1
|
Ashok D, Singh J, Howard HR, Cottam S, Waterhouse A, Bilek MMM. Interfacial engineering for biomolecule immobilisation in microfluidic devices. Biomaterials 2025; 316:123014. [PMID: 39708778 DOI: 10.1016/j.biomaterials.2024.123014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/25/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Microfluidic devices are used for various applications in biology and medicine. From on-chip modelling of human organs for drug screening and fast and straightforward point-of-care (POC) detection of diseases to sensitive biochemical analysis, these devices can be custom-engineered using low-cost techniques. The microchannel interface is essential for these applications, as it is the interface of immobilised biomolecules that promote cell capture, attachment and proliferation, sense analytes and metabolites or provide enzymatic reaction readouts. However, common microfluidic materials do not facilitate the stable immobilisation of biomolecules required for relevant applications, making interfacial engineering necessary to attach biomolecules to the microfluidic surfaces. Interfacial engineering is performed through various immobilisation mechanisms and surface treatment techniques, which suitably modify the surface properties like chemistry and energy to obtain robust biomolecule immobilisation and long-term storage stability suitable for the final application. In this review, we provide an overview of the status of interfacial engineering in microfluidic devices, covering applications, the role of biomolecules, their immobilisation pathways and the influence of microfluidic materials. We then propose treatment techniques to optimise performance for various biological and medical applications and highlight future areas of development.
Collapse
Affiliation(s)
- Deepu Ashok
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, NSW, 2006, Australia; School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia; Heart Research Institute, Newtown, NSW, 2042, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia; School of Physics, Faculty of Science, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Jasneil Singh
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia; Heart Research Institute, Newtown, NSW, 2042, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Henry Robert Howard
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Sophie Cottam
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, NSW, 2006, Australia; School of Physics, Faculty of Science, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Anna Waterhouse
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia.
| | - Marcela M M Bilek
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, NSW, 2006, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia; School of Physics, Faculty of Science, The University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
2
|
Schellberg BG, Koppes AN, Koppes RA. In situ monitoring of barrier function on-chip via automated, non-invasive luminescence sensing. LAB ON A CHIP 2025. [PMID: 40181784 PMCID: PMC11969330 DOI: 10.1039/d4lc01090f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/28/2025] [Indexed: 04/05/2025]
Abstract
Over the past 30 years, organs-on-a-chip (OOCs) have emerged as a robust alternative to address the technological challenges associated with current in vitro and in vivo options. Although OOCs offer improved bio-relevance and controlled complexity, broad adoption has remained limited. Most approaches to characterize on-chip structure and function require human intervention, limiting device translation and feasibility. Here, we introduce a new fiber optic-based sensing platform that enables automated, temporal luminescence sensing on-chip, validated for real-time readout of epithelial and endothelial barrier function under cytokine-induced inflammation. Our platform, capable of at least 1 μM resolution, tracked paracellular transport in situ for 9 days of culture under perfusion on-chip. These results offer an alternative sensing approach for continuous, non-invasive luminescence monitoring in OOCs.
Collapse
Affiliation(s)
- Bryan G Schellberg
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA.
| | - Abigail N Koppes
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA.
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Ryan A Koppes
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA.
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Dornhof J, Kieninger J, Rupitsch SJ, Weltin A. Microsensor systems for cell metabolism - from 2D culture to organ-on-chip (2019-2024). LAB ON A CHIP 2025; 25:1149-1168. [PMID: 39775787 DOI: 10.1039/d4lc00437j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Cell cultures, organs-on-chip and microphysiological systems become increasingly relevant as in vitro models, e.g., in drug development, disease modelling, toxicology or cancer research. It has been underlined repeatedly that culture conditions and metabolic cues have a strong or even essential influence on the reproducibility and validity of such experiments but are often not appropriately measured or controlled. Here we review microsensor systems for cell metabolism for the continuous measurement of culture conditions in microfluidic and lab-on-chip platforms. We identify building blocks, features and essential advantages to underline the relevance of these systems and to derive appropriate requirements for development and practical use. We discuss different formats and geometries of cell culture, microfluidics and the resulting consequences for sensor placement, as the prerequisite for understanding the various approaches and classification of the systems. The major chemical and biosensors based on electrochemical and optical principles are discussed for general understanding and to contextualize current developments. We then review selected recent sensor systems with real-world implementations of sensing in cell cultures and organs-on-chip, employing a helpful characterization. That includes formats and cell models, microfluidic systems and sensor types applied in static and dynamic monitoring of 2D and 3D cell cultures, as well as single spheroids. We discuss notable advances, particularly with respect to sensor performance and the demonstration of long-term continuous measurements. We outline current approaches to system fabrication technologies, material choice, and interfacing, and comment on recent trends. Finally, we conclude with critical remarks on the current state of sensors in cell culture monitoring and identify avenues for future improvements for both developers and users of such systems, which will lead to better and more predictive in vitro models.
Collapse
Affiliation(s)
- Johannes Dornhof
- Laboratory for Electrical Instrumentation and Embedded Systems, IMTEK - Department of Microsystems Engineering, University of Freiburg, Georges-Köhler-Allee 103, 79110 Freiburg, Germany.
| | - Jochen Kieninger
- Laboratory for Electrical Instrumentation and Embedded Systems, IMTEK - Department of Microsystems Engineering, University of Freiburg, Georges-Köhler-Allee 103, 79110 Freiburg, Germany.
| | - Stefan J Rupitsch
- Laboratory for Electrical Instrumentation and Embedded Systems, IMTEK - Department of Microsystems Engineering, University of Freiburg, Georges-Köhler-Allee 103, 79110 Freiburg, Germany.
| | - Andreas Weltin
- Laboratory for Electrical Instrumentation and Embedded Systems, IMTEK - Department of Microsystems Engineering, University of Freiburg, Georges-Köhler-Allee 103, 79110 Freiburg, Germany.
| |
Collapse
|
4
|
Schellberg BG, Koppes RA, Koppes AN. Recent Advances in Integrated Organ-Chip Sensing Toward Robust and User-Friendly Systems. J Biomed Mater Res A 2025; 113:e37876. [PMID: 39893559 DOI: 10.1002/jbm.a.37876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/17/2024] [Accepted: 01/07/2025] [Indexed: 02/04/2025]
Abstract
Organs-on-a-chip (OOC) are an emergent technology that bridge the gap between current in vitro and in vivo models used to inform drug discovery and investigate disease pathophysiology. These systems offer improved bio-relevance and controlled complexity through the integration of physical and/or chemical stimuli matched to physiologically relevant conditions. Although significant advancements have been made toward recreating organ-specific physiology on chip, the methods available to study structure and function of the cell microenvironment are still limited. Established analysis approaches, including fluorescence microscopy, rely on laborious offline workflows that yield limited time-point data. As the OOC field continues to evolve, there is a unique opportunity to engineer improved characterization methods into organ-chip devices. This review provides an overview of current integrated sensing approaches that address current limitations and enable real-time readout of relevant physiological parameters in OOC.
Collapse
Affiliation(s)
- Bryan G Schellberg
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Ryan A Koppes
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Abigail N Koppes
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Naseri P, Seyyedi SM, Hashemi-Tilehnoee M, Naeimi AS. Analysis of magnetic field-induced breakup of ferrofluid droplets in a symmetric Y-junction microchannel. Sci Rep 2024; 14:23763. [PMID: 39390127 PMCID: PMC11467301 DOI: 10.1038/s41598-024-74805-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024] Open
Abstract
This research focuses on the analysis of the breakup of ferrofluid droplets in a symmetric microchannel with a Y-junction microchannel, utilizing computational methods. The study proposes an innovative strategy to enhance the breakup phenomenon by introducing a magnetic field within the branches of the Y-junction microchannel. To verify the obtained results, a comprehensive comparison is conducted, incorporating previous numerical and experimental investigations available in the literature. The outcomes of this comparison demonstrate a significant concurrence between the current findings and the prior studies. The results unequivocally elucidate that the presence of a magnetic field accelerates the fragmentation of the parent droplet in comparison to scenarios without a magnetic field. Furthermore, it is established that the duration required for droplet breakup decreases as the magnetic Bond number increases. Achieved results indicates [Formula: see text] decreases about 3% and 1.5% for L*=3 and L*=4, respectively. It is worth highlighting that this trend is particularly accentuated in the case of smaller non-dimensional lengths, specifically L∗=3.0.
Collapse
Affiliation(s)
- Parviz Naseri
- Department of Mechanical Engineering, Aliabad Katoul branch, Islamic Azad University, Aliabad Katoul, Iran
| | - Seyyed Masoud Seyyedi
- Department of Mechanical Engineering, Aliabad Katoul branch, Islamic Azad University, Aliabad Katoul, Iran.
| | - Mehdi Hashemi-Tilehnoee
- Department of Mechanical Engineering, Aliabad Katoul branch, Islamic Azad University, Aliabad Katoul, Iran
- Centre for Cooperative Research on Alternative Energies (CIC energiGUNE), Basque Research and Technology Alliance (BRTA), Alava Technology Park, Albert Einstein 48, 01510, Vitoria-Gasteiz, Spain
| | - Azadeh Sadat Naeimi
- Department of Physics, Aliabad Katoul branch, Islamic Azad University, Aliabad Katoul, Iran
| |
Collapse
|
6
|
Yuan S, Yuan H, Hay DC, Hu H, Wang C. Revolutionizing Drug Discovery: The Impact of Distinct Designs and Biosensor Integration in Microfluidics-Based Organ-on-a-Chip Technology. BIOSENSORS 2024; 14:425. [PMID: 39329800 PMCID: PMC11430660 DOI: 10.3390/bios14090425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024]
Abstract
Traditional drug development is a long and expensive process with high rates of failure. This has prompted the pharmaceutical industry to seek more efficient drug development frameworks, driving the emergence of organ-on-a-chip (OOC) based on microfluidic technologies. Unlike traditional animal experiments, OOC systems provide a more accurate simulation of human organ microenvironments and physiological responses, therefore offering a cost-effective and efficient platform for biomedical research, particularly in the development of new medicines. Additionally, OOC systems enable quick and real-time analysis, high-throughput experimentation, and automation. These advantages have shown significant promise in enhancing the drug development process. The success of an OOC system hinges on the integration of specific designs, manufacturing techniques, and biosensors to meet the need for integrated multiparameter datasets. This review focuses on the manufacturing, design, sensing systems, and applications of OOC systems, highlighting their design and sensing capabilities, as well as the technical challenges they currently face.
Collapse
Affiliation(s)
- Sheng Yuan
- Centre of Biomedical Systems and Informatics, Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), School of Medicine, International Campus, Zhejiang University, Haining 314400, China
| | - Huipu Yuan
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310020, China
| | - David C. Hay
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK;
| | - Huan Hu
- Zhejiang University-University of Illinois Urbana-Champaign Institute (ZJU-UIUC Institute), International Campus, Zhejiang University, Haining 314400, China
| | - Chaochen Wang
- Centre of Biomedical Systems and Informatics, Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), School of Medicine, International Campus, Zhejiang University, Haining 314400, China
- Department of Gynecology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310020, China
| |
Collapse
|
7
|
Casotti MC, Meira DD, Zetum ASS, Campanharo CV, da Silva DRC, Giacinti GM, da Silva IM, Moura JAD, Barbosa KRM, Altoé LSC, Mauricio LSR, Góes LSBDB, Alves LNR, Linhares SSG, Ventorim VDP, Guaitolini YM, dos Santos EDVW, Errera FIV, Groisman S, de Carvalho EF, de Paula F, de Sousa MVP, Fechine PBA, Louro ID. Integrating frontiers: a holistic, quantum and evolutionary approach to conquering cancer through systems biology and multidisciplinary synergy. Front Oncol 2024; 14:1419599. [PMID: 39224803 PMCID: PMC11367711 DOI: 10.3389/fonc.2024.1419599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Cancer therapy is facing increasingly significant challenges, marked by a wide range of techniques and research efforts centered around somatic mutations, precision oncology, and the vast amount of big data. Despite this abundance of information, the quest to cure cancer often seems more elusive, with the "war on cancer" yet to deliver a definitive victory. A particularly pressing issue is the development of tumor treatment resistance, highlighting the urgent need for innovative approaches. Evolutionary, Quantum Biology and System Biology offer a promising framework for advancing experimental cancer research. By integrating theoretical studies, translational methods, and flexible multidisciplinary clinical research, there's potential to enhance current treatment strategies and improve outcomes for cancer patients. Establishing stronger links between evolutionary, quantum, entropy and chaos principles and oncology could lead to more effective treatments that leverage an understanding of the tumor's evolutionary dynamics, paving the way for novel methods to control and mitigate cancer. Achieving these objectives necessitates a commitment to multidisciplinary and interprofessional collaboration at the heart of both research and clinical endeavors in oncology. This entails dismantling silos between disciplines, encouraging open communication and data sharing, and integrating diverse viewpoints and expertise from the outset of research projects. Being receptive to new scientific discoveries and responsive to how patients react to treatments is also crucial. Such strategies are key to keeping the field of oncology at the forefront of effective cancer management, ensuring patients receive the most personalized and effective care. Ultimately, this approach aims to push the boundaries of cancer understanding, treating it as a manageable chronic condition, aiming to extend life expectancy and enhance patient quality of life.
