology, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milano, Milan, Italy
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione Ca' Granda IRCCS, Policlinico Di Milano, Milan, Italy
| | - Federica Collino
- Laboratory of Translational Research in Paediatric Nephro-Urology, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milano, Milan, Italy
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Torino, via Nizza 52, 10126, Turin, Italy.
| |
Collapse
|
28
|
Del Favero G, Zeugswetter M, Kiss E, Marko D. Endoplasmic Reticulum Adaptation and Autophagic Competence Shape Response to Fluid Shear Stress in T24 Bladder Cancer Cells. Front Pharmacol 2021; 12:647350. [PMID: 34012396 PMCID: PMC8126838 DOI: 10.3389/fphar.2021.647350] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/17/2021] [Indexed: 12/26/2022] Open
Abstract
Accumulation of xenobiotics and waste metabolites in the urinary bladder is constantly accompanied by shear stress originating from the movement of the luminal fluids. Hence, both chemical and physical cues constantly modulate the cellular response in health and disease. In line, bladder cells have to maintain elevated mechanosensory competence together with chemical stress response adaptation potential. However, much of the molecular mechanisms sustaining this plasticity is currently unknown. Taking this as a starting point, we investigated the response of T24 urinary bladder cancer cells to shear stress comparing morphology to functional performance. T24 cells responded to the shear stress protocol (flow speed of 0.03 ml/min, 3 h) by significantly increasing their surface area. When exposed to deoxynivalenol-3-sulfate (DON-3-Sulf), bladder cells increased this response in a concentration-dependent manner (0.1-1 µM). DON-3-Sulf is a urinary metabolite of a very common food contaminant mycotoxin (deoxynivalenol, DON) and was already described to enhance proliferation of cancer cells. Incubation with DON-3-Sulf also caused the enlargement of the endoplasmic reticulum (ER), decreased the lysosomal movement, and increased the formation of actin stress fibers. Similar remodeling of the endoplasmic reticulum and area spread after shear stress were observed upon incubation with the autophagy activator rapamycin (1-100 nM). Performance of experiments in the presence of chloroquine (chloroquine, 30 μM) further contributed to shed light on the mechanistic link between adaptation to the biomechanical stimulation and ER stress response. At the molecular level, we observed that ER reshaping was linked to actin organization, with the two components mutually regulating each other. Indeed, we identified in the ER stress-cytoskeletal rearrangement an important axis defining the physical/chemical response potential of bladder cells and created a workflow for further investigation of urinary metabolites, food constituents, and contaminants, as well as for pharmacological profiling.
Collapse
Affiliation(s)
- Giorgia Del Favero
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria.,Core Facility Multimodal Imaging, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Michael Zeugswetter
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Endre Kiss
- Core Facility Multimodal Imaging, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Doris Marko
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria
| |
Collapse
|
29
|
House A, Atalla I, Lee EJ, Guvendiren M. Designing Biomaterial Platforms for Cardiac Tissue and Disease Modeling. ADVANCED NANOBIOMED RESEARCH 2021; 1:2000022. [PMID: 33709087 PMCID: PMC7942203 DOI: 10.1002/anbr.202000022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Heart disease is one of the leading causes of death in the world. There is a growing demand for in vitro cardiac models that can recapitulate the complex physiology of the cardiac tissue. These cardiac models can provide a platform to better understand the underlying mechanisms of cardiac development and disease and aid in developing novel treatment alternatives and platforms towards personalized medicine. In this review, a summary of engineered cardiac platforms is presented. Basic design considerations for replicating the heart's microenvironment are discussed considering the anatomy of the heart. This is followed by a detailed summary of the currently available biomaterial platforms for modeling the heart tissue in vitro. These in vitro models include 2D surface modified structures, 3D molded structures, porous scaffolds, electrospun scaffolds, bioprinted structures, and heart-on-a-chip devices. The challenges faced by current models and the future directions of in vitro cardiac models are also discussed. Engineered in vitro tissue models utilizing patients' own cells could potentially revolutionize the way we develop treatment and diagnostic alternatives.
