51
|
Injectable Therapeutic Organoids Using Sacrificial Hydrogels. iScience 2020; 23:101052. [PMID: 32353766 PMCID: PMC7191221 DOI: 10.1016/j.isci.2020.101052] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 03/11/2020] [Accepted: 04/03/2020] [Indexed: 12/28/2022] Open
Abstract
Organoids are becoming widespread in drug-screening technologies but have been used sparingly for cell therapy as current approaches for producing self-organized cell clusters lack scalability or reproducibility in size and cellular organization. We introduce a method of using hydrogels as sacrificial scaffolds, which allow cells to form self-organized clusters followed by gentle release, resulting in highly reproducible multicellular structures on a large scale. We demonstrated this strategy for endothelial cells and mesenchymal stem cells to self-organize into blood-vessel units, which were injected into mice, and rapidly formed perfusing vasculature. Moreover, in a mouse model of peripheral artery disease, intramuscular injections of blood-vessel units resulted in rapid restoration of vascular perfusion within seven days. As cell therapy transforms into a new class of therapeutic modality, this simple method—by making use of the dynamic nature of hydrogels—could offer high yields of self-organized multicellular aggregates with reproducible sizes and cellular architectures. Therapeutic, prevascularized organoids were formed in a sacrificial scaffold The organoids are highly reproducible and grown in a high-throughput manner The organoids rapidly formed perfusing vasculature in healthy mice Therapeutic potential was assessed in a mouse model of peripheral artery disease
Collapse
|
52
|
Li J, Wu C, Chu PK, Gelinsky M. 3D printing of hydrogels: Rational design strategies and emerging biomedical applications. MATERIALS SCIENCE AND ENGINEERING: R: REPORTS 2020; 140:100543. [DOI: 10.1016/j.mser.2020.100543] [Citation(s) in RCA: 350] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
53
|
Lin Z, Luo G, Du W, Kong T, Liu C, Liu Z. Recent Advances in Microfluidic Platforms Applied in Cancer Metastasis: Circulating Tumor Cells' (CTCs) Isolation and Tumor-On-A-Chip. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1903899. [PMID: 31747120 DOI: 10.1002/smll.201903899] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/13/2019] [Indexed: 05/03/2023]
Abstract
Cancer remains the leading cause of death worldwide despite the enormous efforts that are made in the development of cancer biology and anticancer therapeutic treatment. Furthermore, recent studies in oncology have focused on the complex cancer metastatic process as metastatic disease contributes to more than 90% of tumor-related death. In the metastatic process, isolation and analysis of circulating tumor cells (CTCs) play a vital role in diagnosis and prognosis of cancer patients at an early stage. To obtain relevant information on cancer metastasis and progression from CTCs, reliable approaches are required for CTC detection and isolation. Additionally, experimental platforms mimicking the tumor microenvironment in vitro give a better understanding of the metastatic microenvironment and antimetastatic drugs' screening. With the advancement of microfabrication and rapid prototyping, microfluidic techniques are now increasingly being exploited to study cancer metastasis as they allow precise control of fluids in small volume and rapid sample processing at relatively low cost and with high sensitivity. Recent advancements in microfluidic platforms utilized in various methods for CTCs' isolation and tumor models recapitulating the metastatic microenvironment (tumor-on-a-chip) are comprehensively reviewed. Future perspectives on microfluidics for cancer metastasis are proposed.
Collapse
Affiliation(s)
- Zhengjie Lin
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Guanyi Luo
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Weixiang Du
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Tiantian Kong
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Changkun Liu
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Zhou Liu
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| |
Collapse
|
54
|
Park E, Kim HK, Jee J, Hahn S, Jeong S, Yoo J. Development of organoid-based drug metabolism model. Toxicol Appl Pharmacol 2019; 385:114790. [DOI: 10.1016/j.taap.2019.114790] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 08/27/2019] [Accepted: 10/22/2019] [Indexed: 12/23/2022]
|
55
|
Sun J, Warden AR, Ding X. Recent advances in microfluidics for drug screening. BIOMICROFLUIDICS 2019; 13:061503. [PMID: 31768197 PMCID: PMC6870548 DOI: 10.1063/1.5121200] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 11/07/2019] [Indexed: 05/03/2023]
Abstract
With ever increasing drug resistance and emergence of new diseases, demand for new drug development is at an unprecedented urgency. This fact has led to extensive recent efforts to develop new drugs and novel techniques for efficient drug screening. However, new drug development is commonly hindered by cost and time span. Thus, developing more accessible, cost-effective methods for drug screening is necessary. Compared with conventional drug screening methods, a microfluidic-based system has superior advantages in sample consumption, reaction time, and cost of the operation. In this paper, the advantages of microfluidic technology in drug screening as well as the critical factors for device design are described. The strategies and applications of microfluidics for drug screening are reviewed. Moreover, current limitations and future prospects for a drug screening microdevice are also discussed.
Collapse
Affiliation(s)
- Jiahui Sun
- State Key Laboratory of Oncogenes and Related Genes, Institute for
Personalized Medicine and School of Biomedical Engineering, Shanghai Jiao Tong
University, Shanghai 200030, China
| | - Antony R. Warden
- State Key Laboratory of Oncogenes and Related Genes, Institute for
Personalized Medicine and School of Biomedical Engineering, Shanghai Jiao Tong
University, Shanghai 200030, China
| | - Xianting Ding
- State Key Laboratory of Oncogenes and Related Genes, Institute for
Personalized Medicine and School of Biomedical Engineering, Shanghai Jiao Tong
University, Shanghai 200030, China
| |
Collapse
|
56
|
Lim KS, Baptista M, Moon S, Woodfield TB, Rnjak-Kovacina J. Microchannels in Development, Survival, and Vascularisation of Tissue Analogues for Regenerative Medicine. Trends Biotechnol 2019; 37:1189-1201. [DOI: 10.1016/j.tibtech.2019.04.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/02/2019] [Accepted: 04/03/2019] [Indexed: 11/26/2022]
|
57
|
Francis L, Greco KV, Boccaccini AR, Roether JJ, English NR, Huang H, Ploeg R, Ansari T. Development of a novel hybrid bioactive hydrogel for future clinical applications. J Biomater Appl 2019; 33:447-465. [PMID: 30223736 DOI: 10.1177/0885328218794163] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Three-dimensional hydrogels are ideal for tissue engineering applications due to their structural integrity and similarity to native soft tissues; however, they can lack mechanical stability. Our objective was to develop a bioactive and mechanically stable hydrogel for clinical application. Auricular cartilage was decellularised using a combination of hypertonic and hypotonic solutions with and without enzymes to produce acellular tissue. Methacryloyl groups were crosslinked with alginate and PVA main chains via 2-aminoethylmathacrylate and the entire macromonomer further crosslinked with the acellular tissue. The resultant hydrogels were characterised for its physicochemical properties (using NMR), in vitro degradation (via GPC analysis), mechanical stability (compression tests) and in vitro biocompatibility (co-culture with bone marrow-derived mesenchymal stem cells). Following decellularisation, the cartilage tissue showed to be acellular at a significant level (DNA content 25.33 ng/mg vs. 351.46 ng/mg control tissue), with good structural and molecular integrity of the retained extra cellular matrix (s-GAG= 0.19 μg/mg vs. 0.65 μg/mg ±0.001 control tissue). Proteomic analysis showed that collagen subtypes and proteoglycans were retained, and SEM and TEM showed preserved matrix ultra-structure. The hybrid hydrogel was successfully cross-linked with biological and polymer components, and it was stable for 30 days in simulated body fluid (poly dispersal index for alginate with tissue was stable at 1.08 and for PVA with tissue was stable at 1.16). It was also mechanically stable (Young's modulus of 0.46 ± 0.31 KPa) and biocompatible, as it was able to support the development of a multi-cellular feature with active cellular proliferation in vitro. We have shown that it is possible to successfully combine biological tissue with both a synthetic and natural polymer and create a hybrid bioactive hydrogel for clinical application.
Collapse
Affiliation(s)
- Lydia Francis
- 1 Tissue Engineering and Regenerative Medicine, Northwick Park Institute for Medical Research (NPIMR), Harrow, UK
| | - Karin V Greco
- 1 Tissue Engineering and Regenerative Medicine, Northwick Park Institute for Medical Research (NPIMR), Harrow, UK
| | - Aldo R Boccaccini
- 2 Department of Materials Science, Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Judith J Roether
- 2 Department of Materials Science, Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Nicholas R English
- 3 Antigen Presentation Research Group, Imperial College London, London, UK
| | - Honglei Huang
- 4 Nuffield Department of Surgical Sciences, Oxford Transplant Centre, University of Oxford, Oxford, UK
| | - R Ploeg
- 4 Nuffield Department of Surgical Sciences, Oxford Transplant Centre, University of Oxford, Oxford, UK
| | - Tahera Ansari
- 1 Tissue Engineering and Regenerative Medicine, Northwick Park Institute for Medical Research (NPIMR), Harrow, UK
| |
Collapse
|
58
|
Wragg NM, Mosqueira D, Blokpeol-Ferreras L, Capel A, Player DJ, Martin NRW, Liu Y, Lewis MP. Development of a 3D Tissue-Engineered Skeletal Muscle and Bone Co-culture System. Biotechnol J 2019; 15:e1900106. [PMID: 31468704 DOI: 10.1002/biot.201900106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 07/05/2019] [Indexed: 12/26/2022]
Abstract
In vitro 3D tissue-engineered (TE) structures have been shown to better represent in vivo tissue morphology and biochemical pathways than monolayer culture, and are less ethically questionable than animal models. However, to create systems with even greater relevance, multiple integrated tissue systems should be recreated in vitro. In the present study, the effects and conditions most suitable for the co-culture of TE skeletal muscle and bone are investigated. High-glucose Dulbecco's modified Eagle medium (HG-DMEM) supplemented with 20% fetal bovine serum followed by HG-DMEM with 2% horse serum is found to enable proliferation of both C2C12 muscle precursor cells and TE85 human osteosarcoma cells, fusion of C2C12s into myotubes, as well as an upregulation of RUNX2/CBFa1 in TE85s. Myotube formation is also evident within indirect contact monolayer cultures. Finally, in 3D co-cultures, TE85 collagen/hydroxyapatite constructs have significantly greater expression of RUNX2/CBFa1 and osteocalcin/BGLAP in the presence of collagen-based C2C12 skeletal muscle constructs; however, fusion within these constructs appears reduced. This work demonstrates the first report of the simultaneous co-culture and differentiation of 3D TE skeletal muscle and bone, and represents a significant step toward a full in vitro 3D musculoskeletal junction model.
