1
|
Kato M, Sato K. A Microfluidic-Based Cell-Stretching Culture Device That Allows for Easy Preparation of Slides for Observation with High-Magnification Objective Lenses. MICROMACHINES 2025; 16:93. [PMID: 39858748 PMCID: PMC11767594 DOI: 10.3390/mi16010093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Microfluidic-based cell-stretching devices are vital for studying the molecular pathways involved in cellular responses to mechanobiological processes. Accurate evaluation of these responses requires detailed observation of cells cultured in this cell-stretching device. This study aimed to develop a method for preparing microscope slides to enable high-magnification imaging of cells in these devices. The key innovation is creating a peelable bond between the cell culture membrane and the upper channel, allowing for easy removal of the upper layer and precise cutting of the membrane for high-magnification microscopy. Using the fabricated device, OP9 cells (15,000 cells/channel) were stretched, and the effects of focal adhesion proteins and the intracellular distribution of YAP1 were examined under a fluorescence microscope with 100× and 60× objectives. Stretch stimulation increased integrinβ1 expression and promoted integrin-vinculin complex formation by approximately 1.4-fold in OP9 cells. Furthermore, YAP1 nuclear localization was significantly enhanced (approximately 1.3-fold) during stretching. This method offers a valuable tool for researchers using microfluidic-based cell-stretching devices. The advancement of imaging techniques in microdevice research is expected to further drive progress in mechanobiology research.
Collapse
Affiliation(s)
| | - Kae Sato
- Department of Chemical and Biological Sciences, Faculty of Science, Japan Women’s University, 2-8-1 Mejirodai, Bunkyo, Tokyo 112-8681, Japan
| |
Collapse
|
2
|
Bathrinarayanan PV, Hallam SM, Grover LM, Vigolo D, Simmons MJH. Microfluidics as a Powerful Tool to Investigate Microvascular Dysfunction in Trauma Conditions: A Review of the State-of-the-Art. Adv Biol (Weinh) 2024; 8:e2400037. [PMID: 39031943 DOI: 10.1002/adbi.202400037] [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: 01/21/2024] [Revised: 04/18/2024] [Indexed: 07/22/2024]
Abstract
Skeletal muscle trauma such as fracture or crush injury can result in a life-threatening condition called acute compartment syndrome (ACS), which involves elevated compartmental pressure within a closed osteo-fascial compartment, leading to collapse of the microvasculature and resulting in necrosis of the tissue due to ischemia. Diagnosis of ACS is complex and controversial due to the lack of standardized objective methods, which results in high rates of misdiagnosis/late diagnosis, leading to permanent neuro-muscular damage. ACS pathophysiology is poorly understood at a cellular level due to the lack of physiologically relevant models. In this context, microfluidics organ-on-chip systems (OOCs) provide an exciting opportunity to investigate the cellular mechanisms of microvascular dysfunction that leads to ACS. In this article, the state-of-the-art OOCs designs and strategies used to investigate microvasculature dysfunction mechanisms is reviewed. The differential effects of hemodynamic shear stress on endothelial cell characteristics such as morphology, permeability, and inflammation, all of which are altered during microvascular dysfunction is highlighted. The article then critically reviews the importance of microfluidics to investigate closely related microvascular pathologies that cause ACS. The article concludes by discussing potential biomarkers of ACS with a special emphasis on glycocalyx and providing a future perspective.
Collapse
Affiliation(s)
- P Vasanthi Bathrinarayanan
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B152TT, UK
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - S M Hallam
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B152TT, UK
| | - L M Grover
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B152TT, UK
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - D Vigolo
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B152TT, UK
- The University of Sydney, School of Biomedical Engineering, Sydney, NSW, 2006, Australia
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| | - M J H Simmons
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B152TT, UK
| |
Collapse
|
3
|
Nguyen HT, Rissanen SL, Peltokangas M, Laakkonen T, Kettunen J, Barthod L, Sivakumar R, Palojärvi A, Junttila P, Talvitie J, Bassis M, Nickels SL, Kalvala S, Ilina P, Tammela P, Lehtonen S, Schwamborn JC, Mosser S, Singh P. Highly scalable and standardized organ-on-chip platform with TEER for biological barrier modeling. Tissue Barriers 2024; 12:2315702. [PMID: 38346163 PMCID: PMC11583584 DOI: 10.1080/21688370.2024.2315702] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 01/02/2024] [Accepted: 01/15/2024] [Indexed: 11/22/2024] Open
Abstract
The development of new therapies is hampered by the lack of predictive, and patient-relevant in vitro models. Organ-on-chip (OOC) technologies can potentially recreate physiological features and hold great promise for tissue and disease modeling. However, the non-standardized design of these chips and perfusion control systems has been a barrier to quantitative high-throughput screening (HTS). Here we present a scalable OOC microfluidic platform for applied kinetic in vitro assays (AKITA) that is applicable for high, medium, and low throughput. Its standard 96-well plate and 384-well plate layouts ensure compatibility with existing laboratory workflows and high-throughput data collection and analysis tools. The AKITA plate is optimized for the modeling of vascularized biological barriers, primarily the blood-brain barrier, skin, and lung, with precise flow control on a custom rocker. The integration of trans-epithelial electrical resistance (TEER) sensors allows rapid and repeated monitoring of barrier integrity over long time periods. Together with automated liquid handling and compound permeability testing analyses, we demonstrate the flexibility of the AKITA platform for establishing human-relevant models for preclinical drug and precision medicine's efficacy, toxicity, and permeability under near-physiological conditions.
Collapse
Affiliation(s)
- Hoang-Tuan Nguyen
- Finnadvance Ltd, Oulu, Finland
- Faculty of Biochemistry and Molecular Medicine, and Biocenter Oulu, University of Oulu, Oulu, Finland
| | | | | | | | | | | | | | | | | | | | - Michele Bassis
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Sarah L. Nickels
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Sara Kalvala
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Polina Ilina
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Päivi Tammela
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Sarka Lehtonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Jens C. Schwamborn
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | | | | |
Collapse
|
4
|
Liu Z, Fan Y, Cui M, Wang X, Zhao P. Investigation of tumour environments through advancements in microtechnology and nanotechnology. Biomed Pharmacother 2024; 178:117230. [PMID: 39116787 DOI: 10.1016/j.biopha.2024.117230] [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: 06/05/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
Cancer has a significant negative social and economic impact on both developed and developing countries. As a result, understanding the onset and progression of cancer is critical for developing therapies that can improve the well-being and health of individuals with cancer. With time, study has revealed, the tumor microenvironment has great influence on this process. Micro and nanoscale engineering techniques can be used to study the tumor microenvironment. Nanoscale and Microscale engineering use Novel technologies and designs with small dimensions to recreate the TME. Knowing how cancer cells interact with one another can help researchers develop therapeutic approaches that anticipate and counteract cancer cells' techniques for evading detection and fighting anti-cancer treatments, such as microfabrication techniques, microfluidic devices, nanosensors, and nanodevices used to study or recreate the tumor microenvironment. Nevertheless, a complicated action just like the growth and in cancer advancement, and their intensive association along the environment around it that has to be studied in more detail.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Radiology, Shengjing Hospital of China Medical University, China
| | - Yan Fan
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Mengyao Cui
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xu Wang
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Pengfei Zhao
- Department of Radiology, Shengjing Hospital of China Medical University, China.
| |
Collapse
|
5
|
Lim J, Fang HW, Bupphathong S, Sung PC, Yeh CE, Huang W, Lin CH. The Edifice of Vasculature-On-Chips: A Focused Review on the Key Elements and Assembly of Angiogenesis Models. ACS Biomater Sci Eng 2024; 10:3548-3567. [PMID: 38712543 PMCID: PMC11167599 DOI: 10.1021/acsbiomaterials.3c01978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/08/2024]
Abstract
The conception of vascularized organ-on-a-chip models provides researchers with the ability to supply controlled biological and physical cues that simulate the in vivo dynamic microphysiological environment of native blood vessels. The intention of this niche research area is to improve our understanding of the role of the vasculature in health or disease progression in vitro by allowing researchers to monitor angiogenic responses and cell-cell or cell-matrix interactions in real time. This review offers a comprehensive overview of the essential elements, including cells, biomaterials, microenvironmental factors, microfluidic chip design, and standard validation procedures that currently govern angiogenesis-on-a-chip assemblies. In addition, we emphasize the importance of incorporating a microvasculature component into organ-on-chip devices in critical biomedical research areas, such as tissue engineering, drug discovery, and disease modeling. Ultimately, advances in this area of research could provide innovative solutions and a personalized approach to ongoing medical challenges.
Collapse
Affiliation(s)
- Joshua Lim
- Graduate
Institute of Nanomedicine and Medical Engineering, College of Biomedical
Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsu-Wei Fang
- High-value
Biomaterials Research and Commercialization Center, National Taipei University of Technology, Taipei 10608, Taiwan
- Department
of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei 10608, Taiwan
- Institute
of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Sasinan Bupphathong
- Graduate
Institute of Nanomedicine and Medical Engineering, College of Biomedical
Engineering, Taipei Medical University, Taipei 11031, Taiwan
- High-value
Biomaterials Research and Commercialization Center, National Taipei University of Technology, Taipei 10608, Taiwan
| | - Po-Chan Sung
- School
of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Chen-En Yeh
- School
of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Wei Huang
- Department
of Orthodontics, Rutgers School of Dental
Medicine, Newark, New Jersey 07103, United States
| | - Chih-Hsin Lin
- Graduate
Institute of Nanomedicine and Medical Engineering, College of Biomedical
Engineering, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
6
|
Hall E, Mendiola K, Lightsey NK, Hanjaya-Putra D. Mimicking blood and lymphatic vasculatures using microfluidic systems. BIOMICROFLUIDICS 2024; 18:031502. [PMID: 38726373 PMCID: PMC11081709 DOI: 10.1063/5.0175154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 04/04/2024] [Indexed: 05/12/2024]
Abstract
The role of the circulatory system, containing the blood and lymphatic vasculatures, within the body, has become increasingly focused on by researchers as dysfunction of either of the systems has been linked to serious complications and disease. Currently, in vivo models are unable to provide the sufficient monitoring and level of manipulation needed to characterize the fluidic dynamics of the microcirculation in blood and lymphatic vessels; thus in vitro models have been pursued as an alternative model. Microfluidic devices have the required properties to provide a physiologically relevant circulatory system model for research as well as the experimental tools to conduct more advanced research analyses of microcirculation flow. In this review paper, the physiological behavior of fluid flow and electrical communication within the endothelial cells of the systems are detailed and discussed to highlight their complexities. Cell co-culturing methods and other relevant organ-on-a-chip devices will be evaluated to demonstrate the feasibility and relevance of the in vitro microfluidic model. Microfluidic systems will be determined as a noteworthy model that can display physiologically relevant flow of the cardiovascular and lymphatic systems, which will enable researchers to investigate the systems' prevalence in diseases and identify potential therapeutics.
Collapse
Affiliation(s)
- Eva Hall
- Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | | | - N. Keilany Lightsey
- Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | | |
Collapse
|
7
|
Garlisi Torales LD, Sempowski BA, Krikorian GL, Woodis KM, Paulissen SM, Smith CL, Sheppard SE. Central conducting lymphatic anomaly: from bench to bedside. J Clin Invest 2024; 134:e172839. [PMID: 38618951 PMCID: PMC11014661 DOI: 10.1172/jci172839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024] Open
Abstract
Central conducting lymphatic anomaly (CCLA) is a complex lymphatic anomaly characterized by abnormalities of the central lymphatics and may present with nonimmune fetal hydrops, chylothorax, chylous ascites, or lymphedema. CCLA has historically been difficult to diagnose and treat; however, recent advances in imaging, such as dynamic contrast magnetic resonance lymphangiography, and in genomics, such as deep sequencing and utilization of cell-free DNA, have improved diagnosis and refined both genotype and phenotype. Furthermore, in vitro and in vivo models have confirmed genetic causes of CCLA, defined the underlying pathogenesis, and facilitated personalized medicine to improve outcomes. Basic, translational, and clinical science are essential for a bedside-to-bench and back approach for CCLA.
