1
|
Salvadori A, Watanabe M, Markovic M, Sudo R, Ovsianikov A. Controlled microvasculature for organ-on-a-chip applications produced by high-definition laser patterning. Biofabrication 2025; 17:035011. [PMID: 40315895 DOI: 10.1088/1758-5090/add37e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 05/02/2025] [Indexed: 05/04/2025]
Abstract
Organs-on-Chips (OoCs) are 3D models aiming to faithfully replicatein vitrospecific functions of human organs or tissues. While promising as an alternative to traditional 2D cell culture and animal models in drug development, controlled realization of complex microvasculature within OoC remains a significant challenge. Here, we demonstrate how femtosecond laser patterning allows to produce hollow microvascular-like channels inside a collagen-based matrix directly within a microfluidic chip. The hydrogel preparation protocol was optimized to maintain structural stability, facilitating successful endothelialization of produced channels. The resulting microvascular structures exhibit notable physiological relevance, as evidenced by the expression of key endothelial markers (ZO-1, and VE-cadherin) and the successful reproduction of the barrier function. Furthermore, tumor necrosis factor-alpha (TNF-α) exposure induces a concentration-dependent increase in vascular permeability and expression of intercellular adhesion molecule-1 (ICAM-1). The proposed method holds the potential to control and faithfully reproduce the vascularization process in OoC platforms, in both physiological and inflammatory conditions.
Collapse
Affiliation(s)
- Alice Salvadori
- Research Group 3D Printing and Biofabrication, Institute of Material Science and Technology, Technische Universität Wien (TU Wien), Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Masafumi Watanabe
- Research Group 3D Printing and Biofabrication, Institute of Material Science and Technology, Technische Universität Wien (TU Wien), Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Japan Society for the Promotion of Science (JSPS) Overseas Research Fellow, Japan
| | - Marica Markovic
- Research Group 3D Printing and Biofabrication, Institute of Material Science and Technology, Technische Universität Wien (TU Wien), Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Ryo Sudo
- Department of System Design Engineering, Keio University, Yokohama, Japan
| | - Aleksandr Ovsianikov
- Research Group 3D Printing and Biofabrication, Institute of Material Science and Technology, Technische Universität Wien (TU Wien), Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
2
|
Sasaki N. Microfluidic devices integrated with track-etched porous membranes for cell-based bioanalysis. ANAL SCI 2025:10.1007/s44211-025-00789-7. [PMID: 40346437 DOI: 10.1007/s44211-025-00789-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 04/29/2025] [Indexed: 05/11/2025]
Abstract
Microfluidic devices integrated with track-etched porous membranes are useful for cell-based bioanalysis. In this mini review, our latest achievements based on membrane-integrated microfluidic devices are reviewed. The membrane-integrated microfluidic devices in parallel configuration have been employed to develop a microfluidic model of microcirculation and for the development of nanomedicines. Co-culture of endothelial cells with tumor cells and parallel permeation assays through keratinocyte cell layer have also been demonstrated. Membrane-integrated microfluidic devices in perpendicular configuration have been employed to simulate extravasation of tumor cells and to evaluate the efficacy of anti-inflammatory drug on the function of keratinocyte cell layer formed on the membrane. These microfluidic devices have unique features and a lot of advantages, so we hope that they are utilized as a powerful tool for cell-based bioanalysis.
Collapse
Affiliation(s)
- Naoki Sasaki
- Department of Chemistry, College of Science, Rikkyo University, 3-34-1 Nishi-Ikebukuro, Toshima, Tokyo, 171-8501, Japan.
| |
Collapse
|
3
|
Staples SCR, Yin H, Sutherland FSK, Prescott EK, Tinney D, Hamilton DW, Goldman D, Poepping TL, Ellis CG, Pickering JG. Intussusceptive angiogenesis-on-a-chip: Evidence for transluminal vascular bridging by endothelial delamination. Proc Natl Acad Sci U S A 2025; 122:e2423700122. [PMID: 40244661 PMCID: PMC12036988 DOI: 10.1073/pnas.2423700122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Intussusceptive angiogenesis is an increasingly recognized vessel duplication process that generates and reshapes microvascular beds. However, the mechanism by which a vessel splits into two is poorly understood. Particularly vexing is formation of the hallmark transluminal endothelial cell bridge. How an endothelial cell comes to cross a flowing lumen rather than line it is enigmatic. To elucidate this, we used a microvessel-on-a-chip strategy, creating a microconduit coherently lined with flow-sensitive endothelial cells but in which transluminal bridges also formed. Bridge morphologies ranged from filamentous strand to multicellular columns with a central extracellular matrix-containing core. These bridge architectures were found to recapitulate those in microvessels in embryos, tumors, diseased organs, and the dermis of patients with limb-threatening ischemia. Time-lapse, multiplane, three-dimensional (3D) microscopy of the microphysiologic conduit revealed that bridges arose from endothelial cells oriented orthogonal to flow that partially released from the wall while retaining attachments at the ends. This delamination process was blocked by hyperactivation of Rho and augmented by interventions that weaken cell-substrate interactions, including inhibiting nonmuscle myosin II and blocking α5ß1 integrin. Thus, endothelial cells can leave their monolayer and transect a flowing lumen through controlled delamination. This previously unrecognized lumen entry program could explain the launch of intussusceptive angiogenesis and opens a framework for intervening.
Collapse
Affiliation(s)
- Sabrina C. R. Staples
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - Hao Yin
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - Frances S. K. Sutherland
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - Emma K. Prescott
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - Dylan Tinney
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - Douglas W. Hamilton
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - Daniel Goldman
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - Tamie L. Poepping
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Physics and Astronomy, Western University, London, ONN6A 3K7, Canada
| | - Christopher G. Ellis
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - J. Geoffrey Pickering
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- London Health Sciences Centre, London, ONN6A 5A5, Canada
| |
Collapse
|
4
|
Xu X, Qiu Y, Chen CY, Carton M, Campbell PMR, Chowdhury AM, Bandyopadhyay BC, Bentley WE, Smith BR, Sochol RD. 3D nanoprinting of PDMS microvessels with tailored tortuosity and microporosity via direct laser writing. LAB ON A CHIP 2025; 25:1947-1958. [PMID: 40104860 PMCID: PMC11921864 DOI: 10.1039/d4lc01051e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/01/2025] [Indexed: 03/20/2025]
Abstract
Microvessels (e.g., capillaries) are ubiquitous throughout human anatomy, yet recreating their three-dimensional (3D) microfluidic and architectural sophistication at biologically accurate length scales has remained a critical challenge. To overcome this barrier, here we report a hybrid additive manufacturing-or "3D printing"-strategy in which "Two-Photon Direct Laser Writing (DLW)" is used to nanoprint microvessels of arbitrary design directly atop "Liquid-Crystal Display (LCD)" 3D-printed microfluidic chips. Fabrication results indicated effective production of 100 μm-diameter 3D polydimethylsiloxane (PDMS) microfluidic vessels with 5 μm-thick walls-featuring arrays of pre-designed 5 μm-diameter micropores-as well as three discrete spiralled, intertwined microvessels. Experimental results with MDA-MB-231 epithelial breast cancer cells revealed the ability for the 3D PDMS microvessels to support cell culture. In combination, these results suggest that the presented strategy for 3D nanoprinting PDMS microvessels with custom-designed architectures and microporosity offers a promising pathway to enable new classes of "organ-on-a-chip (OOC)" systems for wide-ranging biomedical applications.
Collapse
Affiliation(s)
- Xin Xu
- Department of Mechanical Engineering, University of Maryland, College Park, MD, 20742, USA.
| | - Yunxiu Qiu
- Institute for Quantitative Health Science and Engineering, Department of Chemical Engineering and Material Science, Michigan State University, East Lan-sing, MI, 48824, USA
| | - Chen-Yu Chen
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, 20742, USA
| | - Molly Carton
- Department of Mechanical Engineering, University of Maryland, College Park, MD, 20742, USA.
| | - Paige M R Campbell
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - A Muhaymin Chowdhury
- Department of Mechanical Engineering, University of Maryland, College Park, MD, 20742, USA.
| | | | - William E Bentley
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, 20742, USA
| | - Bryan Ronain Smith
- Institute for Quantitative Health Science and Engineering, Department of Chemical Engineering and Material Science, Michigan State University, East Lan-sing, MI, 48824, USA
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Ryan D Sochol
- Department of Mechanical Engineering, University of Maryland, College Park, MD, 20742, USA.
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, 20742, USA
- Maryland Robotics Center, University of Maryland, College Park, MD, 20742, USA
- Institute for Systems Research, University of Maryland, College Park, MD, 20742, USA
| |
Collapse
|
5
|
Li Y, Fu BM. A Cost-Effective and Easy to Assemble 3D Human Microchannel Blood-Brain Barrier Model and Its Application in Tumor Cell Adhesion Under Flow. Cells 2025; 14:456. [PMID: 40136705 PMCID: PMC11941619 DOI: 10.3390/cells14060456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
By utilizing polydimethylsiloxane (PDMS), collagen hydrogel, and a cell line for human cerebral microvascular endothelial cells, we produced a 3D microchannel blood-brain barrier (BBB) model under physiological flow. This 3D BBB has a circular-shaped cross-section and a diameter of ~100 μm, which can properly mimic the cerebral microvessel responsible for material exchange between the circulating blood and brain tissue. The permeability of the 3D microchannel BBB to a small molecule (sodium fluorescein with a molecular weight of 376) and that to a large molecule (Dex-70k) are the same as those of rat cerebral microvessels. This 3D BBB model can replicate the effects of a plasma protein, orosomucoid, a cytokine, vascular endothelial growth factor (VEGF), and an enzyme, heparinase III, on either rat cerebral or mesenteric microvessesels in terms of permeability and the modulation of glycocalyx (heparan sulfate). It can also replicate the adhesion of a breast cancer cell, MDA-MB-231, in rat mesenteric microvessels under no treatment or treatments with VEGF, orosomucoid, and heparinase III. Because of difficulties in accessing human cerebral microvessels, this inexpensive and easy to assemble 3D human BBB model can be applied to investigate BBB-modulating mechanisms in health and in disease and to develop therapeutic interventions targeting tumor metastasis to the brain.
Collapse
Affiliation(s)
| | - Bingmei M. Fu
- Department of Biomedical Engineering, The City College of the City University of New York, New York, NY 10031, USA;
| |
Collapse
|
6
|
Aye SSS, Fang Z, Wu MCL, Lim KS, Ju LA. Integrating microfluidics, hydrogels, and 3D bioprinting for personalized vessel-on-a-chip platforms. Biomater Sci 2025; 13:1131-1160. [PMID: 39834160 DOI: 10.1039/d4bm01354a] [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: 01/22/2025]
Abstract
Thrombosis, a major cause of morbidity and mortality worldwide, presents a complex challenge in cardiovascular medicine due to the intricacy of clotting mechanisms in living organisms. Traditional research approaches, including clinical studies and animal models, often yield conflicting results due to the inability to control variables in these complex systems, highlighting the need for more precise investigative tools. This review explores the evolution of in vitro thrombosis models, from conventional polydimethylsiloxane (PDMS)-based microfluidic devices to advanced hydrogel-based systems and cutting-edge 3D bioprinted vascular constructs. We discuss how these emerging technologies, particularly vessel-on-a-chip platforms, are enabling researchers to control previously unmanageable factors, thereby offering unprecedented opportunities to pinpoint specific clotting mechanisms. While PDMS-based devices offer optical transparency and fabrication ease, their inherent limitations, including non-physiological rigidity and surface properties, have driven the development of hydrogel-based systems that better mimic the extracellular matrix of blood vessels. The integration of microfluidics with biomimetic materials and tissue engineering approaches has led to the development of sophisticated models capable of simulating patient-specific vascular geometries, flow dynamics, and cellular interactions under highly controlled conditions. The advent of 3D bioprinting further enables the creation of complex, multi-layered vascular structures with precise spatial control over geometry and cellular composition. Despite significant progress, challenges remain in achieving long-term stability, incorporating immune components, and translating these models to clinical applications. By providing a comprehensive overview of current advancements and future prospects, this review aims to stimulate further innovation in thrombosis research and accelerate the development of more effective, personalized approaches to thrombosis prevention and treatment.