Collapse
Affiliation(s)
- Nicholas M Wragg
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.,Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, UK
| | - Diogo Mosqueira
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Lia Blokpeol-Ferreras
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Andrew Capel
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Darren J Player
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.,Institute of Orthopaedics and Musculoskeletal Sciences, RNOH University College London, Stanmore, UK
| | - Neil R W Martin
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Yang Liu
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, UK
| | - Mark P Lewis
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| |
Collapse
|
59
|
Screening of perfused combinatorial 3D microenvironments for cell culture. Acta Biomater 2019; 96:222-236. [PMID: 31255663 DOI: 10.1016/j.actbio.2019.06.047] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 06/23/2019] [Accepted: 06/25/2019] [Indexed: 02/08/2023]
Abstract
Biomaterials combining biochemical and biophysical cues to establish close-to-extracellular matrix (ECM) models have been explored for cell expansion and differentiation purposes. Multivariate arrays are used as material-saving and rapid-to-analyze platforms, which enable selecting hit-spotted formulations targeting specific cellular responses. However, these systems often lack the ability to emulate dynamic mechanical aspects that occur in specific biological milieus and affect physiological phenomena including stem cells differentiation, tumor progression, or matrix modulation. We report a tailor-made strategy to address the combined effect of flow and biochemical composition of three-dimensional (3D) biomaterials on cellular response. We suggest a simple-to-implement device comprising (i) a perforated platform accommodating miniaturized 3D biomaterials and (ii) a bioreactor that enables the incorporation of the biomaterial-containing array into a disposable perfusion chamber. The system was upscaled to parallelizable setups, increasing the number of analyzed platforms per independent experiment. As a proof-of-concept, porous chitosan scaffolds with 1 mm diameter were functionalized with combinations of 5 ECM and cell-cell contact-mediating proteins, relevant for bone and dental regeneration, corresponding to 32 protein combinatorial formulations. Mesenchymal stem cells adhesion and production of an early osteogenic marker were assessed on-chip on static and under-flow dynamic perfusion conditions. Different hit-spotted biomaterial formulations were detected for the different flow regimes using direct image analysis. Cell-binding proteins still poorly explored as biomaterials components - amelogenin and E-cadherin - were here shown as relevant cell response modulators. Their combination with ECM cell-binding proteins - fibronectin, vitronectin, and type 1 collagen - rendered specific biomaterial combinations with high cell adhesion and ALP production under flow. The developed versatile system may be targeted at widespread tissue regeneration applications, and as a disease model/drug screening platform. STATEMENT OF SIGNIFICANCE: A perfusion system that enables cell culture in arrays of three-dimensional biomaterials under dynamic flow is reported. The effect of 31 cell-binding protein combinations in the adhesion and alkaline phosphatase (ALP) production of mesenchymal stem cells was assessed using a single bioreactor chamber. Flow perfusion was not only assessed as a classical enhancer/accelerator of cell growth and early osteogenic differentiation. We hypothesized that flow may affect cell-protein interactions, and that key components driving cell response may differ under static or dynamic regimes. Indeed, hit-spotted formulations that elicited highest cell attachment and ALP production on static cell culture differed from the ones detected for dynamic flow assays. The impacting role of poorly studied proteins as E-cadherin and amelogenin as biomaterial components was highlighted.
Collapse
|
60
|
Kejlová K, Dvořáková M, Vavrouš A, Ševčík V, Kanďárová H, Letašiová S, Sosnovcová J, Jírová D. Toxicity of food contact paper evaluated by combined biological and chemical methods. Toxicol In Vitro 2019; 59:26-34. [PMID: 30951805 DOI: 10.1016/j.tiv.2019.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/27/2019] [Accepted: 04/01/2019] [Indexed: 12/16/2022]
Abstract
The study was focused on assessment of potential health risks of paper-based food contact materials (FCMs) in a step-wise approach using three toxicological bioassays in vitro and chemical analyses of migrating contaminants. 3T3 NRU cytotoxicity test showed high sensitivity to detect basal toxicity of FCMs extracts and served as a first-line test for selection of samples for further testing. The reconstructed human intestine model EpiIntestinal showed more realistic tissue response than cell culture monolayer and higher resistance despite prolonged exposure to the selected 6 samples, i.e. negligible decrease of viability and intestinal penetration, nevertheless an increase of IL-8 after exposure to black printed sample extract. Yeast based assays identified weak agonistic/antagonostic activity to human androgen receptor of the black printed sample. In accordance with the biological effects, the targeted LC and GC analytical methods confirmed the presence of high amounts of phthalates, photoinitiators and PAHs that could justify the hazard of the black printed sample. Heavily printed uncoated FCMs are recognized not to be suitable for direct contact with food. The selected bioassays and chemical analyses might be useful tools to detect targeted biological effects of xenobiotics suspected to contribute to human exposure from food.
Collapse
Affiliation(s)
- Kristina Kejlová
- National Institute of Public Health, Šrobárova 48, 100 42 Prague 10, Czech Republic.
| | - Markéta Dvořáková
- National Institute of Public Health, Šrobárova 48, 100 42 Prague 10, Czech Republic; Third Faculty of Medicine, Charles University in Prague, Ruská 87, 100 00 Prague 10, Czech Republic
| | - Adam Vavrouš
- National Institute of Public Health, Šrobárova 48, 100 42 Prague 10, Czech Republic; Faculty of Science, Department of Analytical Chemistry, Charles University in Prague, Hlavova 2030/8, 128 43 Prague 2, Czech Republic
| | - Václav Ševčík
- National Institute of Public Health, Šrobárova 48, 100 42 Prague 10, Czech Republic; Faculty of Science, Department of Analytical Chemistry, Charles University in Prague, Hlavova 2030/8, 128 43 Prague 2, Czech Republic
| | - Helena Kanďárová
- MatTek In Vitro Life Science Laboratories, Mlynské Nivy 73, 82105 Bratislava, Slovak Republic
| | - Silvia Letašiová
- MatTek In Vitro Life Science Laboratories, Mlynské Nivy 73, 82105 Bratislava, Slovak Republic
| | - Jitka Sosnovcová
- National Institute of Public Health, Šrobárova 48, 100 42 Prague 10, Czech Republic
| | - Dagmar Jírová
- National Institute of Public Health, Šrobárova 48, 100 42 Prague 10, Czech Republic
| |
Collapse
|
61
|
Micro Vacuum Chuck and Tensile Test System for Bio-Mechanical Evaluation of 3D Tissue Constructed of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes (hiPS-CM). MICROMACHINES 2019; 10:mi10070487. [PMID: 31331014 PMCID: PMC6680730 DOI: 10.3390/mi10070487] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/15/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022]
Abstract
In this report, we propose a micro vacuum chuck (MVC) which can connect three-dimensional (3D) tissues to a tensile test system by vacuum pressure. Because the MVC fixes the 3D tissue by vacuum pressure generated on multiple vacuum holes, it is expected that the MVC can fix 3D tissue to the system easily and mitigate the damage which can happen by handling during fixing. In order to decide optimum conditions for the size of the vacuum holes and the vacuum pressure, various sized vacuum holes and vacuum pressures were applied to a normal human cardiac fibroblast 3D tissue. From the results, we confirmed that a square shape with 100 µm sides was better for fixing the 3D tissue. Then we mounted our developed MVCs on a specially developed tensile test system and measured the bio-mechanical property (beating force) of cardiac 3D tissue which was constructed of human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CM); the 3D tissue had been assembled by the layer-by-layer (LbL) method. We measured the beating force of the cardiac 3D tissue and confirmed the measured force followed the Frank-Starling relationship. This indicates that the beating property of cardiac 3D tissue obtained by the LbL method was close to that of native cardiac tissue.
Collapse
|
62
|
Fox S, Polak J, Schmid Daners M, Meboldt M. Fabrication of Bioengineered Skin by Injection Molding: A Feasibility Study on Automation. SLAS Technol 2019; 24:506-514. [PMID: 31251675 DOI: 10.1177/2472630319857966] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The use of bioengineered skin has facilitated fundamental and applied research because it enables the investigation of complex interactions between various cell types as well as the extracellular matrix. The predominantly manual fabrication of these living tissues means, however, that their quality, standardization, and production volume are extremely dependent on the technician's experience. Simple laboratory automation could facilitate the use of living tissues by a greater number of research groups. We developed and present here an injection molding technique for the fabrication of bilayered skin equivalents. The tissue was formed automatically by two separate injections into a customized mold to produce the dermal and epidermal skin layers. We demonstrated the biocompatibility of this fabrication process and confirmed the resulting bilayered morphology of the bioengineered skin using histology and immunohistochemistry. Our findings highlight the possibility of fabricating multilayered living tissue by injection molding, suggesting that further investigation into this automation method could result in the rapid and low-cost fabrication of standardized bioengineered skin.
Collapse
Affiliation(s)
- S Fox
- Product Development Group Zurich pd
- z, Department of Mechanical and Process Engineering, ETH Zürich, Zürich, Switzerland
| | - J Polak
- Product Development Group Zurich pd
- z, Department of Mechanical and Process Engineering, ETH Zürich, Zürich, Switzerland
| | - M Schmid Daners
- Product Development Group Zurich pd
- z, Department of Mechanical and Process Engineering, ETH Zürich, Zürich, Switzerland
| | - M Meboldt
- Product Development Group Zurich pd
- z, Department of Mechanical and Process Engineering, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
63
|
Park SB, Lee SY, Jung WH, Lee J, Jeong HG, Hong J, Kang D, Kim KY. Development of in vitro three-dimensional co-culture system for metabolic syndrome therapeutic agents. Diabetes Obes Metab 2019; 21:1146-1157. [PMID: 30609258 DOI: 10.1111/dom.13628] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 12/23/2018] [Accepted: 01/02/2019] [Indexed: 01/08/2023]
Abstract
AIMS There are many obstacles to overcome in the development of new drugs for metabolic diseases, including efficacy and toxicity problems in later stages of drug development. To overcome these problems and predict efficacy and toxicity in early stages, we constructed a new model of insulin resistance in terms of communication between 3T3-L1 adipocytes and RAW264.7 macrophages by three-dimensional (3D) culture. RESULTS In this study, results focused on the functional resemblance between 3D co-culture of adipocytes and macrophages and adipose tissue in diabetic mice. The 3D mono-culture preadipocytes showed good cell viability and induced cell differentiation to adipocytes, without cell confluence or cell-cell contact and interaction. The 3D co-cultured preadipocytes with RAW264.7 macrophages induced greater insulin resistance than two-dimensional and 3D mono-cultured adipocytes. Additionally, we demonstrated that 3D co-culture model had functional metabolic similarity to adipose tissue in diabetic mice. We utilized this 3D co-culture system to screen PPARγ antagonists that might have potential as therapeutic agents for diabetes as demonstrated by an in vivo assay. CONCLUSION This in vitro 3D co-culture system could serve as a next-generation platform to accelerate the development of therapeutics for metabolic diseases.
Collapse
Affiliation(s)
- Sung B Park
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Sun Y Lee
- College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Won H Jung
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Junhee Lee
- Department of Nature-Inspired Nano Convergence System, Korea Institute of Machinery and Materials, Daejeon, Republic of Korea
| | - Hye G Jeong
- Department of Toxicology, College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Jongki Hong
- College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Dukjin Kang
- Center for Bioanalysis, Division of Chemical and Medical Metrology, Korea Research Institute of Standards and Science, Daejeon, Republic of Korea
| | - Ki Y Kim
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
64
|
Savoji H, Mohammadi MH, Rafatian N, Toroghi MK, Wang EY, Zhao Y, Korolj A, Ahadian S, Radisic M. Cardiovascular disease models: A game changing paradigm in drug discovery and screening. Biomaterials 2019; 198:3-26. [PMID: 30343824 PMCID: PMC6397087 DOI: 10.1016/j.biomaterials.2018.09.036] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/11/2018] [Accepted: 09/22/2018] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease is the leading cause of death worldwide. Although investment in drug discovery and development has been sky-rocketing, the number of approved drugs has been declining. Cardiovascular toxicity due to therapeutic drug use claims the highest incidence and severity of adverse drug reactions in late-stage clinical development. Therefore, to address this issue, new, additional, replacement and combinatorial approaches are needed to fill the gap in effective drug discovery and screening. The motivation for developing accurate, predictive models is twofold: first, to study and discover new treatments for cardiac pathologies which are leading in worldwide morbidity and mortality rates; and second, to screen for adverse drug reactions on the heart, a primary risk in drug development. In addition to in vivo animal models, in vitro and in silico models have been recently proposed to mimic the physiological conditions of heart and vasculature. Here, we describe current in vitro, in vivo, and in silico platforms for modelling healthy and pathological cardiac tissues and their advantages and disadvantages for drug screening and discovery applications. We review the pathophysiology and the underlying pathways of different cardiac diseases, as well as the new tools being developed to facilitate their study. We finally suggest a roadmap for employing these non-animal platforms in assessing drug cardiotoxicity and safety.