Collapse
Affiliation(s)
- Matheus Correia Casotti
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | - Débora Dummer Meira
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | | | | | | | - Giulia Maria Giacinti
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | - Iris Moreira da Silva
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | - João Augusto Diniz Moura
- Laboratório de Oncologia Clínica e Experimental, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | - Karen Ruth Michio Barbosa
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | - Lorena Souza Castro Altoé
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | | | | | - Lyvia Neves Rebello Alves
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | | | - Vinícius do Prado Ventorim
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | - Yasmin Moreto Guaitolini
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | | | | | - Sonia Groisman
- Instituto de Biologia Roberto Alcântara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, RJ, Brazil
| | - Elizeu Fagundes de Carvalho
- Instituto de Biologia Roberto Alcântara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, RJ, Brazil
| | - Flavia de Paula
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | | | - Pierre Basílio Almeida Fechine
- Group of Chemistry of Advanced Materials (GQMat), Department of Analytical Chemistry and Physical-Chemistry, Federal University of Ceará (UFC), Fortaleza, CE, Brazil
| | - Iuri Drumond Louro
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| |
Collapse
|
8
|
Spitz S, Schobesberger S, Brandauer K, Ertl P. Sensor-integrated brain-on-a-chip platforms: Improving the predictive validity in neurodegenerative research. Bioeng Transl Med 2024; 9:e10604. [PMID: 38818126 PMCID: PMC11135156 DOI: 10.1002/btm2.10604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/31/2023] [Accepted: 09/12/2023] [Indexed: 06/01/2024] Open
Abstract
Affecting millions of individuals worldwide, neurodegenerative diseases (NDDs) pose a significant and growing health concern in people over the age of 60 years. Contributing to this trend are the steady increase in the aging population coupled with a persistent lack of disease-altering treatment strategies targeting NDDs. The absence of efficient therapeutics can be attributed to high failure rates in clinical trials and the ineptness of animal models in preceding preclinical studies. To that end, in recent years, significant research effort has been dedicated to the development of human cell-based preclinical disease models characterized by a higher degree of predictive validity. However, a key requirement of any in vitro model constitutes the precise knowledge and replication of the target tissues' (patho-)physiological microenvironment. Herein, microphysiological systems have demonstrated superiority over conventional static 2D/3D in vitro cell culture systems, as they allow for the emulation and continuous monitoring of the onset, progression, and remission of disease-associated phenotypes. This review provides an overview of recent advances in the field of NDD research using organ-on-a-chip platforms. Specific focus is directed toward non-invasive sensing strategies encompassing electrical, electrochemical, and optical sensors. Additionally, promising on- and integrable off-chip sensing strategies targeting key analytes in NDDs will be presented and discussed in detail.
Collapse
Affiliation(s)
- Sarah Spitz
- Faculty of Technical ChemistryVienna University of TechnologyViennaAustria
- Present address:
Department of Mechanical Engineering and Biological EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | | | | | - Peter Ertl
- Faculty of Technical ChemistryVienna University of TechnologyViennaAustria
| |
Collapse
|
9
|
Tong W, Shi J, Yu Z, Ran B, Chen H, Zhu Y. High sensitivity and automatic chemiluminescence detection of glucose and lactate using a spin-disc paper-based device. LAB ON A CHIP 2024; 24:810-818. [PMID: 38224458 DOI: 10.1039/d3lc00937h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
This paper reports a spin-disc paper-based device with 10 individual detection units containing electromagnetic modules controlling the sample incubation time before chemiluminescence (CL) signal detection. After the sample was added to the top paper chip and incubated with the enzyme, the electromagnet was turned off to allow contact between the top and bottom paper. The H2O2 generated by the sample flowed vertically to the bottom paper and initiated the oxidase of the luminol to generate the CL signal. After one detection the disc was automatically rotated to the next position to repeat the above detection. The advantage of using the device over the lateral flow and the in situ detection was firstly proved using the detection of H2O2 and the glucose/lactate sample with 5 minute incubation. The CL intensity was increased 300 times/1000 times as the glucose/lactate was incubated for 5 minutes compared to the non-incubated samples. Afterward, the device was employed to separately detect glucose and lactate diluted in PBS, artificial sweat, artificial saliva, and fresh cell culture media. Finally, the device was employed to detect the glucose and lactate in the media collected over the 24 hour culture of PC3 cells. The uptake and production rates of glucose and lactate were correspondingly determined as 0.328 ± 0.015 pmol h-1 per cell and 1.254 ± 0.053 pmol h-1 per cell, respectively. The reported device has wide application potential due to its capabilities in automatic detection of multiple samples with very high sensitivity and small sample volume (down to 0.5 μL).
Collapse
Affiliation(s)
- Wenqiang Tong
- School of Science, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, China
- Center for Microflows and Nanoflows, School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, China
| | - Jiaming Shi
- School of Science, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, China
- Center for Microflows and Nanoflows, School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, China
| | - Zhihang Yu
- School of Science, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, China
- Center for Microflows and Nanoflows, School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, China
| | - Bin Ran
- School of Science, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, China
- Center for Microflows and Nanoflows, School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, China
| | - Huaying Chen
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, China.
- Center for Microflows and Nanoflows, School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, China
| | - Yonggang Zhu
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, China.
- Center for Microflows and Nanoflows, School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, China
| |
Collapse
|
10
|
Fuse H, Kikawada T, Cornette R. Effective methods for immobilization of non-adherent Pv11 cells while maintaining their desiccation tolerance. Cytotechnology 2023; 75:491-503. [PMID: 37841960 PMCID: PMC10575823 DOI: 10.1007/s10616-023-00592-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/24/2023] [Indexed: 10/17/2023] Open
Abstract
Pv11 was derived from embryos of the sleeping chironomid Polypedilum vanderplanki, which displays an extreme form of desiccation tolerance known as anhydrobiosis. Pre-treatment with a high concentration of trehalose allows Pv11 cells to enter anhydrobiosis. In the dry state, Pv11 cells preserve transgenic luciferase while retaining its activity. Thus, these cells could be utilized for dry-preserving antibodies, enzymes, signaling proteins or other valuable biological materials without denaturation. However, Pv11 cells grow in suspension, which limits their applicability; for instance, they cannot be integrated into microfluidic devices or used in devices such as sensor chips. Therefore, in this paper, we developed an effective immobilization system for Pv11 cells that, crucially, allows them to maintain their anhydrobiotic potential even when immobilized. Pv11 cells exhibited a very high adhesion rate with both biocompatible anchor for membrane (BAM) and Cell-Tak coatings, which have been reported to be effective on other cultured cells. We also found that Pv11 cells immobilized well to uncoated glass if handled in serum-free medium. Interestingly, Pv11 cells showed desiccation tolerance when trehalose treatment was done prior to immobilization of the cells. In contrast, trehalose treatment after immobilization of Pv11 cells resulted in a significant decrease in desiccation tolerance. Thus, it is important to induce anhydrobiosis before immobilization. In summary, we report the successful development of a protocol for the dry preservation of immobilized Pv11 cells. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-023-00592-0.
Collapse
Affiliation(s)
- Hiroto Fuse
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5, Kashiwa, Chiba 277-8562 Japan
| | - Takahiro Kikawada
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5, Kashiwa, Chiba 277-8562 Japan
- Division of Biomaterial Sciences, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), 1-2 Owashi, Tsukuba, Ibaraki 305-0851 Japan
| | - Richard Cornette
- Division of Biomaterial Sciences, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), 1-2 Owashi, Tsukuba, Ibaraki 305-0851 Japan
| |
Collapse
|
11
|
Kim H, Kim EJ, Ngo HV, Nguyen HD, Park C, Choi KH, Park JB, Lee BJ. Cellular Efficacy of Fattigated Nanoparticles and Real-Time ROS Occurrence Using Microfluidic Hepatocarcinoma Chip System: Effect of Anticancer Drug Solubility and Shear Stress. Pharmaceuticals (Basel) 2023; 16:1330. [PMID: 37765137 PMCID: PMC10536289 DOI: 10.3390/ph16091330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/09/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
The objective of this study was to evaluate the effectiveness of organ-on-chip system investigating simultaneous cellular efficacy and real-time reactive oxygen species (ROS) occurrence of anticancer drug-loaded nanoparticles (NPs) using hepatocarcinoma cells (HepG2) chip system under static and hepatomimicking shear stress conditions (5 dyne/cm2). Then, the role of hepatomimetic shear stress exposed to HepG2 and drug solubility were compared. The highly soluble doxorubicin (DOX) and poorly soluble paclitaxel (PTX) were chosen. Fattigated NPs (AONs) were formed via self-assembly of amphiphilic albumin (HSA)-oleic acid conjugate (AOC). Then, drug-loaded AONs (DOX-AON or PTX-AON) were exposed to a serum-free HepG2 medium at 37 °C and 5% carbon dioxide for 24 h using a real-time ROS sensor chip-based microfluidic system. The cellular efficacy and simultaneous ROS occurrence of free drugs and drug-loaded AONs were compared. The cellular efficacy of drug-loaded AONs varied in a dose-dependent manner and were consistently correlated with real-time of ROS occurrence. Drug-loaded AONs increased the intracellular fluorescence intensity and decreased the cellular efficacy compared to free drugs under dynamic conditions. The half-maximal inhibitory concentration (IC50) values of free DOX (13.4 μg/mL) and PTX (54.44 μg/mL) under static conditions decreased to 11.79 and 38.43 μg/mL, respectively, under dynamic conditions. Furthermore, DOX- and PTX-AONs showed highly decreased IC50 values of 5.613 and 21.86 μg/mL, respectively, as compared to free drugs under dynamic conditions. It was evident that cellular efficacy and real-time ROS occurrence were well-correlated and highly dependent on the drug-loaded nanostructure, drug solubility and physiological shear stress.
Collapse
Affiliation(s)
- Hoyoung Kim
- College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea; (H.K.); (E.-J.K.); (H.V.N.); (H.D.N.)
| | - Eun-Ji Kim
- College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea; (H.K.); (E.-J.K.); (H.V.N.); (H.D.N.)
| | - Hai V. Ngo
- College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea; (H.K.); (E.-J.K.); (H.V.N.); (H.D.N.)
| | - Hy D. Nguyen
- College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea; (H.K.); (E.-J.K.); (H.V.N.); (H.D.N.)
| | - Chulhun Park
- College of Pharmacy, Jeju National University, Jeju 63243, Republic of Korea;
| | - Kyung Hyun Choi
- Advanced Micro-Mechatronics Lab, Mechatronics Engineering, Jeju National University, Jeju 63243, Republic of Korea;
- BioSpero, Jeju 63309, Republic of Korea
| | - Jun-Bom Park
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea;
| | - Beom-Jin Lee
- College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea; (H.K.); (E.-J.K.); (H.V.N.); (H.D.N.)
| |
Collapse
|
12
|
Zhao C, Wang Z, Tang X, Qin J, Jiang Z. Recent advances in sensor-integrated brain-on-a-chip devices for real-time brain monitoring. Colloids Surf B Biointerfaces 2023; 229:113431. [PMID: 37473652 DOI: 10.1016/j.colsurfb.2023.113431] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/18/2023] [Accepted: 06/26/2023] [Indexed: 07/22/2023]
Abstract
Brain science has remained in the global spotlight as an important field of scientific and technological discovery. Numerous in vitro and in vivo animal studies have been performed to understand the pathological processes involved in brain diseases and develop strategies for their diagnosis and treatment. However, owing to species differences between animals and humans, several drugs have shown high rates of treatment failure in clinical settings, hindering the development of diagnostic and treatment modalities for brain diseases. In this scenario, microfluidic brain-on-a-chip (BOC) devices, which allow the direct use of human tissues for experiments, have emerged as novel tools for effectively avoiding species differences and performing screening for new drugs. Although microfluidic BOC technology has achieved significant progress in recent years, monitoring slight changes in neurochemicals, neurotransmitters, and environmental states in the brain has remained challenging owing to the brain's complex environment. Hence, the integration of BOC with new sensors that have high sensitivity and high selectivity is urgently required for the real-time dynamic monitoring of BOC parameters. As sensor-based technologies for BOC have not been summarized, here, we review the principle, fabrication process, and application-based classification of sensor-integrated BOC, and then summarize the opportunities and challenges for their development. Generally, sensor-integrated BOC enables real-time monitoring and dynamic analysis, accurately measuring minute changes in the brain and thus enabling the realization of in vivo brain analysis and drug development.