Collapse
Affiliation(s)
- San Seint Seint Aye
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia.
| | - Zhongqi Fang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia.
| | - Mike C L Wu
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia.
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia.
| | - Khoon S Lim
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia.
- School of Medical Sciences, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia.
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia.
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW 2006, Australia
- Heart Research Institute, Newtown, NSW 2042, Australia
| |
Collapse
|
7
|
Wu M, Sapin-Minet A, Stefan L, Perrin J, Raeth-Fries I, Gaucher C. Heparinized collagen-based hydrogels for tissue engineering: physical, mechanical and biological properties. Int J Pharm 2025; 670:125126. [PMID: 39719257 DOI: 10.1016/j.ijpharm.2024.125126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/20/2024] [Accepted: 12/21/2024] [Indexed: 12/26/2024]
Abstract
As the main protein forming the vascular extracellular matrix, collagen has a weak antigenicity, making it an attractive candidate for coatings of vascular grafts. In order to bring antithrombotic properties to collagen for obtaining suitable blood compatibility of surfaces and further bioactive molecule carrying capacity, heparinization appears as a method of choice. Thus, in this article, pH-driven self-assembly was used to form collagen-based hydrogels with physical incorporation of heparins, especially low molecular weight heparin or unfractionated heparin at 1 IU/mL and 6 IU/mL. These heparinized hydrogels were evaluated for their physicochemical properties including gelation kinetic, spreading, viscoelasticity, microstructure and heparin quantification, and their biocompatibility such as cytocompatibility and their capacity to release bioactive heparins with antithrombotic properties. The loading capacity of collagen-based hydrogels was higher for unfractionated heparin (60 to 80 %) than for low molecular weight heparin (20 %). Interestingly, the highest concentration (6 IU/mL) of heparin used to form collagen-based hydrogels resulted in the formation of a softer hydrogel owning a better spreadability compared to 1 IU/mL and non heparinized collagen-based hydrogel. The 3D structure observation showed a layered formation with visible pores or spaces between the layers in all types of collagen-based hydrogels. These layers are interconnected by fibrous structures, suggesting a networked architecture. Moreover, the amount of heparin released from collagen-based hydrogel prepared with 6 IU /mL was higher than from those heparinized with 1 IU/mL attested by a delay in blood coagulation (activated partial thromboplastin time and thrombin time) and the abolishment of thrombin generation. Those hydrogels were also biocompatible, with low albumin adsorption and no impact on cell viability. Finally, heparin is retained in these hydrogels for at least seven days after cell seeding, providing the possibility of long-term antithrombotic properties. Thus, we succeeded in develop/obtain coatings with antithrombogenic properties, biocompatible and able to retain heparins at least seven days, making them good candidates with a great potential for the development and application of efficient blood-contacting materials.
Collapse
Affiliation(s)
- Meiling Wu
- Université de Lorraine, CITHEFOR, F-54000 Nancy, France
| | | | - Loïc Stefan
- Université de Lorraine, CNRS, LCPM, F-54000 Nancy, France
| | - Julien Perrin
- Université de Lorraine, CHRU-Nancy, Service d'hématologie biologique, F-54000 Nancy, France; Université de Lorraine, INSERM, DCAC, F-54000 Nancy, France
| | | | | |
Collapse
|
8
|
Jorgensen C, Linville RM, Galea I, Lambden E, Vögele M, Chen C, Troendle EP, Ruggiu F, Ulmschneider MB, Schiøtt B, Lorenz CD. Permeability Benchmarking: Guidelines for Comparing in Silico, in Vitro, and in Vivo Measurements. J Chem Inf Model 2025; 65:1067-1084. [PMID: 39823383 PMCID: PMC11815851 DOI: 10.1021/acs.jcim.4c01815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 01/04/2025] [Accepted: 01/06/2025] [Indexed: 01/19/2025]
Abstract
Permeability is a measure of the degree to which cells can transport molecules across biological barriers. Units of permeability are distance per unit time (typically cm/s), where accurate measurements are needed to define drug delivery in homeostasis and to model dysfunction occurring during disease. This perspective offers a set of community-led guidelines to benchmark permeability data across multidisciplinary approaches and different biological contexts. First, we lay out the analytical framework for three methodologies to calculate permeability: in silico assays using either transition-based counting or the inhomogeneous-solubility diffusion approaches, in vitro permeability assays using cells cultured in 2D or 3D geometries, and in vivo assays utilizing in situ brain perfusion or multiple time-point regression analysis. Then, we demonstrate a systematic benchmarking of in silico to both in vitro and in vivo, depicting the ways in which each benchmarking is sensitive to the choices of assay design. Finally, we outline seven recommendations for best practices in permeability benchmarking and underscore the significance of tailored permeability assays in driving advancements in drug delivery research and development. Our exploration encompasses a discussion of "generic" and tissue-specific biological barriers, including the blood-brain barrier (BBB), which is a major hurdle for the delivery of therapeutic agents into the brain. By addressing challenges in reconciling simulated data with experimental assays, we aim to provide insights essential for optimizing accuracy and reliability in permeability modeling.
Collapse
Affiliation(s)
- Christian Jorgensen
- School
of Medicine, Pharmacy and Biomedical Sciences, Faculty of Science
& Health, University of Portsmouth, Portsmouth PO1 2DT, Hampshire, U.K.
- Dept.
of Chemistry, Aarhus University, Langelandsgade, 140 8000 Aarhus C, Denmark
| | - Raleigh M. Linville
- The
Picower Institute for Learning and Memory, Massachusetts Institute of Technology, 43 Vassar Street, Cambridge, Massachusetts 02139, United States
| | - Ian Galea
- Clinical
Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, U.K.
| | - Edward Lambden
- Dept.
of Chemistry, King’s College London, London WC2R 2LS, U.K.
| | - Martin Vögele
- Department
of Computer Science, Stanford University, Stanford, California 94305, United States
- Department
of Molecular and Cellular Physiology, Stanford
University, Stanford, California 94305, United States
- Institute
for Computational and Mathematical Engineering, Stanford University, Stanford, California 94305, United States
| | - Charles Chen
- Synthetic
Biology Group, Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Evan P. Troendle
- Wellcome−Wolfson
Institute for Experimental Medicine, School of Medicine, Dentistry
and Biomedical Sciences, Queen’s
University Belfast, Belfast, County
Antrim, BT9 7BL, Northern Ireland, U.K.
| | - Fiorella Ruggiu
- Kimia
Therapeutics, 740 Heinz
Avenue, Berkeley, California 94710, United States
| | | | - Birgit Schiøtt
- Dept.
of Chemistry, Aarhus University, Langelandsgade, 140 8000 Aarhus C, Denmark
| | | |
Collapse
|
9
|
Martinelli C, Bocconi A, Milone S, Baldissera T, Cherubin L, Buccioli G, Perottoni S, Conci C, Cerullo G, Osellame R, Chirico G, Jacchetti E, Raimondi MT. A 3D millifluidic model of a dermal perivascular microenvironment on a chip. LAB ON A CHIP 2025; 25:423-439. [PMID: 39757968 PMCID: PMC11701800 DOI: 10.1039/d4lc00898g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/16/2024] [Indexed: 01/07/2025]
Abstract
The process of angiogenesis plays a pivotal role in skin regeneration, ensuring the provision of nutrients and oxygen to the nascent tissue, thanks to the formation of novel microvascular networks supporting functional tissue regeneration. Unfortunately, most of the current therapeutic approaches for skin regeneration lack vascularization, required to promote effective angiogenesis. Thus, in vitro tridimensional models, complemented with specific biochemical signals, can be a valuable tool to unravel the neovascularization mechanisms and develop novel clinical strategies. In this work, we designed and validated a tridimensional microstructured dynamic model of the dermal perivascular microenvironment on a chip. We carried out the fabrication of an array of microstructures by two-photon laser polymerization, then used as a 3D substrate for co-culture of human dermal fibroblasts and endothelial cells. We included the substrate in a miniaturized optically accessible bioreactor (MOAB) which provides the physiological interstitial flow, upon perfusion in the presence or absence of the pro-angiogenic stimuli VEGF and TGF-β1. We determined the parameters to be applied under dynamic conditions by an in silico model simulating individual 3D microenvironments within the bioreactor's chambers. We computed the fluid velocity and wall shear stress acting on endothelial cells along with the oxygen concentration profile, and we chose the most suitable flow rate for maintaining dermal physiological conditions. Experimental results showed the effectiveness of the developed platform as a 3D dynamic model of angiogenesis. This is the first combined experimental and computational study involving chemically stimulated 3D co-cultures for successfully simulating the physiological dermal perivascular microenvironment.
Collapse
Affiliation(s)
- Chiara Martinelli
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza L. da Vinci, 32, 20133 Milan, Italy.
| | - Alberto Bocconi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza L. da Vinci, 32, 20133 Milan, Italy.
| | - Sofia Milone
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza L. da Vinci, 32, 20133 Milan, Italy.
| | - Teresa Baldissera
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza L. da Vinci, 32, 20133 Milan, Italy.
| | - Leonardo Cherubin
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza L. da Vinci, 32, 20133 Milan, Italy.
| | - Giovanni Buccioli
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza L. da Vinci, 32, 20133 Milan, Italy.
| | - Simone Perottoni
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza L. da Vinci, 32, 20133 Milan, Italy.
| | - Claudio Conci
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza L. da Vinci, 32, 20133 Milan, Italy.
| | - Giulio Cerullo
- Institute for Photonics and Nanotechnologies (IFN), CNR and Department of Physics, Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy.
| | - Roberto Osellame
- Institute for Photonics and Nanotechnologies (IFN), CNR and Department of Physics, Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy.
| | - Giuseppe Chirico
- Department of Physics, Università di Milano-Bicocca, Piazza della Scienza, 3, 20126 Milan, Italy.
| | - Emanuela Jacchetti
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza L. da Vinci, 32, 20133 Milan, Italy.
| | - Manuela Teresa Raimondi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza L. da Vinci, 32, 20133 Milan, Italy.
| |
Collapse
|
10
|
Malkani S, Prado O, Stevens KR. Sacrificial Templating for Accelerating Clinical Translation of Engineered Organs. ACS Biomater Sci Eng 2025; 11:1-12. [PMID: 39701582 PMCID: PMC11733865 DOI: 10.1021/acsbiomaterials.4c01824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024]
Abstract
Transplantable engineered organs could one day be used to treat patients suffering from end-stage organ failure. Yet, producing hierarchical vascular networks that sustain the viability and function of cells within human-scale organs remains a major challenge. Sacrificial templating has emerged as a promising biofabrication method that could overcome this challenge. Here, we explore and evaluate various strategies and materials that have been used for sacrificial templating. First, we emphasize fabrication approaches that use highly biocompatible sacrificial reagents and minimize the duration that cells spend in fabrication conditions without oxygen and nutrients. We then discuss strategies to create continuous, hierarchical vascular networks, both using biofabrication alone and using hybrid methods that integrate biologically driven vascular self-assembly into sacrificial templating workflows. Finally, we address the importance of structurally reinforcing engineered vessel walls to achieve stable blood flow in vivo, so that engineered organs remain perfused and functional long after implantation. Together, these sacrificial templating strategies have the potential to overcome many current limitations in biofabrication and accelerate clinical translation of transplantable, fully functional engineered organs to rescue patients from organ failure.