Collapse
Affiliation(s)
- Luciana Daniela Garlisi Torales
- Unit on Vascular Malformations, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Benjamin A. Sempowski
- Unit on Vascular Malformations, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Georgia L. Krikorian
- Unit on Vascular Malformations, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Kristina M. Woodis
- Unit on Vascular Malformations, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Scott M. Paulissen
- Unit on Vascular Malformations, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Christopher L. Smith
- Division of Cardiology, Jill and Mark Fishman Center for Lymphatic Disorders, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Sarah E. Sheppard
- Unit on Vascular Malformations, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| |
Collapse
|
8
|
Chandran Suja V, Qi QM, Halloran K, Zhang J, Shaha S, Prakash S, Kumbhojkar N, Deslandes A, Huille S, Gokarn YR, Mitragotri S. A biomimetic chip to assess subcutaneous bioavailability of monoclonal antibodies in humans. PNAS NEXUS 2023; 2:pgad317. [PMID: 37901442 PMCID: PMC10612570 DOI: 10.1093/pnasnexus/pgad317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/12/2023] [Indexed: 10/31/2023]
Abstract
Subcutaneous (subQ) injection is a common route for delivering biotherapeutics, wherein pharmacokinetics is largely influenced by drug transport in a complex subQ tissue microenvironment. The selection of good drug candidates with beneficial pharmacokinetics for subQ injections is currently limited by a lack of reliable testing models. To address this limitation, we report here a Subcutaneous Co-Culture Tissue-on-a-chip for Injection Simulation (SubCuTIS). SubCuTIS possesses a 3D coculture tissue architecture, and it allows facile quantitative determination of relevant scale independent drug transport rate constants. SubCuTIS captures key in vivo physiological characteristics of the subQ tissues, and it differentiates the transport behavior of various chemically distinct molecules. We supplemented the transport measurements with theoretical modeling, which identified subtle differences in the local absorption rate constants of seven clinically available mAbs. Accounting for first-order proteolytic catabolism, we established a mathematical framework to assess clinical bioavailability using the local absorption rate constants obtained from SubCuTIS. Taken together, the technology described here broadens the applicability of organs-on-chips as a standardized and easy-to-use device for quantitative analysis of subQ drug transport.
Collapse
Affiliation(s)
- Vineeth Chandran Suja
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Qin M Qi
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, MA 02115, USA
| | | | | | - Suyog Shaha
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Supriya Prakash
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Ninad Kumbhojkar
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, MA 02115, USA
| | | | - Sylvain Huille
- Sanofi R&D, Impasse Des Ateliers, Vitry-sur-Seine 94400 France
| | | | - Samir Mitragotri
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, 3 Blackfan Circle, Boston, MA 02115, USA
| |
Collapse
|
9
|
Kutluk H, Bastounis EE, Constantinou I. Integration of Extracellular Matrices into Organ-on-Chip Systems. Adv Healthc Mater 2023; 12:e2203256. [PMID: 37018430 PMCID: PMC11468608 DOI: 10.1002/adhm.202203256] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/20/2023] [Indexed: 04/07/2023]
Abstract
The extracellular matrix (ECM) is a complex, dynamic network present within all tissues and organs that not only acts as a mechanical support and anchorage point but can also direct fundamental cell behavior, function, and characteristics. Although the importance of the ECM is well established, the integration of well-controlled ECMs into Organ-on-Chip (OoC) platforms remains challenging and the methods to modulate and assess ECM properties on OoCs remain underdeveloped. In this review, current state-of-the-art design and assessment of in vitro ECM environments is discussed with a focus on their integration into OoCs. Among other things, synthetic and natural hydrogels, as well as polydimethylsiloxane (PDMS) used as substrates, coatings, or cell culture membranes are reviewed in terms of their ability to mimic the native ECM and their accessibility for characterization. The intricate interplay among materials, OoC architecture, and ECM characterization is critically discussed as it significantly complicates the design of ECM-related studies, comparability between works, and reproducibility that can be achieved across research laboratories. Improving the biomimetic nature of OoCs by integrating properly considered ECMs would contribute to their further adoption as replacements for animal models, and precisely tailored ECM properties would promote the use of OoCs in mechanobiology.
Collapse
Affiliation(s)
- Hazal Kutluk
- Institute of Microtechnology (IMT)Technical University of BraunschweigAlte Salzdahlumer Str. 20338124BraunschweigGermany
- Center of Pharmaceutical Engineering (PVZ)Technical University of BraunschweigFranz‐Liszt‐Str. 35a38106BraunschweigGermany
| | - Effie E. Bastounis
- Institute of Microbiology and Infection Medicine (IMIT)Eberhard Karls University of TübingenAuf der Morgenstelle 28, E872076TübingenGermany
- Cluster of Excellence “Controlling Microbes to Fight Infections” EXC 2124Eberhard Karls University of TübingenAuf der Morgenstelle 2872076TübingenGermany
| | - Iordania Constantinou
- Institute of Microtechnology (IMT)Technical University of BraunschweigAlte Salzdahlumer Str. 20338124BraunschweigGermany
- Center of Pharmaceutical Engineering (PVZ)Technical University of BraunschweigFranz‐Liszt‐Str. 35a38106BraunschweigGermany
| |
Collapse
|
10
|
Ejazi SA, Louisthelmy R, Maisel K. Mechanisms of Nanoparticle Transport across Intestinal Tissue: An Oral Delivery Perspective. ACS NANO 2023. [PMID: 37410891 DOI: 10.1021/acsnano.3c02403] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Oral drug administration has been a popular choice due to patient compliance and limited clinical resources. Orally delivered drugs must circumvent the harsh gastrointestinal (GI) environment to effectively enter the systemic circulation. The GI tract has a number of structural and physiological barriers that limit drug bioavailability including mucus, the tightly regulated epithelial layer, immune cells, and associated vasculature. Nanoparticles have been used to enhance oral bioavailability of drugs, as they can act as a shield to the harsh GI environment and prevent early degradation while also increasing uptake and transport of drugs across the intestinal epithelium. Evidence suggests that different nanoparticle formulations may be transported via different intracellular mechanisms to cross the intestinal epithelium. Despite the existence of a significant body of work on intestinal transport of nanoparticles, many key questions remain: What causes the poor bioavailability of the oral drugs? What factors contribute to the ability of a nanoparticle to cross different intestinal barriers? Do nanoparticle properties such as size and charge influence the type of endocytic pathways taken? In this Review, we summarize the different components of intestinal barriers and the types of nanoparticles developed for oral delivery. In particular, we focus on the various intracellular pathways used in nanoparticle internalization and nanoparticle or cargo translocation across the epithelium. Understanding the gut barrier, nanoparticle characteristics, and transport pathways may lead to the development of more therapeutically useful nanoparticles as drug carriers.
Collapse
Affiliation(s)
- Sarfaraz Ahmad Ejazi
- Fischell Department of Bioengineering, University of Maryland, 3120 A. James Clark Hall, College Park, Maryland 20742, United States
| | - Rebecca Louisthelmy
- Fischell Department of Bioengineering, University of Maryland, 3120 A. James Clark Hall, College Park, Maryland 20742, United States
| | - Katharina Maisel
- Fischell Department of Bioengineering, University of Maryland, 3120 A. James Clark Hall, College Park, Maryland 20742, United States
| |
Collapse
|
11
|
Nazari N, Yun W, Kovscek AR. The motion of long bubbles in microchannels using a meter-long, rectangular capillary on a chip. J Colloid Interface Sci 2023; 638:149-160. [PMID: 36736116 DOI: 10.1016/j.jcis.2023.01.073] [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: 10/14/2022] [Revised: 01/12/2023] [Accepted: 01/14/2023] [Indexed: 01/22/2023]
Abstract
HYPOTHESIS The dynamics of gas-liquid interfaces differs between aqueous surfactant and nanoparticle mixtures in rectangular cross-section capillaries. EXPERIMENTS We designed and fabricated a new microfluidic device with a meter-long channel and a noncircular cross section (35 μm by 100 μm by 1 m) to study the flow behavior of long bubbles in capillaries wetted by water as well as surfactant and nanoparticle solutions. Flow in the novel symmetric loop device maintains essentially straight and inertialess conditions over a wide range of flow rates. FINDINGS The pressure-drop versus velocity relationship of long bubbles in capillaries with noncircular cross sections has been studied theoretically but not extensively validated. The measured pressure drop (normalized with respect to interfacial properties) experienced by bubbles varies as Ca2/3 over the range 10-7<Ca<10-4 where Ca=μUσ. Thus, the drag also scales as Ca2/3. The measurements from our new device thereby confirm previous theoretical relationships of the flow of long gas bubbles wetted by surfactant solutions in noncircular capillaries. Contrary to the hypothesis, the Ca2/3 drag-velocity relationship applies to gas bubbles in deionized water, nanoparticles in brine, surfactant in brine, and nanoparticle-surfactant mixtures in brine for the conditions studied.
Collapse
Affiliation(s)
- Negar Nazari
- Energy Science and Engineering, 367 Panama St., Room 050, Stanford, CA 94305, USA.
| | - Wonjin Yun
- Energy Science and Engineering, 367 Panama St., Room 050, Stanford, CA 94305, USA.
| | - Anthony R Kovscek
- Energy Science and Engineering, 367 Panama St., Room 050, Stanford, CA 94305, USA.
| |
Collapse
|
12
|
Kagawa M, Morioka K, Osashima M, Hemmi A, Yamamoto S, Shoji A, Uchiyama K, Nakajima H. Development of small-sized fluorescence detector for pipette tip-based biosensor for on-site diagnosis. Talanta 2023; 256:124311. [PMID: 36738624 DOI: 10.1016/j.talanta.2023.124311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023]
Abstract
A small-sized fluorescence detector (referred to as a pipette tip [PT]-reader) was developed for a pipette tip-based biosensor. The PT-reader allows us to measure the fluorescence intensity of a solution in a truncated cone-shaped pipette tip with only the tip inserted into the PT-reader. A pipette holder made from a mixture of polydimethylsiloxane (PDMS) and carbon black was capable of the rigorous position arrangement of a truncated cone shaped-pipette tip and the prevention of stray light. The detection performance of the PT-reader was evaluated by measurement of resorufin. The limit of detection (LOD; 3σ) and the relative standard deviation (RSD, n = 4) were estimated to be 0.46 μM and 0.47-4.1%, respectively. This performance was comparable to that of a desktop-type fluorescence microplate reader. In addition, the PT-reader was applied to the quantification of immunoglobulin A (IgA), and the LOD (3σ) of IgA was estimated to be 1.0 ng/mL. The quantitation values of IgA in human saliva obtained by the PT-based enzyme-linked immunosorbent assay (PT-ELISA) were in agreement with those obtained by conventional ELISA. The PT-reader is expected to be useful for low-cost and user-friendly measurements, and the technique of device development proposed in this study will contribute to the progress of on-site medical diagnosis.