Collapse
Affiliation(s)
- Chen Zhao
- School of Medical Technology, School of Life Science, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Zihao Wang
- School of Medical Technology, School of Life Science, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Xiaoying Tang
- School of Medical Technology, School of Life Science, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China.
| | - Jieling Qin
- School of Medical Technology, School of Life Science, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China; Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Zhenqi Jiang
- School of Medical Technology, School of Life Science, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China.
| |
Collapse
|
13
|
Mieville V, Griffioen AW, Benamran D, Nowak-Sliwinska P. Advanced in vitro models for renal cell carcinoma therapy design. Biochim Biophys Acta Rev Cancer 2023; 1878:188942. [PMID: 37343729 DOI: 10.1016/j.bbcan.2023.188942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023]
Abstract
Renal cell carcinoma (RCC) and its principal subtype, clear cell RCC, are the most diagnosed kidney cancer. Despite substantial improvement over the last decades, current pharmacological intervention still fails to achieve long-term therapeutic success. RCC is characterized by a high intra- and inter-tumoral heterogeneity and is heavily influenced by the crosstalk of the cells composing the tumor microenvironment, such as cancer-associated fibroblasts, endothelial cells and immune cells. Moreover, multiple physicochemical properties such as pH, interstitial pressure or oxygenation may also play an important role. These elements are often poorly recapitulated in in vitro models used for drug development. This inadequate recapitulation of the tumor is partially responsible for the current lack of an effective and curative treatment. Therefore, there are needs for more complex in vitro or ex vivo drug screening models. In this review, we discuss the current state-of-the-art of RCC models and suggest strategies for their further development.
Collapse
Affiliation(s)
- Valentin Mieville
- School of Pharmaceutical Sciences, Faculty of Sciences, University of Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland; Translational Research Center in Oncohaematology, Geneva, Switzerland
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Daniel Benamran
- Division of Urology, Geneva University Hospitals, Geneva, Switzerland
| | - Patrycja Nowak-Sliwinska
- School of Pharmaceutical Sciences, Faculty of Sciences, University of Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland; Translational Research Center in Oncohaematology, Geneva, Switzerland.
| |
Collapse
|
14
|
Milton LA, Viglione MS, Ong LJY, Nordin GP, Toh YC. Vat photopolymerization 3D printed microfluidic devices for organ-on-a-chip applications. LAB ON A CHIP 2023; 23:3537-3560. [PMID: 37476860 PMCID: PMC10448871 DOI: 10.1039/d3lc00094j] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
Organs-on-a-chip, or OoCs, are microfluidic tissue culture devices with micro-scaled architectures that repeatedly achieve biomimicry of biological phenomena. They are well positioned to become the primary pre-clinical testing modality as they possess high translational value. Current methods of fabrication have facilitated the development of many custom OoCs that have generated promising results. However, the reliance on microfabrication and soft lithographic fabrication techniques has limited their prototyping turnover rate and scalability. Additive manufacturing, known commonly as 3D printing, shows promise to expedite this prototyping process, while also making fabrication easier and more reproducible. We briefly introduce common 3D printing modalities before identifying two sub-types of vat photopolymerization - stereolithography (SLA) and digital light processing (DLP) - as the most advantageous fabrication methods for the future of OoC development. We then outline the motivations for shifting to 3D printing, the requirements for 3D printed OoCs to be competitive with the current state of the art, and several considerations for achieving successful 3D printed OoC devices touching on design and fabrication techniques, including a survey of commercial and custom 3D printers and resins. In all, we aim to form a guide for the end-user to facilitate the in-house generation of 3D printed OoCs, along with the future translation of these important devices.
Collapse
Affiliation(s)
- Laura A Milton
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia.
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia
| | - Matthew S Viglione
- Department of Electrical and Computer Engineering, Brigham Young University, Provo, Utah, USA.
| | - Louis Jun Ye Ong
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia.
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia
- Max Planck Queensland Centre (MPQC) for the Materials Science of Extracellular Matrices, Queensland University of Technology, Brisbane, Australia
| | - Gregory P Nordin
- Department of Electrical and Computer Engineering, Brigham Young University, Provo, Utah, USA.
| | - Yi-Chin Toh
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia.
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia
- Max Planck Queensland Centre (MPQC) for the Materials Science of Extracellular Matrices, Queensland University of Technology, Brisbane, Australia
- Centre for Microbiome Research, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
15
|
Babaliari E, Ranella A, Stratakis E. Microfluidic Systems for Neural Cell Studies. Bioengineering (Basel) 2023; 10:902. [PMID: 37627787 PMCID: PMC10451731 DOI: 10.3390/bioengineering10080902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/05/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Whereas the axons of the peripheral nervous system (PNS) spontaneously regenerate after an injury, the occurring regeneration is rarely successful because axons are usually directed by inappropriate cues. Therefore, finding successful ways to guide neurite outgrowth, in vitro, is essential for neurogenesis. Microfluidic systems reflect more appropriately the in vivo environment of cells in tissues such as the normal fluid flow within the body, consistent nutrient delivery, effective waste removal, and mechanical stimulation due to fluid shear forces. At the same time, it has been well reported that topography affects neuronal outgrowth, orientation, and differentiation. In this review, we demonstrate how topography and microfluidic flow affect neuronal behavior, either separately or in synergy, and highlight the efficacy of microfluidic systems in promoting neuronal outgrowth.
Collapse
Affiliation(s)
- Eleftheria Babaliari
- Foundation for Research and Technology—Hellas (F.O.R.T.H.), Institute of Electronic Structure and Laser (I.E.S.L.), Vasilika Vouton, 70013 Heraklion, Greece;
| | - Anthi Ranella
- Foundation for Research and Technology—Hellas (F.O.R.T.H.), Institute of Electronic Structure and Laser (I.E.S.L.), Vasilika Vouton, 70013 Heraklion, Greece;
| | - Emmanuel Stratakis
- Foundation for Research and Technology—Hellas (F.O.R.T.H.), Institute of Electronic Structure and Laser (I.E.S.L.), Vasilika Vouton, 70013 Heraklion, Greece;
- Department of Physics, University of Crete, 70013 Heraklion, Greece
| |
Collapse
|
16
|
Yu J, Yin Y, Leng Y, Zhang J, Wang C, Chen Y, Li X, Wang X, Liu H, Liao Y, Jin Y, Zhang Y, Lu K, Wang K, Wang X, Wang L, Zheng F, Gu Z, Li Y, Fan Y. Emerging strategies of engineering retinal organoids and organoid-on-a-chip in modeling intraocular drug delivery: current progress and future perspectives. Adv Drug Deliv Rev 2023; 197:114842. [PMID: 37105398 DOI: 10.1016/j.addr.2023.114842] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/17/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023]
Abstract
Retinal diseases are a rising concern as major causes of blindness in an aging society; therapeutic options are limited, and the precise pathogenesis of these diseases remains largely unknown. Intraocular drug delivery and nanomedicines offering targeted, sustained, and controllable delivery are the most challenging and popular topics in ocular drug development and toxicological evaluation. Retinal organoids (ROs) and organoid-on-a-chip (ROoC) are both emerging as promising in-vitro models to faithfully recapitulate human eyes for retinal research in the replacement of experimental animals and primary cells. In this study, we review the generation and application of ROs resembling the human retina in cell subtypes and laminated structures and introduce the emerging engineered ROoC as a technological opportunity to address critical issues. On-chip vascularization, perfusion, and close inter-tissue interactions recreate physiological environments in vitro, whilst integrating with biosensors facilitates real-time analysis and monitoring during organogenesis of the retina representing engineering efforts in ROoC models. We also emphasize that ROs and ROoCs hold the potential for applications in modeling intraocular drug delivery in vitro and developing next-generation retinal drug delivery strategies.
Collapse
Affiliation(s)
- Jiaheng Yu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yuqi Yin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yubing Leng
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Jingcheng Zhang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Chunyan Wang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Yanyun Chen
- Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xiaorui Li
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Xudong Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Hui Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yulong Liao
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yishan Jin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yihan Zhang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Keyu Lu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Kehao Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China
| | - Xiaofei Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China
| | - Lizhen Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China
| | - Fuyin Zheng
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China.
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Yinghui Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.
| | - Yubo Fan
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China.
| |
Collapse
|
17
|
Zhou L, Liu L, Chang MA, Ma C, Chen W, Chen P. Spatiotemporal dissection of tumor microenvironment via in situ sensing and monitoring in tumor-on-a-chip. Biosens Bioelectron 2023; 225:115064. [PMID: 36680970 PMCID: PMC9918721 DOI: 10.1016/j.bios.2023.115064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
Real-time monitoring in the tumor microenvironment provides critical insights of cancer progression and mechanistic understanding of responses to cancer treatments. However, clinical challenges and significant questions remain regarding assessment of limited clinical tissue samples, establishment of validated, controllable pre-clinical cancer models, monitoring of static versus dynamic markers, and the translation of insights gained from in vitro tumor microenvironments to systematic investigation and understanding in clinical practice. State-of-art tumor-on-a-chip strategies will be reviewed herein, and emerging real-time sensing and monitoring platforms for on-chip analysis of tumor microenvironment will also be examined. The integration of the sensors with tumor-on-a-chip platforms to provide spatiotemporal information of the tumor microenvironment and the associated challenges will be further evaluated. Though optimal integrated systems for in situ monitoring are still in evolution, great promises lie ahead that will open new paradigm for rapid, comprehensive analysis of cancer development and assist clinicians with powerful tools to guide the diagnosis, prognosis and treatment course in cancer.
Collapse
Affiliation(s)
- Lang Zhou
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, 36849, USA
| | - Lunan Liu
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA; Department of Biomedical Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Muammar Ali Chang
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, 36849, USA
| | - Chao Ma
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA; Department of Biomedical Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA; Department of Biomedical Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Pengyu Chen
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, 36849, USA.
| |
Collapse
|
18
|
Shi J, Tong W, Yu Z, Tong L, Chen H, Jin J, Zhu Y. Pollution-Free and Highly Sensitive Lactate Detection in Cell Culture Based on a Microfluidic Chip. MICROMACHINES 2023; 14:770. [PMID: 37421003 DOI: 10.3390/mi14040770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/23/2023] [Accepted: 03/26/2023] [Indexed: 07/09/2023]
Abstract
Cell metabolite detection is important for cell analysis. As a cellular metabolite, lactate and its detection play an important role in disease diagnosis, drug screening and clinical therapeutics. This paper reports a microfluidic chip integrated with a backflow prevention channel for cell culture and lactate detection. It can effectively realize the upstream and downstream separation of the culture chamber and the detection zone, and prevent the pollution of cells caused by the potential backflow of reagent and buffer solutions. Due to such a separation, it is possible to analyze the lactate concentration in the flow process without contamination of cells. With the information of residence time distribution of the microchannel networks and the detected time signal in the detection chamber, it is possible to calculate the lactate concentration as a function of time using the de-convolution method. We have further demonstrated the suitability of this detection method by measuring lactate production in human umbilical vein endothelial cells (HUVEC). The microfluidic chip presented here shows good stability in metabolite quick detection and can work continuously for more than a few days. It sheds new insights into pollution-free and high-sensitivity cell metabolism detection, showing broad application prospects in cell analysis, drug screening and disease diagnosis.
Collapse
Affiliation(s)
- Jiaming Shi
- School of Science, Harbin Institute of Technology (Shenzhen), Shenzhen 518000, China
| | - Wenqiang Tong
- School of Science, Harbin Institute of Technology (Shenzhen), Shenzhen 518000, China
| | - Zhihang Yu
- School of Science, Harbin Institute of Technology (Shenzhen), Shenzhen 518000, China
| | - Lei Tong
- School of Science, Harbin Institute of Technology (Shenzhen), Shenzhen 518000, China
| | - Huaying Chen
- Center for Microflows and Nanoflows, School of Mechanical Engineering and Automation, Harbin Institute of Technology (Shenzhen), Shenzhen 518000, China
| | - Jing Jin
- Center for Microflows and Nanoflows, School of Mechanical Engineering and Automation, Harbin Institute of Technology (Shenzhen), Shenzhen 518000, China
| | - Yonggang Zhu
- School of Science, Harbin Institute of Technology (Shenzhen), Shenzhen 518000, China
- Center for Microflows and Nanoflows, School of Mechanical Engineering and Automation, Harbin Institute of Technology (Shenzhen), Shenzhen 518000, China
| |
Collapse
|
19
|
Sabaté Del Río J, Ro J, Yoon H, Park TE, Cho YK. Integrated technologies for continuous monitoring of organs-on-chips: Current challenges and potential solutions. Biosens Bioelectron 2023; 224:115057. [PMID: 36640548 DOI: 10.1016/j.bios.2022.115057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023]
Abstract
Organs-on-chips (OoCs) are biomimetic in vitro systems based on microfluidic cell cultures that recapitulate the in vivo physicochemical microenvironments and the physiologies and key functional units of specific human organs. These systems are versatile and can be customized to investigate organ-specific physiology, pathology, or pharmacology. They are more physiologically relevant than traditional two-dimensional cultures, can potentially replace the animal models or reduce the use of these models, and represent a unique opportunity for the development of personalized medicine when combined with human induced pluripotent stem cells. Continuous monitoring of important quality parameters of OoCs via a label-free, non-destructive, reliable, high-throughput, and multiplex method is critical for assessing the conditions of these systems and generating relevant analytical data; moreover, elaboration of quality predictive models is required for clinical trials of OoCs. Presently, these analytical data are obtained by manual or automatic sampling and analyzed using single-point, off-chip traditional methods. In this review, we describe recent efforts to integrate biosensing technologies into OoCs for monitoring the physiologies, functions, and physicochemical microenvironments of OoCs. Furthermore, we present potential alternative solutions to current challenges and future directions for the application of artificial intelligence in the development of OoCs and cyber-physical systems. These "smart" OoCs can learn and make autonomous decisions for process optimization, self-regulation, and data analysis.