Collapse
Affiliation(s)
- Sherina Malkani
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
- Department
of Bioengineering, Rice University, Houston, Texas 77005, United States
- Institute
for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Olivia Prado
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
- Institute
for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Kelly R. Stevens
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
- Institute
for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98195, United States
- Department
of Laboratory Medicine and Pathology, University
of Washington, Seattle, Washington 98195, United States
- Brotman
Baty Institute for Precision Medicine, Seattle, Washington 98195, United States
| |
Collapse
|
11
|
Mainik P, Aponte‐Santamaría C, Fladung M, Curticean RE, Wacker I, Hofhaus G, Bastmeyer M, Schröder RR, Gräter F, Blasco E. Responsive 3D Printed Microstructures Based on Collagen Folding and Unfolding. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408597. [PMID: 39604251 PMCID: PMC11753499 DOI: 10.1002/smll.202408597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Indexed: 11/29/2024]
Abstract
Mimicking extracellular matrices holds great potential for tissue engineering in biological and biomedical applications. A key compound for the mechanical stability of these matrices is collagen, which also plays an important role in many intra- and intercellular processes. Two-photon 3D laser printing offers structuring of these matrices with subcellular resolution. So far, efforts on 3D microprinting of collagen have been limited to simple geometries and customized set-ups. Herein, an easily accessible approach is presented using a collagen type I methacrylamide (ColMA) ink system which can be stored at room temperature and be precisely printed using a commercial two-photon 3D laser printer. The formulation and printing parameters are carefully optimized enabling the manufacturing of defined 3D microstructures. Furthermore, these printed microstructures show a fully reversible response upon heating and cooling in multiple cycles, indicating successful collagen folding and unfolding. This experimental observation has been supported by molecular dynamics simulations. Thus, the study opens new perspectives for designing new responsive biomaterials for 4D (micro)printing.
Collapse
Affiliation(s)
- Philipp Mainik
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg University69120HeidelbergGermany
- Organic Chemistry Institute (OCI)Heidelberg University69120HeidelbergGermany
| | | | - Magdalena Fladung
- Cell and NeurobiologyZoological InstituteKarlsruhe Institute of Technology (KIT)76131KarlsruheGermany
| | | | - Irene Wacker
- BioQuantHeidelberg University69120HeidelbergGermany
| | - Götz Hofhaus
- BioQuantHeidelberg University69120HeidelbergGermany
| | - Martin Bastmeyer
- Cell and NeurobiologyZoological InstituteKarlsruhe Institute of Technology (KIT)76131KarlsruheGermany
- Institute for Biological and Chemical Systems – Biological Information Processing (IBCS‐BIP)Karlsruhe Institute of Technology (KIT)76344KarlsruheGermany
| | | | - Frauke Gräter
- Heidelberg Institute for Theoretical Studies (HITS)69118HeidelbergGermany
- Interdisciplinary Center for Scientific Computing (IWR)Heidelberg University69120HeidelbergGermany
| | - Eva Blasco
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg University69120HeidelbergGermany
- Organic Chemistry Institute (OCI)Heidelberg University69120HeidelbergGermany
| |
Collapse
|
12
|
Martins EAG, Deus IA, Gomes MC, Silva AS, Mano JF, Custódio CA. Human Chorionic Membrane-derived Tunable Hydrogels for Vascular Tissue Engineering Strategies. Adv Healthc Mater 2025; 14:e2401510. [PMID: 39101324 DOI: 10.1002/adhm.202401510] [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: 04/24/2024] [Revised: 07/17/2024] [Indexed: 08/06/2024]
Abstract
One of the foremost targets in the advancement of biomaterials to engineer vascularized tissues is not only to replicate the composition of the intended tissue but also to create thicker structures incorporating a vascular network for adequate nutrients and oxygen supply. For the first time, to the best of current knowledge, a clinically relevant biomaterial is developed, demonstrating that hydrogels made from the human decellularized extracellular matrix can exhibit robust mechanical properties (in the kPa range) and angiogenic capabilities simultaneously. These properties enable the culture and organization of human umbilical vein endothelial cells into tubular structures, maintaining their integrity for 14 days in vitro without the need for additional polymers or angiogenesis-related factors. This is achieved by repurposing the placenta chorionic membrane (CM), a medical waste with an exceptional biochemical composition, into a valuable resource for bioengineering purposes. After decellularization, the CM underwent chemical modification with methacryloyl groups, giving rise to methacrylated CM (CMMA). CMMA preserved key proteins, as well as glycosaminoglycans. The resulting hydrogels rapidly photopolymerize and have enhanced strength and customizable mechanical properties. Furthermore, they demonstrate angio-vasculogenic competence in vitro and in vivo, holding significant promise as a humanized platform for the engineering of vascularized tissues.
Collapse
Affiliation(s)
- Elisa A G Martins
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Inês A Deus
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Maria C Gomes
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Ana S Silva
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - João F Mano
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Catarina A Custódio
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| |
Collapse
|
13
|
Power L, Shuhmaher R, Houtz P, Chen J, Rudolph S, Yuen J, Machour M, Levy E, Wu L, Levenberg S, Whalen M, Chen Y, Kaplan DL. 3D Neurovascular Unit Tissue Model to Assess Responses to Traumatic Brain Injury. J Biomed Mater Res A 2025; 113:e37816. [PMID: 39440483 DOI: 10.1002/jbm.a.37816] [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: 07/09/2024] [Revised: 09/26/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
The neurovascular unit (NVU) is a critical interface in the central nervous system that links vascular interactions with glial and neural tissue. Disruption of the NVU has been linked to the onset and progression of neurodegenerative diseases. Despite its significance the NVU remains challenging to study in a physiologically relevant manner. Here, a 3D cell triculture model of the NVU is developed that incorporates human primary brain microvascular endothelial cells, astrocytes, and pericytes into a tissue system that can be sustained in vitro for several weeks. This tissue model helps recapitulate the complexity of the NVU and can be used to interrogate the mechanisms of disease and cell-cell interactions. The NVU tissue model displays elevated cell death and inflammatory responses following mechanical damage, to emulate traumatic brain injury (TBI) under controlled laboratory conditions, including lactate dehydrogenase (LDH) release, elevated inflammatory markers TNF-α and monocyte chemoattractant cytokines MCP-2 and MCP-3 and reduced expression of the tight junction marker ZO-1. This 3D tissue model serves as a tool for deciphering mechanisms of TBIs and immune responses associated with the NVU.
Collapse
Affiliation(s)
- Liam Power
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Rita Shuhmaher
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Philip Houtz
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Jinpeng Chen
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Sara Rudolph
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - John Yuen
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Majd Machour
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Emily Levy
- Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Limin Wu
- Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Shulamit Levenberg
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Michael Whalen
- Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ying Chen
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| |
Collapse
|
14
|
Chernokal B, Ferrick BJ, Gleghorn JP. Zonal patterning of extracellular matrix and stromal cell populations along a perfusable cellular microchannel. LAB ON A CHIP 2024; 24:5238-5250. [PMID: 39479925 PMCID: PMC11525951 DOI: 10.1039/d4lc00579a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/09/2024] [Indexed: 11/02/2024]
Abstract
The spatial organization of biophysical and biochemical cues in the extracellular matrix (ECM) in concert with reciprocal cell-cell signaling is vital to tissue patterning during development. However, elucidating the role an individual microenvironmental factor plays using existing in vivo models is difficult due to their inherent complexity. In this work, we have developed a microphysiological system to spatially pattern the biochemical, biophysical, and stromal cell composition of the ECM along an epithelialized 3D microchannel. This technique is adaptable to multiple hydrogel compositions and scalable to the number of zones patterned. We confirmed that the methodology to create distinct zones resulted in a continuous, annealed hydrogel with regional interfaces that did not hinder the transport of soluble molecules. Further, the interface between hydrogel regions did not disrupt microchannel structure, epithelial lumen formation, or media perfusion through an acellular or cellularized microchannel. Finally, we demonstrated spatially patterned tubulogenic sprouting of a continuous epithelial tube into the surrounding hydrogel confined to local regions with stromal cell populations, illustrating spatial control of cell-cell interactions and signaling gradients. This easy-to-use system has wide utility for modeling three-dimensional epithelial and endothelial tissue interactions with heterogeneous hydrogel compositions and/or stromal cell populations to investigate their mechanistic roles during development, homeostasis, or disease.
Collapse
Affiliation(s)
- Brea Chernokal
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19713, USA.
| | - Bryan J Ferrick
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19713, USA.
| | - Jason P Gleghorn
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19713, USA.
| |
Collapse
|
15
|
Iqbal MZ, Riaz M, Biedermann T, Klar AS. Breathing new life into tissue engineering: exploring cutting-edge vascularization strategies for skin substitutes. Angiogenesis 2024; 27:587-621. [PMID: 38842751 PMCID: PMC11564345 DOI: 10.1007/s10456-024-09928-6] [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: 07/20/2023] [Accepted: 05/02/2024] [Indexed: 06/07/2024]
Abstract
Tissue-engineered skin substitutes (TESS) emerged as a new therapeutic option to improve skin transplantation. However, establishing an adequate and rapid vascularization in TESS is a critical factor for their clinical application and successful engraftment in patients. Therefore, several methods have been applied to improve the vascularization of skin substitutes including (i) modifying the structural and physicochemical properties of dermal scaffolds; (ii) activating biological scaffolds with growth factor-releasing systems or gene vectors; and (iii) developing prevascularized skin substitutes by loading scaffolds with capillary-forming cells. This review provides a detailed overview of the most recent and important developments in the vascularization strategies for skin substitutes. On the one hand, we present cell-based approaches using stem cells, microvascular fragments, adipose tissue derived stromal vascular fraction, endothelial cells derived from blood and skin as well as other pro-angiogenic stimulation methods. On the other hand, we discuss how distinct 3D bioprinting techniques and microfluidics, miRNA manipulation, cell sheet engineering and photosynthetic scaffolds like GelMA, can enhance skin vascularization for clinical applications. Finally, we summarize and discuss the challenges and prospects of the currently available vascularization techniques that may serve as a steppingstone to a mainstream application of skin tissue engineering.
Collapse
Affiliation(s)
- M Zohaib Iqbal
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Mahrukh Riaz
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Agnes S Klar
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland.
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland.
- University of Zurich, Zurich, Switzerland.
| |
Collapse
|
16
|
Agarwal SS, Cortes-Medina M, Holter JC, Avendano A, Tinapple JW, Barlage JM, Menyhert MM, Onua LM, Song JW. Rapid low-cost assembly of modular microvessel-on-a-chip with benchtop xurography. LAB ON A CHIP 2024; 24:5065-5076. [PMID: 39397763 PMCID: PMC11472271 DOI: 10.1039/d4lc00565a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/06/2024] [Indexed: 10/15/2024]
Abstract
Blood and lymphatic vessels in the body are central to molecular and cellular transport, tissue repair, and pathophysiology. Several approaches have been employed for engineering microfabricated blood and lymphatic vessels in vitro, yet traditionally these approaches require specialized equipment, facilities, and research training beyond the capabilities of many biomedical laboratories. Here we present xurography as an inexpensive, accessible, and versatile rapid prototyping technique for engineering cylindrical and lumenized microvessels. Using a benchtop xurographer, or a cutting plotter, we fabricated modular multi-layer poly(dimethylsiloxane) (PDMS)-based microphysiological systems (MPS) that house endothelial-lined microvessels approximately 260 μm in diameter embedded within a user-defined 3-D extracellular matrix (ECM). We validated the vascularized MPS (or vessel-on-a-chip) by quantifying changes in blood vessel permeability due to the pro-angiogenic chemokine CXCL12. Moreover, we demonstrated the reconfigurable versatility of this approach by engineering a total of four distinct vessel-ECM arrangements, which were obtained by only minor adjustments to a few steps of the fabrication process. Several of these arrangements, such as ones that incorporate close-ended vessel structures and spatially distinct ECM compartments along the same microvessel, have not been widely achieved with other microfabrication strategies. Therefore, we anticipate that our low-cost and easy-to-implement fabrication approach will facilitate broader adoption of MPS with customizable vascular architectures and ECM components while reducing the turnaround time required for iterative designs.