Collapse
Affiliation(s)
- Masakazu Kagawa
- Department of Applied Chemistry for Environment, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, 1-1 Minami-osawa, Hachioji, Tokyo, 192-0397, Japan
| | - Kazuhiro Morioka
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan.
| | - Moeko Osashima
- Department of Applied Chemistry for Environment, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, 1-1 Minami-osawa, Hachioji, Tokyo, 192-0397, Japan
| | - Akihide Hemmi
- Mebius Advanced Technology Ltd., 3-31-6 Nishiogi-kita, Suginami-ku, Tokyo, 167-0042, Japan
| | - Shoji Yamamoto
- Department of Applied Chemistry for Environment, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, 1-1 Minami-osawa, Hachioji, Tokyo, 192-0397, Japan
| | - Atsushi Shoji
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Katsumi Uchiyama
- Department of Applied Chemistry for Environment, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, 1-1 Minami-osawa, Hachioji, Tokyo, 192-0397, Japan
| | - Hizuru Nakajima
- Department of Applied Chemistry for Environment, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, 1-1 Minami-osawa, Hachioji, Tokyo, 192-0397, Japan.
| |
Collapse
|
13
|
Bogseth A, Ramirez A, Vaughan E, Maisel K. In Vitro Models of Blood and Lymphatic Vessels-Connecting Tissues and Immunity. Adv Biol (Weinh) 2023; 7:e2200041. [PMID: 35751460 PMCID: PMC9790046 DOI: 10.1002/adbi.202200041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/10/2022] [Indexed: 12/27/2022]
Abstract
Blood and lymphatic vessels are regulators of physiological processes, including oxygenation and fluid transport. Both vessels are ubiquitous throughout the body and are critical for sustaining tissue homeostasis. The complexity of each vessel's processes has limited the understanding of exactly how the vessels maintain their functions. Both vessels have been shown to be involved in the pathogenesis of many diseases, including cancer metastasis, and it is crucial to probe further specific mechanisms involved. In vitro models are developed to better understand blood and lymphatic physiological functions and their mechanisms. In this review, blood and lymphatic in vitro model systems, including 2D and 3D designs made using Transwells, microfluidic devices, organoid cultures, and various other methods, are described. Models studying endothelial cell-extracellular matrix interactions, endothelial barrier properties, transendothelial transport and cell migration, lymph/angiogenesis, vascular inflammation, and endothelial-cancer cell interactions are particularly focused. While the field has made significant progress in modeling and understanding lymphatic and blood vasculature, more models that include coculture of multiple cell types, complex extracellular matrix, and 3D morphologies, particularly for models mimicking disease states, will help further the understanding of the role of blood and lymphatic vasculature in health and disease.
Collapse
Affiliation(s)
- Amanda Bogseth
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Ann Ramirez
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Erik Vaughan
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Katharina Maisel
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| |
Collapse
|
14
|
Mu X, Gerhard-Herman MD, Zhang YS. Building Blood Vessel Chips with Enhanced Physiological Relevance. ADVANCED MATERIALS TECHNOLOGIES 2023; 8:2201778. [PMID: 37693798 PMCID: PMC10489284 DOI: 10.1002/admt.202201778] [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: 10/20/2022] [Indexed: 09/12/2023]
Abstract
Blood vessel chips are bioengineered microdevices, consisting of biomaterials, human cells, and microstructures, which recapitulate essential vascular structure and physiology and allow a well-controlled microenvironment and spatial-temporal readouts. Blood vessel chips afford promising opportunities to understand molecular and cellular mechanisms underlying a range of vascular diseases. The physiological relevance is key to these blood vessel chips that rely on bioinspired strategies and bioengineering approaches to translate vascular physiology into artificial units. Here, we discuss several critical aspects of vascular physiology, including morphology, material composition, mechanical properties, flow dynamics, and mass transport, which provide essential guidelines and a valuable source of bioinspiration for the rational design of blood vessel chips. We also review state-of-art blood vessel chips that exhibit important physiological features of the vessel and reveal crucial insights into the biological processes and disease pathogenesis, including rare diseases, with notable implications for drug screening and clinical trials. We envision that the advances in biomaterials, biofabrication, and stem cells improve the physiological relevance of blood vessel chips, which, along with the close collaborations between clinicians and bioengineers, enable their widespread utility.
Collapse
Affiliation(s)
- Xuan Mu
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Marie Denise Gerhard-Herman
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| |
Collapse
|
15
|
Suarez AC, Hammel JH, Munson JM. Modeling lymphangiogenesis: Pairing in vitro and in vivo metrics. Microcirculation 2023; 30:e12802. [PMID: 36760223 PMCID: PMC10121924 DOI: 10.1111/micc.12802] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 01/20/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023]
Abstract
Lymphangiogenesis is the mechanism by which the lymphatic system develops and expands new vessels facilitating fluid drainage and immune cell trafficking. Models to study lymphangiogenesis are necessary for a better understanding of the underlying mechanisms and to identify or test new therapeutic agents that target lymphangiogenesis. Across the lymphatic literature, multiple models have been developed to study lymphangiogenesis in vitro and in vivo. In vitro, lymphangiogenesis can be modeled with varying complexity, from monolayers to hydrogels to explants, with common metrics for characterizing proliferation, migration, and sprouting of lymphatic endothelial cells (LECs) and vessels. In comparison, in vivo models of lymphangiogenesis often use genetically modified zebrafish and mice, with in situ mouse models in the ear, cornea, hind leg, and tail. In vivo metrics, such as activation of LECs, number of new lymphatic vessels, and sprouting, mirror those most used in vitro, with the addition of lymphatic vessel hyperplasia and drainage. The impacts of lymphangiogenesis vary by context of tissue and pathology. Therapeutic targeting of lymphangiogenesis can have paradoxical effects depending on the pathology including lymphedema, cancer, organ transplant, and inflammation. In this review, we describe and compare lymphangiogenic outcomes and metrics between in vitro and in vivo studies, specifically reviewing only those publications in which both testing formats are used. We find that in vitro studies correlate well with in vivo in wound healing and development, but not in the reproductive tract or the complex tumor microenvironment. Considerations for improving in vitro models are to increase complexity with perfusable microfluidic devices, co-cultures with tissue-specific support cells, the inclusion of fluid flow, and pairing in vitro models of differing complexities. We believe that these changes would strengthen the correlation between in vitro and in vivo outcomes, giving more insight into lymphangiogenesis in healthy and pathological states.
Collapse
Affiliation(s)
- Aileen C. Suarez
- Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA
- Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Department of Biomedical Engineering & Mechanics, Virginia Tech, Blacksburg, VA
| | - Jennifer H. Hammel
- Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA
- Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Department of Biomedical Engineering & Mechanics, Virginia Tech, Blacksburg, VA
| | - Jennifer M. Munson
- Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA
- Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Department of Biomedical Engineering & Mechanics, Virginia Tech, Blacksburg, VA
| |
Collapse
|
16
|
Salipante PF. Microfluidic techniques for mechanical measurements of biological samples. BIOPHYSICS REVIEWS 2023; 4:011303. [PMID: 38505816 PMCID: PMC10903441 DOI: 10.1063/5.0130762] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/30/2022] [Indexed: 03/21/2024]
Abstract
The use of microfluidics to make mechanical property measurements is increasingly common. Fabrication of microfluidic devices has enabled various types of flow control and sensor integration at micrometer length scales to interrogate biological materials. For rheological measurements of biofluids, the small length scales are well suited to reach high rates, and measurements can be made on droplet-sized samples. The control of flow fields, constrictions, and external fields can be used in microfluidics to make mechanical measurements of individual bioparticle properties, often at high sampling rates for high-throughput measurements. Microfluidics also enables the measurement of bio-surfaces, such as the elasticity and permeability properties of layers of cells cultured in microfluidic devices. Recent progress on these topics is reviewed, and future directions are discussed.
Collapse
Affiliation(s)
- Paul F. Salipante
- National Institute of Standards and Technology, Polymers and Complex Fluids Group, Gaithersburg, Maryland 20899, USA
| |
Collapse
|
17
|
Abstract
Despite enormous advances, cardiovascular disorders are still a major threat to global health and are responsible for one-third of deaths worldwide. Research for new therapeutics and the investigation of their effects on vascular parameters is often limited by species-specific pathways and a lack of high-throughput methods. The complex 3-dimensional environment of blood vessels, intricate cellular crosstalks, and organ-specific architectures further complicate the quest for a faithful human in vitro model. The development of novel organoid models of various tissues such as brain, gut, and kidney signified a leap for the field of personalized medicine and disease research. By utilizing either embryonic- or patient-derived stem cells, different developmental and pathological mechanisms can be modeled and investigated in a controlled in vitro environment. We have recently developed self-organizing human capillary blood vessel organoids that recapitulate key processes of vasculogenesis, angiogenesis, and diabetic vasculopathy. Since then, this organoid system has been utilized as a model for other disease processes, refined, and adapted for organ specificity. In this review, we will discuss novel and alternative approaches to blood vessel engineering and explore the cellular identity of engineered blood vessels in comparison to in vivo vasculature. Future perspectives and the therapeutic potential of blood vessel organoids will be discussed.
Collapse
Affiliation(s)
- Kirill Salewskij
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna (K.S., J.M.P.).,Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Austria (K.S.)
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna (K.S., J.M.P.).,Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada (J.M.P.)
| |
Collapse
|
18
|
Sasaki S, Suzuki T, Morikawa K, Matsusaki M, Sato K. Fabrication of a Gelatin-Based Microdevice for Vascular Cell Culture. MICROMACHINES 2022; 14:107. [PMID: 36677169 PMCID: PMC9860854 DOI: 10.3390/mi14010107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 06/17/2023]
Abstract
This study presents a novel technique for fabricating microfluidic devices with microbial transglutaminase-gelatin gels instead of polydimethylsiloxane (PDMS), in which flow culture simulates blood flow and a capillary network is incorporated for assays of vascular permeability or angiogenesis. We developed a gelatin-based device with a coverslip as the bottom, which allows the use of high-magnification lenses with short working distances, and we observed the differences in cell dynamics on gelatin, glass, and PDMS surfaces. The tubes of the gelatin microfluidic channel are designed to be difficult to pull out of the inlet hole, making sample introduction easy, and the gelatin channel can be manipulated from the cell introduction to the flow culture steps in a manner comparable to that of a typical PDMS channel. Human umbilical vein endothelial cells (HUVECs) and normal human dermal fibroblasts (NHDFs) were successfully co-cultured, resulting in structures that mimicked blood vessels with inner diameters ranging from 10 µm to 500 µm. Immunostaining and scanning electron microscopy results showed that the affinity of fibronectin for gelatin was stronger than that for glass or PDMS, making gelatin a suitable substrate for cell adhesion. The ability for microscopic observation at high magnification and the ease of sample introduction make this device easier to use than conventional gelatin microfluidics, and the above-mentioned small modifications in the device structure are important points that improve its convenience as a cell assay device.
Collapse
Affiliation(s)
- Satoko Sasaki
- Department of Chemical and Biological Sciences, Faculty of Science, Japan Women’s University, 2-8-1 Mejirodai, Bunkyo, Tokyo 112-8681, Japan
| | - Tomoko Suzuki
- Department of Chemical and Biological Sciences, Faculty of Science, Japan Women’s University, 2-8-1 Mejirodai, Bunkyo, Tokyo 112-8681, Japan
| | - Kyojiro Morikawa
- Institute of Nanoengineering and Microsystems, Department of Power Mechanical Engineering, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
- Collaborative Research Organization for Micro and Nano Multifunctional Devices, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo 113-8656, Japan
| | - Michiya Matsusaki
- Division of Applied Chemistry, Graduate School of Engineering, Osaka University, 1-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kae Sato
- Department of Chemical and Biological Sciences, Faculty of Science, Japan Women’s University, 2-8-1 Mejirodai, Bunkyo, Tokyo 112-8681, Japan
| |
Collapse
|
19
|
Joddar B, Natividad-Diaz SL, Padilla AE, Esparza AA, Ramirez SP, Chambers DR, Ibaroudene H. Engineering approaches for cardiac organoid formation and their characterization. Transl Res 2022; 250:46-67. [PMID: 35995380 PMCID: PMC10370285 DOI: 10.1016/j.trsl.2022.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/05/2022] [Accepted: 08/15/2022] [Indexed: 11/29/2022]
Abstract
Cardiac organoids are 3-dimensional (3D) structures composed of tissue or niche-specific cells, obtained from diverse sources, encapsulated in either a naturally derived or synthetic, extracellular matrix scaffold, and include exogenous biochemical signals such as essential growth factors. The overarching goal of developing cardiac organoid models is to establish a functional integration of cardiomyocytes with physiologically relevant cells, tissues, and structures like capillary-like networks composed of endothelial cells. These organoids used to model human heart anatomy, physiology, and disease pathologies in vitro have the potential to solve many issues related to cardiovascular drug discovery and fundamental research. The advent of patient-specific human-induced pluripotent stem cell-derived cardiovascular cells provide a unique, single-source approach to study the complex process of cardiovascular disease progression through organoid formation and incorporation into relevant, controlled microenvironments such as microfluidic devices. Strategies that aim to accomplish such a feat include microfluidic technology-based approaches, microphysiological systems, microwells, microarray-based platforms, 3D bioprinted models, and electrospun fiber mat-based scaffolds. This article discusses the engineering or technology-driven practices for making cardiac organoid models in comparison with self-assembled or scaffold-free methods to generate organoids. We further discuss emerging strategies for characterization of the bio-assembled cardiac organoids including electrophysiology and machine-learning and conclude with prospective points of interest for engineering cardiac tissues in vitro.