Collapse
Affiliation(s)
- Andrew House
- Instructive Biomaterials and Additive Manufacturing Laboratory, Otto H. York Chemical and Materials Engineering, 138 York Center, University Heights, Newark, NJ 07102, USA
| | - Iren Atalla
- Instructive Biomaterials and Additive Manufacturing Laboratory, Otto H. York Chemical and Materials Engineering, 138 York Center, University Heights, Newark, NJ 07102, USA
| | - Eun Jung Lee
- Instructive Biomaterials and Additive Manufacturing Laboratory, Otto H. York Chemical and Materials Engineering, 138 York Center, University Heights, Newark, NJ 07102, USA
| | - Murat Guvendiren
- Instructive Biomaterials and Additive Manufacturing Laboratory, Otto H. York Chemical and Materials Engineering, 138 York Center, University Heights, Newark, NJ 07102, USA
| |
Collapse
|
30
|
Tissue Engineering of Oral Mucosa and Salivary Gland: Disease Modeling and Clinical Applications. MICROMACHINES 2020; 11:mi11121066. [PMID: 33266093 PMCID: PMC7761376 DOI: 10.3390/mi11121066] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/25/2020] [Accepted: 11/29/2020] [Indexed: 12/23/2022]
Abstract
Oral mucosa and salivary gland are composed of complex and dynamic networks of extracellular matrix, multiple cell types, vasculature, and various biochemical agents. Two-dimensional (2D) cell culture is commonly used in testing new drugs and experimental therapies. However, 2D cell culture cannot fully replicate the architecture, physiological, and pathological microenvironment of living human oral mucosa and salivary glands. Recent microengineering techniques offer state of the science cell culture models that can recapitulate human organ structures and functions. This narrative review describes emerging in vitro models of oral and salivary gland tissue such as 3D cell culture models, spheroid and organoid models, tissue-on-a-chip, and functional decellularized scaffolds. Clinical applications of these models are also discussed in this review.
Collapse
|
31
|
Mosavati B, Oleinikov AV, Du E. Development of an Organ-on-a-Chip-Device for Study of Placental Pathologies. Int J Mol Sci 2020; 21:E8755. [PMID: 33228194 PMCID: PMC7699553 DOI: 10.3390/ijms21228755] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/01/2020] [Accepted: 11/17/2020] [Indexed: 02/07/2023] Open
Abstract
The human placenta plays a key role in reproduction and serves as a major interface for maternofetal exchange of nutrients. Study of human placenta pathology presents a great experimental challenge because it is not easily accessible. In this paper, a 3D placenta-on-a-chip model is developed by bioengineering techniques to simulate the placental interface between maternal and fetal blood in vitro. In this model, trophoblasts cells and human umbilical vein endothelial cells are cultured on the opposite sides of a porous polycarbonate membrane, which is sandwiched between two microfluidic channels. Glucose diffusion across this barrier is analyzed under shear flow conditions. Meanwhile, a numerical model of the 3D placenta-on-a-chip model is developed. Numerical results of concentration distributions and the convection-diffusion mass transport is compared to the results obtained from the experiments for validation. Finally, effects of flow rate and membrane porosity on glucose diffusion across the placental barrier are studied using the validated numerical model. The placental model developed here provides a potentially helpful tool to study a variety of other processes at the maternal-fetal interface, for example, effects of drugs or infections like malaria on transport of various substances across the placental barrier.
Collapse
Affiliation(s)
- Babak Mosavati
- Department of Ocean and Mechanical Engineering, College of Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL 33431, USA;
| | - Andrew V. Oleinikov
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA;
| | - E. Du
- Department of Ocean and Mechanical Engineering, College of Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL 33431, USA;
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
| |
Collapse
|
32
|
Chen X, Zhang YS, Zhang X, Liu C. Organ-on-a-chip platforms for accelerating the evaluation of nanomedicine. Bioact Mater 2020; 6:1012-1027. [PMID: 33102943 PMCID: PMC7566214 DOI: 10.1016/j.bioactmat.2020.09.022] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/01/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Nanomedicine involves the use of engineered nanoscale materials in an extensive range of diagnostic and therapeutic applications and can be applied to the treatment of many diseases. Despite the rapid progress and tremendous potential of nanomedicine in the past decades, the clinical translational process is still quite slow, owing to the difficulty in understanding, evaluating, and predicting nanomaterial behaviors within the complex environment of human beings. Microfluidics-based organ-on-a-chip (Organ Chip) techniques offer a promising way to resolve these challenges. Sophisticatedly designed Organ Chip enable in vitro simulation of the in vivo microenvironments, thus providing robust platforms for evaluating nanomedicine. Herein, we review recent developments and achievements in Organ Chip models for nanomedicine evaluations, categorized into seven broad sections based on the target organ systems: respiratory, digestive, lymphatic, excretory, nervous, and vascular, as well as coverage on applications relating to cancer. We conclude by providing our perspectives on the challenges and potential future directions for applications of Organ Chip in nanomedicine. Microfluidics-based organ-on-a-chip (Organ Chip) techniques offer a promising way to understand, evaluate, and predict nanomedicine behaviors within the complex environment. Organ Chip models for nanomedicine evaluations are categorized into seven broad sections based on the targeted body systems. Limitations, challenges, and perspectives of Organ Chip for accelerating the assessment of nanomedicine are discussed, respectively.