Collapse
Affiliation(s)
- Shashwat S Agarwal
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH 43210, USA.
| | - Marcos Cortes-Medina
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Jacob C Holter
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Alex Avendano
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Joseph W Tinapple
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Joseph M Barlage
- Department of Biomedical Education and Anatomy, The Ohio State University, Columbus, OH 43210, USA
| | - Miles M Menyhert
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Lotanna M Onua
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Jonathan W Song
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH 43210, USA.
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
17
|
Tran T, Galdina V, Urquidi O, Reis Galvão D, Rieben R, Adachi TBM, Puga Yung GL, Seebach JD. Assessment of NK cytotoxicity and interactions with porcine endothelial cells by live-cell imaging in 2D static and 3D microfluidic systems. Sci Rep 2024; 14:24199. [PMID: 39406778 PMCID: PMC11480498 DOI: 10.1038/s41598-024-75217-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
Natural Killer (NK) cells are pivotal in immune responses to viral infections, malignancies, autoimmune diseases, and transplantation. Assessment of NK cell adhesion, migration, and cytotoxicity is fundamental for in vitro studies. We propose a novel live-cell tracking method that addresses these three major aspects of NK cell function using human NK cells and primary porcine aortic endothelial cells (PAECs) in two-dimensional (2D) static assays and an in-house cylindrical 3D microfluidic system. The results showed a significant increase of NK cytotoxicity against pTNF-activated PAECs, with apoptotic cell death observed in the majority of dead cells, while no difference was observed in the conventional Delfia assay. Computed analysis of NK cell trajectories revealed distinct migratory behaviors, including trajectory length, diameter, average speed, and arrest coefficient. In 3D microfluidic experiments, NK cell attachment to pTNF-activated PAECs substantially increased, accompanied by more dead PAECs compared to control conditions. NK cell trajectories showed versatile migration in various directions and interactions with PAECs. This study uniquely demonstrates NK attachment and killing in a 3D system that mimics blood vessel conditions. Our microscope method offers sensitive single-cell level results, addressing diverse aspects of NK functions. It is adaptable for studying other immune and target cells, providing insights into various biological questions.
Collapse
Affiliation(s)
- Thao Tran
- Department of Medicine, Laboratory of Translational Immunology, Division of Immunology and Allergy, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Viktoriia Galdina
- Department of Medicine, Laboratory of Translational Immunology, Division of Immunology and Allergy, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Oscar Urquidi
- Department of Physical Chemistry, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Daniela Reis Galvão
- Department of Medicine, Laboratory of Translational Immunology, Division of Immunology and Allergy, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Robert Rieben
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Takuji B M Adachi
- Department of Physical Chemistry, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Gisella L Puga Yung
- Department of Medicine, Laboratory of Translational Immunology, Division of Immunology and Allergy, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland.
| | - Jörg D Seebach
- Department of Medicine, Laboratory of Translational Immunology, Division of Immunology and Allergy, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland.
| |
Collapse
|
18
|
Schmid KF, Zeinali S, Moser SK, Dubey C, Schneider S, Deng H, Haefliger S, Marti TM, Guenat OT. Assessing the metastatic potential of circulating tumor cells using an organ-on-chip model. Front Bioeng Biotechnol 2024; 12:1457884. [PMID: 39439549 PMCID: PMC11493642 DOI: 10.3389/fbioe.2024.1457884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024] Open
Abstract
Metastatic lung cancer remains a leading cause of death worldwide, with its intricate metastatic cascade posing significant challenges to researchers and clinicians. Despite substantial progress in understanding this cascade, many aspects remain elusive. Microfluidic-based vasculature-on-chip models have emerged as powerful tools in cancer research, enabling the simulation of specific stages of tumor progression. In this study, we investigate the extravasation behaviors of A549 lung cancer cell subpopulations, revealing distinct differences based on their phenotypes. Our results show that holoclones, which exhibit an epithelial phenotype, do not undergo extravasation. In contrast, paraclones, characterized by a mesenchymal phenotype, demonstrate a notable capacity for extravasation. Furthermore, we observed that paraclones migrate significantly faster than holoclones within the microfluidic model. Importantly, we found that the depletion of vascular endothelial growth factor (VEGF) effectively inhibits the extravasation of paraclones. These findings highlight the utility of microfluidic-based models in replicating key aspects of the metastatic cascade. The insights gained from this study underscore the potential of these models to advance precision medicine by facilitating the assessment of patient-specific cancer cell dynamics and drug responses. This approach could lead to improved strategies for predicting metastatic risk and tailoring personalized cancer therapies, potentially involving the sampling of cancer cells from patients during tumor resection or biopsies.
Collapse
Affiliation(s)
- Karin F. Schmid
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Soheila Zeinali
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Susanne K. Moser
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Christelle Dubey
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Sabine Schneider
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Haibin Deng
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Simon Haefliger
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Thomas M. Marti
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Olivier T. Guenat
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
19
|
Wang Z, Liu X, Shi X, Wang Y. Microvascular network based on the Hilbert curve for nutrient transport in thick tissue. Regen Biomater 2024; 11:rbae094. [PMID: 39350955 PMCID: PMC11441758 DOI: 10.1093/rb/rbae094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/09/2024] [Accepted: 08/04/2024] [Indexed: 10/04/2024] Open
Abstract
To address the uneven nutrient distribution within three-dimensional (3D) tissue models and organoids currently used in medical research, this study introduces a microvascular network based on the Hilbert curve. Our aim was to develop innovative solutions for enhancing nutrient supply in thick tissue models in vitro. By using 3D bioprinting, we engineered microvascular networks of varying Hilbert orders and validated their efficacy in enhancing nutrient uniformity through numerical simulations and experiments. These networks facilitated broader and more uniform nutrient distribution throughout the thick tissue models, particularly the 2° Hilbert microvascular structure, which occupies less space and significantly reduces regions of cellular death. Furthermore, we explored the potential of assembling larger tissue constructs using the 2° Hilbert microvascular network, showcasing its applicability in constructing large-scale biological models. The findings suggest that the 2° Hilbert microvascular structure is particularly effective in ensuring adequate nutrient delivery, thus enhancing the viability and functionality of large-volume tissue models. These innovations hold significant promise for advancing the fields of tissue engineering and regenerative medicine by improving nutrient delivery to in vitro thick tissue block models. This provides a robust foundation for future in vitro research and clinical applications, potentially leading to more effective treatments and interventions in the medical field. The development of these microvascular networks represents a crucial step forward in overcoming the limitations of current 3D tissue models and organoids, paving the way for more sophisticated and reliable biomedical research tools.
Collapse
Affiliation(s)
- Zhenxing Wang
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China
| | - Xuemin Liu
- Department of Gynecology and Obstetrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Xuetao Shi
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China
| | - Yingjun Wang
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
20
|
Zhao N, Pessell AF, Zhu N, Searson PC. Tissue-Engineered Microvessels: A Review of Current Engineering Strategies and Applications. Adv Healthc Mater 2024; 13:e2303419. [PMID: 38686434 PMCID: PMC11338730 DOI: 10.1002/adhm.202303419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 04/10/2024] [Indexed: 05/02/2024]
Abstract
Microvessels, including arterioles, capillaries, and venules, play an important role in regulating blood flow, enabling nutrient and waste exchange, and facilitating immune surveillance. Due to their important roles in maintaining normal function in human tissues, a substantial effort has been devoted to developing tissue-engineered models to study endothelium-related biology and pathology. Various engineering strategies have been developed to recapitulate the structural, cellular, and molecular hallmarks of native human microvessels in vitro. In this review, recent progress in engineering approaches, key components, and culture platforms for tissue-engineered human microvessel models is summarized. Then, tissue-specific models, and the major applications of tissue-engineered microvessels in development, disease modeling, drug screening and delivery, and vascularization in tissue engineering, are reviewed. Finally, future research directions for the field are discussed.
Collapse
Affiliation(s)
- Nan Zhao
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Alexander F Pessell
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Ninghao Zhu
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| |
Collapse
|
21
|
Martier A, Chen Z, Schaps H, Mondrinos MJ, Fang JS. Capturing physiological hemodynamic flow and mechanosensitive cell signaling in vessel-on-a-chip platforms. Front Physiol 2024; 15:1425618. [PMID: 39135710 PMCID: PMC11317428 DOI: 10.3389/fphys.2024.1425618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/10/2024] [Indexed: 08/15/2024] Open
Abstract
Recent advances in organ chip (or, "organ-on-a-chip") technologies and microphysiological systems (MPS) have enabled in vitro investigation of endothelial cell function in biomimetic three-dimensional environments under controlled fluid flow conditions. Many current organ chip models include a vascular compartment; however, the design and implementation of these vessel-on-a-chip components varies, with consequently varied impact on their ability to capture and reproduce hemodynamic flow and associated mechanosensitive signaling that regulates key characteristics of healthy, intact vasculature. In this review, we introduce organ chip and vessel-on-a-chip technology in the context of existing in vitro and in vivo vascular models. We then briefly discuss the importance of mechanosensitive signaling for vascular development and function, with focus on the major mechanosensitive signaling pathways involved. Next, we summarize recent advances in MPS and organ chips with an integrated vascular component, with an emphasis on comparing both the biomimicry and adaptability of the diverse approaches used for supporting and integrating intravascular flow. We review current data showing how intravascular flow and fluid shear stress impacts vessel development and function in MPS platforms and relate this to existing work in cell culture and animal models. Lastly, we highlight new insights obtained from MPS and organ chip models of mechanosensitive signaling in endothelial cells, and how this contributes to a deeper understanding of vessel growth and function in vivo. We expect this review will be of broad interest to vascular biologists, physiologists, and cardiovascular physicians as an introduction to organ chip platforms that can serve as viable model systems for investigating mechanosensitive signaling and other aspects of vascular physiology.
Collapse
Affiliation(s)
- A. Martier
- Department of Biomedical Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, United States
| | - Z. Chen
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, New Orleans, LA, United States
| | - H. Schaps
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, New Orleans, LA, United States
| | - M. J. Mondrinos
- Department of Biomedical Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, United States
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, United States
| | - J. S. Fang
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, New Orleans, LA, United States
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, United States
| |
Collapse
|
22
|
Yadav R, Mahajan S, Singh H, Mehra NK, Madan J, Doijad N, Singh PK, Guru SK. Emerging In Vitro and In Vivo Models: Hope for the Better Understanding of Cancer Progression and Treatment. Adv Biol (Weinh) 2024; 8:e2300487. [PMID: 38581078 DOI: 10.1002/adbi.202300487] [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: 09/12/2023] [Revised: 03/04/2024] [Indexed: 04/07/2024]
Abstract
Various cancer models have been developed to aid the understanding of the underlying mechanisms of tumor development and evaluate the effectiveness of various anticancer drugs in preclinical studies. These models accurately reproduce the critical stages of tumor initiation and development to mimic the tumor microenvironment better. Using these models for target validation, tumor response evaluation, resistance modeling, and toxicity comprehension can significantly enhance the drug development process. Herein, various in vivo or animal models are presented, typically consisting of several mice and in vitro models ranging in complexity from transwell models to spheroids and CRISPR-Cas9 technologies. While in vitro models have been used for decades and dominate the early stages of drug development, they are still limited primary to simplistic tests based on testing on a single cell type cultivated in Petri dishes. Recent advancements in developing new cancer therapies necessitate the generation of complicated animal models that accurately mimic the tumor's complexity and microenvironment. Mice make effective tumor models as they are affordable, have a short reproductive cycle, exhibit rapid tumor growth, and are simple to manipulate genetically. Human cancer mouse models are crucial to understanding the neoplastic process and basic and clinical research improvements. The following review summarizes different in vitro and in vivo metastasis models, their advantages and disadvantages, and their ability to serve as a model for cancer research.