Collapse
Affiliation(s)
- Binata Joddar
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL); Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas; Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas.
| | - Sylvia L Natividad-Diaz
- Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas; Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas
| | - Andie E Padilla
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL); Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas
| | - Aibhlin A Esparza
- Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas
| | - Salma P Ramirez
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL); Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas
| | | | | |
Collapse
|
20
|
Mayo LN, Kutys ML. Conversation before crossing: dissecting metastatic tumor-vascular interactions in microphysiological systems. Am J Physiol Cell Physiol 2022; 323:C1333-C1344. [PMID: 36121131 PMCID: PMC9602802 DOI: 10.1152/ajpcell.00173.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 01/12/2023]
Abstract
Tumor metastasis via the circulation requires crossing the vascular barrier twice: first, during intravasation when tumor cells disseminate from the primary site through proximal vasculature, and second, during extravasation, when tumor cells exit the circulation to form distant metastatic seeds. During these key metastatic events, chemomechanical signaling between tumor cells and endothelial cells elicits reciprocal changes in cell morphology and behavior that are necessary to breach the vessel wall. Existing experimental systems have provided a limited understanding of the diverse mechanisms underlying tumor-endothelial interactions during intravasation and extravasation. Recent advances in microphysiological systems have revolutionized the ability to generate miniaturized human tissues with tailored three-dimensional architectures, physiological cell interfaces, and precise chemical and physical microenvironments. By doing so, microphysiological systems enable experimental access to complex morphogenic processes associated with human tumor progression with unprecedented resolution and biological control. Here, we discuss recent examples in which microphysiological systems have been leveraged to reveal new mechanistic insight into cellular and molecular control systems operating at the tumor-endothelial interface during intravasation and extravasation.
Collapse
Affiliation(s)
- Lakyn N Mayo
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, California
- UCSF-UC Berkeley Joint Graduate Program in Bioengineering, University of California San Francisco, San Francisco, California
- Medical Scientist Training Program, University of California San Francisco, San Francisco, California
| | - Matthew L Kutys
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, California
- UCSF-UC Berkeley Joint Graduate Program in Bioengineering, University of California San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| |
Collapse
|
21
|
Jia F, Gao Y, Wang H. Recent Advances in Drug Delivery System Fabricated by Microfluidics for Disease Therapy. Bioengineering (Basel) 2022; 9:625. [PMID: 36354536 PMCID: PMC9687342 DOI: 10.3390/bioengineering9110625] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/16/2022] [Accepted: 10/26/2022] [Indexed: 09/08/2024] Open
Abstract
Traditional drug therapy faces challenges such as drug distribution throughout the body, rapid degradation and excretion, and extensive adverse reactions. In contrast, micro/nanoparticles can controllably deliver drugs to target sites to improve drug efficacy. Unlike traditional large-scale synthetic systems, microfluidics allows manipulation of fluids at the microscale and shows great potential in drug delivery and precision medicine. Well-designed microfluidic devices have been used to fabricate multifunctional drug carriers using stimuli-responsive materials. In this review, we first introduce the selection of materials and processing techniques for microfluidic devices. Then, various well-designed microfluidic chips are shown for the fabrication of multifunctional micro/nanoparticles as drug delivery vehicles. Finally, we describe the interaction of drugs with lymphatic vessels that are neglected in organs-on-chips. Overall, the accelerated development of microfluidics holds great potential for the clinical translation of micro/nanoparticle drug delivery systems for disease treatment.
Collapse
Affiliation(s)
- Fuhao Jia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanbing Gao
- Troop 96901 of the Chinese People’s Liberation Army, Beijing 100094, China
| | - Hai Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
22
|
Wan Z, Zhong AX, Zhang S, Pavlou G, Coughlin MF, Shelton SE, Nguyen HT, Lorch JH, Barbie DA, Kamm RD. A Robust Method for Perfusable Microvascular Network Formation In Vitro. SMALL METHODS 2022; 6:e2200143. [PMID: 35373502 PMCID: PMC9844969 DOI: 10.1002/smtd.202200143] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/03/2022] [Indexed: 05/02/2023]
Abstract
Micropost-based microfluidic devices are widely used for microvascular network (MVN) formation in diverse research fields. However, consistently generating perfusable MVNs of physiological morphology and dimension has proven to be challenging. Here, how initial seeding parameters determine key characteristics of MVN formation is investigated and a robust two-step seeding strategy to generate perfusable physiological MVNs in microfluidic devices is established.
Collapse
Affiliation(s)
- Zhengpeng Wan
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Amy X Zhong
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Shun Zhang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Georgios Pavlou
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Mark F Coughlin
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Sarah E Shelton
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Huu Tuan Nguyen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Jochen H Lorch
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - David A Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Roger D Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
23
|
Kolarzyk AM, Wong G, Lee E. Lymphatic Tissue and Organ Engineering for In Vitro Modeling and In Vivo Regeneration. Cold Spring Harb Perspect Med 2022; 12:a041169. [PMID: 35288402 PMCID: PMC9435571 DOI: 10.1101/cshperspect.a041169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The lymphatic system has an important role in maintaining fluid homeostasis and transporting immune cells and biomolecules, such as dietary fat, metabolic products, and antigens in different organs and tissues. Therefore, impaired lymphatic vessel function and/or lymphatic vessel deficiency can lead to numerous human diseases. The discovery of lymphatic endothelial markers and prolymphangiogenic growth factors, along with a growing number of in vitro and in vivo models and technologies has expedited research in lymphatic tissue and organ engineering, advancing therapeutic strategies. In this article, we describe lymphatic tissue and organ engineering in two- and three-dimensional culture systems and recently developed microfluidics and organ-on-a-chip systems in vitro. Next, we discuss advances in lymphatic tissue and organ engineering in vivo, focusing on biomaterial and scaffold engineering and their applications for lymphatic vessels and lymphoid organ regeneration. Last, we provide expert perspective and prospects in the field of lymphatic tissue engineering.
Collapse
Affiliation(s)
- Anna M Kolarzyk
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York 14853, USA
- Biomedical and Biological Sciences PhD Program, Ithaca, New York 14853, USA
| | - Gigi Wong
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York 14853, USA
- Biological Sciences, Cornell University, Ithaca, New York 14853, USA
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York 14853, USA
- Biomedical and Biological Sciences PhD Program, Ithaca, New York 14853, USA
| |
Collapse
|
24
|
Ersland E, Ebrahim N, Mwizerwa O, Oba T, Oku K, Nishino M, Hikimoto D, Miyoshi H, Tomotoshi K, Rahmanian O, Ekwueme E, Neville C, Sundback C. Human Vascular Wall Microfluidic Model for Preclinical Evaluation of Drug-Induced Vascular Injury. Tissue Eng Part C Methods 2022; 28:83-92. [PMID: 35114818 PMCID: PMC9022170 DOI: 10.1089/ten.tec.2021.0227] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Drug-induced vascular injury (DIVI) in preclinical animal models often leads to candidate compound termination during drug development. DIVI has not been documented in human clinical trials with drugs that cause DIVI in preclinical animals. A robust human preclinical assay for DIVI is needed as an early vascular injury screen. A human vascular wall microfluidic tissue chip was developed with a human umbilical vein endothelial cell (HUVEC)-umbilical artery smooth muscle cell (vascular smooth muscle cell, VSMC) bilayer matured under physiological shear stress. Optimized temporal flow profiles produced HUVEC-VSMC bilayers with quiescent endothelial cell (EC) monolayers, EC tight junctions, and contractile VSMC morphology. Dose-response testing (3-30 μM concentration) was conducted with minoxidil and tadalafil vasodilators. Both drugs have demonstrated preclinical DIVI but lack clinical evidence. The permeability of severely damaged engineered bilayers (30 μM tadalafil) was 4.1 times that of the untreated controls. Immunohistochemical protein assays revealed contrasting perspectives on tadalafil and minoxidil-induced damage. Tadalafil impacted the endothelial monolayer with minor injury to the contractile VSMCs, whereas minoxidil demonstrated minor EC barrier injury but damaged VSMCs and activated ECs in a dose-response manner. This proof-of-concept human vascular wall bilayer model of DIVI is a critical step toward developing a preclinical human screening assay for drug development. Impact statement More than 90% of drug candidates fail during clinical trials due to human efficacy and toxicity concerns. Preclinical studies rely heavily on animal models, although animal toxicity and drug metabolism responses often differ from humans. During the drug development process, perfused in vitro human tissue chips could model the clinical drug response and potential toxicity of candidate compounds. Our long-term objective is to develop a human vascular wall tissue chip to screen for drug-induced vascular injury. Its application could ultimately reduce drug development delays and costs, and improve patient safety.
Collapse
Affiliation(s)
- Erik Ersland
- Department of Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Neven Ebrahim
- Department of Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA.,Department of Anatomy and Embryology, Mansoura University, Mansoura, Egypt
| | - Olive Mwizerwa
- Department of Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Takahiro Oba
- Bioscience and Engineering Laboratories, FUJIFILM Corporation, Kanagawa, Japan
| | - Keisuke Oku
- Bioscience and Engineering Laboratories, FUJIFILM Corporation, Kanagawa, Japan
| | - Masafumi Nishino
- Bioscience and Engineering Laboratories, FUJIFILM Corporation, Kanagawa, Japan
| | - Daichi Hikimoto
- Bioscience and Engineering Laboratories, FUJIFILM Corporation, Kanagawa, Japan
| | - Hayato Miyoshi
- Bioscience and Engineering Laboratories, FUJIFILM Corporation, Kanagawa, Japan
| | - Kimihiko Tomotoshi
- Bioscience and Engineering Laboratories, FUJIFILM Corporation, Kanagawa, Japan
| | - Omid Rahmanian
- Department of Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Emmanuel Ekwueme
- Department of Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Craig Neville
- Department of Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Cathryn Sundback
- Department of Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA.,Address correspondence to: Cathryn Sundback, ScD, Department of Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 01451, USA
| |
Collapse
|
25
|
From organ-on-chip to body-on-chip: The next generation of microfluidics platforms for in vitro drug efficacy and toxicity testing. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 187:41-91. [PMID: 35094781 DOI: 10.1016/bs.pmbts.2021.07.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The high failure rate in drug development is often attributed to the lack of accurate pre-clinical models that may lead to false discoveries and inconclusive data when the compounds are eventually tested in clinical phase. With the evolution of cell culture technologies, drug testing systems have widely improved, and today, with the emergence of microfluidics devices, drug screening seems to be at the dawn of an important revolution. An organ-on-chip allows the culture of living cells in continuously perfused microchambers to reproduce physiological functions of a particular tissue or organ. The advantages of such systems are not only their ability to recapitulate the complex biochemical interactions between different human cell types but also to incorporate physical forces, including shear stress and mechanical stretching or compression. To improve this model, and to reproduce the absorption, distribution, metabolism, and elimination process of an exogenous compound, organ-on-chips can even be linked fluidically to mimic physiological interactions between different organs, leading to the development of body-on-chips. Although these technologies are still at a young age and need to address a certain number of limitations, they already demonstrated their relevance to study the effect of drugs or toxins on organs, displaying a similar response to what is observed in vivo. The purpose of this review is to present the evolution from organ-on-chip to body-on-chip, examine their current use for drug testing and discuss their advantages and future challenges they will face in order to become an essential pillar of pharmaceutical research.