Collapse
Affiliation(s)
- Xi Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, United States
| | - Xinping Zhang
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| |
Collapse
|
33
|
Abstract
AbstractMicrofluidic channels moulded from the soft polymer poly(dimethylsiloxane) (PDMS) are widely used as a platform for mimicking biological environments, and can be used for the simulation of fluid filled structures such as blood and lung vessels. The control of pressure and flow rate within these structures is vital to mimic physiological conditions. The flexibility of PDMS leads to pressure-induced deformation under flow, leading to variable flow profiles along a device. Here, we investigate the change in Young’s modulus of microfluidic channels due to infiltration of mineral oil, a PDMS permeable fluid, and how this affects the resulting pressure profile using a novel pressure measurement method. We found a 53% decrease in Young’s modulus of PDMS due to mineral oil absorption over the course of 3 h accounted for lower internal pressure and larger channel deformation compared to fresh PDMS at a given flow rate. Confocal fluorescence microscopy used to image channel profiles before and after the introduction of mineral oil showed a change in pressure-induced deformation after infiltration of the oil. Atomic force microscopy (AFM) nanoindentation was used to measure Young’s modulus of PDMS before ($$2.80 \pm 0.03$$
2.80
±
0.03
MPa) and after ($$1.32 \pm 0.04$$
1.32
±
0.04
MPa) mineral oil absorption. Raman spectroscopy showed the infiltration of mineral oil into PDMS from channel walls and revealed the diffusion coefficient of mineral oil in PDMS.
Collapse
|
34
|
Shirazi J, Donzanti MJ, Nelson KM, Zurakowski R, Fromen CA, Gleghorn JP. Significant Unresolved Questions and Opportunities for Bioengineering in Understanding and Treating COVID-19 Disease Progression. Cell Mol Bioeng 2020; 13:259-284. [PMID: 32837585 PMCID: PMC7384395 DOI: 10.1007/s12195-020-00637-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/14/2020] [Indexed: 12/19/2022] Open
Abstract
COVID-19 is a disease that manifests itself in a multitude of ways across a wide range of tissues. Many factors are involved, and though impressive strides have been made in studying this novel disease in a very short time, there is still a great deal that is unknown about how the virus functions. Clinical data has been crucial for providing information on COVID-19 progression and determining risk factors. However, the mechanisms leading to the multi-tissue pathology are yet to be fully established. Although insights from SARS-CoV-1 and MERS-CoV have been valuable, it is clear that SARS-CoV-2 is different and merits its own extensive studies. In this review, we highlight unresolved questions surrounding this virus including the temporal immune dynamics, infection of non-pulmonary tissue, early life exposure, and the role of circadian rhythms. Risk factors such as sex and exposure to pollutants are also explored followed by a discussion of ways in which bioengineering approaches can be employed to help understand COVID-19. The use of sophisticated in vitro models can be employed to interrogate intercellular interactions and also to tease apart effects of the virus itself from the resulting immune response. Additionally, spatiotemporal information can be gleaned from these models to learn more about the dynamics of the virus and COVID-19 progression. Application of advanced tissue and organ system models into COVID-19 research can result in more nuanced insight into the mechanisms underlying this condition and elucidate strategies to combat its effects.
Collapse
Affiliation(s)
- Jasmine Shirazi
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE 19716 USA
| | - Michael J. Donzanti
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE 19716 USA
| | - Katherine M. Nelson
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716 USA
| | - Ryan Zurakowski
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE 19716 USA
| | - Catherine A. Fromen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716 USA
| | - Jason P. Gleghorn
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE 19716 USA
| |
Collapse
|
35
|
Guenat OT, Geiser T, Berthiaume F. Clinically Relevant Tissue Scale Responses as New Readouts from Organs-on-a-Chip for Precision Medicine. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2020; 13:111-133. [PMID: 31961712 DOI: 10.1146/annurev-anchem-061318-114919] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Organs-on-chips (OOC) are widely seen as being the next generation in vitro models able to accurately recreate the biochemical-physical cues of the cellular microenvironment found in vivo. In addition, they make it possible to examine tissue-scale functional properties of multicellular systems dynamically and in a highly controlled manner. Here we summarize some of the most remarkable examples of OOC technology's ability to extract clinically relevant tissue-level information. The review is organized around the types of OOC outputs that can be measured from the cultured tissues and transferred to clinically meaningful information. First, the creation of functional tissues-on-chip is discussed, followed by the presentation of tissue-level readouts specific to OOC, such as morphological changes, vessel formation and function, tissue properties, and metabolic functions. In each case, the clinical relevance of the extracted information is highlighted.