Collapse
Affiliation(s)
- Rachana Yadav
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, 500037, India
| | - Hoshiyar Singh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Neelesh Kumar Mehra
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, 500037, India
| | - Jitender Madan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, 500037, India
| | - Nandkumar Doijad
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, 500037, India
| | - Santosh Kumar Guru
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| |
Collapse
|
23
|
Hansen SH. TruD technology for the study of epi- and endothelial tubes in vitro. PLoS One 2024; 19:e0301099. [PMID: 38728291 PMCID: PMC11086873 DOI: 10.1371/journal.pone.0301099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 03/11/2024] [Indexed: 05/12/2024] Open
Abstract
Beyond the smallest organisms, animals rely on tubes to transport cells, oxygen, nutrients, waste products, and a great variety of secretions. The cardiovascular system, lungs, gastrointestinal and genitourinary tracts, as well as major exocrine glands, are all composed of tubes. Paradoxically, despite their ubiquitous importance, most existing devices designed to study tubes are relatively complex to manufacture and/or utilize. The present work describes a simple method for generating tubes in vitro using nothing more than a low-cost 3D printer along with general lab supplies. The technology is termed "TruD", an acronym for true dimensional. Using this technology, it is readily feasible to cast tubes embedded in ECM with easy access to the lumen. The design is modular to permit more complex tube arrangements and to sustain flow. Importantly, by virtue of its simplicity, TruD technology enables typical molecular cell biology experiments where multiple conditions are assayed in replicate.
Collapse
Affiliation(s)
- Steen H. Hansen
- Department of Pediatrics, Division of Gastroenterology, GI Cell Biology Laboratory, Hepatology and Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
24
|
Yin H, Wang Y, Liu N, Zhong S, Li L, Zhang Q, Liu Z, Yue T. Advances in the Model Structure of In Vitro Vascularized Organ-on-a-Chip. CYBORG AND BIONIC SYSTEMS 2024; 5:0107. [PMID: 40353137 PMCID: PMC12063728 DOI: 10.34133/cbsystems.0107] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 02/29/2024] [Indexed: 05/14/2025] Open
Abstract
Microvasculature plays a crucial role in human physiology and is closely related to various human diseases. Building in vitro vascular networks is essential for studying vascular tissue behavior with repeatable morphology and signaling conditions. Engineered 3D microvascular network models, developed through advanced microfluidic-based techniques, provide accurate and reproducible platforms for studying the microvasculature in vitro, an essential component for designing organ-on-chips to achieve greater biological relevance. By optimizing the microstructure of microfluidic devices to closely mimic the in vivo microenvironment, organ-specific models with healthy and pathological microvascular tissues can be created. This review summarizes recent advancements in in vitro strategies for constructing microvascular tissue and microfluidic devices. It discusses the static vascularization chips' classification, structural characteristics, and the various techniques used to build them: growing blood vessels on chips can be either static or dynamic, and in vitro blood vessels can be grown in microchannels, elastic membranes, and hydrogels. Finally, the paper discusses the application scenarios and key technical issues of existing vascularization chips. It also explores the potential for a novel organoid chip vascularization approach that combines organoids and organ chips to generate better vascularization chips.
Collapse
Affiliation(s)
- Hongze Yin
- School of Mechatronic Engineering and Automation,
Shanghai University, Shanghai 200444, China
| | - Yue Wang
- School of Future Technology,
Shanghai University, Shanghai, China
| | - Na Liu
- School of Mechatronic Engineering and Automation,
Shanghai University, Shanghai 200444, China
- Shanghai Key Laboratory of Intelligent Manufacturing and Robotics,
Shanghai University, Shanghai 200444, China
- Shanghai Institute of Intelligent Science and Technology,
Tongji University, Shanghai, China
| | - Songyi Zhong
- School of Mechatronic Engineering and Automation,
Shanghai University, Shanghai 200444, China
- Shanghai Key Laboratory of Intelligent Manufacturing and Robotics,
Shanghai University, Shanghai 200444, China
| | - Long Li
- School of Mechatronic Engineering and Automation,
Shanghai University, Shanghai 200444, China
- School of Future Technology,
Shanghai University, Shanghai, China
| | - Quan Zhang
- School of Mechatronic Engineering and Automation,
Shanghai University, Shanghai 200444, China
- School of Future Technology,
Shanghai University, Shanghai, China
- Shanghai Key Laboratory of Intelligent Manufacturing and Robotics,
Shanghai University, Shanghai 200444, China
| | - Zeyang Liu
- Department of Bioengineering,
University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Tao Yue
- School of Mechatronic Engineering and Automation,
Shanghai University, Shanghai 200444, China
- School of Future Technology,
Shanghai University, Shanghai, China
- Shanghai Key Laboratory of Intelligent Manufacturing and Robotics,
Shanghai University, Shanghai 200444, China
- Shanghai Institute of Intelligent Science and Technology,
Tongji University, Shanghai, China
| |
Collapse
|
25
|
Khanna A, Oropeza BP, Huang NF. Cardiovascular human organ-on-a-chip platform for disease modeling, drug development, and personalized therapy. J Biomed Mater Res A 2024; 112:512-523. [PMID: 37668192 PMCID: PMC11089005 DOI: 10.1002/jbm.a.37602] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/16/2023] [Accepted: 08/17/2023] [Indexed: 09/06/2023]
Abstract
Cardiovascular organ-on-a-chip (OoC) devices are composed of engineered or native functional tissues that are cultured under controlled microenvironments inside microchips. These systems employ microfabrication and tissue engineering techniques to recapitulate human physiology. This review focuses on human OoC systems to model cardiovascular diseases, to perform drug screening, and to advance personalized medicine. We also address the challenges in the generation of organ chips that can revolutionize the large-scale application of these systems for drug development and personalized therapy.
Collapse
Affiliation(s)
| | - Beu P. Oropeza
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, California, USA
- Center for Tissue Regeneration, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Ngan F. Huang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, California, USA
- Center for Tissue Regeneration, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Department of Chemical Engineering, Stanford University, Stanford, California, USA
| |
Collapse
|
26
|
Konopka J, Żuchowska A, Jastrzębska E. Vascularized tumor-on-chip microplatforms for the studies of neovasculature as hope for more effective cancer treatments. Biosens Bioelectron 2024; 249:115986. [PMID: 38194813 DOI: 10.1016/j.bios.2023.115986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/11/2024]
Abstract
Angiogenesis is the development of new blood vessels from pre-existing vasculature. Multiple factors control its course. Disorders of the distribution of angiogenic agents are responsible for development of solid tumors and its metastases. Understanding of the molecular interactions regulating pathological angiogenesis will allow for development of more effective, even personalized treatment. A simulation of angiogenesis under microflow conditions is a promising alternative to previous studies conducted on animals and on 2D cell cultures. In this review, we summarize what has been discovered so far in the field of vascularized tumor-on-a-chip platforms. For this purpose, we describe different vascularization techniques used in microfluidics, present various attempts to induce angiogenesis-on-a-chip and report some approaches to recapitulate vascularized tumor microenvironment under microflow conditions.
Collapse
Affiliation(s)
- Joanna Konopka
- Warsaw University of Technology, Faculty of Chemistry, Medical Biotechnology, 00-664, Warsaw, Poland
| | - Agnieszka Żuchowska
- Warsaw University of Technology, Faculty of Chemistry, Medical Biotechnology, 00-664, Warsaw, Poland
| | - Elżbieta Jastrzębska
- Warsaw University of Technology, Faculty of Chemistry, Medical Biotechnology, 00-664, Warsaw, Poland.
| |
Collapse
|
27
|
Laowpanitchakorn P, Zeng J, Piantino M, Uchida K, Katsuyama M, Matsusaki M. Biofabrication of engineered blood vessels for biomedical applications. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2024; 25:2330339. [PMID: 38633881 PMCID: PMC11022926 DOI: 10.1080/14686996.2024.2330339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/10/2024] [Indexed: 04/19/2024]
Abstract
To successfully engineer large-sized tissues, establishing vascular structures is essential for providing oxygen, nutrients, growth factors and cells to prevent necrosis at the core of the tissue. The diameter scale of the biofabricated vasculatures should range from 100 to 1,000 µm to support the mm-size tissue while being controllably aligned and spaced within the diffusion limit of oxygen. In this review, insights regarding biofabrication considerations and techniques for engineered blood vessels will be presented. Initially, polymers of natural and synthetic origins can be selected, modified, and combined with each other to support maturation of vascular tissue while also being biocompatible. After they are shaped into scaffold structures by different fabrication techniques, surface properties such as physical topography, stiffness, and surface chemistry play a major role in the endothelialization process after transplantation. Furthermore, biological cues such as growth factors (GFs) and endothelial cells (ECs) can be incorporated into the fabricated structures. As variously reported, fabrication techniques, especially 3D printing by extrusion and 3D printing by photopolymerization, allow the construction of vessels at a high resolution with diameters in the desired range. Strategies to fabricate of stable tubular structures with defined channels will also be discussed. This paper provides an overview of the many advances in blood vessel engineering and combinations of different fabrication techniques up to the present time.
Collapse
Affiliation(s)
| | - Jinfeng Zeng
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| | - Marie Piantino
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
- The Consortium for Future Innovation by Cultured Meat, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| | - Kentaro Uchida
- Materials Solution Department, Product Analysis Center, Panasonic Holdings Corporation, Kadoma, Osaka, Japan
| | - Misa Katsuyama
- Materials Solution Department, Product Analysis Center, Panasonic Holdings Corporation, Kadoma, Osaka, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
- The Consortium for Future Innovation by Cultured Meat, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
28
|
Vo Q, Carlson KA, Chiknas PM, Brocker CN, DaSilva L, Clark E, Park SK, Ajiboye AS, Wier EM, Benam KH. On-Chip Reconstitution of Uniformly Shear-Sensing 3D Matrix-embedded Multicellular Blood Microvessel. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2304630. [PMID: 38465199 PMCID: PMC10923530 DOI: 10.1002/adfm.202304630] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Indexed: 03/12/2024]
Abstract
Preclinical human-relevant modeling of organ-specific vasculature offers a unique opportunity to recreate pathophysiological intercellular, tissue-tissue, and cell-matrix interactions for a broad range of applications. Here, we present a reliable, and simply reproducible process for constructing user-controlled long rounded extracellular matrix (ECM)-embedded vascular microlumens on-chip for endothelization and co-culture with stromal cells obtained from human lung. We demonstrate the critical impact of microchannel cross-sectional geometry and length on uniform distribution and magnitude of vascular wall shear stress, which is key when emulating in vivo-observed blood flow biomechanics in health and disease. In addition, we provide an optimization protocol for multicellular culture and functional validation of the system. Moreover, we show the ability to finely tune rheology of the three-dimensional natural matrix surrounding the vascular microchannel to match pathophysiological stiffness. In summary, we provide the scientific community with a matrix-embedded microvasculature on-chip populated with all-primary human-derived pulmonary endothelial cells and fibroblasts to recapitulate and interrogate lung parenchymal biology, physiological responses, vascular biomechanics, and disease biogenesis in vitro. Such a mix-and-match synthetic platform can be feasibly adapted to study blood vessels, matrix, and ECM-embedded cells in other organs and be cellularized with additional stromal cells.