Collapse
|
26
|
SATO K. Development of Microvascular Devices and Their Application to Bioanalytical Chemistry. BUNSEKI KAGAKU 2022. [DOI: 10.2116/bunsekikagaku.71.53] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Kae SATO
- Department of Chemical and Biological Sciences, Japan Women's University
| |
Collapse
|
27
|
Gao F, Sun H, Li X, He P. Leveraging avidin-biotin interaction to quantify permeability property of microvessels-on-a-chip networks. Am J Physiol Heart Circ Physiol 2022; 322:H71-H86. [PMID: 34767485 PMCID: PMC8698539 DOI: 10.1152/ajpheart.00478.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Microvessels-on-a-chip have enabled in vitro studies to closely simulate in vivo microvessel environment. However, assessing microvessel permeability, a functional measure of microvascular exchange, has not been attainable in nonpermeable microfluidic platforms. This study developed a new approach that enables permeability coefficients (Ps) to be quantified in microvessels developed in nonpermeable chip platforms by integrating avidin-biotin technology. Microvessels were developed on biotinylated fibronectin-coated microfluidic channels. Solute transport was assessed by perfusing microvessels with fluorescence-labeled avidin. Avidin molecules that crossed endothelium were captured by substrate biotin and recorded with real-time confocal images. The Ps was derived from the rate of avidin-biotin accumulation at the substrate relative to solute concentration difference across microvessel wall. Avidin tracers with different physiochemical properties were used to characterize the barrier properties of the microvessel wall. The measured baseline Ps and inflammatory mediator-induced increases in Ps and endothelial cell (EC) [Ca2+]i resembled those observed in intact microvessels. Importantly, the spatial accumulation of avidin-biotin at substrate defines the transport pathways. Glycocalyx layer is well formed on endothelium and its degradation increased transcellular transport without affecting EC junctions. This study demonstrated that in vitro microvessels developed in this simply designed microfluidics structurally possess in vivo-like glycocalyx layer and EC junctions and functionally recapitulate basal barrier properties and stimuli-induced responses observed in intact microvessels. This new approach overcomes the limitations of nonpermeable microfluidics and provides an easily executed highly reproducible in vitro microvessel model with in vivo microvessel functionality, suitable for a wide range of applications in blood and vascular research and drug development.NEW & NOTEWORTHY Our study developed a novel method that allows permeability coefficient to be measured in microvessels developed in nonpermeable microfluidic platforms using avidin-biotin technology. It overcomes the major limitation of nonpermeable microfluidic system and provides a simply designed easily executed and highly reproducible in vitro microvessel model with permeability accessibility. This model with in vivo-like endothelial junctions, glycocalyx, and permeability properties advances microfluidics in microvascular research, suitable for a wide range of biomedical and clinical applications.
Collapse
|
28
|
Selahi A, Fernando T, Chakraborty S, Muthuchamy M, Zawieja DC, Jain A. Lymphangion-chip: a microphysiological system which supports co-culture and bidirectional signaling of lymphatic endothelial and muscle cells. LAB ON A CHIP 2021; 22:121-135. [PMID: 34850797 PMCID: PMC9761984 DOI: 10.1039/d1lc00720c] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The pathophysiology of several lymphatic diseases, such as lymphedema, depends on the function of lymphangions that drive lymph flow. Even though the signaling between the two main cellular components of a lymphangion, endothelial cells (LECs) and muscle cells (LMCs), is responsible for crucial lymphatic functions, there are no in vitro models that have included both cell types. Here, a fabrication technique (gravitational lumen patterning or GLP) is developed to create a lymphangion-chip. This organ-on-chip consists of co-culture of a monolayer of endothelial lumen surrounded by multiple and uniformly thick layers of muscle cells. The platform allows construction of a wide range of luminal diameters and muscular layer thicknesses, thus providing a toolbox to create variable anatomy. In this device, lymphatic muscle cells align circumferentially while endothelial cells aligned axially under flow, as only observed in vivo in the past. This system successfully characterizes the dynamics of cell size, density, growth, alignment, and intercellular gap due to co-culture and shear. Finally, exposure to pro-inflammatory cytokines reveals that the device could facilitate the regulation of endothelial barrier function through the lymphatic muscle cells. Therefore, this bioengineered platform is suitable for use in preclinical research of lymphatic and blood mechanobiology, inflammation, and translational outcomes.
Collapse
Affiliation(s)
- Amirali Selahi
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 101 Bizzell Street College Station, TX, 77843, USA.
| | - Teshan Fernando
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 101 Bizzell Street College Station, TX, 77843, USA.
| | - Sanjukta Chakraborty
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, TX, USA
| | - Mariappan Muthuchamy
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, TX, USA
| | - David C Zawieja
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, TX, USA
| | - Abhishek Jain
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 101 Bizzell Street College Station, TX, 77843, USA.
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, TX, USA
- Department of Cardiovascular Sciences, Houston Methodist Academic Institute, Houston, TX, USA
| |
Collapse
|
29
|
Trapecar M. Multiorgan microphysiological systems as tools to interrogate interorgan crosstalk and complex diseases. FEBS Lett 2021; 596:681-695. [PMID: 34923635 DOI: 10.1002/1873-3468.14260] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022]
Abstract
Metabolic and inflammatory disorders such as autoimmune and neurodegenerative diseases are increasing at alarming rates. Many of these are not tissue-specific occurrences but complex, systemic pathologies of unknown origin for which no cure exists. Such complexity obscures causal relationships among factors regulating disease progression. Emerging technologies mimicking human physiology, such as microphysiological systems (MPSs), offer new possibilities to provide clarity in systemic metabolic and inflammatory diseases. Controlled interaction of multiple MPSs and the scalability of biological complexity in MPSs, supported by continuous multiomic monitoring, might hold the key to identifying novel relationships between interorgan crosstalk, metabolism, and immunity. In this perspective, I aim to discuss the current state of modeling multiorgan physiology and evaluate current opportunities and challenges.
Collapse
Affiliation(s)
- Martin Trapecar
- Department of Medicine, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
30
|
Bazban-Shotorbani S, Gavins F, Kant K, Dufva M, Kamaly N. A Biomicrofluidic Screening Platform for Dysfunctional Endothelium‐Targeted Nanoparticles and Therapeutics. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Salime Bazban-Shotorbani
- Department of Health Technology DTU Health Tech Technical University of Denmark Lyngby 2800 Kgs. Denmark
- Department of Chemistry Molecular Sciences Research Hub (MSRH) Imperial College London London W12 0BZ UK
| | - Felicity Gavins
- Department of Life Sciences Centre for Inflammation Research and Translational Medicine (CIRTM) Brunel University London London UB8 3PH UK
| | - Krishna Kant
- Department of Physical Chemistry Biomedical Research Center of Galicia (CINBIO) University of Vigo Vigo 36310 Spain
| | - Martin Dufva
- Department of Health Technology DTU Health Tech Technical University of Denmark Lyngby 2800 Kgs. Denmark
| | - Nazila Kamaly
- Department of Chemistry Molecular Sciences Research Hub (MSRH) Imperial College London London W12 0BZ UK
| |
Collapse
|
31
|
Singh YP, Moses JC, Bhardwaj N, Mandal BB. Overcoming the Dependence on Animal Models for Osteoarthritis Therapeutics - The Promises and Prospects of In Vitro Models. Adv Healthc Mater 2021; 10:e2100961. [PMID: 34302436 DOI: 10.1002/adhm.202100961] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/10/2021] [Indexed: 12/19/2022]
Abstract
Osteoarthritis (OA) is a musculoskeletal disease characterized by progressive degeneration of osteochondral tissues. Current treatment is restricted to the reduction of pain and loss of function of the joint. To better comprehend the OA pathophysiological conditions, several models are employed, however; there is no consensus on a suitable model. In this review, different in vitro models being developed for possible therapeutic intervention of OA are outlined. Herein, various in vitro OA models starting from 2D model, co-culture model, 3D models, dynamic culture model to advanced technologies-based models such as 3D bioprinting, bioassembly, organoids, and organ-on-chip-based models are discussed with their advantages and disadvantages. Besides, different growth factors, cytokines, and chemicals being utilized for induction of OA condition are reviewed in detail. Furthermore, there is focus on scrutinizing different molecular and possible therapeutic targets for better understanding the mechanisms and OA therapeutics. Finally, the underlying challenges associated with in vitro models are discussed followed by future prospective. Taken together, a comprehensive overview of in vitro OA models, factors to induce OA-like conditions, and intricate molecular targets with the potential to develop personalized osteoarthritis therapeutics in the future with clinical translation is provided.
Collapse
Affiliation(s)
- Yogendra Pratap Singh
- Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| | - Joseph Christakiran Moses
- Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| | - Nandana Bhardwaj
- Department of Science and Mathematics Indian Institute of Information Technology Guwahati Bongora Guwahati Assam 781015 India
| | - Biman B. Mandal
- Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
- Centre for Nanotechnology Indian Institute of Technology Guwahati Guwahati Assam 781039 India
- School of Health Sciences and Technology Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| |
Collapse
|
32
|
Dellaquila A, Le Bao C, Letourneur D, Simon‐Yarza T. In Vitro Strategies to Vascularize 3D Physiologically Relevant Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100798. [PMID: 34351702 PMCID: PMC8498873 DOI: 10.1002/advs.202100798] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/23/2021] [Indexed: 05/04/2023]
Abstract
Vascularization of 3D models represents a major challenge of tissue engineering and a key prerequisite for their clinical and industrial application. The use of prevascularized models built from dedicated materials could solve some of the actual limitations, such as suboptimal integration of the bioconstructs within the host tissue, and would provide more in vivo-like perfusable tissue and organ-specific platforms. In the last decade, the fabrication of vascularized physiologically relevant 3D constructs has been attempted by numerous tissue engineering strategies, which are classified here in microfluidic technology, 3D coculture models, namely, spheroids and organoids, and biofabrication. In this review, the recent advancements in prevascularization techniques and the increasing use of natural and synthetic materials to build physiological organ-specific models are discussed. Current drawbacks of each technology, future perspectives, and translation of vascularized tissue constructs toward clinics, pharmaceutical field, and industry are also presented. By combining complementary strategies, these models are envisioned to be successfully used for regenerative medicine and drug development in a near future.