Collapse
Affiliation(s)
- Olivier T Guenat
- ARTORG Center for Biomedical Engineering Research, Medical Faculty, University of Bern, CH-3008 Bern, Switzerland;
- Department of Pulmonary Medicine, University Hospital and University of Bern, CH-3008 Bern, Switzerland
- Thoracic Surgery Department, University Hospital of Bern, Switzerland
| | - Thomas Geiser
- Department of Pulmonary Medicine, University Hospital and University of Bern, CH-3008 Bern, Switzerland
| | - François Berthiaume
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey 08854, USA
| |
Collapse
|
36
|
Del Favero G, Kraegeloh A. Integrating Biophysics in Toxicology. Cells 2020; 9:E1282. [PMID: 32455794 PMCID: PMC7290780 DOI: 10.3390/cells9051282] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/10/2020] [Accepted: 05/15/2020] [Indexed: 12/20/2022] Open
Abstract
Integration of biophysical stimulation in test systems is established in diverse branches of biomedical sciences including toxicology. This is largely motivated by the need to create novel experimental setups capable of reproducing more closely in vivo physiological conditions. Indeed, we face the need to increase predictive power and experimental output, albeit reducing the use of animals in toxicity testing. In vivo, mechanical stimulation is essential for cellular homeostasis. In vitro, diverse strategies can be used to model this crucial component. The compliance of the extracellular matrix can be tuned by modifying the stiffness or through the deformation of substrates hosting the cells via static or dynamic strain. Moreover, cells can be cultivated under shear stress deriving from the movement of the extracellular fluids. In turn, introduction of physical cues in the cell culture environment modulates differentiation, functional properties, and metabolic competence, thus influencing cellular capability to cope with toxic insults. This review summarizes the state of the art of integration of biophysical stimuli in model systems for toxicity testing, discusses future challenges, and provides perspectives for the further advancement of in vitro cytotoxicity studies.
Collapse
Affiliation(s)
- Giorgia Del Favero
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währinger Straße 38-40, 1090 Vienna, Austria
- Core Facility Multimodal Imaging, Faculty of Chemistry, University of Vienna Währinger Straße 38-40, 1090 Vienna, Austria
| | - Annette Kraegeloh
- INM—Leibniz-Institut für Neue Materialien GmbH, Campus D2 2, 66123 Saarbrücken, Germany;
| |
Collapse
|
37
|
Sateesh J, Guha K, Dutta A, Sengupta P, Agarwal A, Srinivasa Rao K. Recreating the size-dependent reabsorption function of proximal convoluted tubule towards artificial kidney applications: Structural analysis and computational study. Artif Organs 2020; 44:E369-E381. [PMID: 32219877 DOI: 10.1111/aor.13683] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/07/2020] [Accepted: 03/18/2020] [Indexed: 01/01/2023]
Abstract
Human kidneys tend to be affected adversely and fail to function more often than any other organ in the body because of diet, heredity, and lifestyle of a person. Dialysis is the technique presently in use for replacing the failed kidney function but it is packed with painfulness, bulkiness, and is costly also. There is a growing need for development of an artificial kidney that eradicates the problems associated with dialysis. This article proposes a structure that mimics the most important aspect of the human kidney: the size-dependent reabsorption of endothelial cells in the proximal convoluted tubule (PCT). The proposed structure consists of transporting channels connecting blood tubules surrounded on both sides of a main tubule. Geometries of the channels are analyzed for optimum flow by varying angles with respect to the main tubule. The analytical formulae have been developed by considering proper boundary conditions governing the flow in the structure, which makes the model as robust, concise, and realistic as the actual PCT. The mathematical model is validated against the benchmark FEM tool COMSOL Multiphysics and the results seem to be satisfactory. This article concludes, that slant channels possess a considerably higher average flow velocity of 5.39 × 10-5 m/s (≈52% reabsorption rate) than straight channels with 4.77 × 10-5 m/s (≈46% reabsorption rate) which is closer to the actual PCT reabsorption rate of 60%. The proposed model is first of its kind in nature among the reported works which creates and exhibits simulation environment of PCT reabsorption function supported by mathematical formulation and also can be useful to study and develop artificial kidney in near future.
Collapse
Affiliation(s)
- Jasti Sateesh
- National MEMS Design Centre, Department of Electronics and Communication Engineering, National Institute of Technology Silchar, Assam, India
| | - Koushik Guha
- National MEMS Design Centre, Department of Electronics and Communication Engineering, National Institute of Technology Silchar, Assam, India
| | - Arindam Dutta
- RG Stone Urology & Laparoscopic Hospital, Kolkata, India
| | | | - Ajay Agarwal
- Smart Sensors Area and Head, Nano Biosensors Group, CSIR-CEERI, Pilani, India
| | - Karumuri Srinivasa Rao
- MEMS Research Centre, Department of Electronics and Communication Engineering, KLEF, Guntur, India
| |
Collapse
|
38
|
Three-dimensional cell-printing of advanced renal tubular tissue analogue. Biomaterials 2020; 232:119734. [DOI: 10.1016/j.biomaterials.2019.119734] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 12/13/2019] [Accepted: 12/25/2019] [Indexed: 12/20/2022]
|
39
|
Pemathilaka RL, Reynolds DE, Hashemi NN. Drug transport across the human placenta: review of placenta-on-a-chip and previous approaches. Interface Focus 2019; 9:20190031. [PMID: 31485316 PMCID: PMC6710654 DOI: 10.1098/rsfs.2019.0031] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2019] [Indexed: 12/20/2022] Open
Abstract
In the past few decades, the placenta became a very controversial topic that has had many researchers and pharmacists discussing the significance of the effects of pharmaceutical drug intake and how it is a possible leading cause towards birth defects. The creation of an in vitro microengineered model of the placenta can be used to replicate the interactions between the mother and fetus, specifically pharmaceutical drug intake reactions. As the field of nanotechnology significantly continues growing, nanotechnology will become more apparent in the study of medicine and other scientific disciplines, specifically microengineering applications. This review is based on past and current research that compares the feasibility and testing of the placenta-on-a-chip microengineered model to the previous and underdeveloped in vivo and ex vivo approaches. The testing of the practicality and effectiveness of the in vitro, in vivo and ex vivo models requires the experimentation of prominent pharmaceutical drugs that most mothers consume during pregnancy. In this case, these drugs need to be studied and tested more often. However, there are challenges associated with the in vitro, in vivo and ex vivo processes when developing a practical placental model, which are discussed in further detail.