Collapse
Affiliation(s)
- Jonathan Sabaté Del Río
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea
| | - Jooyoung Ro
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea; Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Heejeong Yoon
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Tae-Eun Park
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea.
| | - Yoon-Kyoung Cho
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea; Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea.
| |
Collapse
|
20
|
Biosensor integrated tissue chips and their applications on Earth and in space. Biosens Bioelectron 2023; 222:114820. [PMID: 36527831 PMCID: PMC10143284 DOI: 10.1016/j.bios.2022.114820] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 09/24/2022] [Accepted: 10/13/2022] [Indexed: 12/27/2022]
Abstract
The development of space exploration technologies has positively impacted everyday life on Earth in terms of communication, environmental, social, and economic perspectives. The human body constantly fluctuates during spaceflight, even for a short-term mission. Unfortunately, technology is evolving faster than humans' ability to adapt, and many therapeutics entering clinical trials fail even after being subjected to vigorous in vivo testing due to toxicity and lack of efficacy. Therefore, tissue chips (also mentioned as organ-on-a-chip) with biosensors are being developed to compensate for the lack of relevant models to help improve the drug development process. There has been a push to monitor cell and tissue functions, based on their biological signals and utilize the integration of biosensors into tissue chips in space to monitor and assess cell microenvironment in real-time. With the collaboration between the Center for the Advancement of Science in Space (CASIS), the National Aeronautics and Space Administration (NASA) and other partners, they are providing the opportunities to study the effects of microgravity environment has on the human body. Institutions such as the National Institute of Health (NIH) and National Science Foundation (NSF) are partnering with CASIS and NASA to utilize tissue chips onboard the International Space Station (ISS). This article reviews the endless benefits of space technology, the development of integrated biosensors in tissue chips and their applications to better understand human biology, physiology, and diseases in space and on Earth, followed by future perspectives of tissue chip applications on Earth and in space.
Collapse
|
21
|
Ngo H, Amartumur S, Tran VTA, Tran M, Diep YN, Cho H, Lee LP. In Vitro Tumor Models on Chip and Integrated Microphysiological Analysis Platform (MAP) for Life Sciences and High-Throughput Drug Screening. BIOSENSORS 2023; 13:231. [PMID: 36831997 PMCID: PMC9954135 DOI: 10.3390/bios13020231] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/23/2023] [Accepted: 01/31/2023] [Indexed: 06/18/2023]
Abstract
The evolution of preclinical in vitro cancer models has led to the emergence of human cancer-on-chip or microphysiological analysis platforms (MAPs). Although it has numerous advantages compared to other models, cancer-on-chip technology still faces several challenges such as the complexity of the tumor microenvironment and integrating multiple organs to be widely accepted in cancer research and therapeutics. In this review, we highlight the advancements in cancer-on-chip technology in recapitulating the vital biological features of various cancer types and their applications in life sciences and high-throughput drug screening. We present advances in reconstituting the tumor microenvironment and modeling cancer stages in breast, brain, and other types of cancer. We also discuss the relevance of MAPs in cancer modeling and precision medicine such as effect of flow on cancer growth and the short culture period compared to clinics. The advanced MAPs provide high-throughput platforms with integrated biosensors to monitor real-time cellular responses applied in drug development. We envision that the integrated cancer MAPs has a promising future with regard to cancer research, including cancer biology, drug discovery, and personalized medicine.
Collapse
Affiliation(s)
- Huyen Ngo
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sarnai Amartumur
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Van Thi Ai Tran
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Minh Tran
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yen N. Diep
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hansang Cho
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Luke P. Lee
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Bioengineering, University of California at Berkeley, Berkeley, CA 94720, USA
- Department of Electrical Engineering and Computer Science, University of California at Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
22
|
Johnson A, Reimer S, Childres R, Cupp G, Kohs TCL, McCarty OJT, Kang Y(A. The Applications and Challenges of the Development of In Vitro Tumor Microenvironment Chips. Cell Mol Bioeng 2023; 16:3-21. [PMID: 36660587 PMCID: PMC9842840 DOI: 10.1007/s12195-022-00755-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 12/07/2022] [Indexed: 12/27/2022] Open
Abstract
The tumor microenvironment (TME) plays a critical, yet mechanistically elusive role in tumor development and progression, as well as drug resistance. To better understand the pathophysiology of the complex TME, a reductionist approach has been employed to create in vitro microfluidic models called "tumor chips". Herein, we review the fabrication processes, applications, and limitations of the tumor chips currently under development for use in cancer research. Tumor chips afford capabilities for real-time observation, precise control of microenvironment factors (e.g. stromal and cellular components), and application of physiologically relevant fluid shear stresses and perturbations. Applications for tumor chips include drug screening and toxicity testing, assessment of drug delivery modalities, and studies of transport and interactions of immune cells and circulating tumor cells with primary tumor sites. The utility of tumor chips is currently limited by the ability to recapitulate the nuances of tumor physiology, including extracellular matrix composition and stiffness, heterogeneity of cellular components, hypoxic gradients, and inclusion of blood cells and the coagulome in the blood microenvironment. Overcoming these challenges and improving the physiological relevance of in vitro tumor models could provide powerful testing platforms in cancer research and decrease the need for animal and clinical studies.
Collapse
Affiliation(s)
- Annika Johnson
- Department of Mechanical, Civil, and Biomedical Engineering, George Fox University, 414 N. Meridian Street, #6088, Newberg, OR 97132 USA
| | - Samuel Reimer
- Department of Mechanical, Civil, and Biomedical Engineering, George Fox University, 414 N. Meridian Street, #6088, Newberg, OR 97132 USA
| | - Ryan Childres
- Department of Mechanical, Civil, and Biomedical Engineering, George Fox University, 414 N. Meridian Street, #6088, Newberg, OR 97132 USA
| | - Grace Cupp
- Department of Mechanical, Civil, and Biomedical Engineering, George Fox University, 414 N. Meridian Street, #6088, Newberg, OR 97132 USA
| | - Tia C. L. Kohs
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239 USA
| | - Owen J. T. McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239 USA
- Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97201 USA
| | - Youngbok (Abraham) Kang
- Department of Mechanical, Civil, and Biomedical Engineering, George Fox University, 414 N. Meridian Street, #6088, Newberg, OR 97132 USA
| |
Collapse
|
23
|
Cho S, Lee S, Ahn SI. Design and engineering of organ-on-a-chip. Biomed Eng Lett 2023; 13:97-109. [PMID: 36620430 PMCID: PMC9806813 DOI: 10.1007/s13534-022-00258-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/21/2022] [Accepted: 12/20/2022] [Indexed: 01/03/2023] Open
Abstract
Organ-on-a-chip (OOC) is an emerging interdisciplinary technology that reconstitutes the structure, function, and physiology of human tissues as an alternative to conventional preclinical models for drug screening. Over the last decade, substantial progress has been made in mimicking tissue- and organ-level functions on chips through technical advances in biomaterials, stem cell engineering, microengineering, and microfluidic technologies. Structural and engineering constituents, as well as biological components, are critical factors to be considered to reconstitute the tissue function and microenvironment on chips. In this review, we highlight critical engineering technologies for reconstructing the tissue microarchitecture and dynamic spatiotemporal microenvironment in OOCs. We review the technological advances in the field of OOCs for a range of applications, including systemic analysis tools that can be integrated with OOCs, multiorgan-on-chips, and large-scale manufacturing. We then discuss the challenges and future directions for the development of advanced end-user-friendly OOC systems for a wide range of applications.
Collapse
Affiliation(s)
- Sujin Cho
- School of Mechanical Engineering, Pusan National University, Busan, 46241 Republic of Korea
| | - Sumi Lee
- School of Mechanical Engineering, Pusan National University, Busan, 46241 Republic of Korea
| | - Song Ih Ahn
- School of Mechanical Engineering, Pusan National University, Busan, 46241 Republic of Korea
| |
Collapse
|
24
|
Pal A, Kaswan K, Barman SR, Lin YZ, Chung JH, Sharma MK, Liu KL, Chen BH, Wu CC, Lee S, Choi D, Lin ZH. Microfluidic nanodevices for drug sensing and screening applications. Biosens Bioelectron 2023; 219:114783. [PMID: 36257116 PMCID: PMC9533638 DOI: 10.1016/j.bios.2022.114783] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 09/18/2022] [Accepted: 10/01/2022] [Indexed: 11/03/2022]
Abstract
The outbreak of pandemics (e.g., severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2 in 2019), influenza A viruses (H1N1 in 2009), etc.), and worldwide spike in the aging population have created unprecedented urgency for developing new drugs to improve disease treatment. As a result, extensive efforts have been made to design novel techniques for efficient drug monitoring and screening, which form the backbone of drug development. Compared to traditional techniques, microfluidics-based platforms have emerged as promising alternatives for high-throughput drug screening due to their inherent miniaturization characteristics, low sample consumption, integration, and compatibility with diverse analytical strategies. Moreover, the microfluidic-based models utilizing human cells to produce in-vitro biomimetics of the human body pave new ways to predict more accurate drug effects in humans. This review provides a comprehensive summary of different microfluidics-based drug sensing and screening strategies and briefly discusses their advantages. Most importantly, an in-depth outlook of the commonly used detection techniques integrated with microfluidic chips for highly sensitive drug screening is provided. Then, the influence of critical parameters such as sensing materials and microfluidic platform geometries on screening performance is summarized. This review also outlines the recent applications of microfluidic approaches for screening therapeutic and illicit drugs. Moreover, the current challenges and the future perspective of this research field is elaborately highlighted, which we believe will contribute immensely towards significant achievements in all aspects of drug development.
Collapse
Affiliation(s)
- Arnab Pal
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan; International Intercollegiate PhD Program, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Kuldeep Kaswan
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan; International Intercollegiate PhD Program, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Snigdha Roy Barman
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan; International Intercollegiate PhD Program, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yu-Zih Lin
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Jun-Hsuan Chung
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Manish Kumar Sharma
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Kuei-Lin Liu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Bo-Huan Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan; International Intercollegiate PhD Program, National Tsing Hua University, Hsinchu, 30013, Taiwan; Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, 333, Taiwan
| | - Chih-Cheng Wu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan; Center of Quality Management, National Taiwan University Hospital, Hsinchu Branch, Hsinchu, 30059, Taiwan; College of Medicine, National Taiwan University, Taipei, 10051, Taiwan; Institute of Cellular and System Medicine, National Health Research Institute, Zhunan, 35053, Taiwan
| | - Sangmin Lee
- School of Mechanical Engineering, Chung-Ang University, Seoul, 06974, South Korea.
| | - Dongwhi Choi
- Department of Mechanical Engineering (Integrated Engineering Program), Kyung Hee University, Gyeonggi, 17104, South Korea.
| | - Zong-Hong Lin
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan; International Intercollegiate PhD Program, National Tsing Hua University, Hsinchu, 30013, Taiwan; Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan; Department of Chemistry, National Tsing Hua University, Hsinchu, 30013, Taiwan; Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 30013, Taiwan; Department of Mechanical Engineering (Integrated Engineering Program), Kyung Hee University, Gyeonggi, 17104, South Korea.
| |
Collapse
|
25
|
Gilbert DF, Friedrich O, Wiest J. Assaying Proliferation Characteristics of Cells Cultured Under Static Versus Periodic Conditions. Methods Mol Biol 2023; 2644:35-45. [PMID: 37142914 DOI: 10.1007/978-1-0716-3052-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Two-dimensional in vitro culture models are widely being employed for assessing a vast variety of biological questions in different scientific fields. Common in vitro culture models are typically maintained under static conditions, where the surrounding culture medium is replaced every few days-typically every 48 to 72 h-with the aim to remove metabolites and to replenish nutrients. Although this approach is sufficient for supporting cellular survival and proliferation, static culture conditions do mostly not reflect the in vivo situation where cells are continuously being perfused by extracellular fluid, and thus, create a less-physiological environment. In order to evaluate whether the proliferation characteristics of cells in 2D culture maintained under static conditions differ from cells kept in a dynamic environment, in this chapter, we provide a protocol for differential analysis of cellular growth under static versus pulsed-perfused conditions, mimicking continuous replacement of extracellular fluid in the physiological environment. The protocol involves long-term life-cell high-content time-lapse imaging of fluorescent cells at 37 °C and ambient CO2 concentration using multi-parametric biochips applicable for microphysiological analysis of cellular vitality. We provide instructions and useful information for (i) the culturing of cells in biochips, (ii) setup of cell-laden biochips for culturing cells under static and pulsed-perfused conditions, (iii) long-term life-cell high-content time-lapse imaging of fluorescent cells in biochips, and (iv) quantification of cellular proliferation from image series generated from imaging of differentially cultured cells.