Collapse
Affiliation(s)
- Alessandra Dellaquila
- Université de ParisINSERM U1148X Bichat HospitalParisF‐75018France
- Elvesys Microfluidics Innovation CenterParis75011France
- Biomolecular PhotonicsDepartment of PhysicsUniversity of BielefeldBielefeld33615Germany
| | - Chau Le Bao
- Université de ParisINSERM U1148X Bichat HospitalParisF‐75018France
- Université Sorbonne Paris NordGalilée InstituteVilletaneuseF‐93430France
| | | | | |
Collapse
|
33
|
Alderfer L, Hall E, Hanjaya-Putra D. Harnessing biomaterials for lymphatic system modulation. Acta Biomater 2021; 133:34-45. [PMID: 34118451 PMCID: PMC9113193 DOI: 10.1016/j.actbio.2021.06.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/20/2021] [Accepted: 06/01/2021] [Indexed: 12/20/2022]
Abstract
The lymphatic system plays an integral part in regulating immune cell trafficking and the transport of macromolecules. However, its influence on disease progression and drug uptake is understood less than that of the vascular system. To bridge this knowledge gap, biomaterials can be used to investigate the lymphatic system and to provide novel understanding into complex disease processes, including cancer metastasis and inflammation. Insight gained from these mechanistic studies can be further used to design innovative biomaterials to modulate the immune system, improve drug delivery, and promote tissue regeneration. This review article focuses on recent advances in (i) biomaterials used for lymphatic vessel formation, (ii) models for studying lymphatic-immune cells interactions, (iii) pharmaceuticals and their interactions with the lymphatic system, (iv) and strategies for drug delivery via the lymphatic system. Finally, several challenges regarding adopting biomaterials for immunomodulation and future perspectives are discussed. STATEMENT OF SIGNIFICANCE: The lymphatic system plays an integral part in regulating immune cell trafficking and the transport of macromolecules. However, its influence on disease progression and drug uptake is understood less than that of the vascular system. This review article focuses on recent progresses in biomaterials to investigate the lymphatic system and to provide novel understanding into complex disease states. Insight gained from these mechanistic studies can be further used to design innovative biomaterials to modulate the immune system, improve drug delivery, and promote tissue regeneration. Finally, a number of challenges in adopting biomaterials for immunomodulation and future perspectives are discussed.
Collapse
|
34
|
Yu H, Kang D, Whang M, Kim T, Kim J. A Microfluidic Model Artery for Studying the Mechanobiology of Endothelial Cells. Adv Healthc Mater 2021; 10:e2100508. [PMID: 34297476 DOI: 10.1002/adhm.202100508] [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: 03/16/2021] [Revised: 06/25/2021] [Indexed: 11/07/2022]
Abstract
Recent vascular mechanobiology studies find that endothelial cells (ECs) convert multiple mechanical forces into functional responses in a nonadditive way, suggesting that signaling pathways such as those regulating cytoskeleton may be shared among the processes of converting individual forces. However, previous in vitro EC-culture platforms are inherent with extraneous mechanical components, which may saturate or insufficiently activate the shared signaling pathways and accordingly, may misguide EC mechanobiological responses being investigated. Here, a more physiologically relevant model artery is reported that accurately reproduces most of the mechanical forces found in vivo, which can be individually varied in any combination to pathological levels to achieve diseased states. Arterial geometries of normal and diseased states are also realized. By mimicking mechanical microenvironments of early-stage atherosclerosis, it is demonstrated that the elevated levels of the different types of stress experienced by ECs strongly correlate with the disruption of barrier integrity, suggesting that boundaries of an initial lesion could be sites for efficient disease progression.
Collapse
Affiliation(s)
- Hyeonji Yu
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 04107, Republic of Korea
| | - Dongwon Kang
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 04107, Republic of Korea
| | - Minji Whang
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 04107, Republic of Korea
| | - Taeyoung Kim
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 04107, Republic of Korea
| | - Jungwook Kim
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 04107, Republic of Korea
| |
Collapse
|
35
|
Rhyou J, Youn J, Eom S, Kim DS. Facile Fabrication of Electrospun Nanofiber Membrane-Integrated PDMS Microfluidic Chip via Silver Nanowires-Uncured PDMS Adhesive Layer. ACS Macro Lett 2021; 10:965-970. [PMID: 35549208 DOI: 10.1021/acsmacrolett.1c00256] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Although direct electrospinning has been frequently utilized to develop a nanofiber membrane-integrated microfluidic chip, the dielectric substrate material retards the deposition of electrospun nanofibers on the substrate, and the rough surface formed by deposited nanofibers hinders the successful sealing. In this study we introduce a facile fabrication process of an electrospun nanofiber membrane-integrated polydimethylsiloxane (PDMS) microfluidic chip, called a NFM-PDMS chip, by applying the functional layer. The functional layer consists of a silver nanowires (AgNWs)-embedded uncured PDMS adhesive layer (SNUP), which not only effectively concentrates the electric field toward the PDMS substrate, but also provides a smooth surface for robust sealing. The AgNWs in the SNUP play a crucial role as a grounded collector and enable approximately 4× faster electrospinning than the conventional method, forming a free-standing nanofiber membrane. The uncured PDMS adhesive layer in the SNUP maintains the smooth surface after electrospinning and allows the rapid and leakage-free bonding of the NFM-PDMS chip using plasma treatment. A practical application of the NFM-PDMS chip is demonstrated by culturing the human keratinocyte cell line, HaCaT cells. The HaCaT cells are well grown on the free-standing nanofiber membrane under dynamic flow conditions, maintaining good viability over 95% for 7 days of culture.
Collapse
Affiliation(s)
- Junyeol Rhyou
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Jaeseung Youn
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Seongsu Eom
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Dong Sung Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
- Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, 50,
Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|
36
|
Hammel JH, Cook SR, Belanger MC, Munson JM, Pompano RR. Modeling Immunity In Vitro: Slices, Chips, and Engineered Tissues. Annu Rev Biomed Eng 2021; 23:461-491. [PMID: 33872520 PMCID: PMC8277680 DOI: 10.1146/annurev-bioeng-082420-124920] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Modeling immunity in vitro has the potential to be a powerful tool for investigating fundamental biological questions, informing therapeutics and vaccines, and providing new insight into disease progression. There are two major elements to immunity that are necessary to model: primary immune tissues and peripheral tissues with immune components. Here, we systematically review progress made along three strategies to modeling immunity: ex vivo cultures, which preserve native tissue structure; microfluidic devices, which constitute a versatile approach to providing physiologically relevant fluid flow and environmental control; and engineered tissues, which provide precise control of the 3D microenvironment and biophysical cues. While many models focus on disease modeling, more primary immune tissue models are necessary to advance the field. Moving forward, we anticipate that the expansion of patient-specific models may inform why immunity varies from patient to patient and allow for the rapid comprehension and treatment of emerging diseases, such as coronavirus disease 2019.
Collapse
Affiliation(s)
- Jennifer H Hammel
- Fralin Biomedical Research Institute and Department of Biomedical Engineering and Mechanics, Virginia Tech, Roanoke, Virginia 24016, USA;
| | - Sophie R Cook
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA
| | - Maura C Belanger
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA
| | - Jennifer M Munson
- Fralin Biomedical Research Institute and Department of Biomedical Engineering and Mechanics, Virginia Tech, Roanoke, Virginia 24016, USA;
| | - Rebecca R Pompano
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22904, USA;
- Carter Immunology Center and UVA Cancer Center, University of Virginia School of Medicine, Charlottesville, Virginia 22903, USA
| |
Collapse
|
37
|
Frenkel N, Poghosyan S, Alarcón CR, García SB, Queiroz K, van den Bent L, Laoukili J, Rinkes IB, Vulto P, Kranenburg O, Hagendoorn J. Long-Lived Human Lymphatic Endothelial Cells to Study Lymphatic Biology and Lymphatic Vessel/Tumor Coculture in a 3D Microfluidic Model. ACS Biomater Sci Eng 2021; 7:3030-3042. [PMID: 34185991 DOI: 10.1021/acsbiomaterials.0c01378] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The lymphatic system is essential in maintaining tissue fluid homeostasis as well as antigen and immune cell transport to lymph nodes. Moreover, lymphatic vasculature plays an important role in various pathological processes, such as cancer. Fundamental to this research field are representative in vitro models. Here we present a microfluidic lymphatic vessel model to study lymphangiogenesis and its interaction with colon cancer organoids using a newly developed lymphatic endothelial cell (LEC) line. We generated immortalized human LECs by lentiviral transduction of human telomerase (hTERT) and BMI-1 expression cassettes into primary LECs. Immortalized LECs showed an increased growth potential, reduced senescence, and elongated lifespan with maintenance of typical LEC morphology and marker expression for over 12 months while remaining nontransformed. Immortalized LECs were introduced in a microfluidic chip, comprising a free-standing extracellular matrix, where they formed a perfusable vessel-like structure against the extracellular matrix. A gradient of lymphangiogenic factors over the extracellular matrix gel induced the formation of luminated sprouts. Adding mouse colon cancer organoids adjacent to the lymphatic vessel resulted in a stable long-lived coculture model in which cancer cell-induced lymphangiogenesis and cancer cell motility can be investigated. Thus, the development of a stable immortalized lymphatic endothelial cell line in a membrane-free, perfused microfluidic chip yields a highly standardized lymphangiogenesis and lymphatic vessel-tumor cell coculture assay.
Collapse
Affiliation(s)
- Nicola Frenkel
- UMC Utrecht Cancer Center, University Medical Center Utrecht and Utrecht University, Heidelberglaan 100, Utrecht 3584CX, The Netherlands
| | - Susanna Poghosyan
- UMC Utrecht Cancer Center, University Medical Center Utrecht and Utrecht University, Heidelberglaan 100, Utrecht 3584CX, The Netherlands
| | - Carmen Rubio Alarcón
- UMC Utrecht Cancer Center, University Medical Center Utrecht and Utrecht University, Heidelberglaan 100, Utrecht 3584CX, The Netherlands
| | | | | | - Lotte van den Bent
- UMC Utrecht Cancer Center, University Medical Center Utrecht and Utrecht University, Heidelberglaan 100, Utrecht 3584CX, The Netherlands
| | - Jamila Laoukili
- UMC Utrecht Cancer Center, University Medical Center Utrecht and Utrecht University, Heidelberglaan 100, Utrecht 3584CX, The Netherlands
| | - Inne Borel Rinkes
- UMC Utrecht Cancer Center, University Medical Center Utrecht and Utrecht University, Heidelberglaan 100, Utrecht 3584CX, The Netherlands
| | - Paul Vulto
- Mimetas BV, JH Oortweg 19, Leiden, The Netherlands
| | - Onno Kranenburg
- UMC Utrecht Cancer Center, University Medical Center Utrecht and Utrecht University, Heidelberglaan 100, Utrecht 3584CX, The Netherlands
| | - Jeroen Hagendoorn
- UMC Utrecht Cancer Center, University Medical Center Utrecht and Utrecht University, Heidelberglaan 100, Utrecht 3584CX, The Netherlands
| |
Collapse
|
38
|
Llenas M, Paoli R, Feiner-Gracia N, Albertazzi L, Samitier J, Caballero D. Versatile Vessel-on-a-Chip Platform for Studying Key Features of Blood Vascular Tumors. Bioengineering (Basel) 2021; 8:bioengineering8060081. [PMID: 34207754 PMCID: PMC8226980 DOI: 10.3390/bioengineering8060081] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/26/2021] [Accepted: 06/04/2021] [Indexed: 12/19/2022] Open
Abstract
Tumor vessel-on-a-chip systems have attracted the interest of the cancer research community due to their ability to accurately recapitulate the multiple dynamic events of the metastatic cascade. Vessel-on-a-chip microfluidic platforms have been less utilized for investigating the distinctive features and functional heterogeneities of tumor-derived vascular networks. In particular, vascular tumors are characterized by the massive formation of thrombi and severe bleeding, a rare and life-threatening situation for which there are yet no clear therapeutic guidelines. This is mainly due to the lack of technological platforms capable of reproducing these characteristic traits of the pathology in a simple and well-controlled manner. Herein, we report the fabrication of a versatile tumor vessel-on-a-chip platform to reproduce, investigate, and characterize the massive formation of thrombi and hemorrhage on-chip in a fast and easy manner. Despite its simplicity, this method offers multiple advantages to recapitulate the pathophysiological events of vascular tumors, and therefore, may find useful applications in the field of vascular-related diseases, while at the same time being an alternative to more complex approaches.
Collapse
Affiliation(s)
- Marina Llenas
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 15–21, 08028 Barcelona, Spain; (M.L.); (R.P.); (N.F.-G.)