Collapse
Affiliation(s)
| | - David E. Reynolds
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA
| | - Nicole N. Hashemi
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA
- Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
40
|
Kankala RK, Wang SB, Chen AZ. Microengineered Organ-on-a-chip Platforms towards Personalized Medicine. Curr Pharm Des 2019; 24:5354-5366. [DOI: 10.2174/1381612825666190222143542] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/12/2019] [Indexed: 11/22/2022]
Abstract
Current preclinical drug evaluation strategies that are explored to predict the pharmacological parameters,
as well as toxicological issues, utilize traditional oversimplified cell cultures and animal models. However,
these traditional approaches are time-consuming, and cannot reproduce the functions of the complex biological
tissue architectures. On the other hand, the obtained data from animal models cannot be precisely extrapolated to
humans because it sometimes results in the distinct safe starting doses for clinical trials due to vast differences in
their genomes. To address these limitations, the microengineered, biomimetic organ-on-a-chip platforms fabricated
using advanced materials that are interconnected using the microfluidic circuits, can stanchly reiterate or
mimic the complex tissue-organ level structures including the cellular architecture and physiology, compartmentalization
and interconnectivity of human organ platforms. These innovative and cost-effective systems potentially
enable the prediction of the responses toward pharmaceutical compounds and remarkable advances in
materials and microfluidics technology, which can rapidly progress the drug development process. In this review,
we emphasize the integration of microfluidic models with the 3D simulations from tissue engineering to fabricate
organ-on-a-chip platforms, which explicitly fulfill the demand of creating the robust models for preclinical testing
of drugs. At first, we give a brief overview of the limitations associated with the current drug development pipeline
that includes drug screening methods, in vitro molecular assays, cell culture platforms and in vivo models.
Further, we discuss various organ-on-a-chip platforms, highlighting their benefits and performance in the preclinical
stages. Next, we aim to emphasize their current applications toward pharmaceutical benefits including the
drug screening as well as toxicity testing, and advances in personalized precision medicine as well as potential
challenges for their commercialization. We finally recapitulate with the lessons learned and the outlook highlighting
the future directions for accelerating the clinical translation of delivery systems.
Collapse
Affiliation(s)
- Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China
| | - Shi-Bin Wang
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China
| | - Ai-Zheng Chen
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, China
| |
Collapse
|
41
|
O'Grady B, Balikov DA, Wang JX, Neal EK, Ou YC, Bardhan R, Lippmann ES, Bellan LM. Spatiotemporal control and modeling of morphogen delivery to induce gradient patterning of stem cell differentiation using fluidic channels. Biomater Sci 2019; 7:1358-1371. [PMID: 30778445 PMCID: PMC6485939 DOI: 10.1039/c8bm01199k] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The process of cell differentiation in a developing embryo is influenced by numerous factors, including various biological molecules whose presentation varies dramatically over space and time. These morphogens regulate cell fate based on concentration profiles, thus creating discrete populations of cells and ultimately generating large, complex tissues and organs. Recently, several in vitro platforms have attempted to recapitulate the complex presentation of extrinsic signals found in nature. However, it has been a challenge to design versatile platforms that can dynamically control morphogen gradients over extended periods of time. To address some of these issues, we introduce a platform using channels patterned in hydrogels to deliver multiple morphogens to cells in a 3D scaffold, thus creating a spectrum of cell phenotypes based on the resultant morphogen gradients. The diffusion coefficient of a common small molecule morphogen, retinoic acid (RA), was measured within our hydrogel platform using Raman spectroscopy and its diffusion in our platform's geometry was modeled using finite element analysis. The predictive model of spatial gradients was validated in a cell-free hydrogel, and temporal control of morphogen gradients was then demonstrated using a reporter cell line that expresses green fluorescent protein in the presence of RA. Finally, the utility of this approach for regulating cell phenotype was demonstrated by generating opposing morphogen gradients to create a spectrum of mesenchymal stem cell differentiation states.