Collapse
Affiliation(s)
- Daniel F Gilbert
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering (CBI), Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany.
| | - Oliver Friedrich
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering (CBI), Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | | |
Collapse
|
26
|
Imparato G, Urciuolo F, Mazio C, Netti PA. Capturing the spatial and temporal dynamics of tumor stroma for on-chip optimization of microenvironmental targeting nanomedicine. LAB ON A CHIP 2022; 23:25-43. [PMID: 36305728 DOI: 10.1039/d2lc00611a] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Malignant cells grow in a complex microenvironment that plays a key role in cancer progression. The "dynamic reciprocity" existing between cancer cells and their microenvironment is involved in cancer differentiation, proliferation, invasion, metastasis, and drug response. Therefore, understanding the molecular mechanisms underlying the crosstalk between cancer cells and their surrounding tissue (i.e., tumor stroma) and how this interplay affects the disease progression is fundamental to design and validate novel nanotherapeutic approaches. As an important regulator of tumor progression, metastasis and therapy resistance, the extracellular matrix of tumors, the acellular component of the tumor microenvironment, has been identified as very promising target of anticancer treatment, revolutionizing the traditional therapeutic paradigm that sees the neoplastic cells as the preferential objective to fight cancer. To design and to validate such a target therapy, advanced 3D preclinical models are necessary to correctly mimic the complex, dynamic and heterogeneous tumor microenvironment. In addition, the recent advancement in microfluidic technology allows fine-tuning and controlling microenvironmental parameters in tissue-on-chip devices in order to emulate the in vivo conditions. In this review, after a brief description of the origin of tumor microenvironment heterogeneity, some examples of nanomedicine approaches targeting the tumor microenvironment have been reported. Further, how advanced 3D bioengineered tumor models coupled with a microfluidic device can improve the design and testing of anti-cancer nanomedicine targeting the tumor microenvironment has been discussed. We highlight that the presence of a dynamic extracellular matrix, able to capture the spatiotemporal heterogeneity of tumor stroma, is an indispensable requisite for tumor-on-chip model and nanomedicine testing.
Collapse
Affiliation(s)
- Giorgia Imparato
- Center for Advanced Biomaterials for Health Care@CRIB Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci n. 53, 80125 Napoli, Italy.
| | - Francesco Urciuolo
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) and Interdisciplinary Research Centre on Biomaterials (CRIB), University of Napoli Federico II, P.le Tecchio 80, 80125 Napoli, Italy
| | - Claudia Mazio
- Center for Advanced Biomaterials for Health Care@CRIB Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci n. 53, 80125 Napoli, Italy.
| | - Paolo A Netti
- Center for Advanced Biomaterials for Health Care@CRIB Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci n. 53, 80125 Napoli, Italy.
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) and Interdisciplinary Research Centre on Biomaterials (CRIB), University of Napoli Federico II, P.le Tecchio 80, 80125 Napoli, Italy
| |
Collapse
|
27
|
Condelipes PGM, Fontes PM, Godinho-Santos A, Brás EJS, Marques V, Afonso MB, Rodrigues CMP, Chu V, Gonçalves J, Conde JP. Towards personalized antibody cancer therapy: development of a microfluidic cell culture device for antibody selection. LAB ON A CHIP 2022; 22:4717-4728. [PMID: 36349999 DOI: 10.1039/d2lc00918h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Antibody therapy has been one of the most successful therapies for a wide range of diseases, including cancer. One way of expediting antibody therapy development is through phage display technology. Here, by screening thousands of randomly assembled peptide sequences, it is possible to identify potential therapeutic candidates. Conventional screening technologies do not accommodate perfusion through the system, as is the case of standard plate-based cultures. This leads to a poor translation of the experimental results obtained in vitro when moving to a more physiologically relevant setting, such as the case of preclinical animal models or clinical trials. Microfluidics is a technology that can improve screening efficacy by replicating more physiologically relevant conditions such as shear stress. In this work, a polydimethylsiloxane/polystyrene-based microfluidic system for a continuously perfused culture of cancer cells is reported. Human colorectal adenocarcinoma cells (HCT116) expressing CXCR4 were used as a cell target. Fluorescently labeled M13 phages anti-CXCR4 were used to study the efficiency of the microfluidic system as a tool to study the binding kinetics of the engineered bacteriophages. Using our microfluidic platform, we estimated a dissociation constant of 0.45 pM for the engineered phage. Additionally, a receptor internalization assay was developed using SDF-1α to verify phage specificity to the CXCR4 receptor. Upon receptor internalization there was a signal reduction, proving that the anti-CXCR4 fluorescently labelled M13 phages bound specifically to the CXCR4 receptor. The simplicity and ease of use of the microfluidic device design presented in this work can form the basis of a generic platform that facilitates the study and optimization of therapies based on interaction with biological entities such as mammalian cells.
Collapse
Affiliation(s)
- Pedro G M Condelipes
- Instituto de Engenharia de Sistemas e Computadores - Microsistemas e Nanotecnologias (INESC MN), Lisbon, Portugal
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.
| | - Pedro Mendes Fontes
- Instituto de Engenharia de Sistemas e Computadores - Microsistemas e Nanotecnologias (INESC MN), Lisbon, Portugal
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Godinho-Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Eduardo J S Brás
- Instituto de Engenharia de Sistemas e Computadores - Microsistemas e Nanotecnologias (INESC MN), Lisbon, Portugal
- IBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Vanda Marques
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Marta B Afonso
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Cecília M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Virginia Chu
- Instituto de Engenharia de Sistemas e Computadores - Microsistemas e Nanotecnologias (INESC MN), Lisbon, Portugal
| | - João Gonçalves
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - João Pedro Conde
- Instituto de Engenharia de Sistemas e Computadores - Microsistemas e Nanotecnologias (INESC MN), Lisbon, Portugal
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
28
|
Dornhof J, Zieger V, Kieninger J, Frejek D, Zengerle R, Urban GA, Kartmann S, Weltin A. Bioprinting-based automated deposition of single cancer cell spheroids into oxygen sensor microelectrode wells. LAB ON A CHIP 2022; 22:4369-4381. [PMID: 36254669 DOI: 10.1039/d2lc00705c] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Three-dimensional (3D) cell agglomerates, such as microtissues, organoids, and spheroids, become increasingly relevant in biomedicine. They can provide in vitro models that recapitulate functions of the original tissue in the body and have applications in cancer research. For example, they are widely used in organ-on-chip systems. Microsensors can provide essential real-time information on cell metabolism as well as the reliability and quality of culture conditions. The combination of sensors and 3D cell cultures, especially single spheroids, is challenging in terms of reproducible formation, manipulation, and access to spheroids, precise positioning near sensors, and high cell-to-volume ratios to obtain meaningful biosignals in the most parallel approach possible. To overcome this challenge, we combined state-of-the-art bioprinting techniques to automatically print tumour spheroids directly into microwells of a chip-based electrochemical oxygen sensor array. We demonstrated highly accurate and reproducible spheroid formation (diameter of approx. 200 μm) and a spheroid deposition precision of 25 μm within a volume of 22 nl per droplet. Microstructures and hydrogel-coated microwells allowed the placement of single MCF-7 breast cancer spheroids close to the sensor electrodes. The microelectrode wells were sealed for oxygen measurements within a 55 nl volume for fast concentration changes. Accurate and stable amperometric oxygen sensor performance was demonstrated from atmospheric to anoxic regions. Cellular respiration rates from single tumour spheroids in the range of 450-850 fmol min-1 were determined, and alterations of cell metabolism upon drug exposure were shown. Our results uniquely combine bioprinting with 3D cell culture monitoring and demonstrate the much-needed effort for facilitation, parallelization, sensor integration, and drug delivery in 3D cell culture and organ-on-chip experiments. The workflow has a high degree of automation and potential for scalability. In order to achieve greater flexibility in the automation of spheroid formation and trapping, we employed a method based on drop-on-demand liquid handling systems, instead of the typical on-chip approach commonly used in microfluidics. Its relevance ranges from fundamental metabolic research over standardization of cell culture experiments and toxicological studies, to personalized medicine, e.g. patient-specific chemotherapy.
Collapse
Affiliation(s)
- Johannes Dornhof
- Laboratory for Sensors, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany.
| | - Viktoria Zieger
- Laboratory for MEMS Applications, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
| | - Jochen Kieninger
- Laboratory for Sensors, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany.
| | | | - Roland Zengerle
- Laboratory for MEMS Applications, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
- Hahn-Schickard, Freiburg, Germany
| | - Gerald A Urban
- Laboratory for Sensors, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany.
| | - Sabrina Kartmann
- Laboratory for MEMS Applications, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
- Hahn-Schickard, Freiburg, Germany
| | - Andreas Weltin
- Laboratory for Sensors, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
29
|
Mukherjee P, Park SH, Pathak N, Patino CA, Bao G, Espinosa HD. Integrating Micro and Nano Technologies for Cell Engineering and Analysis: Toward the Next Generation of Cell Therapy Workflows. ACS NANO 2022; 16:15653-15680. [PMID: 36154011 DOI: 10.1021/acsnano.2c05494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The emerging field of cell therapy offers the potential to treat and even cure a diverse array of diseases for which existing interventions are inadequate. Recent advances in micro and nanotechnology have added a multitude of single cell analysis methods to our research repertoire. At the same time, techniques have been developed for the precise engineering and manipulation of cells. Together, these methods have aided the understanding of disease pathophysiology, helped formulate corrective interventions at the cellular level, and expanded the spectrum of available cell therapeutic options. This review discusses how micro and nanotechnology have catalyzed the development of cell sorting, cellular engineering, and single cell analysis technologies, which have become essential workflow components in developing cell-based therapeutics. The review focuses on the technologies adopted in research studies and explores the opportunities and challenges in combining the various elements of cell engineering and single cell analysis into the next generation of integrated and automated platforms that can accelerate preclinical studies and translational research.
Collapse
Affiliation(s)
- Prithvijit Mukherjee
- Department of Mechanical Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, Illinois 60208, United States
| | - So Hyun Park
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, Texas 77030, United States
| | - Nibir Pathak
- Department of Mechanical Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, Illinois 60208, United States
| | - Cesar A Patino
- Department of Mechanical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Gang Bao
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, Texas 77030, United States
| | - Horacio D Espinosa
- Department of Mechanical Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
30
|
Mou L, Mandal K, Mecwan MM, Hernandez AL, Maity S, Sharma S, Herculano RD, Kawakita S, Jucaud V, Dokmeci MR, Khademhosseini A. Integrated biosensors for monitoring microphysiological systems. LAB ON A CHIP 2022; 22:3801-3816. [PMID: 36074812 PMCID: PMC9635816 DOI: 10.1039/d2lc00262k] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Microphysiological systems (MPSs), also known as organ-on-a-chip models, aim to recapitulate the functional components of human tissues or organs in vitro. Over the last decade, with the advances in biomaterials, 3D bioprinting, and microfluidics, numerous MPSs have emerged with applications to study diseased and healthy tissue models. Various organs have been modeled using MPS technology, such as the heart, liver, lung, and blood-brain barrier. An important aspect of in vitro modeling is the accurate phenotypical and functional characterization of the modeled organ. However, most conventional characterization methods are invasive and destructive and do not allow continuous monitoring of the cells in culture. On the other hand, microfluidic biosensors enable in-line, real-time sensing of target molecules with an excellent limit of detection and in a non-invasive manner, thereby effectively overcoming the limitation of the traditional techniques. Consequently, microfluidic biosensors have been increasingly integrated into MPSs and used for in-line target detection. This review discusses the state-of-the-art microfluidic biosensors by providing specific examples, detailing their main advantages in monitoring MPSs, and highlighting current developments in this field. Finally, we describe the remaining challenges and potential future developments to advance the current state-of-the-art in integrated microfluidic biosensors.