- Department of Electronics and Biomedical Engineering, University of Barcelona, C. Martí i Franqués 1, 08028 Barcelona, Spain
| | - Roberto Paoli
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 15–21, 08028 Barcelona, Spain; (M.L.); (R.P.); (N.F.-G.)
- Department of Electronics and Biomedical Engineering, University of Barcelona, C. Martí i Franqués 1, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Natalia Feiner-Gracia
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 15–21, 08028 Barcelona, Spain; (M.L.); (R.P.); (N.F.-G.)
| | - Lorenzo Albertazzi
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 15–21, 08028 Barcelona, Spain; (M.L.); (R.P.); (N.F.-G.)
- Department of Biomedical Engineering and Institute of Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5612AZ Eindhoven, The Netherlands
- Correspondence: (L.A.); (J.S.); (D.C.)
| | - Josep Samitier
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 15–21, 08028 Barcelona, Spain; (M.L.); (R.P.); (N.F.-G.)
- Department of Electronics and Biomedical Engineering, University of Barcelona, C. Martí i Franqués 1, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
- Correspondence: (L.A.); (J.S.); (D.C.)
| | - David Caballero
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 15–21, 08028 Barcelona, Spain; (M.L.); (R.P.); (N.F.-G.)
- Department of Electronics and Biomedical Engineering, University of Barcelona, C. Martí i Franqués 1, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
- Correspondence: (L.A.); (J.S.); (D.C.)
| |
Collapse
|
39
|
Wasson EM, Dubbin K, Moya ML. Go with the flow: modeling unique biological flows in engineered in vitro platforms. LAB ON A CHIP 2021; 21:2095-2120. [PMID: 34008661 DOI: 10.1039/d1lc00014d] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Interest in recapitulating in vivo phenomena in vitro using organ-on-a-chip technology has grown rapidly and with it, attention to the types of fluid flow experienced in the body has followed suit. These platforms offer distinct advantages over in vivo models with regards to human relevance, cost, and control of inputs (e.g., controlled manipulation of biomechanical cues from fluid perfusion). Given the critical role biophysical forces play in several tissues and organs, it is therefore imperative that engineered in vitro platforms capture the complex, unique flow profiles experienced in the body that are intimately tied with organ function. In this review, we outline the complex and unique flow regimes experienced by three different organ systems: blood vasculature, lymphatic vasculature, and the intestinal system. We highlight current state-of-the-art platforms that strive to replicate physiological flows within engineered tissues while introducing potential limitations in current approaches.
Collapse
Affiliation(s)
- Elisa M Wasson
- Material Engineering Division, Lawrence Livermore National Laboratory, 7000 East Ave L-222, Livermore, CA 94551, USA.
| | - Karen Dubbin
- Material Engineering Division, Lawrence Livermore National Laboratory, 7000 East Ave L-222, Livermore, CA 94551, USA.
| | - Monica L Moya
- Material Engineering Division, Lawrence Livermore National Laboratory, 7000 East Ave L-222, Livermore, CA 94551, USA.
| |
Collapse
|
40
|
Azizgolshani H, Coppeta JR, Vedula EM, Marr EE, Cain BP, Luu RJ, Lech MP, Kann SH, Mulhern TJ, Tandon V, Tan K, Haroutunian NJ, Keegan P, Rogers M, Gard AL, Baldwin KB, de Souza JC, Hoefler BC, Bale SS, Kratchman LB, Zorn A, Patterson A, Kim ES, Petrie TA, Wiellette EL, Williams C, Isenberg BC, Charest JL. High-throughput organ-on-chip platform with integrated programmable fluid flow and real-time sensing for complex tissue models in drug development workflows. LAB ON A CHIP 2021; 21:1454-1474. [PMID: 33881130 DOI: 10.1039/d1lc00067e] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Drug development suffers from a lack of predictive and human-relevant in vitro models. Organ-on-chip (OOC) technology provides advanced culture capabilities to generate physiologically appropriate, human-based tissue in vitro, therefore providing a route to a predictive in vitro model. However, OOC technologies are often created at the expense of throughput, industry-standard form factors, and compatibility with state-of-the-art data collection tools. Here we present an OOC platform with advanced culture capabilities supporting a variety of human tissue models including liver, vascular, gastrointestinal, and kidney. The platform has 96 devices per industry standard plate and compatibility with contemporary high-throughput data collection tools. Specifically, we demonstrate programmable flow control over two physiologically relevant flow regimes: perfusion flow that enhances hepatic tissue function and high-shear stress flow that aligns endothelial monolayers. In addition, we integrate electrical sensors, demonstrating quantification of barrier function of primary gut colon tissue in real-time. We utilize optical access to the tissues to directly quantify renal active transport and oxygen consumption via integrated oxygen sensors. Finally, we leverage the compatibility and throughput of the platform to screen all 96 devices using high content screening (HCS) and evaluate gene expression using RNA sequencing (RNA-seq). By combining these capabilities in one platform, physiologically-relevant tissues can be generated and measured, accelerating optimization of an in vitro model, and ultimately increasing predictive accuracy of in vitro drug screening.
Collapse
Affiliation(s)
- H Azizgolshani
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - J R Coppeta
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - E M Vedula
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - E E Marr
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - B P Cain
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - R J Luu
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - M P Lech
- Pfizer, Inc., 1 Portland Street, Cambridge, MA 02139, USA
| | - S H Kann
- Draper, 555 Technology Square, Cambridge, MA 02139, USA. and Department of Mechanical Engineering, Boston University, 110 Cummington Mall, Boston, MA 02215, USA
| | - T J Mulhern
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - V Tandon
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - K Tan
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | | | - P Keegan
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - M Rogers
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - A L Gard
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - K B Baldwin
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - J C de Souza
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - B C Hoefler
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - S S Bale
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - L B Kratchman
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - A Zorn
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - A Patterson
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - E S Kim
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - T A Petrie
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - E L Wiellette
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - C Williams
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - B C Isenberg
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| | - J L Charest
- Draper, 555 Technology Square, Cambridge, MA 02139, USA.
| |
Collapse
|
41
|
Lim J, Ching H, Yoon JK, Jeon NL, Kim Y. Microvascularized tumor organoids-on-chips: advancing preclinical drug screening with pathophysiological relevance. NANO CONVERGENCE 2021; 8:12. [PMID: 33846849 PMCID: PMC8042002 DOI: 10.1186/s40580-021-00261-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/17/2021] [Indexed: 05/06/2023]
Abstract
Recent developments of organoids engineering and organ-on-a-chip microfluidic technologies have enabled the recapitulation of the major functions and architectures of microscale human tissue, including tumor pathophysiology. Nevertheless, there remain challenges in recapitulating the complexity and heterogeneity of tumor microenvironment. The integration of these engineering technologies suggests a potential strategy to overcome the limitations in reconstituting the perfusable microvascular system of large-scale tumors conserving their key functional features. Here, we review the recent progress of in vitro tumor-on-a-chip microfluidic technologies, focusing on the reconstruction of microvascularized organoid models to suggest a better platform for personalized cancer medicine.
Collapse
Affiliation(s)
- Jungeun Lim
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- George W, Woodruff School of Mechanical Engineering, Georgia Institute of Technology, North Ave NW, Atlanta, GA, 30332, USA
| | - Hanna Ching
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jeong-Kee Yoon
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Noo Li Jeon
- George W, Woodruff School of Mechanical Engineering, Georgia Institute of Technology, North Ave NW, Atlanta, GA, 30332, USA
- Institute of Advanced Machinery and Design, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - YongTae Kim
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, 08826, Republic of Korea.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| |
Collapse
|
42
|
Bioengineered in vitro models of leukocyte-vascular interactions. Biochem Soc Trans 2021; 49:693-704. [PMID: 33843967 DOI: 10.1042/bst20200620] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 03/12/2021] [Accepted: 03/18/2021] [Indexed: 01/13/2023]
Abstract
Leukocytes continuously circulate our body through the blood and lymphatic vessels. To survey invaders or abnormalities and defend our body against them, blood-circulating leukocytes migrate from the blood vessels into the interstitial tissue space (leukocyte extravasation) and exit the interstitial tissue space through draining lymphatic vessels (leukocyte intravasation). In the process of leukocyte trafficking, leukocytes recognize and respond to multiple biophysical and biochemical cues in these vascular microenvironments to determine adequate migration and adhesion pathways. As leukocyte trafficking is an essential part of the immune system and is involved in numerous immune diseases and related immunotherapies, researchers have attempted to identify the key biophysical and biochemical factors that might be responsible for leukocyte migration, adhesion, and trafficking. Although intravital live imaging of in vivo animal models has been remarkably advanced and utilized, bioengineered in vitro models that recapitulate complicated in vivo vascular structure and microenvironments are needed to better understand leukocyte trafficking since these in vitro models better allow for spatiotemporal analyses of leukocyte behaviors, decoupling of interdependent biological factors, better controlling of experimental parameters, reproducible experiments, and quantitative cellular analyses. This review discusses bioengineered in vitro model systems that are developed to study leukocyte interactions with complex microenvironments of blood and lymphatic vessels. This review focuses on the emerging concepts and methods in generating relevant biophysical and biochemical cues. Finally, the review concludes with expert perspectives on the future research directions for investigating leukocyte and vascular biology using the in vitro models.
Collapse
|
43
|
Wu Y, Fortunato GM, Okesola BO, Brocchetti FLPD, Suntornnond R, Connelly J, De Maria C, Rodriguez-Cabello JC, Vozzi G, Wang W, Mata A. An interfacial self-assembling bioink for the manufacturing of capillary-like structures with tuneable and anisotropic permeability. Biofabrication 2021; 13. [PMID: 33561850 DOI: 10.1088/1758-5090/abe4c3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/09/2021] [Indexed: 12/28/2022]
Abstract
Self-assembling bioinks offer the possibility to biofabricate with molecular precision, hierarchical control, and biofunctionality. For this to become a reality with widespread impact, it is essential to engineer these ink systems ensuring reproducibility and providing suitable standardization. We have reported a self-assembling bioink based on disorder-to-order transitions of an elastin-like recombinamer (ELR) to co-assemble with graphene oxide (GO). Here, we establish reproducible processes, optimize printing parameters for its use as a bioink, describe new advantages that the self-assembling bioink can provide, and demonstrate how to fabricate novel structures with physiological relevance. We fabricate capillary-like structures with resolutions down to ∼10µm in diameter and ∼2µm thick tube walls and use both experimental and finite element analysis to characterize the printing conditions, underlying interfacial diffusion-reaction mechanism of assembly, printing fidelity, and material porosity and permeability. We demonstrate the capacity to modulate the pore size and tune the permeability of the resulting structures with and without human umbilical vascular endothelial cells. Finally, the potential of the ELR-GO bioink to enable supramolecular fabrication of biomimetic structures was demonstrated by printing tubes exhibiting walls with progressively different structure and permeability.