Collapse
Affiliation(s)
- Brian O'Grady
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN, USA.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Sakolish CM, Philip B, Mahler GJ. A human proximal tubule-on-a-chip to study renal disease and toxicity. BIOMICROFLUIDICS 2019; 13:014107. [PMID: 30867877 PMCID: PMC6404920 DOI: 10.1063/1.5083138] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 01/17/2019] [Indexed: 05/08/2023]
Abstract
Renal disease is a global problem with unsustainable health-care costs. There currently exists a lack of accurate human renal disease models that take into account the complex microenvironment of these tissues. Here, we present a reusable microfluidic model of the human proximal tubule and glomerulus, which allows for the growth of renal epithelial cells in a variety of conditions that are representative of renal disease states including altered glomerular filtration rate, hyperglycemia, nephrolithiasis, and drug-induced nephrotoxicity (cisplatin and cyclosporine). Cells were exposed to these conditions under fluid flow or in traditional static cultures to determine the effects of a dynamic microenvironment on the pathogenesis of these renal disease states. The results indicate varying stress-related responses (α-smooth muscle actin (α-SMA) expression, alkaline phosphatase activity, fibronectin, and neutrophil gelatinase-associated lipocalin secretion) to each of these conditions when comparing cells that had been grown in static and dynamic conditions, potentially indicating more realistic and sensitive predictions of human responses and a requirement for a more complex "fit for purpose" model.
Collapse
Affiliation(s)
| | - Brian Philip
- Department of Biomedical Engineering, Binghamton University, Binghamton, New York 13902, USA
| | - Gretchen J. Mahler
- Department of Biomedical Engineering, Binghamton University, Binghamton, New York 13902, USA
- Author to whom correspondence should be addressed: . Tel.: (607) 777-5238
| |
Collapse
|
43
|
He Z, Ranganathan N, Li P. Evaluating nanomedicine with microfluidics. NANOTECHNOLOGY 2018; 29:492001. [PMID: 30215611 DOI: 10.1088/1361-6528/aae18a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Nanomedicines are engineered nanoscale structures that have an extensive range of application in the diagnosis and therapy of many diseases. Despite the rapid progress in and tremendous potential of nanomedicines, their clinical translational process is still slow, owing to the difficulty in understanding, evaluating, and predicting their behavior in complex living organisms. Microfluidic techniques offer a promising way to resolve these challenges. Carefully designed microfluidic chips enable in vivo microenvironment simulation and high-throughput analysis, thus providing robust platforms for nanomedicine evaluation. Here, we summarize the recent developments and achievements in microfluidic methods for nanomedicine evaluation, categorized into four sections based on their target systems: single cell, multicellular system, organ, and organism levels. Finally, we provide our perspectives on the challenges and future directions of microfluidics-based nanomedicine evaluation.
Collapse
Affiliation(s)
- Ziyi He
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, WV, United States of America
| | | | | |
Collapse
|
44
|
In vitro and ex vivo systems at the forefront of infection modeling and drug discovery. Biomaterials 2018; 198:228-249. [PMID: 30384974 PMCID: PMC7172914 DOI: 10.1016/j.biomaterials.2018.10.030] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 10/05/2018] [Accepted: 10/23/2018] [Indexed: 12/11/2022]
Abstract
Bacterial infections and antibiotic resistant bacteria have become a growing problem over the past decade. As a result, the Centers for Disease Control predict more deaths resulting from microorganisms than all cancers combined by 2050. Currently, many traditional models used to study bacterial infections fail to precisely replicate the in vivo bacterial environment. These models often fail to incorporate fluid flow, bio-mechanical cues, intercellular interactions, host-bacteria interactions, and even the simple inclusion of relevant physiological proteins in culture media. As a result of these inadequate models, there is often a poor correlation between in vitro and in vivo assays, limiting therapeutic potential. Thus, the urgency to establish in vitro and ex vivo systems to investigate the mechanisms underlying bacterial infections and to discover new-age therapeutics against bacterial infections is dire. In this review, we present an update of current in vitro and ex vivo models that are comprehensively changing the landscape of traditional microbiology assays. Further, we provide a comparative analysis of previous research on various established organ-disease models. Lastly, we provide insight on future techniques that may more accurately test new formulations to meet the growing demand of antibiotic resistant bacterial infections.
Collapse
|
45
|
Han S, Kim J, Li R, Ma A, Kwan V, Luong K, Sohn LL. Hydrophobic Patterning-Based 3D Microfluidic Cell Culture Assay. Adv Healthc Mater 2018; 7:e1800122. [PMID: 29700986 PMCID: PMC6342489 DOI: 10.1002/adhm.201800122] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/20/2018] [Indexed: 01/11/2023]
Abstract
Engineering physiologically relevant in vitro models of human organs remains a fundamental challenge. Despite significant strides made within the field, many promising organ-on-a-chip models fall short in recapitulating cellular interactions with neighboring cell types, surrounding extracellular matrix (ECM), and exposure to soluble cues due, in part, to the formation of artificial structures that obstruct >50% of the surface area of the ECM. Here, a 3D cell culture platform based upon hydrophobic patterning of hydrogels that is capable of precisely generating a 3D ECM within a microfluidic channel with an interaction area >95% is reported. In this study, for demonstrative purposes, type I collagen (COL1), Matrigel (MAT), COL1/MAT mixture, hyaluronic acid, and cell-laden MAT are formed in the device. Three potential applications are demonstrated, including creating a 3D endothelium model, studying the interstitial migration of cancer cells, and analyzing stem cell differentiation in a 3D environment. The hydrophobic patterned-based 3D cell culture device provides the ease-of-fabrication and flexibility necessary for broad potential applications in organ-on-a-chip platforms.