Collapse
Affiliation(s)
- Quoc Vo
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kaely A. Carlson
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Peter M. Chiknas
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Chad N. Brocker
- Center for Tobacco Products, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Luis DaSilva
- Center for Tobacco Products, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Erica Clark
- Center for Tobacco Products, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Sang Ki Park
- Center for Tobacco Products, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - A. Seun Ajiboye
- Center for Tobacco Products, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Eric M. Wier
- Center for Tobacco Products, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Kambez H. Benam
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
29
|
Moisa SM, Burlacu A, Butnariu LI, Vasile CM, Brinza C, Spoiala EL, Maștaleru A, Leon MM, Rosu ST, Vatasescu R, Cinteză EE. Nanotechnology Innovations in Pediatric Cardiology and Cardiovascular Medicine: A Comprehensive Review. Biomedicines 2024; 12:185. [PMID: 38255290 PMCID: PMC10813221 DOI: 10.3390/biomedicines12010185] [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: 11/29/2023] [Revised: 01/09/2024] [Accepted: 01/13/2024] [Indexed: 01/24/2024] Open
Abstract
(1) Background: Nanomedicine, incorporating various nanoparticles and nanomaterials, offers significant potential in medical practice. Its clinical adoption, however, faces challenges like safety concerns, regulatory hurdles, and biocompatibility issues. Despite these, recent advancements have led to the approval of many nanotechnology-based products, including those for pediatric use. (2) Methods: Our approach included reviewing clinical, preclinical, and animal studies, as well as literature reviews from the past two decades and ongoing trials. (3) Results: Nanotechnology has introduced innovative solutions in cardiovascular care, particularly in managing myocardial ischemia. Key developments include drug-eluting stents, nitric oxide-releasing coatings, and the use of magnetic nanoparticles in cardiomyocyte transplantation. These advancements are pivotal for early detection and treatment. In cardiovascular imaging, nanotechnology enables noninvasive assessments. In pediatric cardiology, it holds promise in assisting the development of biological conduits, synthetic valves, and bioartificial grafts for congenital heart defects, and offers new treatments for conditions like dilated cardiomyopathy and pulmonary hypertension. (4) Conclusions: Nanomedicine presents groundbreaking solutions for cardiovascular diseases in both adults and children. It has the potential to transform cardiac care, from enhancing myocardial ischemia treatment and imaging techniques to addressing congenital heart issues. Further research and guideline development are crucial for optimizing its clinical application and revolutionizing patient care.
Collapse
Affiliation(s)
- Stefana Maria Moisa
- Department of Pediatrics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania (E.L.S.)
- “Sfanta Maria” Clinical Emergency Hospital for Children, 700309 Iasi, Romania (S.T.R.)
| | - Alexandru Burlacu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Institute of Cardiovascular Diseases “Prof. Dr. George I.M. Georgescu”, 700503 Iasi, Romania
| | - Lacramioara Ionela Butnariu
- “Sfanta Maria” Clinical Emergency Hospital for Children, 700309 Iasi, Romania (S.T.R.)
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Corina Maria Vasile
- Pediatric and Adult Congenital Cardiology Department, Centre Hospitalier Universitaire de Bordeaux, 33000 Bordeaux, France;
| | - Crischentian Brinza
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Institute of Cardiovascular Diseases “Prof. Dr. George I.M. Georgescu”, 700503 Iasi, Romania
| | - Elena Lia Spoiala
- Department of Pediatrics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania (E.L.S.)
| | - Alexandra Maștaleru
- Department of Medical Specialties I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.M.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Maria Magdalena Leon
- Department of Medical Specialties I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.M.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Solange Tamara Rosu
- “Sfanta Maria” Clinical Emergency Hospital for Children, 700309 Iasi, Romania (S.T.R.)
- Department of Nursing, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Radu Vatasescu
- Cardio-Thoracic Department, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Clinical Emergency Hospital, 050098 Bucharest, Romania
| | - Eliza Elena Cinteză
- Department of Pediatrics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Pediatric Cardiology, “Marie Curie” Emergency Children’s Hospital, 041451 Bucharest, Romania
| |
Collapse
|
30
|
Tan ML, Jenkins-Johnston N, Huang S, Schutrum B, Vadhin S, Adhikari A, Williams RM, Zipfel WR, Lammerding J, Varner JD, Fischbach C. Endothelial cells metabolically regulate breast cancer invasion toward a microvessel. APL Bioeng 2023; 7:046116. [PMID: 38058993 PMCID: PMC10697723 DOI: 10.1063/5.0171109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/06/2023] [Indexed: 12/08/2023] Open
Abstract
Breast cancer metastasis is initiated by invasion of tumor cells into the collagen type I-rich stroma to reach adjacent blood vessels. Prior work has identified that metabolic plasticity is a key requirement of tumor cell invasion into collagen. However, it remains largely unclear how blood vessels affect this relationship. Here, we developed a microfluidic platform to analyze how tumor cells invade collagen in the presence and absence of a microvascular channel. We demonstrate that endothelial cells secrete pro-migratory factors that direct tumor cell invasion toward the microvessel. Analysis of tumor cell metabolism using metabolic imaging, metabolomics, and computational flux balance analysis revealed that these changes are accompanied by increased rates of glycolysis and oxygen consumption caused by broad alterations of glucose metabolism. Indeed, restricting glucose availability decreased endothelial cell-induced tumor cell invasion. Our results suggest that endothelial cells promote tumor invasion into the stroma due, in part, to reprogramming tumor cell metabolism.
Collapse
Affiliation(s)
- Matthew L. Tan
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Niaa Jenkins-Johnston
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Sarah Huang
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Brittany Schutrum
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Sandra Vadhin
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Abhinav Adhikari
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Rebecca M. Williams
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Warren R. Zipfel
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Jan Lammerding
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Jeffrey D. Varner
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, USA
| | | |
Collapse
|
31
|
Li X, Coates DE. Hollow channels scaffold in bone regenerative: a review. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2023; 34:1702-1715. [PMID: 36794303 DOI: 10.1080/09205063.2023.2181066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/17/2023]
Abstract
Bone substitute materials have been extensively used for bone regeneration over the past 50 years. The development of novel materials, fabrication technologies and the incorporation and release of regenerative cytokines, growth factors, cells and antimicrobials has been driven by the rapid development in the field of additive manufacturing technology. There are still however, significant challenges that need addressing, including ways to better mediate the rapid vascularization of bone scaffolds to enhance subsequent regeneration and osteogenesis. Increasing construct porosity can accelerate the development of blood vessels in the scaffold, but doing so also weakens the constructs mechanical properties. A novel design for promoting rapid vascularization is to fabricate custom-made hollow channels as bone scaffolds. Summarized here are the current developments in hollow channels scaffold, including their biological attributes, physio-chemical properties, and effects on regeneration. An overview of recent developments in scaffold fabrication as they relate to hollow channel constructs and their structural features will be introduced with an emphasis on attributes that enhance new bone and vessel formation. Furthermore, the potential to enhance angiogenesis and osteogenesis by replicating the structure of real bone will be highlighted.
Collapse
Affiliation(s)
- Xiao Li
- University of Otago, Dunedin, New Zealand
| | - Dawn Elizabeth Coates
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| |
Collapse
|
32
|
Cooley MB, Wulftange WJ, Wegierak D, Goreke U, Abenojar EC, Gurkan UA, Exner AA. Real-time imaging of nanobubble ultrasound contrast agent flow, extravasation, and diffusion through an extracellular matrix using a microfluidic model. LAB ON A CHIP 2023; 23:3453-3466. [PMID: 37424286 PMCID: PMC11684791 DOI: 10.1039/d3lc00514c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Lipid shell-stabilized nanoparticles with a perfluorocarbon gas-core, or nanobubbles, have recently attracted attention as a new contrast agent for molecular ultrasound imaging and image-guided therapy. Due to their small size (∼275 nm diameter) and flexible shell, nanobubbles have been shown to extravasate through hyperpermeable vasculature (e.g., in tumors). However, little is known about the dynamics and depth of extravasation of intact, acoustically active nanobubbles. Accordingly, in this work, we developed a microfluidic chip with a lumen and extracellular matrix (ECM) and imaging method that allows real-time imaging and characterization of the extravasation process with high-frequency ultrasound. The microfluidic device has a lumen and is surrounded by an extracellular matrix with tunable porosity. The combination of ultrasound imaging and the microfluidic chip advantageously produces real-time images of the entire length and depth of the matrix. This captures the matrix heterogeneity, offering advantages over other imaging techniques with smaller fields of view. Results from this study show that nanobubbles diffuse through a 1.3 μm pore size (2 mg mL-1) collagen I matrix 25× faster with a penetration depth that was 0.19 mm deeper than a 3.7 μm (4 mg mL-1) matrix. In the 3.7 μm pore size matrix, nanobubbles diffused 92× faster than large nanobubbles (∼875 nm diameter). Decorrelation time analysis was successfully used to differentiate flowing and extra-luminally diffusing nanobubbles. In this work, we show for the first time that combination of an ultrasound-capable microfluidic chip and real-time imaging provided valuable insight into spatiotemporal nanoparticle movement through a heterogeneous extracellular matrix. This work could help accurately predict parameters (e.g., injection dosage) that improve translation of nanoparticles from in vitro to in vivo environments.
Collapse
Affiliation(s)
- Michaela B Cooley
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - William J Wulftange
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Dana Wegierak
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Utku Goreke
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| | - Eric C Abenojar
- Department of Radiology, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| | - Umut A Gurkan
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| | - Agata A Exner
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Department of Radiology, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| |
Collapse
|
33
|
Fernandes J, Karra N, Swindle EJ, Morgan H. Droplet fluidics for time-dependent analysis of barrier permeability in an epithelial barrier on chip system. RSC Adv 2023; 13:14494-14500. [PMID: 37179995 PMCID: PMC10173818 DOI: 10.1039/d3ra00470h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
A droplet generator has been developed that interfaces with a barrier-on-chip platform for temporal analyte compartmentalisation and analysis. Droplets are generated every 20 minutes in 8 separate parallel microchannels, with an average droplet volume of 9.47 ± 0.6 μL, allowing simultaneous analysis of 8 different experiments. The device was tested using an epithelial barrier model by monitoring the diffusion of a fluorescent high molecular weight dextran molecule. The epithelial barrier was perturbed using detergent leading to a peak at 3-4 hours, correlating with simulations. For the untreated (control) a constant, very low level of dextran diffusion was observed. The epithelial cell barrier properties were also continuously measured using electrical impedance spectroscopy to extract an equivalent trans epithelial resistance.
Collapse
Affiliation(s)
- Joao Fernandes
- Electronics and Computer Science, Faculty of Physical Sciences and Engineering, University of Southampton UK
| | - Nikita Karra
- Electronics and Computer Science, Faculty of Physical Sciences and Engineering, University of Southampton UK
| | - Emily J Swindle
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton UK
- Institute for Life Sciences, University of Southampton UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton UK
| | - Hywel Morgan
- Electronics and Computer Science, Faculty of Physical Sciences and Engineering, University of Southampton UK
- Institute for Life Sciences, University of Southampton UK
| |
Collapse
|
34
|
Shevchuk O, Palii S, Pak A, Chantada N, Seoane N, Korda M, Campos-Toimil M, Álvarez E. Vessel-on-a-Chip: A Powerful Tool for Investigating Endothelial COVID-19 Fingerprints. Cells 2023; 12:cells12091297. [PMID: 37174696 PMCID: PMC10177552 DOI: 10.3390/cells12091297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/21/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023] Open
Abstract
Coronavirus disease (COVID-19) causes various vascular and blood-related reactions, including exacerbated responses. The role of endothelial cells in this acute response is remarkable and may remain important beyond the acute phase. As we move into a post-COVID-19 era (where most people have been or will be infected by the SARS-CoV-2 virus), it is crucial to define the vascular consequences of COVID-19, including the long-term effects on the cardiovascular system. Research is needed to determine whether chronic endothelial dysfunction following COVID-19 could lead to an increased risk of cardiovascular and thrombotic events. Endothelial dysfunction could also serve as a diagnostic and therapeutic target for post-COVID-19. This review covers these topics and examines the potential of emerging vessel-on-a-chip technology to address these needs. Vessel-on-a-chip would allow for the study of COVID-19 pathophysiology in endothelial cells, including the analysis of SARS-CoV-2 interactions with endothelial function, leukocyte recruitment, and platelet activation. "Personalization" could be implemented in the models through induced pluripotent stem cells, patient-specific characteristics, or genetic modified cells. Adaptation for massive testing under standardized protocols is now possible, so the chips could be incorporated for the personalized follow-up of the disease or its sequalae (long COVID) and for the research of new drugs against COVID-19.