Collapse
Affiliation(s)
- Houman Savoji
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Mohammad Hossein Mohammadi
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada; Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Naimeh Rafatian
- Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Masood Khaksar Toroghi
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada
| | - Erika Yan Wang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada
| | - Yimu Zhao
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada
| | - Anastasia Korolj
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada
| | - Samad Ahadian
- Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada; Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada.
| |
Collapse
|
65
|
Zhao Y, Kankala RK, Wang SB, Chen AZ. Multi-Organs-on-Chips: Towards Long-Term Biomedical Investigations. Molecules 2019; 24:E675. [PMID: 30769788 PMCID: PMC6412790 DOI: 10.3390/molecules24040675] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/06/2019] [Accepted: 02/11/2019] [Indexed: 12/12/2022] Open
Abstract
With advantageous features such as minimizing the cost, time, and sample size requirements, organ-on-a-chip (OOC) systems have garnered enormous interest from researchers for their ability for real-time monitoring of physical parameters by mimicking the in vivo microenvironment and the precise responses of xenobiotics, i.e., drug efficacy and toxicity over conventional two-dimensional (2D) and three-dimensional (3D) cell cultures, as well as animal models. Recent advancements of OOC systems have evidenced the fabrication of 'multi-organ-on-chip' (MOC) models, which connect separated organ chambers together to resemble an ideal pharmacokinetic and pharmacodynamic (PK-PD) model for monitoring the complex interactions between multiple organs and the resultant dynamic responses of multiple organs to pharmaceutical compounds. Numerous varieties of MOC systems have been proposed, mainly focusing on the construction of these multi-organ models, while there are only few studies on how to realize continual, automated, and stable testing, which still remains a significant challenge in the development process of MOCs. Herein, this review emphasizes the recent advancements in realizing long-term testing of MOCs to promote their capability for real-time monitoring of multi-organ interactions and chronic cellular reactions more accurately and steadily over the available chip models. Efforts in this field are still ongoing for better performance in the assessment of preclinical attributes for a new chemical entity. Further, we give a brief overview on the various biomedical applications of long-term testing in MOCs, including several proposed applications and their potential utilization in the future. Finally, we summarize with perspectives.
Collapse
Affiliation(s)
- Yi Zhao
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China.
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, China.
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China.
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, China.
| | - Shi-Bin Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China.
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, China.
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China.
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, China.
| |
Collapse
|
66
|
Shehzad A, Ravinayagam V, AlRumaih H, Aljafary M, Almohazey D, Almofty S, Al-Rashid NA, Al-Suhaimi EA. Application of Three-dimensional (3D) Tumor Cell Culture Systems and Mechanism of Drug Resistance. Curr Pharm Des 2019; 25:3599-3607. [PMID: 31612821 DOI: 10.2174/1381612825666191014163923] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 09/29/2019] [Indexed: 12/12/2022]
Abstract
The in-vitro experimental model for the development of cancer therapeutics has always been challenging. Recently, the scientific revolution has improved cell culturing techniques by applying three dimensional (3D) culture system, which provides a similar physiologically relevant in-vivo model for studying various diseases including cancer. In particular, cancer cells exhibiting in-vivo behavior in a model of 3D cell culture is a more accurate cell culture model to test the effectiveness of anticancer drugs or characterization of cancer cells in comparison with two dimensional (2D) monolayer. This study underpins various factors that cause resistance to anticancer drugs in forms of spheroids in 3D in-vitro cell culture and also outlines key challenges and possible solutions for the future development of these systems.
Collapse
Affiliation(s)
- Adeeb Shehzad
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Vijaya Ravinayagam
- Scientific Research & Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Hamad AlRumaih
- College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Meneerah Aljafary
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Dana Almohazey
- Stem Cell Research Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Sarah Almofty
- Stem Cell Research Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Noor A Al-Rashid
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Ebtesam A Al-Suhaimi
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
- Stem Cell Research Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
67
|
Thermally-triggered fabrication of cell sheets for tissue engineering and regenerative medicine. Adv Drug Deliv Rev 2019; 138:276-292. [PMID: 30639258 DOI: 10.1016/j.addr.2019.01.004] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 12/24/2018] [Accepted: 01/04/2019] [Indexed: 12/13/2022]
Abstract
Cell transplantation is a promising approach for promoting tissue regeneration in the treatment of damaged tissues or organs. Although cells have conventionally been delivered by direct injection to damaged tissues, cell injection has limited efficiency to deliver therapeutic cells to the target sites. Progress in tissue engineering has moved scaffold-based cell/tissue delivery into the mainstream of tissue regeneration. A variety of scaffolds can be fabricated from natural or synthetic polymers to provide the appropriate culture conditions for cell growth and achieve in-vitro tissue formation. Tissue engineering has now become the primary approach for cell-based therapies. However, there are still serious limitations, particularly for engineering of cell-dense tissues. "Cell sheet engineering" is a scaffold-free tissue technology that holds even greater promise in the field of tissue engineering and regenerative medicine. Thermoresponsive poly(N-isopropylacrylamide)-grafted surfaces allow the fabrication of a tissue-like cell monolayer, a "cell sheet", and efficiently delivers this cell-dense tissue to damaged sites without the use of scaffolds. At present, this unique approach has been applied to human clinical studies in regenerative medicine. Furthermore, this thermally triggered cell manipulation system allows us to produce various types of 3D tissue models not only for regenerative medicine but also for tissue modeling, which can be used for drug discovery. Here, new cell sheet-based technologies are described including vascularization for scaled-up 3D tissue constructs, induced pluripotent stem (iPS) cell technology for human cell sheet fabrication and microfabrication for arranging tissue microstructures, all of which are expected to produce more complex tissues based on cell sheet tissue engineering.
Collapse
|
68
|
Wang W, Zhuang X, Sun H, Dong L, Liu Z, Du G, Ye L, Tian J. Pharmacokinetics of S-EPA, and its inhibition on indoleamine 2,3-dioxgenase: a case of sulfur-substitution affecting distributions in blood cells. Xenobiotica 2018; 49:1338-1343. [PMID: 30507339 DOI: 10.1080/00498254.2018.1546024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
1. S-EPA is a sulfur-substitution analog of epacadostat (EPA), an effective small molecule indoleamine 2,3-dioxgenase1 (IDO) inhibitor. By in vitro and in vivo experiments, pharmacokinetic differences of two closely related analogs, S-EPA and EPA was investigated in this study. 2. Liver microsomes clearance experiments showed S-EPA had comparable metabolic stability with EPA in rat and human liver microsomes. The whole blood distribution experiments showed the distribution ratio of S-EPA in blood cells to plasma in mice, rats, dogs and monkey was 1.2, 4.8, 2.2 and 40.6, respectively. While the distribution ratio of EPA ranged from 0.94 to 1.30 in mice, rats, dogs and was 3.1 in monkeys. 3. The pharmacokinetic study in rats showed the exposure (AUClast) of S-EPA in plasma and blood cells was 1.7-fold and 3.9-fold higher than that of EPA, respectively. Moreover, the exposure ratio of S-EPA in blood cells to plasma was 3.7, while the ratio of EPA was 1.6. 4. In CT26 tumor bearing mice, the IDO inhibition of S-EPA and EPA on plasma or tumor kynurenine was generally consistent. And the inhibition ratio could reach at more than 50% at 3 h after single dose, at least lasting up to 8 h.
Collapse
Affiliation(s)
- Wenyan Wang
- a Ministry of Education, School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong Yantai University , Yantai , PR China
| | - Xuhui Zhuang
- a Ministry of Education, School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong Yantai University , Yantai , PR China
| | - Heyuan Sun
- a Ministry of Education, School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong Yantai University , Yantai , PR China
| | - Lina Dong
- a Ministry of Education, School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong Yantai University , Yantai , PR China
| | - Zongliang Liu
- a Ministry of Education, School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong Yantai University , Yantai , PR China
| | - Guangying Du
- a Ministry of Education, School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong Yantai University , Yantai , PR China
| | - Liang Ye
- b Institute of Toxcicology, Binzhou Medical University , Yantai , PR China
| | - Jingwei Tian
- a Ministry of Education, School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong Yantai University , Yantai , PR China
| |
Collapse
|
69
|
Shanti A, Teo J, Stefanini C. In Vitro Immune Organs-on-Chip for Drug Development: A Review. Pharmaceutics 2018; 10:E278. [PMID: 30558264 PMCID: PMC6320867 DOI: 10.3390/pharmaceutics10040278] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 11/12/2018] [Accepted: 11/21/2018] [Indexed: 12/15/2022] Open
Abstract
The current drug development practice lacks reliable and sensitive techniques to evaluate the immunotoxicity of drug candidates, i.e., their effect on the human immune system. This, in part, has resulted in a high attrition rate for novel drugs candidates. Organ-on-chip devices have emerged as key tools that permit the study of human physiology in controlled in vivo simulating environments. Furthermore, there has been a growing interest in developing the so called "body-on-chip" devices to better predict the systemic effects of drug candidates. This review describes existing biomimetic immune organs-on-chip, highlights their physiological relevance to drug development and discovery and emphasizes the need for developing comprehensive immune system-on-chip models. Such immune models can enhance the performance of novel drug candidates during clinical trials and contribute to reducing the high attrition rate as well as the high cost associated with drug development.
Collapse
Affiliation(s)
- Aya Shanti
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, P.O. Box 127788, UAE.
| | - Jeremy Teo
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi, P.O. Box 129188, UAE.
- Department of Mechanical and Aerospace Engineering, New York University, New York City, P.O. Box 903, NY 10276-0903, USA.
| | - Cesare Stefanini
- Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, P.O. Box 127788, UAE.
| |
Collapse
|
70
|
Qiu Y, Ning D, Zhang P, Curly S, Qiao Y, Ma L, Su M. Three-dimensional microtissues as an in vitro model for personalized radiation therapy. Analyst 2018; 142:3605-3612. [PMID: 28812074 DOI: 10.1039/c7an00794a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This paper describes the use of 3D microtissues as an intermediate model between the 2D cell culture and the animal model to assess radiation-induced cellular and DNA damage in the context of personalized radiation therapy. An agarose microwell array was used to generate 3D microtissues with controlled size and shape. The microtissues were exposed to X-ray radiation of various doses, and the radiation damage to cells was examined using a variety of techniques with different end points. Damage to cell membranes and reduction in metabolic activity were examined with the MTT assay and dye inclusion assay. DNA damage was tested with the micronucleus assay, γ-H2AX immunostaining, and HaloChip assay. 3D microtissues exposed to X-rays are smaller compared to unexposed ones in extended cultures, indicating that X-ray radiation can retard the growth of cells in 3D microtissues, where cells at the outer shells of microtissues can prevent further damage to those inside.