Collapse
Affiliation(s)
- Sewoon Han
- The California Institute for Quantitative Biosciences, Stanley Hall, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Junghyun Kim
- Department of Mechanical Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Rui Li
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Alice Ma
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Vincent Kwan
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kevin Luong
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Lydia L. Sohn
- Department of Mechanical Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
46
|
Kenney RM, Lloyd CC, Whitman NA, Lockett MR. 3D cellular invasion platforms: how do paper-based cultures stack up? Chem Commun (Camb) 2018. [PMID: 28621775 DOI: 10.1039/c7cc02357j] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cellular invasion is the gateway to metastasis, which is the leading cause of cancer-related deaths. Invasion is driven by a number of chemical and mechanical stresses that arise in the tumor microenvironment. In vitro assays are needed for the systematic study of cancer progress. To be truly predictive, these assays must generate tissue-like environments that can be experimentally controlled and manipulated. While two-dimensional (2D) monolayer cultures are easily assembled and evaluated, they lack the extracellular components needed to assess invasion. Three-dimensional (3D) cultures are better suited for invasion studies because they generate cellular phenotypes that are more representative of those found in vivo. This feature article provides an overview of four invasion platforms. We focus on paper-based cultures, an emerging 3D culture platform capable of generating tissue-like structures and quantifying cellular invasion. Paper-based cultures are as easily assembled and analyzed as monolayers, but provide an experimentally powerful platform capable of supporting: co-cultures and representative extracellular environments; experimentally controlled gradients; readouts capable of quantifying, discerning, and separating cells based on their invasiveness. With a series of examples we highlight the potential of paper-based cultures, and discuss how they stack up against other invasion platforms.
Collapse
Affiliation(s)
- Rachael M Kenney
- Department of Chemistry, University of North Carolina at Chapel Hill, Kenan and Caudill Laboratories, 125 South Road, Chapel Hill, NC 27599-3290, USA.
| | | | | | | |
Collapse
|
47
|
Nagamine K, Sato H, Kai H, Kaji H, Kanzaki M, Nishizawa M. Contractile Skeletal Muscle Cells Cultured with a Conducting Soft Wire for Effective, Selective Stimulation. Sci Rep 2018; 8:2253. [PMID: 29396483 PMCID: PMC5797109 DOI: 10.1038/s41598-018-20729-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 01/23/2018] [Indexed: 01/04/2023] Open
Abstract
Contractile skeletal muscle cells were cultured so as to wrap around an electrode wire to enable their selective stimulation even when they were co-cultured with other electrically-excitable cells. Since the electrode wire was composed of the conducting polymer poly(3,4-ethylenedioxythiophene) (PEDOT) and polyurethane (PU), which is soft and highly capacitive (~10 mF cm-2), non-faradaic electrical stimulation with charge/discharge currents could be applied to the surrounding cells without causing significant damage even for longer periods (more than a week). The advantage of this new culture system was demonstrated in the study of chemotactic interaction of monocytes and skeletal muscle cells via myokines.
Collapse
Affiliation(s)
- Kuniaki Nagamine
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, 6-6-01 Aramaki, Aoba-ku, Sendai, 980-8579, Japan
| | - Hirotaka Sato
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, 6-6-01 Aramaki, Aoba-ku, Sendai, 980-8579, Japan
| | - Hiroyuki Kai
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, 6-6-01 Aramaki, Aoba-ku, Sendai, 980-8579, Japan
| | - Hirokazu Kaji
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, 6-6-01 Aramaki, Aoba-ku, Sendai, 980-8579, Japan
| | - Makoto Kanzaki
- Department of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, 6-6-04 Aoba-ku, Sendai, 980-8579, Japan
| | - Matsuhiko Nishizawa
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, 6-6-01 Aramaki, Aoba-ku, Sendai, 980-8579, Japan.