Collapse
Affiliation(s)
- Lei Mou
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
- Department of Clinical Laboratory, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, No. 63 Duobao Road, Liwan District, Guangzhou, Guangdong, P. R. China
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
| | - Marvin Magan Mecwan
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
| | - Ana Lopez Hernandez
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
| | - Surjendu Maity
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
| | - Saurabh Sharma
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
| | - Rondinelli Donizetti Herculano
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
- Department of Bioprocess and Biotechnology Engineering, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP 14801-902, Brazil
| | - Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
| | - Mehmet Remzi Dokmeci
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
| |
Collapse
|
31
|
Wongsa C, Udomsom S, Budwong A, Kiwfo K, Grudpan K, Paengnakorn P. Sequential Injection Amperometric System Coupling with Bioreactor for In-Line Glucose Monitoring in Cell Culture Application. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27196665. [PMID: 36235202 PMCID: PMC9573359 DOI: 10.3390/molecules27196665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/28/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022]
Abstract
We proposed a specially designed sequential injection (SI) amperometric system coupling with a bioreactor for in-line glucose monitoring in cell culture. The system is composed of three main parts which are the bioreactor, SI system, and electrochemical detection unit. The bioreactor accommodates six individual cell culture units which can be operated separately under different conditions. The SI system enables automatic in-line sampling and in-line sample dilution, with a specially designed mixing unit; therefore, it has the benefits of fast analysis time and less contamination risk. The use of 3D-printed microfluidic components, a mixing channel, and a flow cell helped to reduce operational time and sample volume. A disposable screen-printed electrode (SPE), modified with glucose oxidase (GOD), carbon nanotube, and gold nanoparticle, was used for detection. The developed system provided a linear range up to 3.8 mM glucose in cell culture media. In order to work with cell culture in higher glucose media, the in-line sample dilution can be applied. The developed SI system was demonstrated with mouse fibroblast (L929) cell culture. The results show that glucose concentration obtained from the SI system is comparable with that obtained from the conventional colorimetric method. This work can be further developed and applied for in vitro cell-based experiments in biomedical research.
Collapse
Affiliation(s)
- Chanyanut Wongsa
- Biomedical Engineering Institute, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Suruk Udomsom
- Biomedical Engineering Institute, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Apiwat Budwong
- Biomedical Engineering Institute, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kanokwan Kiwfo
- Center of Excellence for Innovation in Analytical Science and Technology for Biodiversity-Based Economic and Society (I-ANALY-S-T_B.BES-CMU), Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kate Grudpan
- Center of Excellence for Innovation in Analytical Science and Technology for Biodiversity-Based Economic and Society (I-ANALY-S-T_B.BES-CMU), Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Pathinan Paengnakorn
- Biomedical Engineering Institute, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence for Innovation in Analytical Science and Technology for Biodiversity-Based Economic and Society (I-ANALY-S-T_B.BES-CMU), Chiang Mai University, Chiang Mai 50200, Thailand
- Correspondence:
| |
Collapse
|
32
|
Kann SH, Shaughnessey EM, Coppeta JR, Azizgolshani H, Isenberg BC, Vedula EM, Zhang X, Charest JL. Measurement of oxygen consumption rates of human renal proximal tubule cells in an array of organ-on-chip devices to monitor drug-induced metabolic shifts. MICROSYSTEMS & NANOENGINEERING 2022; 8:109. [PMID: 36187891 PMCID: PMC9519964 DOI: 10.1038/s41378-022-00442-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 06/16/2023]
Abstract
Measurement of cell metabolism in moderate-throughput to high-throughput organ-on-chip (OOC) systems would expand the range of data collected for studying drug effects or disease in physiologically relevant tissue models. However, current measurement approaches rely on fluorescent imaging or colorimetric assays that are focused on endpoints, require labels or added substrates, and lack real-time data. Here, we integrated optical-based oxygen sensors in a high-throughput OOC platform and developed an approach for monitoring cell metabolic activity in an array of membrane bilayer devices. Each membrane bilayer device supported a culture of human renal proximal tubule epithelial cells on a porous membrane suspended between two microchannels and exposed to controlled, unidirectional perfusion and physiologically relevant shear stress for several days. For the first time, we measured changes in oxygen in a membrane bilayer format and used a finite element analysis model to estimate cell oxygen consumption rates (OCRs), allowing comparison with OCRs from other cell culture systems. Finally, we demonstrated label-free detection of metabolic shifts in human renal proximal tubule cells following exposure to FCCP, a drug known for increasing cell oxygen consumption, as well as oligomycin and antimycin A, drugs known for decreasing cell oxygen consumption. The capability to measure cell OCRs and detect metabolic shifts in an array of membrane bilayer devices contained within an industry standard microtiter plate format will be valuable for analyzing flow-responsive and physiologically complex tissues during drug development and disease research.
Collapse
Affiliation(s)
- Samuel H. Kann
- Draper Scholar, 555 Technology Square, Cambridge, MA 02139 USA
- Department of Mechanical Engineering, Boston University, 110 Cummington Mall, Boston, MA 02215 USA
| | - Erin M. Shaughnessey
- Draper Scholar, 555 Technology Square, Cambridge, MA 02139 USA
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155 USA
| | | | | | | | | | - Xin Zhang
- Department of Mechanical Engineering, Boston University, 110 Cummington Mall, Boston, MA 02215 USA
| | - Joseph L. Charest
- Draper, 555 Technology Square, Cambridge, MA 02139 USA
- Present Address: Biogen, 225 Binney Street, Cambridge, MA 02142 USA
| |
Collapse
|
33
|
Giampetruzzi L, Blasi L, Barca A, Sciurti E, Verri T, Casino F, Siciliano P, Francioso L. Advances in Trans-Epithelial Electrical Resistance (TEER) monitoring integration in an Intestinal Barrier-on-Chip (IBoC) platform with microbubbles-tolerant analytical method. SENSING AND BIO-SENSING RESEARCH 2022. [DOI: 10.1016/j.sbsr.2022.100512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
34
|
Seo Y, Bang S, Son J, Kim D, Jeong Y, Kim P, Yang J, Eom JH, Choi N, Kim HN. Brain physiome: A concept bridging in vitro 3D brain models and in silico models for predicting drug toxicity in the brain. Bioact Mater 2022; 13:135-148. [PMID: 35224297 PMCID: PMC8843968 DOI: 10.1016/j.bioactmat.2021.11.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/01/2021] [Accepted: 11/06/2021] [Indexed: 12/12/2022] Open
Abstract
In the last few decades, adverse reactions to pharmaceuticals have been evaluated using 2D in vitro models and animal models. However, with increasing computational power, and as the key drivers of cellular behavior have been identified, in silico models have emerged. These models are time-efficient and cost-effective, but the prediction of adverse reactions to unknown drugs using these models requires relevant experimental input. Accordingly, the physiome concept has emerged to bridge experimental datasets with in silico models. The brain physiome describes the systemic interactions of its components, which are organized into a multilevel hierarchy. Because of the limitations in obtaining experimental data corresponding to each physiome component from 2D in vitro models and animal models, 3D in vitro brain models, including brain organoids and brain-on-a-chip, have been developed. In this review, we present the concept of the brain physiome and its hierarchical organization, including cell- and tissue-level organizations. We also summarize recently developed 3D in vitro brain models and link them with the elements of the brain physiome as a guideline for dataset collection. The connection between in vitro 3D brain models and in silico modeling will lead to the establishment of cost-effective and time-efficient in silico models for the prediction of the safety of unknown drugs.
Collapse
Affiliation(s)
- Yoojin Seo
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Seokyoung Bang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jeongtae Son
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Dongsup Kim
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Yong Jeong
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Pilnam Kim
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jihun Yang
- Next&Bio Inc., Seoul, 02841, Republic of Korea
| | - Joon-Ho Eom
- Medical Device Research Division, National Institute of Food and Drug Safety Evaluation, Cheongju, 28159, Republic of Korea
| | - Nakwon Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Hong Nam Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, Republic of Korea
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul, 03722, Republic of Korea
| |
Collapse
|
35
|
Wu CC, Wang CJ, Chang CLT, Shiku H, Wang YR, Yan JD, Ding SJ. Dissolved Oxygen-Sensing Chip Integrating an Open Container Connected with a Position-Raised Channel for Estimation of Cellular Mitochondrial Activity. ACS Sens 2022; 7:1808-1818. [PMID: 35748570 DOI: 10.1021/acssensors.1c02287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The measurement of oxygen consumption of adherent cells is a profoundly important issue for estimating the bioenergetic health and metabolism activity of cells. The study describes the construction of a microfluidic chip consisting of an open container connected with a position-raised channel and dissolved oxygen (DO)-sensing gold ultramicroelectrodes for quantifying the oxygen consumption rate (OCR) of adherent cells. The microfluidic chip design can reduce the action of shear force on the adherent cells during medium replacement. The residual concentration of analytes in the open container was only 4.4% after solution replacement via the position-raised channel. The DO reduction current measured by ultramicroelectrodes averaged in the range of 40-60 s presented high reproducibility with a 1.1% relative standard deviation suitable for OCR calculation. After short-term (90 min) cultivation, the microfluidic chip can monitor the time-dependent change in the OCR of 3T3-L1 cells for several hours by repeatedly replacing the culture medium or with the stimulation of different mitochondrial inhibitors. The presented microfluidic cell-based chip has great promise for drug screening and chemosensitivity testing by measuring OCR to evaluate the mitochondrial function of adherent cells.
Collapse
Affiliation(s)
- Ching-Chou Wu
- Department of Bio-Industrial Mechatronics Engineering, National Chung Hsing University, Taichung City 402, Taiwan.,Innovation and Development Center of Sustainable Agriculture, National Chung Hsing University, Taichung City 402, Taiwan
| | - Chieh-Jen Wang
- Department of Bio-Industrial Mechatronics Engineering, National Chung Hsing University, Taichung City 402, Taiwan
| | | | - Hitoshi Shiku
- Graduate School of Engineering, Tohoku University, 6-6-11 Aramaki-aza Aoba, Aoba-ku, Sendai 980-8579, Japan
| | - Yu-Ren Wang
- Department of Bio-Industrial Mechatronics Engineering, National Chung Hsing University, Taichung City 402, Taiwan
| | - Jia-De Yan
- Department of Bio-Industrial Mechatronics Engineering, National Chung Hsing University, Taichung City 402, Taiwan
| | - Shinn-Jyh Ding
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung City 402, Taiwan, ROC.,Institute of Oral Science, Chung Shan Medical University, Taichung City 402, Taiwan
| |
Collapse
|
36
|
A three-dimensional electrochemical biosensor integrated with hydrogel for cells culture and lactate release monitoring. J Electroanal Chem (Lausanne) 2022. [DOI: 10.1016/j.jelechem.2022.116338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
37
|
Mai P, Hampl J, Baca M, Brauer D, Singh S, Weise F, Borowiec J, Schmidt A, Küstner JM, Klett M, Gebinoga M, Schroeder IS, Markert UR, Glahn F, Schumann B, Eckstein D, Schober A. MatriGrid® Based Biological Morphologies: Tools for 3D Cell Culturing. Bioengineering (Basel) 2022; 9:bioengineering9050220. [PMID: 35621498 PMCID: PMC9138054 DOI: 10.3390/bioengineering9050220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 02/06/2023] Open
Abstract
Recent trends in 3D cell culturing has placed organotypic tissue models at another level. Now, not only is the microenvironment at the cynosure of this research, but rather, microscopic geometrical parameters are also decisive for mimicking a tissue model. Over the years, technologies such as micromachining, 3D printing, and hydrogels are making the foundation of this field. However, mimicking the topography of a particular tissue-relevant substrate can be achieved relatively simply with so-called template or morphology transfer techniques. Over the last 15 years, in one such research venture, we have been investigating a micro thermoforming technique as a facile tool for generating bioinspired topographies. We call them MatriGrid®s. In this research account, we summarize our learning outcome from this technique in terms of the influence of 3D micro morphologies on different cell cultures that we have tested in our laboratory. An integral part of this research is the evolution of unavoidable aspects such as possible label-free sensing and fluidic automatization. The development in the research field is also documented in this account.