Collapse
Affiliation(s)
- Yuting Qiu
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
| | | | | | | | | | | | | |
Collapse
|
71
|
Park SM, Lee SJ, Lim J, Kim BC, Han SJ, Kim DS. Versatile Fabrication of Size- and Shape-Controllable Nanofibrous Concave Microwells for Cell Spheroid Formation. ACS APPLIED MATERIALS & INTERFACES 2018; 10:37878-37885. [PMID: 30360112 DOI: 10.1021/acsami.8b15821] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Although the microfabrication techniques for microwells enabled to guide physiologically relevant three-dimensional cell spheroid formation, there have been substantial interests to more closely mimic nano/microtopographies of in vivo cellular microenvironment. Here, we developed a versatile fabrication process for nanofibrous concave microwells (NCMs) with a controllable size and shape. The key to the fabrication process was the use of an array of hemispherical convex electrolyte solution drops as the grounded collector for electrospinning, which greatly improved the degree of freedom of the size, shape, and curvature of an NCM. A polymer substrate with through-holes was prepared for the electrolyte solution to come out through the hole and to naturally form a convex shape because of surface tension. Subsequent electrolyte-assisted electrospinning process enabled to achieve various arrays of NCMs of triangular, rectangular, and circular shapes with sizes ranging from 1000 μm down to 250 μm. As one example of biomedical applications, the formation of human hepatoma cell line (HepG2) spheroids was demonstrated on the NCMs. The results indicated that the NCM enabled uniform, size-controllable spheroid formation of HepG2 cells, resulting in 1.5 times higher secretion of albumin from HepG2 cells on the NCM on day 14 compared with those on a nanofibrous flat microwell as a control.
Collapse
Affiliation(s)
- Sang Min Park
- Department of Mechanical Engineering , Pohang University of Science and Technology (POSTECH) , 77 Cheongam-ro , Pohang , Gyeongbuk 37673 , South Korea
| | - Seong Jin Lee
- Department of Mechanical Engineering , Pohang University of Science and Technology (POSTECH) , 77 Cheongam-ro , Pohang , Gyeongbuk 37673 , South Korea
| | - Jiwon Lim
- Department of Mechanical Engineering , Pohang University of Science and Technology (POSTECH) , 77 Cheongam-ro , Pohang , Gyeongbuk 37673 , South Korea
| | - Bum Chang Kim
- Department of Mechanical Engineering , Pohang University of Science and Technology (POSTECH) , 77 Cheongam-ro , Pohang , Gyeongbuk 37673 , South Korea
| | - Seon Jin Han
- Department of Mechanical Engineering , Pohang University of Science and Technology (POSTECH) , 77 Cheongam-ro , Pohang , Gyeongbuk 37673 , South Korea
| | - Dong Sung Kim
- Department of Mechanical Engineering , Pohang University of Science and Technology (POSTECH) , 77 Cheongam-ro , Pohang , Gyeongbuk 37673 , South Korea
| |
Collapse
|
72
|
Soares RM, Siqueira NM, Prabhakaram MP, Ramakrishna S. Electrospinning and electrospray of bio-based and natural polymers for biomaterials development. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 92:969-982. [DOI: 10.1016/j.msec.2018.08.004] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 07/12/2018] [Accepted: 08/02/2018] [Indexed: 01/13/2023]
|
73
|
Uhl C, Shi W, Liu Y. Organ-on-Chip Devices Toward Applications in Drug Development and Screening. J Med Device 2018. [DOI: 10.1115/1.4040272] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
As a necessary pathway to man-made organs, organ-on-chips (OOC), which simulate the activities, mechanics, and physiological responses of real organs, have attracted plenty of attention over the past decade. As the maturity of three-dimensional (3D) cell-culture models and microfluidics advances, the study of OOCs has made significant progress. This review article provides a comprehensive overview and classification of OOC microfluidics. Specifically, the review focuses on OOC systems capable of being used in preclinical drug screening and development. Additionally, the review highlights the strengths and weaknesses of each OOC system toward the goal of improved drug development and screening. The various OOC systems investigated throughout the review include, blood vessel, lung, liver, and tumor systems and the potential benefits, which each provides to the growing challenge of high-throughput drug screening. Published OOC systems have been reviewed over the past decade (2007–2018) with focus given mainly to more recent advances and improvements within each organ system. Each OOC system has been reviewed on how closely and realistically it is able to mimic its physiological counterpart, the degree of information provided by the system toward the ultimate goal of drug development and screening, how easily each system would be able to transition to large scale high-throughput drug screening, and what further improvements to each system would help to improve the functionality, realistic nature of the platform, and throughput capacity. Finally, a summary is provided of where the broad field of OOCs appears to be headed in the near future along with suggestions on where future efforts should be focused for optimized performance of OOC systems in general.
Collapse
Affiliation(s)
- Christopher Uhl
- Department of Bioengineering, Lehigh University, Bethlehem, PA 18015
| | - Wentao Shi
- Department of Bioengineering, Lehigh University, Bethlehem, PA 18015
| | - Yaling Liu
- Department of Bioengineering, Lehigh University, Bethlehem, PA 18015
- Department of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, PA 18015 e-mail:
| |
Collapse
|
74
|
Takahashi H, Shimizu T, Okano T. Engineered Human Contractile Myofiber Sheets as a Platform for Studies of Skeletal Muscle Physiology. Sci Rep 2018; 8:13932. [PMID: 30224737 PMCID: PMC6141563 DOI: 10.1038/s41598-018-32163-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/03/2018] [Indexed: 12/25/2022] Open
Abstract
Skeletal muscle physiology and the mechanisms of muscle diseases can be effectively studied by an in-vitro tissue model produced by muscle tissue engineering. Engineered human cell-based tissues are required more than ever because of the advantages they bring as tissue models in research studies. This study reports on a production method of a human skeletal myofiber sheet that demonstrates biomimetic properties including the aligned structure of myofibers, basement membrane-like structure of the extracellular matrix, and unidirectional contractile ability. The contractile ability and drug responsibility shown in this study indicate that this engineered muscle tissue has potential as a human cell-based tissue model for clinically relevant in-vitro studies in muscle physiology and drug discovery. Moreover, this engineered tissue can be used to better understand the relationships between mechanical stress and myogenesis, including muscle growth and regeneration. In this study, periodic exercise induced by continuous electrical pulse stimulation enhanced the contractile ability of the engineered myofibers and the secretion of interleukin-6 (IL-6) and vascular endothelial growth factor (VEGF) from the exercising myofibers. Since the physiology of skeletal muscle is directly related to mechanical stress, these features point to application as a tissue model and platform for future biological studies of skeletal muscle including muscle metabolism, muscle atrophy and muscle regeneration.
Collapse
Affiliation(s)
- Hironobu Takahashi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan.
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
75
|
Abstract
To perform the drug screening, planar cultured cell models are commonly applied to test efficacy and toxicity of drugs. However, planar cultured cells are different from the human 3D organs or tissues in vivo. To simulate the human 3D organs or tissues, 3D spheroids are developed by culturing a small aggregate of cells which reside around the extracellular matrix and interact with other cells in liquid media. Here we apply lung carcinoma cell lines to engineer the 3D lung cancer spheroid-based biosensor using the interdigitated electrodes for drug efficacy evaluation. The results show 3D spheroid had higher drug resistance than the planar cell model. The anticarcinogen inhibition on different 3D lung cancer spheroid models (A549, H1299, H460) can be quantitatively evaluated by electric impedance sensing. Besides, we delivered combination of anticarcinogens treatments to A549 spheroids which is commonly used in clinic treatment, and found the synergistic effect of cisplatin plus etoposide had higher drug response. To simultaneously test the drug efficacy and side effects on multi-organ model with circulatory system, a connected multiwell interdigitated electrode arraywas applied to culture different organoid spheroids. Overall, the organization of 3D cancer spheroids-based biosensor, which has higher predictive value for drug discovery and personalized medicine screening, is expected to be well applied in the area of pharmacy and clinical medicine.
Collapse
|
76
|
Macdougall LJ, Wiley KL, Kloxin AM, Dove AP. Design of synthetic extracellular matrices for probing breast cancer cell growth using robust cyctocompatible nucleophilic thiol-yne addition chemistry. Biomaterials 2018; 178:435-447. [PMID: 29773227 PMCID: PMC6699181 DOI: 10.1016/j.biomaterials.2018.04.046] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/27/2018] [Accepted: 04/23/2018] [Indexed: 12/16/2022]
Abstract
Controlled, three-dimensional (3D) cell culture systems are of growing interest for both tissue regeneration and disease, including cancer, enabling hypothesis testing about the effects of microenvironment cues on a variety of cellular processes, including aspects of disease progression. In this work, we encapsulate and culture in three dimensions different cancer cell lines in a synthetic extracellular matrix (ECM), using mild and efficient chemistry. Specifically, harnessing the nucleophilic addition of thiols to activated alkynes, we have created hydrogel-based materials with multifunctional poly(ethylene glycol) (PEG) and select biomimetic peptides. These materials have definable, controlled mechanical properties (G' = 4-10 kPa) and enable facile incorporation of pendant peptides for cell adhesion, relevant for mimicking soft tissues, where polymer architecture allows tuning of matrix degradation. These matrices rapidly formed in the presence of sensitive breast cancer cells (MCF-7) for successful encapsulation with high cell viability, greatly improved relative to that observed with the more widely used radically-initiated thiol-ene crosslinking chemistry. Furthermore, controlled matrix degradation by both bulk and local mechanisms, ester hydrolysis of the polymer network and cell-driven enzymatic hydrolysis of cell-degradable peptide, allowed cell proliferation and the formation of cell clusters within these thiol-yne hydrogels. These studies demonstrate the importance of chemistry in ECM mimics and the potential thiol-yne chemistry has as a crosslinking reaction for the encapsulation and culture of cells, including those sensitive to radical crosslinking pathways.
Collapse
Affiliation(s)
- Laura J Macdougall
- Department of Chemistry, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Katherine L Wiley
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| | - April M Kloxin
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA; Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | - Andrew P Dove
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
77
|
Jensen G, Morrill C, Huang Y. 3D tissue engineering, an emerging technique for pharmaceutical research. Acta Pharm Sin B 2018; 8:756-766. [PMID: 30258764 PMCID: PMC6148716 DOI: 10.1016/j.apsb.2018.03.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 03/09/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022] Open
Abstract
Tissue engineering and the tissue engineering model have shown promise in improving methods of drug delivery, drug action, and drug discovery in pharmaceutical research for the attenuation of the central nervous system inflammatory response. Such inflammation contributes to the lack of regenerative ability of neural cells, as well as the temporary and permanent loss of function associated with neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and traumatic brain injury. This review is focused specifically on the recent advances in the tissue engineering model made by altering scaffold biophysical and biochemical properties for use in the treatment of neurodegenerative diseases. A portion of this article will also be spent on the review of recent progress made in extracellular matrix decellularization as a new and innovative scaffold for disease treatment.
Collapse
Affiliation(s)
| | | | - Yu Huang
- Department of Biological Engineering, Utah State University, Logan, UT, 84322, USA
| |
Collapse
|
78
|
Maleiner B, Tomasch J, Heher P, Spadiut O, Rünzler D, Fuchs C. The Importance of Biophysical and Biochemical Stimuli in Dynamic Skeletal Muscle Models. Front Physiol 2018; 9:1130. [PMID: 30246791 PMCID: PMC6113794 DOI: 10.3389/fphys.2018.01130] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 07/30/2018] [Indexed: 12/31/2022] Open
Abstract
Classical approaches to engineer skeletal muscle tissue based on current regenerative and surgical procedures still do not meet the desired outcome for patient applications. Besides the evident need to create functional skeletal muscle tissue for the repair of volumetric muscle defects, there is also growing demand for platforms to study muscle-related diseases, such as muscular dystrophies or sarcopenia. Currently, numerous studies exist that have employed a variety of biomaterials, cell types and strategies for maturation of skeletal muscle tissue in 2D and 3D environments. However, researchers are just at the beginning of understanding the impact of different culture settings and their biochemical (growth factors and chemical changes) and biophysical cues (mechanical properties) on myogenesis. With this review we intend to emphasize the need for new in vitro skeletal muscle (disease) models to better recapitulate important structural and functional aspects of muscle development. We highlight the importance of choosing appropriate system components, e.g., cell and biomaterial type, structural and mechanical matrix properties or culture format, and how understanding their interplay will enable researchers to create optimized platforms to investigate myogenesis in healthy and diseased tissue. Thus, we aim to deliver guidelines for experimental designs to allow estimation of the potential influence of the selected skeletal muscle tissue engineering setup on the myogenic outcome prior to their implementation. Moreover, we offer a workflow to facilitate identifying and selecting different analytical tools to demonstrate the successful creation of functional skeletal muscle tissue. Ultimately, a refinement of existing strategies will lead to further progression in understanding important aspects of muscle diseases, muscle aging and muscle regeneration to improve quality of life of patients and enable the establishment of new treatment options.