Collapse
Affiliation(s)
- Yuanhao Wu
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, United Kingdom.,Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom.,Institute of Bioengineering, Queen Mary University of London, London E1 4NS, United Kingdom.,School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, United Kingdom
| | - Gabriele Maria Fortunato
- Research Center 'E. Piaggio' and Dipartimento di Ingegneria dell'Informazione, University of Pisa, Largo Lucio Lazzarino, Pisa 1-56122, Italy
| | - Babatunde O Okesola
- Institute of Bioengineering, Queen Mary University of London, London E1 4NS, United Kingdom.,School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, United Kingdom
| | | | - Ratima Suntornnond
- CREATE LAB, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, United Kingdom
| | - John Connelly
- CREATE LAB, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, United Kingdom
| | - Carmelo De Maria
- Research Center 'E. Piaggio' and Dipartimento di Ingegneria dell'Informazione, University of Pisa, Largo Lucio Lazzarino, Pisa 1-56122, Italy
| | | | - Giovanni Vozzi
- Research Center 'E. Piaggio' and Dipartimento di Ingegneria dell'Informazione, University of Pisa, Largo Lucio Lazzarino, Pisa 1-56122, Italy
| | - Wen Wang
- Institute of Bioengineering, Queen Mary University of London, London E1 4NS, United Kingdom.,School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, United Kingdom
| | - Alvaro Mata
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, United Kingdom.,Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom.,Institute of Bioengineering, Queen Mary University of London, London E1 4NS, United Kingdom.,School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, United Kingdom.,Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| |
Collapse
|
44
|
Filelfi SL, Onorato A, Brix B, Goswami N. Lymphatic Senescence: Current Updates and Perspectives. BIOLOGY 2021; 10:biology10040293. [PMID: 33916784 PMCID: PMC8066652 DOI: 10.3390/biology10040293] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/23/2021] [Accepted: 03/29/2021] [Indexed: 12/14/2022]
Abstract
Simple Summary The lymphatic system is involved in tissue homeostasis, immune processes as well as transport of lipids, proteins and pathogens. Aging affects all physiological systems. However, it is not well studied how aging affects the lymphatic vasculature. Therefore, this review aims at investigating how senescence could lead to changes in the structure and function of the lymphatic vessels. We report that lymphatic senescence is associated with alterations in lymphatic muscles and nerve fibers, lymphatic endothelial cells membrane dysfunction, as well as changes in lymphatic pump, acute inflammation responses and immune function. Abstract Lymphatic flow is necessary for maintenance of vital physiological functions in humans and animals. To carry out optimal lymphatic flow, adequate contractile activity of the lymphatic collectors is necessary. Like in all body systems, aging has also an effect on the lymphatic system. However, limited knowledge is available on how aging directly affects the lymphatic system anatomy, physiology and function. We investigated how senescence leads to alterations in morphology and function of the lymphatic vessels. We used the strategy of a review to summarize the scientific literature of studies that have been published in the area of lymphatic senescence. Searches were carried out on PubMed and Web of Science using predefined search queries. We obtained an initial set of 1060 publications. They were filtered to 114 publications based on strict inclusion and exclusion criteria. Finally, the most appropriate 57 studies that specifically addressed lymphatic senescence have been selected for the preparation of this review. Analysis of the literature showed that lymphatic senescence is associated with alterations in lymphatic muscles and nerve fibers, lymphatic glycocalyx function of lymphatic endothelial cells, effects of chronic ultraviolet light exposure and oxidative stress as well as changes in lymphatic pump, acute inflammation responses and immune function. The current review underscores the relevance of the understudied area of lymphatic senescence. Continued research on the impact of aging on the structure and function of the lymphatic vasculature is needed to provide further insights to develop innovative clinical diagnostic—and treatment—modalities as well as to reduce the morbidity associated with diseases related to the lymphatic system.
Collapse
Affiliation(s)
- Sebastian Lucio Filelfi
- Physiology Division, Otto Loewi Research Center, Medical University of Graz, 8036 Graz, Austria; (S.L.F.); (B.B.)
| | - Alberto Onorato
- Oncology Reference Centre, Institute of Hospitalization and Care with Scientific Characterization, 33081 Aviano, Italy;
| | - Bianca Brix
- Physiology Division, Otto Loewi Research Center, Medical University of Graz, 8036 Graz, Austria; (S.L.F.); (B.B.)
| | - Nandu Goswami
- Physiology Division, Otto Loewi Research Center, Medical University of Graz, 8036 Graz, Austria; (S.L.F.); (B.B.)
- Department of Health Sciences, Alma Mater Europeae Maribor, 2000 Maribor, Slovenia
- Correspondence: ; Tel.: +43-3857-3852
| |
Collapse
|
45
|
Lee S, Kang H, Park D, Yu J, Koh SK, Cho D, Kim D, Kang K, Jeon NL. Modeling 3D Human Tumor Lymphatic Vessel Network Using High‐Throughput Platform. Adv Biol (Weinh) 2021. [DOI: 10.1002/adbi.202000195] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Somin Lee
- Interdisciplinary Program for Bioengineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - Habin Kang
- Interdisciplinary Program for Bioengineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - Dohyun Park
- Department of Mechanical Engineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - James Yu
- Interdisciplinary Program for Bioengineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - Seung Kwon Koh
- Department of Health Sciences and Technology SAIHST Sungkyunkwan University 115, Irwon‐ro, Gangnam‐gu Seoul 06355 Republic of Korea
| | - Duck Cho
- Department of Health Sciences and Technology SAIHST Sungkyunkwan University 115, Irwon‐ro, Gangnam‐gu Seoul 06355 Republic of Korea
- Department of Laboratory Medicine and Genetics Samsung Medical Center Sungkyunkwan University School of Medicine 115, Irwon‐ro, Gangnam‐gu Seoul 06355 Republic of Korea
| | - Da‐Hyun Kim
- Adult Stem Cell Research Center and Research Institute for Veterinary Science College of Veterinary Medicine Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - Kyung‐Sun Kang
- Adult Stem Cell Research Center and Research Institute for Veterinary Science College of Veterinary Medicine Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - Noo Li Jeon
- Interdisciplinary Program for Bioengineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
- Department of Mechanical Engineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
- Institute of Advanced Machinery and Design Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
- Institute of BioEngineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| |
Collapse
|
46
|
Moses SR, Adorno JJ, Palmer AF, Song JW. Vessel-on-a-chip models for studying microvascular physiology, transport, and function in vitro. Am J Physiol Cell Physiol 2021; 320:C92-C105. [PMID: 33176110 PMCID: PMC7846973 DOI: 10.1152/ajpcell.00355.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/20/2020] [Accepted: 11/08/2020] [Indexed: 12/15/2022]
Abstract
To understand how the microvasculature grows and remodels, researchers require reproducible systems that emulate the function of living tissue. Innovative contributions toward fulfilling this important need have been made by engineered microvessels assembled in vitro with microfabrication techniques. Microfabricated vessels, commonly referred to as "vessels-on-a-chip," are from a class of cell culture technologies that uniquely integrate microscale flow phenomena, tissue-level biomolecular transport, cell-cell interactions, and proper three-dimensional (3-D) extracellular matrix environments under well-defined culture conditions. Here, we discuss the enabling attributes of microfabricated vessels that make these models more physiological compared with established cell culture techniques and the potential of these models for advancing microvascular research. This review highlights the key features of microvascular transport and physiology, critically discusses the strengths and limitations of different microfabrication strategies for studying the microvasculature, and provides a perspective on current challenges and future opportunities for vessel-on-a-chip models.
Collapse
Affiliation(s)
- Savannah R Moses
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Jonathan J Adorno
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
| | - Andre F Palmer
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Jonathan W Song
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, Ohio
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
47
|
Lee CS, Leong KW. Advances in microphysiological blood-brain barrier (BBB) models towards drug delivery. Curr Opin Biotechnol 2020; 66:78-87. [PMID: 32711361 PMCID: PMC7744339 DOI: 10.1016/j.copbio.2020.06.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 12/22/2022]
Abstract
Though the blood-brain barrier (BBB) is vital for the maintenance of brain homeostasis, it also accounts for a high attrition rate of therapies targeting the central nervous system (CNS). The challenge of delivery across the BBB is attributed to a combination of low permeability through an endothelium closely knit by tight and adherens junctions, extremely low rates of endothelial transcytosis, and efflux transporters. In the past decade, enormous research efforts have been spent to develop BBB penetration strategies using biochemical or physical stimuli, aided by BBB-on-chips or microphysiological BBB models to facilitate in vitro studies. Here, we discuss recent advances in BBB-chip technology that have enabled effective preclinical screenings of brain targeting therapeutics and external stimulation, such as sonoporation and electroporation, for improved BBB penetration.
Collapse
Affiliation(s)
- Caleb S Lee
- Columbia University, NY, NY, 10027, United States
| | - Kam W Leong
- Columbia University, NY, NY, 10027, United States.
| |
Collapse
|
48
|
Maharjan S, Cecen B, Zhang YS. 3D Immunocompetent Organ-on-a-Chip Models. SMALL METHODS 2020; 4:2000235. [PMID: 33072861 PMCID: PMC7567338 DOI: 10.1002/smtd.202000235] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Indexed: 05/15/2023]
Abstract
In recent years, engineering of various human tissues in microphysiologically relevant platforms, known as organs-on-chips (OOCs), has been explored to establish in vitro tissue models that recapitulate the microenvironments found in native organs and tissues. However, most of these models have overlooked the important roles of immune cells in maintaining tissue homeostasis under physiological conditions and in modulating the tissue microenvironments during pathophysiology. Significantly, gradual progress is being made in the development of more sophisticated microphysiologically relevant human-based OOC models that allow the studies of the key biophysiological aspects of specific tissues or organs, interactions between cells (parenchymal, vascular, and immune cells) and their extracellular matrix molecules, effects of native tissue architectures (geometry, dynamic flow or mechanical forces) on tissue functions, as well as unravelling the mechanism underlying tissue-specific diseases and drug testing. In this Progress Report, we discuss the different components of the immune system, as well as immune OOC platforms and immunocompetent OOC approaches that have simulated one or more components of the immune system. We also outline the challenges to recreate a fully functional tissue system in vitro with a focus on the incorporation of the immune system.
Collapse
Affiliation(s)
- Sushila Maharjan
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Berivan Cecen
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| |
Collapse
|
49
|
Fathi P, Holland G, Pan D, Esch MB. Lymphatic Vessel on a Chip with Capability for Exposure to Cyclic Fluidic Flow. ACS APPLIED BIO MATERIALS 2020; 3:6697-6707. [DOI: 10.1021/acsabm.0c00609] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Parinaz Fathi
- Biomedical Technologies Group, Microsystems and Nanotechnology Division, Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
- Departments of Bioengineering, Materials Science and Engineering, and Beckman Institute, University of Illinois at Urbana-Champaign, Champaign, Illinois 61801, United States
- Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, Illinois 61801, United States
| | - Glenn Holland
- Photonics and Plasmonics Group, Microsystems and Nanotechnology Division, Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Dipanjan Pan
- Departments of Bioengineering, Materials Science and Engineering, and Beckman Institute, University of Illinois at Urbana-Champaign, Champaign, Illinois 61801, United States
- Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, Illinois 61801, United States
| | - Mandy B. Esch
- Biomedical Technologies Group, Microsystems and Nanotechnology Division, Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| |
Collapse
|
50
|
Virumbrales-Muñoz M, Ayuso JM, Gong MM, Humayun M, Livingston MK, Lugo-Cintrón KM, McMinn P, Álvarez-García YR, Beebe DJ. Microfluidic lumen-based systems for advancing tubular organ modeling. Chem Soc Rev 2020; 49:6402-6442. [PMID: 32760967 PMCID: PMC7521761 DOI: 10.1039/d0cs00705f] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Microfluidic lumen-based systems are microscale models that recapitulate the anatomy and physiology of tubular organs. These technologies can mimic human pathophysiology and predict drug response, having profound implications for drug discovery and development. Herein, we review progress in the development of microfluidic lumen-based models from the 2000s to the present. The core of the review discusses models for mimicking blood vessels, the respiratory tract, the gastrointestinal tract, renal tubules, and liver sinusoids, and their application to modeling organ-specific diseases. We also highlight emerging application areas, such as the lymphatic system, and close the review discussing potential future directions.
Collapse
Affiliation(s)
- María Virumbrales-Muñoz
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - José M Ayuso
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA and Morgridge Institute for Research, Madison, WI, USA
| | - Max M Gong
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA and Department of Biomedical Engineering, Trine University, Angola, IN, USA
| | - Mouhita Humayun
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Megan K Livingston
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Karina M Lugo-Cintrón
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Patrick McMinn
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Yasmín R Álvarez-García
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA and Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| |
Collapse
|