Collapse
Affiliation(s)
- Oksana Shevchuk
- Department of Pharmacology and Clinical Pharmacology, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine
| | - Svitlana Palii
- Department of Pharmacology and Clinical Pharmacology, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine
| | - Anastasiia Pak
- Department of Medical Biochemistry, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine
| | - Nuria Chantada
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Nuria Seoane
- Physiology and Pharmacology of Chronic Diseases (FIFAEC) Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Mykhaylo Korda
- Department of Medical Biochemistry, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine
| | - Manuel Campos-Toimil
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Physiology and Pharmacology of Chronic Diseases (FIFAEC) Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Ezequiel Álvarez
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIBERCV, Institute of Health Carlos III, 28220 Madrid, Spain
| |
Collapse
|
35
|
Ferrari D, Sengupta A, Heo L, Pethö L, Michler J, Geiser T, de Jesus Perez VA, Kuebler WM, Zeinali S, Guenat OT. Effects of biomechanical and biochemical stimuli on angio- and vasculogenesis in a complex microvasculature-on-chip. iScience 2023; 26:106198. [PMID: 36879808 PMCID: PMC9985038 DOI: 10.1016/j.isci.2023.106198] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/31/2022] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
The endothelium of blood vessels is a vital organ that reacts differently to subtle changes in stiffness and mechanical forces exerted on its environment (extracellular matrix (ECM)). Upon alteration of these biomechanical cues, endothelial cells initiate signaling pathways that govern vascular remodeling. The emerging organs-on-chip technologies allow the mimicking of complex microvasculature networks, identifying the combined or singular effects of these biomechanical or biochemical stimuli. Here, we present a microvasculature-on-chip model to investigate the singular effect of ECM stiffness and mechanical cyclic stretch on vascular development. Following two different approaches for vascular growth, the effect of ECM stiffness on sprouting angiogenesis and the effect of cyclic stretch on endothelial vasculogenesis are studied. Our results indicate that ECM hydrogel stiffness controls the size of the patterned vasculature and the density of sprouting angiogenesis. RNA sequencing shows that the cellular response to stretching is characterized by the upregulation of certain genes such as ANGPTL4+5, PDE1A, and PLEC.
Collapse
Affiliation(s)
- Dario Ferrari
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Arunima Sengupta
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Lyong Heo
- Stanford Center for Genomics and Personalized Medicine, Palo Alto, CA, USA
| | - Laszlo Pethö
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Laboratory for Mechanics of Materials and Nanostructures, Thun, Switzerland
| | - Johann Michler
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Laboratory for Mechanics of Materials and Nanostructures, Thun, Switzerland
| | - Thomas Geiser
- Department of Pulmonary Medicine, Inselspital, University Hospital of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Vinicio A. de Jesus Perez
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University Medical Center, Stanford, CA, USA
| | - Wolfgang M. Kuebler
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Soheila Zeinali
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Olivier T. Guenat
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
- Department of Pulmonary Medicine, Inselspital, University Hospital of Bern, Bern, Switzerland
- Department of General Thoracic Surgery, Inselspital, University Hospital of Bern, Bern, Switzerland
| |
Collapse
|
36
|
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
|
37
|
You S, Xiang Y, Hwang HH, Berry DB, Kiratitanaporn W, Guan J, Yao E, Tang M, Zhong Z, Ma X, Wangpraseurt D, Sun Y, Lu TY, Chen S. High cell density and high-resolution 3D bioprinting for fabricating vascularized tissues. SCIENCE ADVANCES 2023; 9:eade7923. [PMID: 36812321 PMCID: PMC9946358 DOI: 10.1126/sciadv.ade7923] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/24/2023] [Indexed: 06/18/2023]
Abstract
Three-dimensional (3D) bioprinting techniques have emerged as the most popular methods to fabricate 3D-engineered tissues; however, there are challenges in simultaneously satisfying the requirements of high cell density (HCD), high cell viability, and fine fabrication resolution. In particular, bioprinting resolution of digital light processing-based 3D bioprinting suffers with increasing bioink cell density due to light scattering. We developed a novel approach to mitigate this scattering-induced deterioration of bioprinting resolution. The inclusion of iodixanol in the bioink enables a 10-fold reduction in light scattering and a substantial improvement in fabrication resolution for bioinks with an HCD. Fifty-micrometer fabrication resolution was achieved for a bioink with 0.1 billion per milliliter cell density. To showcase the potential application in tissue/organ 3D bioprinting, HCD thick tissues with fine vascular networks were fabricated. The tissues were viable in a perfusion culture system, with endothelialization and angiogenesis observed after 14 days of culture.
Collapse
Affiliation(s)
- Shangting You
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yi Xiang
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Henry H. Hwang
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - David B. Berry
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Orthopedic Surgery, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wisarut Kiratitanaporn
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jiaao Guan
- Department of Electrical and Computer Engineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Emmie Yao
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Min Tang
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Zheng Zhong
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Xinyue Ma
- School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Daniel Wangpraseurt
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
- Scripps Institution of Oceanography, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yazhi Sun
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ting-yu Lu
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Shaochen Chen
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Electrical and Computer Engineering, University of California, San Diego, La Jolla, CA 92093, USA
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
38
|
Cabral KA, Srivastava V, Graham AJ, Coyle MC, Stashko C, Weaver V, Gartner ZJ. Programming the Self-Organization of Endothelial Cells into Perfusable Microvasculature. Tissue Eng Part A 2023; 29:80-92. [PMID: 36181350 PMCID: PMC10266707 DOI: 10.1089/ten.tea.2022.0072] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 09/15/2022] [Indexed: 11/12/2022] Open
Abstract
The construction of three-dimensional (3D) microvascular networks with defined structures remains challenging. Emerging bioprinting strategies provide a means of patterning endothelial cells (ECs) into the geometry of 3D microvascular networks, but the microenvironmental cues necessary to promote their self-organization into cohesive and perfusable microvessels are not well known. To this end, we reconstituted microvessel formation in vitro by patterning thin lines of closely packed ECs fully embedded within a 3D extracellular matrix (ECM) and observed how different microenvironmental parameters influenced EC behaviors and their self-organization into microvessels. We found that the inclusion of fibrillar matrices, such as collagen I, into the ECM positively influenced cell condensation into extended geometries such as cords. We also identified the presence of a high-molecular-weight protein(s) in fetal bovine serum that negatively influenced EC condensation. This component destabilized cord structure by promoting cell protrusions and destabilizing cell-cell adhesions. Endothelial cords cultured in the presence of fibrillar collagen and in the absence of this protein activity were able to polarize, lumenize, incorporate mural cells, and support fluid flow. These optimized conditions allowed for the construction of branched and perfusable microvascular networks directly from patterned cells in as little as 3 days. These findings reveal important design principles for future microvascular engineering efforts based on bioprinting and micropatterning techniques. Impact statement Bioprinting is a potential strategy to achieve microvascularization in engineered tissues. However, the controlled self-organization of patterned endothelial cells into perfusable microvasculature remains challenging. We used DNA Programmed Assembly of Cells to create cell-dense, capillary-sized cords of endothelial cells with complete control over their structure. We optimized the matrix and media conditions to promote self-organization and maturation of these endothelial cords into stable and perfusable microvascular networks.
Collapse
Affiliation(s)
- Katelyn A. Cabral
- Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, Berkeley, California, USA
| | - Vasudha Srivastava
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
| | - Austin J. Graham
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
- Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, California, USA
| | - Maxwell C. Coyle
- Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley, California, USA
| | - Connor Stashko
- Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, Berkeley, California, USA
| | - Valerie Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Zev J. Gartner
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
- Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, California, USA
- Center for Cellular Construction, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
39
|
Experimental Models of In Vitro Blood-Brain Barrier for CNS Drug Delivery: An Evolutionary Perspective. Int J Mol Sci 2023; 24:ijms24032710. [PMID: 36769032 PMCID: PMC9916529 DOI: 10.3390/ijms24032710] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Central nervous system (CNS) disorders represent one of the leading causes of global health burden. Nonetheless, new therapies approved against these disorders are among the lowest compared to their counterparts. The absence of reliable and efficient in vitro blood-brain barrier (BBB) models resembling in vivo barrier properties stands out as a significant roadblock in developing successful therapy for CNS disorders. Therefore, advancement in the creation of robust and sensitive in vitro BBB models for drug screening might allow us to expedite neurological drug development. This review discusses the major in vitro BBB models developed as of now for exploring the barrier properties of the cerebral vasculature. Our main focus is describing existing in vitro models, including the 2D transwell models covering both single-layer and co-culture models, 3D organoid models, and microfluidic models with their construction, permeability measurement, applications, and limitations. Although microfluidic models are better at recapitulating the in vivo properties of BBB than other models, significant gaps still exist for their use in predicting the performance of neurotherapeutics. However, this comprehensive account of in vitro BBB models can be useful for researchers to create improved models in the future.
Collapse
|
40
|
de Silva L, Bernal PN, Rosenberg A, Malda J, Levato R, Gawlitta D. Biofabricating the vascular tree in engineered bone tissue. Acta Biomater 2023; 156:250-268. [PMID: 36041651 DOI: 10.1016/j.actbio.2022.08.051] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 01/18/2023]
Abstract
The development of tissue engineering strategies for treatment of large bone defects has become increasingly relevant, given the growing demand for bone substitutes. Native bone is composed of a dense vascular network necessary for the regulation of bone development, regeneration and homeostasis. A major obstacle in fabricating living, clinically relevant-sized bone mimics (1-10 cm3) is the limited supply of nutrients, including oxygen to the core of the construct. Therefore, strategies to support vascularization are pivotal for the development of tissue engineered bone constructs. Creating a functional bone construct integrated with a vascular network, capable of delivering the necessary nutrients for optimal tissue development is imperative for translation into the clinics. The vascular system is composed of a complex network that runs throughout the body in a tree-like hierarchical branching fashion. A significant challenge for tissue engineering approaches lies in mimicking the intricate, multi-scale structures consisting of larger vessels (macro-vessels) which interconnect with multiple sprouting vessels (microvessels) in a closed network. The advent of biofabrication has enabled complex, out of plane channels to be generated and has laid the groundwork for the creation of multi-scale vasculature in recent years. This review highlights the key state-of-the-art achievements for the development of vascular networks of varying scales in the field of biofabrication with a particular focus for its application in developing a functional tissue engineered bone construct. STATEMENT OF SIGNIFICANCE: There is a growing need for bone substitutes to overcome the limited supply of patient-derived bone. Bone tissue engineering aims to overcome this by combining stem cells with scaffolds to restore missing bone. The current bottleneck in upscaling is the lack of an integrated vascular network, required for the delivery of nutrients to cells. 3D bioprinting techniques has enabled the creation of complex hollow structures of varying dimensions that resemble native blood vessels. The convergence of multiple materials, cell types and fabrication approaches, opens the possibility of developing clinically-relevant sized vascularized bone constructs. This review provides an up-to-date insight of the technologies currently available for the generation of complex vascular networks, with a focus on their application in bone tissue engineering.