Collapse
Affiliation(s)
- Babette Maleiner
- Department of Biochemical Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria.,The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Janine Tomasch
- Department of Biochemical Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria.,The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Philipp Heher
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Ludwig Boltzmann Institute for Experimental and Clinical Traumatology/AUVA Research Center, Vienna, Austria.,Trauma Care Consult GmbH, Vienna, Austria
| | - Oliver Spadiut
- Institute of Chemical Engineering, Vienna University of Technology, Vienna, Austria
| | - Dominik Rünzler
- Department of Biochemical Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria.,The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Christiane Fuchs
- Department of Biochemical Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria.,The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
79
|
Schöneberg J, De Lorenzi F, Theek B, Blaeser A, Rommel D, Kuehne AJC, Kießling F, Fischer H. Engineering biofunctional in vitro vessel models using a multilayer bioprinting technique. Sci Rep 2018; 8:10430. [PMID: 29992981 PMCID: PMC6041340 DOI: 10.1038/s41598-018-28715-0] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 06/27/2018] [Indexed: 02/07/2023] Open
Abstract
Recent advances in the field of bioprinting have led to the development of perfusable complex structures. However, most of the existing printed vascular channels lack the composition or key structural and physiological features of natural blood vessels or they make use of more easily printable but less biocompatible hydrogels. Here, we use a drop-on-demand bioprinting technique to generate in vitro blood vessel models, consisting of a continuous endothelium imitating the tunica intima, an elastic smooth muscle cell layer mimicking the tunica media, and a surrounding fibrous and collagenous matrix of fibroblasts mimicking the tunica adventitia. These vessel models with a wall thickness of up to 425 µm and a diameter of about 1 mm were dynamically cultivated in fluidic bioreactors for up to three weeks under physiological flow conditions. High cell viability (>83%) after printing and the expression of VE-Cadherin, smooth muscle actin, and collagen IV were observed throughout the cultivation period. It can be concluded that the proposed novel technique is suitable to achieve perfusable vessel models with a biofunctional multilayer wall composition. Such structures hold potential for the creation of more physiologically relevant in vitro disease models suitable especially as platforms for the pre-screening of drugs.
Collapse
Affiliation(s)
- Jan Schöneberg
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany
| | - Federica De Lorenzi
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| | - Benjamin Theek
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| | - Andreas Blaeser
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany
| | - Dirk Rommel
- DWI - Leibniz Institute for Interactive Materials, RWTH Aachen University, Aachen, Germany
| | - Alexander J C Kuehne
- DWI - Leibniz Institute for Interactive Materials, RWTH Aachen University, Aachen, Germany
| | - Fabian Kießling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| | - Horst Fischer
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany.
| |
Collapse
|
80
|
Svensson M, Chen P. Human Organotypic Respiratory Models. Curr Top Microbiol Immunol 2018:29-54. [PMID: 29808337 DOI: 10.1007/82_2018_91] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Biomedical research aiming to understand the molecular basis of human lung tissue development, homeostasis and disease, or to develop new therapies for human respiratory diseases, requires models that faithfully recapitulate the human condition. This has stimulated biologists and engineers to develop in vitro organotypic models mimicking human respiratory tissues. In this chapter, we provide examples of different types of model systems ranging from simple unicellular cultures to more complex multicellular systems. The models contain, in varying degree, cell types present in real tissue in combination with different extracellular matrix components that can critically affect cell phenotype and function. We also describe how organotypic respiratory models can be combined with human innate immune cells, to better recapitulate tissue inflammation, a key component in, for example, infectious diseases. These models have the potential to provide new insights into lung physiology, tissue infection and inflammation, disease mechanisms, as well as provide a platform for identification of novel targets and screening of candidate drugs in human lung disorders.
Collapse
Affiliation(s)
- Mattias Svensson
- F59, Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, 141 86, Stockholm, Sweden.
| | - Puran Chen
- F59, Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, 141 86, Stockholm, Sweden
| |
Collapse
|
81
|
Russo C, Lewis EEL, Flint L, Clench MR. Mass Spectrometry Imaging of 3D Tissue Models. Proteomics 2018; 18:e1700462. [PMID: 29687599 DOI: 10.1002/pmic.201700462] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 04/12/2018] [Indexed: 12/15/2022]
Abstract
A 3D cell culture is an artificially created environment in which cells are permitted to grow/interact with their surroundings in all three dimensions. Derived from 3D cell culture, organoids are generally small-scale constructs of cells that are fabricated in the laboratory to serve as 3D representations of in vivo tissues and organs. Due to regulatory, economic and societal issues concerning the use of animals in scientific research, it seems clear that the use of 3D cell culture and organoids in for example early stage studies of drug efficacy and toxicity will increase. The combination of such 3D tissue models with mass spectrometry imaging provides a label-free methodology for the study of drug absorption/penetration, drug efficacy/toxicity, and drug biotransformation. In this article, some of the successes achieved to date and challenges to be overcome before this methodology is more widely adopted are discussed.
Collapse
Affiliation(s)
- Cristina Russo
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, S1 WB Sheffield, UK
| | - Emily E L Lewis
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, S1 WB Sheffield, UK.,Innovenn UK Ltd., National Agri-Food Innovation Campus, Sand Hutton, YO41 1LZ York, UK
| | - Lucy Flint
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, S1 WB Sheffield, UK
| | - Malcolm R Clench
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, S1 WB Sheffield, UK
| |
Collapse
|
82
|
Jones JM, Player DJ, Martin NRW, Capel AJ, Lewis MP, Mudera V. An Assessment of Myotube Morphology, Matrix Deformation, and Myogenic mRNA Expression in Custom-Built and Commercially Available Engineered Muscle Chamber Configurations. Front Physiol 2018; 9:483. [PMID: 29867538 PMCID: PMC5951956 DOI: 10.3389/fphys.2018.00483] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 04/16/2018] [Indexed: 12/28/2022] Open
Abstract
There are several three-dimensional (3D) skeletal muscle (SkM) tissue engineered models reported in the literature. 3D SkM tissue engineering (TE) aims to recapitulate the structure and function of native (in vivo) tissue, within an in vitro environment. This requires the differentiation of myoblasts into aligned multinucleated myotubes surrounded by a biologically representative extracellular matrix (ECM). In the present work, a new commercially available 3D SkM TE culture chamber manufactured from polyether ether ketone (PEEK) that facilitates suitable development of these myotubes is presented. To assess the outcomes of the myotubes within these constructs, morphological, gene expression, and ECM remodeling parameters were compared against a previously published custom-built model. No significant differences were observed in the morphological and gene expression measures between the newly introduced and the established construct configuration, suggesting biological reproducibility irrespective of manufacturing process. However, TE SkM fabricated using the commercially available PEEK chambers displayed reduced variability in both construct attachment and matrix deformation, likely due to increased reproducibility within the manufacturing process. The mechanical differences between systems may also have contributed to such differences, however, investigation of these variables was beyond the scope of the investigation. Though more expensive than the custom-built models, these PEEK chambers are also suitable for multiple use after autoclaving. As such this would support its use over the previously published handmade culture chamber system, particularly when seeking to develop higher-throughput systems or when experimental cost is not a factor.
Collapse
Affiliation(s)
- Julia M Jones
- Division of Surgery and Interventional Science, Institute of Orthopaedics and Musculoskeletal Science, University College London, London, United Kingdom.,School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Darren J Player
- Division of Surgery and Interventional Science, Institute of Orthopaedics and Musculoskeletal Science, University College London, London, United Kingdom.,School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Neil R W Martin
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Andrew J Capel
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Mark P Lewis
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Vivek Mudera
- Division of Surgery and Interventional Science, Institute of Orthopaedics and Musculoskeletal Science, University College London, London, United Kingdom
| |
Collapse
|
83
|
Kundu A, Ausaf T, Rajaraman S. 3D Printing, Ink Casting and Micromachined Lamination (3D PICLμM): A Makerspace Approach to the Fabrication of Biological Microdevices. MICROMACHINES 2018; 9:E85. [PMID: 30393360 PMCID: PMC6187583 DOI: 10.3390/mi9020085] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/07/2018] [Accepted: 02/11/2018] [Indexed: 11/17/2022]
Abstract
We present a novel benchtop-based microfabrication technology: 3D printing, ink casting, micromachined lamination (3D PICLμM) for rapid prototyping of lab-on-a-chip (LOC) and biological devices. The technology uses cost-effective, makerspace-type microfabrication processes, all of which are ideally suited for low resource settings, and utilizing a combination of these processes, we have demonstrated the following devices: (i) 2D microelectrode array (MEA) targeted at in vitro neural and cardiac electrophysiology, (ii) microneedle array targeted at drug delivery through a transdermal route and (iii) multi-layer microfluidic chip targeted at multiplexed assays for in vitro applications. The 3D printing process has been optimized for printing angle, temperature of the curing process and solvent polishing to address various biofunctional considerations of the three demonstrated devices. We have depicted that the 3D PICLμM process has the capability to fabricate 30 μm sized MEAs (average 1 kHz impedance of 140 kΩ with a double layer capacitance of 3 μF), robust and reliable microneedles having 30 μm radius of curvature and ~40 N mechanical fracture strength and microfluidic devices having 150 μm wide channels and 400 μm fluidic vias capable of fluid mixing and transmitted light microparticle visualization. We believe our 3D PICLμM is ideally suited for applications in areas such as electrophysiology, drug delivery, disease in a dish, organ on a chip, environmental monitoring, agricultural therapeutic delivery and genomic testing.
Collapse
Affiliation(s)
- Avra Kundu
- NanoScience Technology Center (NSTC), University of Central Florida, Orlando, FL 32826, USA.
| | - Tariq Ausaf
- NanoScience Technology Center (NSTC), University of Central Florida, Orlando, FL 32826, USA.
- Department of Electrical & Computer Engineering, University of Central Florida, Orlando, FL 32826, USA.
| | - Swaminathan Rajaraman
- NanoScience Technology Center (NSTC), University of Central Florida, Orlando, FL 32826, USA.
- Department of Electrical & Computer Engineering, University of Central Florida, Orlando, FL 32826, USA.
- Bridging the Innovation Development Gap (BRIDG), Neo City, FL 34744, USA.