Collapse
Affiliation(s)
- Patrick Mai
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Jörg Hampl
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
- Correspondence: (J.H.); (A.S.); Tel.: +49-3677-6933387 (A.S.)
| | - Martin Baca
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Dana Brauer
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Sukhdeep Singh
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Frank Weise
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Justyna Borowiec
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - André Schmidt
- Placenta Lab, Department of Obstetrics, Jena University Hospital, 07747 Jena, Germany; (A.S.); (U.R.M.)
| | - Johanna Merle Küstner
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Maren Klett
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Michael Gebinoga
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Insa S. Schroeder
- Biophysics Division, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany;
| | - Udo R. Markert
- Placenta Lab, Department of Obstetrics, Jena University Hospital, 07747 Jena, Germany; (A.S.); (U.R.M.)
| | - Felix Glahn
- Institute of Environmental Toxicology, Martin-Luther-University Halle-Wittenberg, 06097 Halle, Germany; (F.G.); (B.S.); (D.E.)
| | - Berit Schumann
- Institute of Environmental Toxicology, Martin-Luther-University Halle-Wittenberg, 06097 Halle, Germany; (F.G.); (B.S.); (D.E.)
| | - Diana Eckstein
- Institute of Environmental Toxicology, Martin-Luther-University Halle-Wittenberg, 06097 Halle, Germany; (F.G.); (B.S.); (D.E.)
| | - Andreas Schober
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
- Correspondence: (J.H.); (A.S.); Tel.: +49-3677-6933387 (A.S.)
| |
Collapse
|
38
|
Park SJ, Jung TH, Kim JH, Lee KY, Kim J, Ju J, Moon SH. In silico design and fabrication of an SFI chip-based microspheroid culture system. Biomater Sci 2022; 10:2991-3005. [PMID: 35521942 DOI: 10.1039/d2bm00250g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The emergence of microfluidic devices and computational fluid dynamics (CFD) has propelled the need for next-generation biomimetic cell culture platforms that are flexible for monitoring and regulation. Therefore, this study evaluated a CFD application in an in silico-designed and spheroid-based flow integration 3D cell culture chip (SFI chip) to illustrate cell culture, drug screening, cytokine delivery, and differentiation of cells in a platform that partially recapitulates the natural environment. Our results show that a flow rate of 0.05 mL h-1 or less induced no physical stress in the SFI chip (15 mm), and uniform cell spheroids (approximately 200 μm) were formed across the platform. The cultured cells were tested in several experimental contexts (co-culture, drug screening, cytokine delivery, and differentiation), demonstrating the usefulness of computational simulation in expediting discovery and simple and effective means to scale the production of standardized cell spheroids cultured under dynamic and natural conditions. Advanced cell culture technologies can be used to accelerate research and discovery and the preclinical and clinical development of cell and cell-free therapies for urgent medical needs.
Collapse
Affiliation(s)
- Soon-Jung Park
- Department of Medicine, Konkuk University School of Medicine, Seoul, Republic of Korea.,Stem Cell Research Institute, T&R Biofab Co. Ltd, Siheung, Republic of Korea.
| | - Taek-Hee Jung
- Department of Medicine, Konkuk University School of Medicine, Seoul, Republic of Korea.,Stem Cell Research Institute, T&R Biofab Co. Ltd, Siheung, Republic of Korea.
| | - Jong Hyun Kim
- Department of Biological Science, Hyupsung University, Hwasung, Republic of Korea
| | - Kyoung-Yong Lee
- Carbon Neutral Technology R&D Department, Korea Institute of Industrial Technology (KITECH), Cheonan, Republic of Korea
| | - Jeongyun Kim
- Department of Physics, College of Science & Technology, Dankook University, Cheonan, Chungnam, 31116, Republic of Korea.
| | - Jongil Ju
- Department of Physics, College of Science & Technology, Dankook University, Cheonan, Chungnam, 31116, Republic of Korea. .,Department of R&D, ABM Scientific Co., Cheonan, Republic of Korea
| | - Sung-Hwan Moon
- Department of Medicine, Konkuk University School of Medicine, Seoul, Republic of Korea.,Stem Cell Research Institute, T&R Biofab Co. Ltd, Siheung, Republic of Korea. .,Department of Animal Biotechnology, Sangji University, Wonju, Republic of Korea
| |
Collapse
|
39
|
Automated Analysis of Acetaminophen Toxicity on 3D HepaRG Cell Culture in Microbioreactor. Bioengineering (Basel) 2022; 9:bioengineering9050196. [PMID: 35621474 PMCID: PMC9137798 DOI: 10.3390/bioengineering9050196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 11/16/2022] Open
Abstract
Real-time monitoring of bioanalytes in organotypic cell cultivation devices is a major research challenge in establishing stand-alone diagnostic systems. Presently, no general technical facility is available that offers a plug-in system for bioanalytics in diversely available organotypic culture models. Therefore, each analytical device has to be tuned according to the microfluidic and interface environment of the 3D in vitro system. Herein, we report the design and function of a 3D automated culture and analysis device (3D-ACAD) which actively perfuses a custom-made 3D microbioreactor, samples the culture medium and simultaneously performs capillary-based flow ELISA. A microstructured MatriGrid® has been explored as a 3D scaffold for culturing HepaRG cells, with albumin investigated as a bioanalytical marker using flow ELISA. We investigated the effect of acetaminophen (APAP) on the albumin secretion of HepaRG cells over 96 h and compared this with the albumin secretion of 2D monolayer HepaRG cultures. Automated on-line monitoring of albumin secretion in the 3D in vitro mode revealed that the application of hepatotoxic drug-like APAP results in decreased albumin secretion. Furthermore, a higher sensitivity of the HepaRG cell culture in the automated 3D-ACAD system to APAP was observed compared to HepaRG cells cultivated as a monolayer. The results support the use of the 3D-ACAD model as a stand-alone device, working in real time and capable of analyzing the condition of the cell culture by measuring a functional analyte. Information obtained from our system is compared with conventional cell culture and plate ELISA, the results of which are presented herein.
Collapse
|
40
|
De Stefano P, Bianchi E, Dubini G. The impact of microfluidics in high-throughput drug-screening applications. BIOMICROFLUIDICS 2022; 16:031501. [PMID: 35646223 PMCID: PMC9142169 DOI: 10.1063/5.0087294] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/02/2022] [Indexed: 05/05/2023]
Abstract
Drug discovery is an expensive and lengthy process. Among the different phases, drug discovery and preclinical trials play an important role as only 5-10 of all drugs that begin preclinical tests proceed to clinical trials. Indeed, current high-throughput screening technologies are very expensive, as they are unable to dispense small liquid volumes in an accurate and quick way. Moreover, despite being simple and fast, drug screening assays are usually performed under static conditions, thus failing to recapitulate tissue-specific architecture and biomechanical cues present in vivo even in the case of 3D models. On the contrary, microfluidics might offer a more rapid and cost-effective alternative. Although considered incompatible with high-throughput systems for years, technological advancements have demonstrated how this gap is rapidly reducing. In this Review, we want to further outline the role of microfluidics in high-throughput drug screening applications by looking at the multiple strategies for cell seeding, compartmentalization, continuous flow, stimuli administration (e.g., drug gradients or shear stresses), and single-cell analyses.
Collapse
Affiliation(s)
- Paola De Stefano
- Laboratory of Biological Structure Mechanics, Department of Chemistry, Materials and Chemical Engineering “G. Natta,” Politecnico di Milano, Italy
| | - Elena Bianchi
- Laboratory of Biological Structure Mechanics, Department of Chemistry, Materials and Chemical Engineering “G. Natta,” Politecnico di Milano, Italy
| | - Gabriele Dubini
- Laboratory of Biological Structure Mechanics, Department of Chemistry, Materials and Chemical Engineering “G. Natta,” Politecnico di Milano, Italy
| |
Collapse
|
41
|
Udomsom S, Budwong A, Wongsa C, Sangngam P, Baipaywad P, Manaspon C, Auephanwiriyakul S, Theera-Umpon N, Paengnakorn P. Automatic Programmable Bioreactor with pH Monitoring System for Tissue Engineering Application. Bioengineering (Basel) 2022; 9:bioengineering9050187. [PMID: 35621465 PMCID: PMC9138136 DOI: 10.3390/bioengineering9050187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 12/17/2022] Open
Abstract
Tissue engineering technology has been advanced and applied to various applications in the past few years. The presence of a bioreactor is one key factor to the successful development of advanced tissue engineering products. In this work, we developed a programmable bioreactor with a controlling program that allowed each component to be automatically operated. Moreover, we developed a new pH sensor for non-contact and real-time pH monitoring. We demonstrated that the prototype bioreactor could facilitate automatic cell culture of L929 cells. It showed that the cell viability was greater than 80% and cell proliferation was enhanced compared to that of the control obtained by a conventional cell culture procedure. This result suggests the possibility of a system that could be potentially useful for medical and industrial applications, including cultured meat, drug testing, etc.
Collapse
Affiliation(s)
- Suruk Udomsom
- Biomedical Engineering Institute, Chiang Mai University, Chiang Mai 50200, Thailand; (S.U.); (A.B.); (C.W.); (P.S.); (P.B.); (C.M.); (S.A.); (N.T.-U.)
| | - Apiwat Budwong
- Biomedical Engineering Institute, Chiang Mai University, Chiang Mai 50200, Thailand; (S.U.); (A.B.); (C.W.); (P.S.); (P.B.); (C.M.); (S.A.); (N.T.-U.)
| | - Chanyanut Wongsa
- Biomedical Engineering Institute, Chiang Mai University, Chiang Mai 50200, Thailand; (S.U.); (A.B.); (C.W.); (P.S.); (P.B.); (C.M.); (S.A.); (N.T.-U.)
| | - Pakorn Sangngam
- Biomedical Engineering Institute, Chiang Mai University, Chiang Mai 50200, Thailand; (S.U.); (A.B.); (C.W.); (P.S.); (P.B.); (C.M.); (S.A.); (N.T.-U.)
| | - Phornsawat Baipaywad
- Biomedical Engineering Institute, Chiang Mai University, Chiang Mai 50200, Thailand; (S.U.); (A.B.); (C.W.); (P.S.); (P.B.); (C.M.); (S.A.); (N.T.-U.)
| | - Chawan Manaspon
- Biomedical Engineering Institute, Chiang Mai University, Chiang Mai 50200, Thailand; (S.U.); (A.B.); (C.W.); (P.S.); (P.B.); (C.M.); (S.A.); (N.T.-U.)
| | - Sansanee Auephanwiriyakul
- Biomedical Engineering Institute, Chiang Mai University, Chiang Mai 50200, Thailand; (S.U.); (A.B.); (C.W.); (P.S.); (P.B.); (C.M.); (S.A.); (N.T.-U.)
- Department of Computational Engineering, Faculty of Engineering, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Theera-Umpon
- Biomedical Engineering Institute, Chiang Mai University, Chiang Mai 50200, Thailand; (S.U.); (A.B.); (C.W.); (P.S.); (P.B.); (C.M.); (S.A.); (N.T.-U.)
- Department of Electrical Engineering, Faculty of Engineering, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Pathinan Paengnakorn
- Biomedical Engineering Institute, Chiang Mai University, Chiang Mai 50200, Thailand; (S.U.); (A.B.); (C.W.); (P.S.); (P.B.); (C.M.); (S.A.); (N.T.-U.)
- Center of Excellence for Innovation in Analytical Science and Technology, Chiang Mai University, Chiang Mai 50200, Thailand
- Correspondence:
| |
Collapse
|
42
|
Kavand H, Nasiri R, Herland A. Advanced Materials and Sensors for Microphysiological Systems: Focus on Electronic and Electrooptical Interfaces. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107876. [PMID: 34913206 DOI: 10.1002/adma.202107876] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/07/2021] [Indexed: 06/14/2023]
Abstract
Advanced in vitro cell culture systems or microphysiological systems (MPSs), including microfluidic organ-on-a-chip (OoC), are breakthrough technologies in biomedicine. These systems recapitulate features of human tissues outside of the body. They are increasingly being used to study the functionality of different organs for applications such as drug evolutions, disease modeling, and precision medicine. Currently, developers and endpoint users of these in vitro models promote how they can replace animal models or even be a better ethically neutral and humanized alternative to study pathology, physiology, and pharmacology. Although reported models show a remarkable physiological structure and function compared to the conventional 2D cell culture, they are almost exclusively based on standard passive polymers or glass with none or minimal real-time stimuli and readout capacity. The next technology leap in reproducing in vivo-like functionality and real-time monitoring of tissue function could be realized with advanced functional materials and devices. This review describes the currently reported electronic and optical advanced materials for sensing and stimulation of MPS models. In addition, an overview of multi-sensing for Body-on-Chip platforms is given. Finally, one gives the perspective on how advanced functional materials could be integrated into in vitro systems to precisely mimic human physiology.