| |
Collapse
|
48
|
Plegue TJ, Kovach KM, Thompson AJ, Potkay JA. Stability of Polyethylene Glycol and Zwitterionic Surface Modifications in PDMS Microfluidic Flow Chambers. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:492-502. [PMID: 29231737 DOI: 10.1021/acs.langmuir.7b03095] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Blood-material interactions are crucial to the lifetime, safety, and overall success of blood contacting devices. Hydrophilic polymer coatings have been employed to improve device lifetime by shielding blood contacting materials from the natural foreign body response, primarily the intrinsic pathway of the coagulation cascade. These coatings have the ability to repel proteins, cells, bacteria, and other micro-organisms. Coatings are desired to have long-term stability, so that the nonthrombogenic and nonfouling effects gained are long lasting. Unfortunately, there exist limited studies which investigate their stability under dynamic flow conditions as encountered in a physiological setting. In addition, direct comparisons between multiple coatings are lacking in the literature. In this study, we investigate the stability of polyethylene glycol (PEG), zwitterionic sulfobetaine silane (SBSi), and zwitterionic polyethylene glycol sulfobetaine silane (PEG-SBSi) grafted by a room temperature, sequential flow chemistry process on polydimethylsiloxane (PDMS) over time under ambient, static fluid (no flow), and physiologically relevant flow conditions and compare the results to uncoated PDMS controls. PEG, SBSi, and PEG-SBSi coatings maintained contact angles below 20° for up to 35 days under ambient conditions. SBSi and PEG-SBSi showed increased stability and hydrophilicity after 7 days under static conditions. They also retained contact angles ≤40° for all shear rates after 7 days under flow, demonstrating their potential for long-term stability. The effectiveness of the coatings to resist platelet adhesion was also studied under physiological flow conditions. PEG showed a 69% reduction in adhered platelets, PEG-SBSi a significant 80% reduction, and SBSi a significant 96% reduction compared to uncoated control samples, demonstrating their potential applicability for blood contacting applications. In addition, the presented coatings and their stability under shear may be of interest in other applications including marine coatings, lab on a chip devices, and contact lenses, where it is desirable to reduce surface fouling due to proteins, cells, and other organisms.
Collapse
Affiliation(s)
- Thomas J Plegue
- VA Ann Arbor Healthcare System , Ann Arbor, Michigan 48105, United States
| | - Kyle M Kovach
- Department of Biomedical Engineering, Case Western Reserve University , Cleveland, Ohio 44106, United States
| | - Alex J Thompson
- VA Ann Arbor Healthcare System , Ann Arbor, Michigan 48105, United States
- Department of Surgery, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Joseph A Potkay
- VA Ann Arbor Healthcare System , Ann Arbor, Michigan 48105, United States
- Department of Surgery, University of Michigan , Ann Arbor, Michigan 48109, United States
| |
Collapse
|
49
|
Ahadian S, Civitarese R, Bannerman D, Mohammadi MH, Lu R, Wang E, Davenport-Huyer L, Lai B, Zhang B, Zhao Y, Mandla S, Korolj A, Radisic M. Organ-On-A-Chip Platforms: A Convergence of Advanced Materials, Cells, and Microscale Technologies. Adv Healthc Mater 2018; 7. [PMID: 29034591 DOI: 10.1002/adhm.201700506] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/15/2017] [Indexed: 12/11/2022]
Abstract
Significant advances in biomaterials, stem cell biology, and microscale technologies have enabled the fabrication of biologically relevant tissues and organs. Such tissues and organs, referred to as organ-on-a-chip (OOC) platforms, have emerged as a powerful tool in tissue analysis and disease modeling for biological and pharmacological applications. A variety of biomaterials are used in tissue fabrication providing multiple biological, structural, and mechanical cues in the regulation of cell behavior and tissue morphogenesis. Cells derived from humans enable the fabrication of personalized OOC platforms. Microscale technologies are specifically helpful in providing physiological microenvironments for tissues and organs. In this review, biomaterials, cells, and microscale technologies are described as essential components to construct OOC platforms. The latest developments in OOC platforms (e.g., liver, skeletal muscle, cardiac, cancer, lung, skin, bone, and brain) are then discussed as functional tools in simulating human physiology and metabolism. Future perspectives and major challenges in the development of OOC platforms toward accelerating clinical studies of drug discovery are finally highlighted.
Collapse
Affiliation(s)
- Samad Ahadian
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Robert Civitarese
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Dawn Bannerman
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Mohammad Hossein Mohammadi
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Rick Lu
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Erika Wang
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Locke Davenport-Huyer
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Ben Lai
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Boyang Zhang
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Yimu Zhao
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Serena Mandla
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Anastasia Korolj
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| |
Collapse
|
50
|
Organoids, organs-on-chips and other systems, and microbiota. Emerg Top Life Sci 2017; 1:385-400. [PMID: 33525777 PMCID: PMC7289039 DOI: 10.1042/etls20170047] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 10/11/2017] [Accepted: 10/16/2017] [Indexed: 12/15/2022]
Abstract
The human gut microbiome is considered an organ in its entirety and has been the subject of extensive research due to its role in physiology, metabolism, digestion, and immune regulation. Disequilibria of the normal microbiome have been associated with the development of several gastrointestinal diseases, but the exact underlying interactions are not well understood. Conventional in vivo and in vitro modelling systems fail to faithfully recapitulate the complexity of the human host–gut microbiome, emphasising the requirement for novel systems that provide a platform to study human host–gut microbiome interactions with a more holistic representation of the human in vivo microenvironment. In this review, we outline the progression and applications of new and old modelling systems with particular focus on their ability to model and to study host–microbiome cross-talk.
Collapse
|