- Department of Material Science & Engineering, University of Central Florida, Orlando, FL 32826, USA.
| |
Collapse
|
84
|
Turker E, Arslan-Yildiz A. Recent Advances in Magnetic Levitation: A Biological Approach from Diagnostics to Tissue Engineering. ACS Biomater Sci Eng 2018; 4:787-799. [DOI: 10.1021/acsbiomaterials.7b00700] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Esra Turker
- Department of Bioengineering, Izmir Institute of Technology (IZTECH), 35430 Izmir, Turkey
| | - Ahu Arslan-Yildiz
- Department of Bioengineering, Izmir Institute of Technology (IZTECH), 35430 Izmir, Turkey
| |
Collapse
|
85
|
Pridgeon CS, Schlott C, Wong MW, Heringa MB, Heckel T, Leedale J, Launay L, Gryshkova V, Przyborski S, Bearon RN, Wilkinson EL, Ansari T, Greenman J, Hendriks DFG, Gibbs S, Sidaway J, Sison-Young RL, Walker P, Cross MJ, Park BK, Goldring CEP. Innovative organotypic in vitro models for safety assessment: aligning with regulatory requirements and understanding models of the heart, skin, and liver as paradigms. Arch Toxicol 2018; 92:557-569. [PMID: 29362863 PMCID: PMC5818581 DOI: 10.1007/s00204-018-2152-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 11/27/2017] [Indexed: 01/02/2023]
Abstract
The development of improved, innovative models for the detection of toxicity of drugs, chemicals, or chemicals in cosmetics is crucial to efficiently bring new products safely to market in a cost-effective and timely manner. In addition, improvement in models to detect toxicity may reduce the incidence of unexpected post-marketing toxicity and reduce or eliminate the need for animal testing. The safety of novel products of the pharmaceutical, chemical, or cosmetics industry must be assured; therefore, toxicological properties need to be assessed. Accepted methods for gathering the information required by law for approval of substances are often animal methods. To reduce, refine, and replace animal testing, innovative organotypic in vitro models have emerged. Such models appear at different levels of complexity ranging from simpler, self-organized three-dimensional (3D) cell cultures up to more advanced scaffold-based co-cultures consisting of multiple cell types. This review provides an overview of recent developments in the field of toxicity testing with in vitro models for three major organ types: heart, skin, and liver. This review also examines regulatory aspects of such models in Europe and the UK, and summarizes best practices to facilitate the acceptance and appropriate use of advanced in vitro models.
Collapse
Affiliation(s)
- Chris S Pridgeon
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK
| | - Constanze Schlott
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK
| | - Min Wei Wong
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK
| | - Minne B Heringa
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Tobias Heckel
- Dr. Johannes Heidenhain GmbH, Dr.-Johannes-Heidenhain-Straße 5, 83301, Traunreut, Germany
| | - Joe Leedale
- Department of Mathematical Sciences, University of Liverpool, Liverpool, L69 7ZL, UK
| | | | - Vitalina Gryshkova
- Investigative Toxicology, Department of Non-Clinical Development, UCB Biopharma SPRL, 1420, Braine L'Alleud, Belgium
| | | | - Rachel N Bearon
- Department of Mathematical Sciences, University of Liverpool, Liverpool, L69 7ZL, UK
| | - Emma L Wilkinson
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK
| | - Tahera Ansari
- Northwick Park Institute for Medical Research, Northwick Park and St Mark's Hospital, Middlesex, HA1 3UJ, UK
| | - John Greenman
- School of Life Sciences, University of Hull, Hull, HU6 7RX, UK
| | - Delilah F G Hendriks
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Sue Gibbs
- Department of Dermatology, VU University Medical Center, Amsterdam, The Netherlands
- Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, The Netherlands
| | | | - Rowena L Sison-Young
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK
| | - Paul Walker
- Cyprotex Discovery Ltd, Cheshire, SK10 4TG, UK
| | - Mike J Cross
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK
| | - B Kevin Park
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK
| | - Chris E P Goldring
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK.
| |
Collapse
|
86
|
Osaki T, Shin Y, Sivathanu V, Campisi M, Kamm RD. In Vitro Microfluidic Models for Neurodegenerative Disorders. Adv Healthc Mater 2018; 7. [PMID: 28881425 DOI: 10.1002/adhm.201700489] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 07/18/2017] [Indexed: 01/09/2023]
Abstract
Microfluidic devices enable novel means of emulating neurodegenerative disease pathophysiology in vitro. These organ-on-a-chip systems can potentially reduce animal testing and substitute (or augment) simple 2D culture systems. Reconstituting critical features of neurodegenerative diseases in a biomimetic system using microfluidics can thereby accelerate drug discovery and improve our understanding of the mechanisms of several currently incurable diseases. This review describes latest advances in modeling neurodegenerative diseases in the central nervous system and the peripheral nervous system. First, this study summarizes fundamental advantages of microfluidic devices in the creation of compartmentalized cell culture microenvironments for the co-culture of neurons, glial cells, endothelial cells, and skeletal muscle cells and in their recapitulation of spatiotemporal chemical gradients and mechanical microenvironments. Then, this reviews neurodegenerative-disease-on-a-chip models focusing on Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Finally, this study discusses about current drawbacks of these models and strategies that may overcome them. These organ-on-chip technologies can be useful to be the first line of testing line in drug development and toxicology studies, which can contribute significantly to minimize the phase of animal testing steps.
Collapse
Affiliation(s)
- Tatsuya Osaki
- Department of Mechanical EngineeringMassachusetts Institutes of Technology 500 Technology Square MIT Building, Room NE47‐321 Cambridge MA 02139 USA
| | - Yoojin Shin
- Department of Mechanical EngineeringMassachusetts Institutes of Technology 500 Technology Square MIT Building, Room NE47‐321 Cambridge MA 02139 USA
| | - Vivek Sivathanu
- Department of Mechanical EngineeringMassachusetts Institutes of Technology 500 Technology Square MIT Building, Room NE47‐321 Cambridge MA 02139 USA
| | - Marco Campisi
- Department of Mechanical and Aerospace EngineeringPolitecnico di Torino Corso Duca degli Abruzzi 24 10129 Torino Italy
| | - Roger D. Kamm
- Department of Mechanical EngineeringMassachusetts Institutes of Technology 500 Technology Square MIT Building, Room NE47‐321 Cambridge MA 02139 USA
- Department of Biological EngineeringMassachusetts Institutes of Technology 500 Technology Square, MIT Building, Room NE47‐321 Cambridge MA 02139 USA
| |
Collapse
|
87
|
Türker E, Demirçak N, Arslan-Yildiz A. Scaffold-free three-dimensional cell culturing using magnetic levitation. Biomater Sci 2018; 6:1745-1753. [DOI: 10.1039/c8bm00122g] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Magnetic levitation platform ensures a scaffold-free 3D cell culture formation by utilizing Gadolinium(iii) chelates, which provide paramagnetic environment for levitation; therefore, the cells are assembled into complex 3D structures.
Collapse
Affiliation(s)
- Esra Türker
- Department of Bioengineering
- Izmir Institute of Technology (IZTECH)
- Izmir
- Turkey
| | - Nida Demirçak
- Department of Bioengineering
- Izmir Institute of Technology (IZTECH)
- Izmir
- Turkey
| | - Ahu Arslan-Yildiz
- Department of Bioengineering
- Izmir Institute of Technology (IZTECH)
- Izmir
- Turkey
| |
Collapse
|
88
|
Methods to Evaluate Cell Growth, Viability, and Response to Treatment in a Tissue Engineered Breast Cancer Model. Sci Rep 2017; 7:14167. [PMID: 29074857 PMCID: PMC5658356 DOI: 10.1038/s41598-017-14326-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/09/2017] [Indexed: 01/01/2023] Open
Abstract
The use of in vitro, engineered surrogates in the field of cancer research is of interest for studies involving mechanisms of growth and metastasis, and response to therapeutic intervention. While biomimetic surrogates better model human disease, their complex composition and dimensionality make them challenging to evaluate in a real-time manner. This feature has hindered the broad implementation of these models, particularly in drug discovery. Herein, several methods and approaches for the real-time, non-invasive analysis of cell growth and response to treatment in tissue-engineered, three-dimensional models of breast cancer are presented. The tissue-engineered surrogates used to demonstrate these methods consist of breast cancer epithelial cells and fibroblasts within a three dimensional volume of extracellular matrix and are continuously perfused with nutrients via a bioreactor system. Growth of the surrogates over time was measured using optical in vivo (IVIS) imaging. Morphologic changes in specific cell populations were evaluated by multi-photon confocal microscopy. Response of the surrogates to treatment with paclitaxel was measured by optical imaging and by analysis of lactate dehydrogenase and caspase-cleaved cytokeratin 18 in the perfused medium. Each method described can be repeatedly performed during culture, allowing for real-time, longitudinal analysis of cell populations within engineered tumor models.
Collapse
|
89
|
Tsamandouras N, Chen WLK, Edington CD, Stokes CL, Griffith LG, Cirit M. Integrated Gut and Liver Microphysiological Systems for Quantitative In Vitro Pharmacokinetic Studies. AAPS JOURNAL 2017; 19:1499-1512. [PMID: 28752430 DOI: 10.1208/s12248-017-0122-4] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/08/2017] [Indexed: 01/05/2023]
Abstract
Investigation of the pharmacokinetics (PK) of a compound is of significant importance during the early stages of drug development, and therefore several in vitro systems are routinely employed for this purpose. However, the need for more physiologically realistic in vitro models has recently fueled the emerging field of tissue-engineered 3D cultures, also referred to as organs-on-chips, or microphysiological systems (MPSs). We have developed a novel fluidic platform that interconnects multiple MPSs, allowing PK studies in multi-organ in vitro systems along with the collection of high-content quantitative data. This platform was employed here to integrate a gut and a liver MPS together in continuous communication, and investigate simultaneously different PK processes taking place after oral drug administration in humans (e.g., intestinal permeability, hepatic metabolism). Measurement of tissue-specific phenotypic metrics indicated that gut and liver MPSs can be fluidically coupled with circulating common medium without compromising their functionality. The PK of diclofenac and hydrocortisone was investigated under different experimental perturbations, and results illustrate the robustness of this integrated system for quantitative PK studies. Mechanistic model-based analysis of the obtained data allowed the derivation of the intrinsic parameters (e.g., permeability, metabolic clearance) associated with the PK processes taking place in each MPS. Although these processes were not substantially affected by the gut-liver interaction, our results indicate that inter-MPS communication can have a modulating effect (hepatic metabolism upregulation). We envision that our integrative approach, which combines multi-cellular tissue models, multi-MPS platforms, and quantitative mechanistic modeling, will have broad applicability in pre-clinical drug development.