Collapse
Affiliation(s)
- Hanie Kavand
- Division of Micro- and Nanosystems, Department of Intelligent Systems, KTH Royal Institute of Technology, Malvinas Väg 10 pl 5, Stockholm, 100 44, Sweden
| | - Rohollah Nasiri
- AIMES, Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, Solnavägen 9/B8, Solna, 171 65, Sweden
- Division of Nanobiotechnology, Department of Protein Science, KTH Royal Institute of Technology, Tomtebodavägen 23a, Solna, 171 65, Sweden
| | - Anna Herland
- Division of Micro- and Nanosystems, Department of Intelligent Systems, KTH Royal Institute of Technology, Malvinas Väg 10 pl 5, Stockholm, 100 44, Sweden
- AIMES, Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, Solnavägen 9/B8, Solna, 171 65, Sweden
- Division of Nanobiotechnology, Department of Protein Science, KTH Royal Institute of Technology, Tomtebodavägen 23a, Solna, 171 65, Sweden
| |
Collapse
|
43
|
|
44
|
Dsouza VL, Kuthethur R, Kabekkodu SP, Chakrabarty S. Organ-on-Chip platforms to study tumor evolution and chemosensitivity. Biochim Biophys Acta Rev Cancer 2022; 1877:188717. [PMID: 35304293 DOI: 10.1016/j.bbcan.2022.188717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 10/18/2022]
Abstract
Despite tremendous advancements in oncology research and therapeutics, cancer remains a primary cause of death worldwide. One of the significant factors in this critical challenge is a precise diagnosis and limited knowledge on how the tumor microenvironment (TME) behaves to the treatment and its role in chemo-resistance. Therefore, it is critical to understand the contribution of a heterogeneous TME in cancer drug response in individual patients for effective therapy management. Micro-physiological systems along with tissue engineering have facilitated the development of more physiologically relevant platforms, known as Organ-on-Chips (OoC). OoC platforms recapitulate the critical hallmarks of the TME in vitro and subsequently abet in sensitivity and efficacy testing of anti-cancer drugs before clinical trials. The OoC platforms incorporating conventional in vitro models enable researchers to control the cellular, molecular, chemical, and biophysical parameters of the TME in precise combinations while analyzing how they contribute to tumor progression and therapy response. This review discusses the application of OoC platforms integrated with conventional 2D cell lines, 3D organoids and spheroid models, and the organotypic tissue slices, including patient-derived and xenograft tumor slice cultures in cancer treatment responses. We summarize the relevance and drawbacks of conventional in vitro models in assessing cancer treatment response, challenges and limitations associated with OoC models, and future opportunities enabled by the OoC technologies towards developing personalized cancer diagnostics and therapeutics.
Collapse
Affiliation(s)
- Venzil Lavie Dsouza
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Raviprasad Kuthethur
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Sanjiban Chakrabarty
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| |
Collapse
|
45
|
Fedi A, Vitale C, Giannoni P, Caluori G, Marrella A. Biosensors to Monitor Cell Activity in 3D Hydrogel-Based Tissue Models. SENSORS (BASEL, SWITZERLAND) 2022; 22:1517. [PMID: 35214418 PMCID: PMC8879987 DOI: 10.3390/s22041517] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/06/2022] [Accepted: 02/09/2022] [Indexed: 12/13/2022]
Abstract
Three-dimensional (3D) culture models have gained relevant interest in tissue engineering and drug discovery owing to their suitability to reproduce in vitro some key aspects of human tissues and to provide predictive information for in vivo tests. In this context, the use of hydrogels as artificial extracellular matrices is of paramount relevance, since they allow closer recapitulation of (patho)physiological features of human tissues. However, most of the analyses aimed at characterizing these models are based on time-consuming and endpoint assays, which can provide only static and limited data on cellular behavior. On the other hand, biosensing systems could be adopted to measure on-line cellular activity, as currently performed in bi-dimensional, i.e., monolayer, cell culture systems; however, their translation and integration within 3D hydrogel-based systems is not straight forward, due to the geometry and materials properties of these advanced cell culturing approaches. Therefore, researchers have adopted different strategies, through the development of biochemical, electrochemical and optical sensors, but challenges still remain in employing these devices. In this review, after examining recent advances in adapting existing biosensors from traditional cell monolayers to polymeric 3D cells cultures, we will focus on novel designs and outcomes of a range of biosensors specifically developed to provide real-time analysis of hydrogel-based cultures.
Collapse
Affiliation(s)
- Arianna Fedi
- National Research Council of Italy, Institute of Electronics, Computer and Telecommunication Engineering (IEIIT), 16149 Genoa, Italy; (A.F.); (C.V.)
- Department of Computer Science, Bioengineering, Robotics and Systems Engineering (DIBRIS), University of Genoa, 16126 Genoa, Italy
| | - Chiara Vitale
- National Research Council of Italy, Institute of Electronics, Computer and Telecommunication Engineering (IEIIT), 16149 Genoa, Italy; (A.F.); (C.V.)
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy;
| | - Paolo Giannoni
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy;
| | - Guido Caluori
- IHU LIRYC, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux Université, 33600 Pessac, France;
- INSERM UMR 1045, Cardiothoracic Research Center of Bordeaux, University of Bordeaux, 33600 Pessac, France
| | - Alessandra Marrella
- National Research Council of Italy, Institute of Electronics, Computer and Telecommunication Engineering (IEIIT), 16149 Genoa, Italy; (A.F.); (C.V.)
| |
Collapse
|
46
|
Kabay G, Manz A, Dincer C. Microfluidic Roadmap for Translational Nanotheranostics. SMALL METHODS 2022; 6:e2101217. [PMID: 34957704 DOI: 10.1002/smtd.202101217] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/22/2021] [Indexed: 06/14/2023]
Abstract
Nanotheranostic materials (NTMs) shed light on the mechanisms responsible for complex diseases such as cancer because they enable making a diagnosis, monitoring the disease progression, and applying a targeted therapy simultaneously. However, several issues such as the reproducibility and mass production of NTMs hamper their application for clinical practice. To address these issues and facilitate the clinical application of NTMs, microfluidic systems have been increasingly used. This perspective provides a glimpse into the current state-of-art of NTM research, emphasizing the methods currently employed at each development stage of NTMs and the related open problems. This work reviews microfluidic technologies used to develop NTMs, ranging from the fabrication and testing of a single NTM up to their manufacturing on a large scale. Ultimately, a step-by-step vision on the future development of NTMs for clinical practice enabled by microfluidics techniques is provided.
Collapse
Affiliation(s)
- Gozde Kabay
- University of Freiburg, Department of Microsystems Engineering (IMTEK), 79110, Freiburg, Germany
- University of Freiburg, FIT Freiburg Center for Interactive Materials and Bioinspired Technologies, 79110, Freiburg, Germany
| | - Andreas Manz
- Korea Institute of Science and Technology (KIST) in Europe, 66123, Saarbrücken, Germany
| | - Can Dincer
- University of Freiburg, Department of Microsystems Engineering (IMTEK), 79110, Freiburg, Germany
- University of Freiburg, FIT Freiburg Center for Interactive Materials and Bioinspired Technologies, 79110, Freiburg, Germany
| |
Collapse
|
47
|
Dornhof J, Kieninger J, Muralidharan H, Maurer J, Urban GA, Weltin A. Microfluidic organ-on-chip system for multi-analyte monitoring of metabolites in 3D cell cultures. LAB ON A CHIP 2022; 22:225-239. [PMID: 34851349 DOI: 10.1039/d1lc00689d] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Three-dimensional cell cultures using patient-derived stem cells are essential in vitro models for a more efficient and individualized cancer therapy. Currently, culture conditions and metabolite concentrations, especially hypoxia, are often not accessible continuously and in situ within microphysiological systems. However, understanding and standardizing the cellular microenvironment are the key to successful in vitro models. We developed a microfluidic organ-on-chip platform for matrix-based, heterogeneous 3D cultures with fully integrated electrochemical chemo- and biosensor arrays for the energy metabolites oxygen, lactate, and glucose. Advanced microstructures allow straightforward cell matrix integration with standard laboratory equipment, compartmentalization, and microfluidic access. Single, patient-derived, triple-negative breast cancer stem cells develop into tumour organoids in a heterogeneous spheroid culture on-chip. Our system allows unprecedented control of culture conditions, including hypoxia, and simultaneous verification by integrated sensors. Beyond previous works, our results demonstrate precise and reproducible on-chip multi-analyte metabolite monitoring under dynamic conditions from a matrix-based culture over more than one week. Responses to alterations in culture conditions and cancer drug exposure, such as metabolite consumption and production rates, could be accessed quantitatively and in real-time, in contrast to endpoint analyses. Our approach highlights the importance of continuous, in situ metabolite monitoring in 3D cell cultures regarding the standardization and control of culture conditions, and drug screening in cancer research. Overall, the results underline the potential of microsensors in organ-on-chip systems for successful application, e.g. in personalized medicine.
Collapse
Affiliation(s)
- Johannes Dornhof
- Laboratory for Sensors, IMTEK - Department of Microsystems Engineering, University of Freiburg, Georges-Köhler-Allee 103, 79110 Freiburg, Germany.
| | - Jochen Kieninger
- Laboratory for Sensors, IMTEK - Department of Microsystems Engineering, University of Freiburg, Georges-Köhler-Allee 103, 79110 Freiburg, Germany.
| | - Harshini Muralidharan
- Molecular Gynecology, Clinic for Gynecology and Obstetrics, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Jochen Maurer
- Molecular Gynecology, Clinic for Gynecology and Obstetrics, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Gerald A Urban
- Laboratory for Sensors, IMTEK - Department of Microsystems Engineering, University of Freiburg, Georges-Köhler-Allee 103, 79110 Freiburg, Germany.
| | - Andreas Weltin
- Laboratory for Sensors, IMTEK - Department of Microsystems Engineering, University of Freiburg, Georges-Köhler-Allee 103, 79110 Freiburg, Germany.
| |
Collapse
|
48
|
Caballero D, Reis RL, Kundu SC. Current Trends in Microfluidics and Biosensors for Cancer Research Applications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1379:81-112. [DOI: 10.1007/978-3-031-04039-9_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
49
|
Lopez-Muñoz GA, Mughal S, Ramón-Azcón J. Sensors and Biosensors in Organs-on-a-Chip Platforms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1379:55-80. [DOI: 10.1007/978-3-031-04039-9_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
50
|
Rousset N, Sandoval RL, Modena MM, Hierlemann A, Misun PM. Modeling and measuring glucose diffusion and consumption by colorectal cancer spheroids in hanging drops using integrated biosensors. MICROSYSTEMS & NANOENGINEERING 2022; 8:14. [PMID: 35136653 PMCID: PMC8803859 DOI: 10.1038/s41378-021-00348-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 10/21/2021] [Accepted: 11/28/2021] [Indexed: 05/02/2023]
Abstract
As 3D in vitro tissue models become more pervasive, their built-in nutrient, metabolite, compound, and waste gradients increase biological relevance at the cost of analysis simplicity. Investigating these gradients and the resulting metabolic heterogeneity requires invasive and time-consuming methods. An alternative is using electrochemical biosensors and measuring concentrations around the tissue model to obtain size-dependent metabolism data. With our hanging-drop-integrated enzymatic glucose biosensors, we conducted current measurements within hanging-drop compartments hosting spheroids formed from the human colorectal carcinoma cell line HCT116. We developed a physics-based mathematical model of analyte consumption and transport, considering (1) diffusion and enzymatic conversion of glucose to form hydrogen peroxide (H2O2) by the glucose-oxidase-based hydrogel functionalization of our biosensors at the microscale; (2) H2O2 oxidation at the electrode surface, leading to amperometric H2O2 readout; (3) glucose diffusion and glucose consumption by cancer cells in a spherical tissue model at the microscale; (4) glucose and H2O2 transport in our hanging-drop compartments at the macroscale; and (5) solvent evaporation, leading to glucose and H2O2 upconcentration. Our model relates the measured currents to the glucose concentrations generating the currents. The low limit of detection of our biosensors (0.4 ± 0.1 μM), combined with our current-fitting method, enabled us to reveal glucose dynamics within our system. By measuring glucose dynamics in hanging-drop compartments populated by cancer spheroids of various sizes, we could infer glucose distributions within the spheroid, which will help translate in vitro 3D tissue model results to in vivo.
Collapse
Affiliation(s)
- Nassim Rousset
- ETH Zürich, Department of Biosystems Science and Engineering, Bio Engineering Laboratory, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Rubén López Sandoval
- ETH Zürich, Department of Biosystems Science and Engineering, Bio Engineering Laboratory, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Mario Matteo Modena
- ETH Zürich, Department of Biosystems Science and Engineering, Bio Engineering Laboratory, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Andreas Hierlemann
- ETH Zürich, Department of Biosystems Science and Engineering, Bio Engineering Laboratory, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Patrick M. Misun
- ETH Zürich, Department of Biosystems Science and Engineering, Bio Engineering Laboratory, Mattenstrasse 26, CH-4058 Basel, Switzerland
| |
Collapse
|