Collapse
Affiliation(s)
- Nikolaos Tsamandouras
- Department of Biological Engineering, Massachusetts Institute of Technology, Room 16-429, Building 16, 77 Massachusetts Avenue, Cambridge, Massachusetts, 02139, USA
| | - Wen Li Kelly Chen
- Department of Biological Engineering, Massachusetts Institute of Technology, Room 16-429, Building 16, 77 Massachusetts Avenue, Cambridge, Massachusetts, 02139, USA
| | - Collin D Edington
- Department of Biological Engineering, Massachusetts Institute of Technology, Room 16-429, Building 16, 77 Massachusetts Avenue, Cambridge, Massachusetts, 02139, USA
| | | | - Linda G Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Room 16-429, Building 16, 77 Massachusetts Avenue, Cambridge, Massachusetts, 02139, USA
| | - Murat Cirit
- Department of Biological Engineering, Massachusetts Institute of Technology, Room 16-429, Building 16, 77 Massachusetts Avenue, Cambridge, Massachusetts, 02139, USA.
| |
Collapse
|
90
|
Kuen J, Darowski D, Kluge T, Majety M. Pancreatic cancer cell/fibroblast co-culture induces M2 like macrophages that influence therapeutic response in a 3D model. PLoS One 2017; 12:e0182039. [PMID: 28750018 PMCID: PMC5531481 DOI: 10.1371/journal.pone.0182039] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 07/11/2017] [Indexed: 12/13/2022] Open
Abstract
Pancreatic cancer (PC) remains one of the most challenging solid tumors to treat with a high unmet medical need as patients poorly respond to standard-of-care-therapies. Prominent desmoplastic reaction involving cancer-associated fibroblasts (CAFs) and the immune cells in the tumor microenvironment (TME) and their cross-talk play a significant role in tumor immune escape and progression. To identify the key cellular mechanisms induce an immunosuppressive tumor microenvironment, we established 3D co-culture model with pancreatic cancer cells, CAFs and monocytes. Using this model, we analyzed the influence of tumor cells and fibroblasts on monocytes and their immune suppressive phenotype. Phenotypic characterization of the monocytes after 3D co-culture with tumor/fibroblast spheroids was performed by analyzing the expression of defined cell surface markers and soluble factors. Functionality of these monocytes and their ability to influence T cell phenotype and proliferation was investigated. 3D co-culture of monocytes with pancreatic cancer cells and fibroblasts induced the production of immunosuppressive cytokines which are known to promote polarization of M2 like macrophages and myeloid derived suppressive cells (MDSCs). These co-culture spheroid polarized monocyte derived macrophages (MDMs) were poorly differentiated and had an M2 phenotype. The immunosuppressive function of these co-culture spheroids polarized MDMs was demonstrated by their ability to inhibit CD4+ and CD8+ T cell activation and proliferation in vitro, which we could partially reverse by 3D co-culture spheroid treatment with therapeutic molecules that are able to re-activated spheroid polarized MDMs or block immune suppressive factors such as Arginase-I.
Collapse
Affiliation(s)
- Janina Kuen
- Discovery Oncology, Roche Innovation Center Munich, Roche Pharma Research and Early development, Penzberg, Germany
| | - Diana Darowski
- Discovery Oncology, Roche Innovation Center Munich, Roche Pharma Research and Early development, Penzberg, Germany
| | - Tobias Kluge
- Discovery Oncology, Roche Innovation Center Munich, Roche Pharma Research and Early development, Penzberg, Germany
| | - Meher Majety
- Discovery Oncology, Roche Innovation Center Munich, Roche Pharma Research and Early development, Penzberg, Germany
- * E-mail:
| |
Collapse
|
91
|
Peng W, Datta P, Ayan B, Ozbolat V, Sosnoski D, Ozbolat IT. 3D bioprinting for drug discovery and development in pharmaceutics. Acta Biomater 2017; 57:26-46. [PMID: 28501712 DOI: 10.1016/j.actbio.2017.05.025] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 05/05/2017] [Accepted: 05/09/2017] [Indexed: 02/08/2023]
Abstract
Successful launch of a commercial drug requires significant investment of time and financial resources wherein late-stage failures become a reason for catastrophic failures in drug discovery. This calls for infusing constant innovations in technologies, which can give reliable prediction of efficacy, and more importantly, toxicology of the compound early in the drug discovery process before clinical trials. Though computational advances have resulted in more rationale in silico designing, in vitro experimental studies still require gaining industry confidence and improving in vitro-in vivo correlations. In this quest, due to their ability to mimic the spatial and chemical attributes of native tissues, three-dimensional (3D) tissue models have now proven to provide better results for drug screening compared to traditional two-dimensional (2D) models. However, in vitro fabrication of living tissues has remained a bottleneck in realizing the full potential of 3D models. Recent advances in bioprinting provide a valuable tool to fabricate biomimetic constructs, which can be applied in different stages of drug discovery research. This paper presents the first comprehensive review of bioprinting techniques applied for fabrication of 3D tissue models for pharmaceutical studies. A comparative evaluation of different bioprinting modalities is performed to assess the performance and ability of fabricating 3D tissue models for pharmaceutical use as the critical selection of bioprinting modalities indeed plays a crucial role in efficacy and toxicology testing of drugs and accelerates the drug development cycle. In addition, limitations with current tissue models are discussed thoroughly and future prospects of the role of bioprinting in pharmaceutics are provided to the reader. STATEMENT OF SIGNIFICANCE Present advances in tissue biofabrication have crucial role to play in aiding the pharmaceutical development process achieve its objectives. Advent of three-dimensional (3D) models, in particular, is viewed with immense interest by the community due to their ability to mimic in vivo hierarchical tissue architecture and heterogeneous composition. Successful realization of 3D models will not only provide greater in vitro-in vivo correlation compared to the two-dimensional (2D) models, but also eventually replace pre-clinical animal testing, which has their own shortcomings. Amongst all fabrication techniques, bioprinting- comprising all the different modalities (extrusion-, droplet- and laser-based bioprinting), is emerging as the most viable fabrication technique to create the biomimetic tissue constructs. Notwithstanding the interest in bioprinting by the pharmaceutical development researchers, it can be seen that there is a limited availability of comparative literature which can guide the proper selection of bioprinting processes and associated considerations, such as the bioink selection for a particular pharmaceutical study. Thus, this work emphasizes these aspects of bioprinting and presents them in perspective of differential requirements of different pharmaceutical studies like in vitro predictive toxicology, high-throughput screening, drug delivery and tissue-specific efficacies. Moreover, since bioprinting techniques are mostly applied in regenerative medicine and tissue engineering, a comparative analysis of similarities and differences are also expounded to help researchers make informed decisions based on contemporary literature.
Collapse
|
92
|
Miles JR, Laughlin TD, Sargus-Patino CN, Pannier AK. In vitro porcine blastocyst development in three-dimensional alginate hydrogels. Mol Reprod Dev 2017; 84:775-787. [PMID: 28407335 DOI: 10.1002/mrd.22814] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 04/07/2017] [Indexed: 11/08/2022]
Abstract
Appropriate embryonic and fetal development significantly impact pregnancy success and, therefore, the efficiency of swine production. The pre-implantation period of porcine pregnancy is characterized by several developmental hallmarks, which are initiated by the dramatic morphological change that occurs as pig blastocysts elongate from spherical to filamentous blastocysts. Deficiencies in blastocyst elongation contribute to approximately 20% of embryonic loss, and have a direct influence on within-litter birth weight variation. Although factors identified within the uterine environment may play a role in blastocyst elongation, little is known about the exact mechanisms by which porcine (or other species') blastocysts initiate and progress through the elongation process. This is partly due to the difficulty of replicating elongation in vitro, which would allow for its study in a controlled environment and in real-time. We developed a three dimensional (3-D) culture system using alginate hydrogel matrices that can encapsulate pig blastocysts, maintain viability and blastocyst architecture, and facilitate reproducible morphological changes with corresponding expression of steroidogenic enzyme transcripts and estrogen production, consistent with the initiation of elongation in vivo. This review highlights key aspects of the pre-implantation period of porcine pregnancy and the difficulty of studying blastocyst elongation in vivo or by using in vitro systems. This review also provides insights on the utility of 3-D hydrogels to study blastocyst elongation continuously and in real-time as a complementary and confirmatory approach to in vivo analysis.
Collapse
Affiliation(s)
- Jeremy R Miles
- USDA, U.S. Meat Animal Research Center (USMARC), Clay Center, Nebraska
| | - Taylor D Laughlin
- Department of Biological Systems Engineering, University of Nebraska-Lincoln (UNL), Lincoln, Nebraska
| | - Catherine N Sargus-Patino
- Department of Biological Systems Engineering, University of Nebraska-Lincoln (UNL), Lincoln, Nebraska
| | - Angela K Pannier
- Department of Biological Systems Engineering, University of Nebraska-Lincoln (UNL), Lincoln, Nebraska
| |
Collapse
|
93
|
Watson PMD, Kavanagh E, Allenby G, Vassey M. Bioengineered 3D Glial Cell Culture Systems and Applications for Neurodegeneration and Neuroinflammation. SLAS DISCOVERY 2017; 22:583-601. [PMID: 28346104 DOI: 10.1177/2472555217691450] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Neurodegeneration and neuroinflammation are key features in a range of chronic central nervous system (CNS) diseases such as Alzheimer's and Parkinson's disease, as well as acute conditions like stroke and traumatic brain injury, for which there remains significant unmet clinical need. It is now well recognized that current cell culture methodologies are limited in their ability to recapitulate the cellular environment that is present in vivo, and there is a growing body of evidence to show that three-dimensional (3D) culture systems represent a more physiologically accurate model than traditional two-dimensional (2D) cultures. Given the complexity of the environment from which cells originate, and their various cell-cell and cell-matrix interactions, it is important to develop models that can be controlled and reproducible for drug discovery. 3D cell models have now been developed for almost all CNS cell types, including neurons, astrocytes, microglia, and oligodendrocyte cells. This review will highlight a number of current and emerging techniques for the culture of astrocytes and microglia, glial cell types with a critical role in neurodegenerative and neuroinflammatory conditions. We describe recent advances in glial cell culture using electrospun polymers and hydrogel macromolecules, and highlight how these novel culture environments influence astrocyte and microglial phenotypes in vitro, as compared to traditional 2D systems. These models will be explored to illuminate current trends in the techniques used to create 3D environments for application in research and drug discovery focused on astrocytes and microglial cells.
Collapse
|
94
|
Vernetti L, Gough A, Baetz N, Blutt S, Broughman JR, Brown JA, Foulke-Abel J, Hasan N, In J, Kelly E, Kovbasnjuk O, Repper J, Senutovitch N, Stabb J, Yeung C, Zachos NC, Donowitz M, Estes M, Himmelfarb J, Truskey G, Wikswo JP, Taylor DL. Functional Coupling of Human Microphysiology Systems: Intestine, Liver, Kidney Proximal Tubule, Blood-Brain Barrier and Skeletal Muscle. Sci Rep 2017; 7:42296. [PMID: 28176881 PMCID: PMC5296733 DOI: 10.1038/srep42296] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 12/20/2016] [Indexed: 12/12/2022] Open
Abstract
Organ interactions resulting from drug, metabolite or xenobiotic transport between organs are key components of human metabolism that impact therapeutic action and toxic side effects. Preclinical animal testing often fails to predict adverse outcomes arising from sequential, multi-organ metabolism of drugs and xenobiotics. Human microphysiological systems (MPS) can model these interactions and are predicted to dramatically improve the efficiency of the drug development process. In this study, five human MPS models were evaluated for functional coupling, defined as the determination of organ interactions via an in vivo-like sequential, organ-to-organ transfer of media. MPS models representing the major absorption, metabolism and clearance organs (the jejunum, liver and kidney) were evaluated, along with skeletal muscle and neurovascular models. Three compounds were evaluated for organ-specific processing: terfenadine for pharmacokinetics (PK) and toxicity; trimethylamine (TMA) as a potentially toxic microbiome metabolite; and vitamin D3. We show that the organ-specific processing of these compounds was consistent with clinical data, and discovered that trimethylamine-N-oxide (TMAO) crosses the blood-brain barrier. These studies demonstrate the potential of human MPS for multi-organ toxicity and absorption, distribution, metabolism and excretion (ADME), provide guidance for physically coupling MPS, and offer an approach to coupling MPS with distinct media and perfusion requirements.
Collapse
Affiliation(s)
- Lawrence Vernetti
- University of Pittsburgh, Drug Discovery Institute Pittsburgh, PA, USA.,Department of Computational and Systems Biology, University of Pittsburgh, Baltimore, PA, USA
| | - Albert Gough
- University of Pittsburgh, Drug Discovery Institute Pittsburgh, PA, USA.,Department of Computational and Systems Biology, University of Pittsburgh, Baltimore, PA, USA
| | - Nicholas Baetz
- Departments of Physiology and Medicine, GI Division, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sarah Blutt
- Departments of Molecular Virology and Microbiology and Medicine, Baylor College of Medicine, Houston, TX, USA
| | - James R Broughman
- Departments of Molecular Virology and Microbiology and Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jacquelyn A Brown
- Department of Physics and Astronomy, Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN, USA
| | - Jennifer Foulke-Abel
- Departments of Physiology and Medicine, GI Division, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nesrin Hasan
- Departments of Physiology and Medicine, GI Division, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Julie In
- Departments of Physiology and Medicine, GI Division, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Edward Kelly
- Department of Pharmaceutics, University of Washington, WA, USA
| | - Olga Kovbasnjuk
- Departments of Physiology and Medicine, GI Division, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jonathan Repper
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nina Senutovitch
- University of Pittsburgh, Drug Discovery Institute Pittsburgh, PA, USA
| | - Janet Stabb
- Departments of Physiology and Medicine, GI Division, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Catherine Yeung
- Department of Pharmacy, University of Washington, WA, USA.,Kidney Research Institute, University of Washington, WA, USA
| | - Nick C Zachos
- Departments of Physiology and Medicine, GI Division, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mark Donowitz
- Departments of Physiology and Medicine, GI Division, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mary Estes
- Departments of Molecular Virology and Microbiology and Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jonathan Himmelfarb
- Kidney Research Institute, University of Washington, WA, USA.,Department of Medicine, University of Washington, WA, USA
| | - George Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - John P Wikswo
- Department of Physics and Astronomy, Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - D Lansing Taylor
- University of Pittsburgh, Drug Discovery Institute Pittsburgh, PA, USA.,Department of Computational and Systems Biology, University of Pittsburgh, Baltimore, PA, USA.,University of Pittsburgh Cancer Institute, PA, USA
| |
Collapse
|
95
|
Hardwick RN, Viergever C, Chen AE, Nguyen DG. 3D bioengineered tissues: From advancements in in vitro safety to new horizons in disease modeling. Clin Pharmacol Ther 2017; 101:453-457. [PMID: 27864923 DOI: 10.1002/cpt.569] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 10/31/2016] [Accepted: 11/09/2016] [Indexed: 01/04/2023]
Abstract
Research aimed at more fully emulating human biology in vitro has rapidly progressed in recent years with advancements in 3D tissue engineering and microphysiological systems. The initial target of such systems has been directed towards drug and chemical safety assessment, with the goal of improving sensitivity and predictive capabilities. Here we discuss recent developments of in vitro organ culture systems, and their future applications in modeling human disease.
Collapse
Affiliation(s)
- R N Hardwick
- Organovo Holdings Inc, San Diego, California, USA
| | - C Viergever
- Organovo Holdings Inc, San Diego, California, USA
| | - A E Chen
- Organovo Holdings Inc, San Diego, California, USA
| | - D G Nguyen
- Organovo Holdings Inc, San Diego, California, USA
| |
Collapse
|
96
|
Vernetti LA, Vogt A, Gough A, Taylor DL. Evolution of Experimental Models of the Liver to Predict Human Drug Hepatotoxicity and Efficacy. Clin Liver Dis 2017; 21:197-214. [PMID: 27842772 PMCID: PMC6325638 DOI: 10.1016/j.cld.2016.08.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
In this article, we review the past applications of in vitro models in identifying human hepatotoxins and then focus on the use of multiscale experimental models in drug development, including the use of zebrafish and human cell-based, 3-dimensional, microfluidic systems of liver functions as key components in applying Quantitative Systems Pharmacology (QSP). We have implemented QSP as a platform to improve the rate of success in the process of drug discovery and development of therapeutics.
Collapse
Affiliation(s)
- Lawrence A Vernetti
- Department of Computational and Systems Biology, University of Pittsburgh Drug Discovery Institute, Biomedical Science Tower 200 Lothrop Street, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| | - Andreas Vogt
- Department of Computational and Systems Biology, University of Pittsburgh Drug Discovery Institute, Biomedical Science Tower 200 Lothrop Street, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Albert Gough
- Department of Computational and Systems Biology, University of Pittsburgh Drug Discovery Institute, Biomedical Science Tower 200 Lothrop Street, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - D Lansing Taylor
- Department of Computational and Systems Biology, University of Pittsburgh Drug Discovery Institute, Biomedical Science Tower 200 Lothrop Street, University of Pittsburgh, Pittsburgh, PA 15260, USA; University of Pittsburgh Cancer Institute, 5150 Centre Avenue, Pittsburgh, PA 15232, USA
| |
Collapse
|
97
|
Bardsley K, Deegan AJ, El Haj A, Yang Y. Current State-of-the-Art 3D Tissue Models and Their Compatibility with Live Cell Imaging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1035:3-18. [PMID: 29080127 DOI: 10.1007/978-3-319-67358-5_1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mammalian cells grow within a complex three-dimensional (3D) microenvironment where multiple cells are organized and surrounded by extracellular matrix (ECM). The quantity and types of ECM components, alongside cell-to-cell and cell-to-matrix interactions dictate cellular differentiation, proliferation and function in vivo. To mimic natural cellular activities, various 3D tissue culture models have been established to replace conventional two dimensional (2D) culture environments. Allowing for both characterization and visualization of cellular activities within possibly bulky 3D tissue models presents considerable challenges due to the increased thickness and subsequent light scattering features of such 3D models. In this chapter, state-of-the-art methodologies used to establish 3D tissue models are discussed, first with a focus on both scaffold-free and scaffold-based 3D tissue model formation. Following on, multiple 3D live cell imaging systems, mainly optical imaging modalities, are introduced. Their advantages and disadvantages are discussed, with the aim of stimulating more research in this highly demanding research area.
Collapse
Affiliation(s)
- Katie Bardsley
- Institute for Science and Technology in Medicine, Keele University, Stoke-on-Trent, ST4 7QB, UK
| | - Anthony J Deegan
- Institute for Science and Technology in Medicine, Keele University, Stoke-on-Trent, ST4 7QB, UK
| | - Alicia El Haj
- Institute for Science and Technology in Medicine, Keele University, Stoke-on-Trent, ST4 7QB, UK
| | - Ying Yang
- Institute for Science and Technology in Medicine, Keele University, Stoke-on-Trent, ST4 7QB, UK.
| |
Collapse
|
98
|
Coppeta JR, Mescher MJ, Isenberg BC, Spencer AJ, Kim ES, Lever AR, Mulhern TJ, Prantil-Baun R, Comolli JC, Borenstein JT. A portable and reconfigurable multi-organ platform for drug development with onboard microfluidic flow control. LAB ON A CHIP 2016; 17:134-144. [PMID: 27901159 PMCID: PMC5177565 DOI: 10.1039/c6lc01236a] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The drug development pipeline is severely limited by a lack of reliable tools for prediction of human clinical safety and efficacy profiles for compounds at the pre-clinical stage. Here we present the design and implementation of a platform technology comprising multiple human cell-based tissue models in a portable and reconfigurable format that supports individual organ function and crosstalk for periods of up to several weeks. Organ perfusion and crosstalk are enabled by a precision flow control technology based on electromagnetic actuators embedded in an arrayed format on a microfluidic platform. We demonstrate two parallel circuits of connected airway and liver modules on a platform containing 62 electromagnetic microactuators, with precise and controlled flow rates as well as functional biological metrics over a two week time course. Technical advancements enabled by this platform include the use of non-sorptive construction materials, enhanced scalability, portability, flow control, and usability relative to conventional flow control modes (such as capillary action, pressure heads, or pneumatic air lines), and a reconfigurable and modular organ model format with common fluidic port architecture. We demonstrate stable biological function for multiple pairs of airway-liver models for periods of 2 weeks in the platform, with precise control over fluid levels, temperature, flow rate and oxygenation in order to support relevant use cases involving drug toxicity, efficacy testing, and organ-organ interaction.
Collapse
Affiliation(s)
- J R Coppeta
- Materials and Microfabrication Directorate, Draper, Cambridge, MA 02139, USA.
| | - M J Mescher
- Materials and Microfabrication Directorate, Draper, Cambridge, MA 02139, USA.
| | - B C Isenberg
- Materials and Microfabrication Directorate, Draper, Cambridge, MA 02139, USA.
| | - A J Spencer
- Materials and Microfabrication Directorate, Draper, Cambridge, MA 02139, USA.
| | - E S Kim
- Materials and Microfabrication Directorate, Draper, Cambridge, MA 02139, USA.
| | - A R Lever
- Materials and Microfabrication Directorate, Draper, Cambridge, MA 02139, USA.
| | - T J Mulhern
- Materials and Microfabrication Directorate, Draper, Cambridge, MA 02139, USA.
| | - R Prantil-Baun
- Materials and Microfabrication Directorate, Draper, Cambridge, MA 02139, USA.
| | - J C Comolli
- Materials and Microfabrication Directorate, Draper, Cambridge, MA 02139, USA.
| | - J T Borenstein
- Materials and Microfabrication Directorate, Draper, Cambridge, MA 02139, USA.
| |
Collapse
|
99
|
Rossow T, Lienemann PS, Mooney DJ. Cell Microencapsulation by Droplet Microfluidic Templating. MACROMOL CHEM PHYS 2016. [DOI: 10.1002/macp.201600380] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Torsten Rossow
- John A. Paulson School of Engineering and Applied Sciences; Harvard University; Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering; Cambridge MA 02138 USA
| | - Philipp S. Lienemann
- John A. Paulson School of Engineering and Applied Sciences; Harvard University; Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering; Cambridge MA 02138 USA
| | - David J. Mooney
- John A. Paulson School of Engineering and Applied Sciences; Harvard University; Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering; Cambridge MA 02138 USA
| |
Collapse
|
100
|
Portillo-Lara R, Annabi N. Microengineered cancer-on-a-chip platforms to study the metastatic microenvironment. LAB ON A CHIP 2016; 16:4063-4081. [PMID: 27605305 DOI: 10.1039/c6lc00718j] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
More than 90% of cancer-related deaths can be attributed to the occurrence of metastatic diseases. Recent studies have highlighted the importance of the multicellular, biochemical and biophysical stimuli from the tumor microenvironment during carcinogenesis, treatment failure, and metastasis. Therefore, there is a need for experimental platforms that are able to recapitulate the complex pathophysiological features of the metastatic microenvironment. Recent advancements in biomaterials, microfluidics, and tissue engineering have led to the development of living multicellular microculture systems, which are maintained in controllable microenvironments and function with organ level complexity. The applications of these "on-chip" technologies for detection, separation, characterization and three dimensional (3D) propagation of cancer cells have been extensively reviewed in previous works. In this contribution, we focus on integrative microengineered platforms that allow the study of multiple aspects of the metastatic microenvironment, including the physicochemical cues from the tumor associated stroma, the heterocellular interactions that drive trans-endothelial migration and angiogenesis, the environmental stresses that metastatic cancer cells encounter during migration, and the physicochemical gradients that direct cell motility and invasion. We discuss the application of these systems as in vitro assays to elucidate fundamental mechanisms of cancer metastasis, as well as their use as human relevant platforms for drug screening in biomimetic microenvironments. We then conclude with our commentaries on current progress and future perspectives of microengineered systems for fundamental and translational cancer research.
Collapse
Affiliation(s)
- R Portillo-Lara
- Department of Chemical Engineering, Northeastern University, 451 Snell Engineering Building, 360 Huntington Ave, Boston, MA 02115, USA. and Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey, Mexico
| | - N Annabi
- Department of Chemical Engineering, Northeastern University, 451 Snell Engineering Building, 360 Huntington Ave, Boston, MA 02115, USA. and Biomaterials Innovation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA and Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|