Collapse
Affiliation(s)
- Leanne de Silva
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, the Netherlands.
| | - Paulina N Bernal
- Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, the Netherlands; Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands
| | - Ajw Rosenberg
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands
| | - Jos Malda
- Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, the Netherlands; Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands; Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CT, the Netherlands
| | - Riccardo Levato
- Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, the Netherlands; Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands; Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CT, the Netherlands
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, the Netherlands
| |
Collapse
|
41
|
Hydrogel-Based Tissue-Mimics for Vascular Regeneration and Tumor Angiogenesis. Regen Med 2023. [DOI: 10.1007/978-981-19-6008-6_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
42
|
Blood brain barrier-on-a-chip to model neurological diseases. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
43
|
Sano T, Nakajima T, Senda KA, Nakano S, Yamato M, Ikeda Y, Zeng H, Kawabe JI, Matsunaga YT. Image-based crosstalk analysis of cell-cell interactions during sprouting angiogenesis using blood-vessel-on-a-chip. Stem Cell Res Ther 2022; 13:532. [PMID: 36575469 PMCID: PMC9795717 DOI: 10.1186/s13287-022-03223-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 12/15/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Sprouting angiogenesis is an important mechanism for morphogenetic phenomena, including organ development, wound healing, and tissue regeneration. In regenerative medicine, therapeutic angiogenesis is a clinical solution for recovery from ischemic diseases. Mesenchymal stem cells (MSCs) have been clinically used given their pro-angiogenic effects. MSCs are reported to promote angiogenesis by differentiating into pericytes or other vascular cells or through cell-cell communication using multiple protein-protein interactions. However, how MSCs physically contact and move around ECs to keep the sprouting angiogenesis active remains unknown. METHODS We proposed a novel framework of EC-MSC crosstalk analysis using human umbilical vein endothelial cells (HUVECs) and MSCs obtained from mice subcutaneous adipose tissue on a 3D in vitro model, microvessel-on-a-chip, which allows cell-to-tissue level study. The microvessels were fabricated and cultured for 10 days in a collagen matrix where MSCs were embedded. RESULTS Immunofluorescence imaging using a confocal laser microscope showed that MSCs smoothed the surface of the microvessel and elongated the angiogenic sprouts by binding to the microvessel's specific microstructures. Additionally, three-dimensional modeling of HUVEC-MSC intersections revealed that MSCs were selectively located around protrusions or roots of angiogenic sprouts, whose surface curvature was excessively low or high, respectively. CONCLUSIONS The combination of our microvessel-on-a-chip system for 3D co-culture and image-based crosstalk analysis demonstrated that MSCs are selectively localized to concave-convex surfaces on scaffold structures and that they are responsible for the activation and stabilization of capillary vessels.
Collapse
Affiliation(s)
- Takanori Sano
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505 Japan
| | - Tadaaki Nakajima
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505 Japan ,grid.268441.d0000 0001 1033 6139Department of Science, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027 Japan
| | - Koharu Alicia Senda
- Hiroo Gakuen Junior and Senior High School, 5-1-14 Minami Azabu, Minato-ku, Tokyo, 106-0047 Japan
| | - Shizuka Nakano
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505 Japan
| | - Mizuho Yamato
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505 Japan
| | - Yukinori Ikeda
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505 Japan
| | - Hedele Zeng
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505 Japan
| | - Jun-ichi Kawabe
- grid.252427.40000 0000 8638 2724Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-higashi, Asahikawa, Hokkaido 078-8510 Japan
| | - Yukiko T. Matsunaga
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505 Japan
| |
Collapse
|
44
|
de Graaf MNS, Vivas A, Kasi DG, van den Hil FE, van den Berg A, van der Meer AD, Mummery CL, Orlova VV. Multiplexed fluidic circuit board for controlled perfusion of 3D blood vessels-on-a-chip. LAB ON A CHIP 2022; 23:168-181. [PMID: 36484766 PMCID: PMC9764810 DOI: 10.1039/d2lc00686c] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/21/2022] [Indexed: 06/11/2023]
Abstract
Three-dimensional (3D) blood vessels-on-a-chip (VoC) models integrate the biological complexity of vessel walls with dynamic microenvironmental cues, such as wall shear stress (WSS) and circumferential strain (CS). However, these parameters are difficult to control and are often poorly reproducible due to the high intrinsic diameter variation of individual 3D-VoCs. As a result, the throughput of current 3D systems is one-channel-at-a-time. Here, we developed a fluidic circuit board (FCB) for simultaneous perfusion of up to twelve 3D-VoCs using a single set of control parameters. By designing the internal hydraulic resistances in the FCB appropriately, it was possible to provide a pre-set WSS to all connected 3D-VoCs, despite significant variation in lumen diameters. Using this FCB, we found that variation of CS or WSS induce morphological changes to human induced pluripotent stem cell (hiPSC)-derived endothelial cells (ECs) and conclude that control of these parameters using a FCB is necessary to study 3D-VOCs.
Collapse
Affiliation(s)
- Mees N S de Graaf
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Aisen Vivas
- Applied Stem Cell Technologies, University of Twente, 7500AE Enschede, The Netherlands
- BIOS Lab on a Chip Group, MESA+ Institute for Nanotechnology, Technical Medical Centre, Max Planck Institute for Complex Fluid Dynamics, University of Twente, 7500AE Enschede, The Netherlands
| | - Dhanesh G Kasi
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Neurology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Francijna E van den Hil
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Albert van den Berg
- BIOS Lab on a Chip Group, MESA+ Institute for Nanotechnology, Technical Medical Centre, Max Planck Institute for Complex Fluid Dynamics, University of Twente, 7500AE Enschede, The Netherlands
| | | | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
- Applied Stem Cell Technologies, University of Twente, 7500AE Enschede, The Netherlands
| | - Valeria V Orlova
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| |
Collapse
|
45
|
Varhue WB, Rane A, Castellanos-Sanchez R, Peirce SM, Christ G, Swami NS. Perfusable cell-laden micropatterned hydrogels for delivery of spatiotemporal vascular-like cues to tissues. ORGANS-ON-A-CHIP 2022; 4:100017. [PMID: 36865345 PMCID: PMC9977322 DOI: 10.1016/j.ooc.2022.100017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The integration of vasculature at physiological scales within 3D cultures of cell-laden hydrogels for the delivery of spatiotemporal mass transport, chemical and mechanical cues, is a stepping-stone towards building in vitro tissue models that recapitulate in vivo cues. To address this challenge, we present a versatile method to micropattern adjoining hydrogel shells with a perfusable channel or lumen core, for enabling facile integration with fluidic control systems, on one hand, and to cell-laden biomaterial interfaces, on the other hand. This microfluidic imprint lithography methodology utilizes the high tolerance and reversible nature of the bond alignment process to lithographically position multiple layers of imprints within a microfluidic device for sequential filling and patterning of hydrogel lumen structures with single or multiple shells. Through fluidic interfacing of the structures, the ability to deliver physiologically relevant mechanical cues for recapitulating cyclical stretch on the hydrogel shell and shear stress on endothelial cells in the lumen are validated. We envision application of this platform for recapitulation of the bio-functionality and topology of micro-vasculatures, alongside the ability to deliver transport and mechanical cues, as needed for 3D culture to construct in vitro tissue models.
Collapse
Affiliation(s)
- Walter B. Varhue
- Electrical and Computer Engineering, University of Virginia, Charlottesville, VA, 22904, USA
| | - Aditya Rane
- Chemistry, University of Virginia, Charlottesville, VA, 22904, USA
| | | | - Shayn M. Peirce
- Biomedical Engineering, University of Virginia, Charlottesville, VA, 22904, USA
| | - George Christ
- Biomedical Engineering, University of Virginia, Charlottesville, VA, 22904, USA
| | - Nathan S. Swami
- Electrical and Computer Engineering, University of Virginia, Charlottesville, VA, 22904, USA
- Chemistry, University of Virginia, Charlottesville, VA, 22904, USA
- Corresponding author. University of Virginia, 351 McCormick Rd, Charlottesville, VA, 22904, USA. (N.S. Swami)
| |
Collapse
|
46
|
Camman M, Joanne P, Brun J, Marcellan A, Dumont J, Agbulut O, Hélary C. Anisotropic dense collagen hydrogels with two ranges of porosity to mimic the skeletal muscle extracellular matrix. BIOMATERIALS ADVANCES 2022; 144:213219. [PMID: 36481519 DOI: 10.1016/j.bioadv.2022.213219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 11/08/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
Despite the crucial role of the extracellular matrix (ECM) in the organotypic organization and function of skeletal muscles, most 3D models do not mimic its specific characteristics, namely its biochemical composition, stiffness, anisotropy, and porosity. Here, a novel 3D in vitro model of muscle ECM was developed reproducing these four crucial characteristics of the native ECM. An anisotropic hydrogel mimicking the muscle fascia was obtained thanks to unidirectional 3D printing of dense collagen with aligned collagen fibrils. The space between the different layers was tuned to generate an intrinsic network of pores (100 μm) suitable for nutrient and oxygen diffusion. By modulating the gelling conditions, the mechanical properties of the construct reached those measured in the physiological muscle ECM. This artificial matrix was thus evaluated for myoblast differentiation. The addition of large channels (600 μm) by molding permitted to create a second range of porosity suitable for cell colonization without altering the physical properties of the hydrogel. Skeletal myoblasts embedded in Matrigel®, seeded within the channels, organized in 3D, and differentiated into multinucleated myotubes. These results show that porous and anisotropic dense collagen hydrogels are promising biomaterials to model skeletal muscle ECM.
Collapse
Affiliation(s)
- Marie Camman
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, CNRS, UMR 7574, F-75005, Paris, France; Institut de Biologie Paris-Seine (IBPS), Sorbonne Université, CNRS, UMR 8256, Inserm U1164, Biological Adaptation and Ageing, F-75005, Paris, France
| | - Pierre Joanne
- Institut de Biologie Paris-Seine (IBPS), Sorbonne Université, CNRS, UMR 8256, Inserm U1164, Biological Adaptation and Ageing, F-75005, Paris, France
| | - Julie Brun
- Sciences et Ingénierie de la Matière Molle, ESPCI Paris, Université PSL, CNRS, Sorbonne Université, F-75005, Paris, France
| | - Alba Marcellan
- Sciences et Ingénierie de la Matière Molle, ESPCI Paris, Université PSL, CNRS, Sorbonne Université, F-75005, Paris, France
| | - Julien Dumont
- CIRB Microscopy facility, Collège de France, CNRS, UMR 7241, Inserm U1050, F-75005, Paris, France
| | - Onnik Agbulut
- Institut de Biologie Paris-Seine (IBPS), Sorbonne Université, CNRS, UMR 8256, Inserm U1164, Biological Adaptation and Ageing, F-75005, Paris, France.
| | - Christophe Hélary
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, CNRS, UMR 7574, F-75005, Paris, France.
| |
Collapse
|
47
|
Wang X, Chan V, Corridon PR. Acellular Tissue-Engineered Vascular Grafts from Polymers: Methods, Achievements, Characterization, and Challenges. Polymers (Basel) 2022; 14:4825. [PMID: 36432950 PMCID: PMC9695055 DOI: 10.3390/polym14224825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/03/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022] Open
Abstract
Extensive and permanent damage to the vasculature leading to different pathogenesis calls for developing innovative therapeutics, including drugs, medical devices, and cell therapies. Innovative strategies to engineer bioartificial/biomimetic vessels have been extensively exploited as an effective replacement for vessels that have seriously malfunctioned. However, further studies in polymer chemistry, additive manufacturing, and rapid prototyping are required to generate highly engineered vascular segments that can be effectively integrated into the existing vasculature of patients. One recently developed approach involves designing and fabricating acellular vessel equivalents from novel polymeric materials. This review aims to assess the design criteria, engineering factors, and innovative approaches for the fabrication and characterization of biomimetic macro- and micro-scale vessels. At the same time, the engineering correlation between the physical properties of the polymer and biological functionalities of multiscale acellular vascular segments are thoroughly elucidated. Moreover, several emerging characterization techniques for probing the mechanical properties of tissue-engineered vascular grafts are revealed. Finally, significant challenges to the clinical transformation of the highly promising engineered vessels derived from polymers are identified, and unique perspectives on future research directions are presented.
Collapse
Affiliation(s)
- Xinyu Wang
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Vincent Chan
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Peter R. Corridon
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Center for Biotechnology, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| |
Collapse
|
48
|
LaMontagne E, Muotri AR, Engler AJ. Recent advancements and future requirements in vascularization of cortical organoids. Front Bioeng Biotechnol 2022; 10:1048731. [PMID: 36406234 PMCID: PMC9669755 DOI: 10.3389/fbioe.2022.1048731] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/18/2022] [Indexed: 07/23/2023] Open
Abstract
The fields of tissue engineering and disease modeling have become increasingly cognizant of the need to create complex and mature structures in vitro to adequately mimic the in vivo niche. Specifically for neural applications, human brain cortical organoids (COs) require highly stratified neurons and glial cells to generate synaptic functions, and to date, most efforts achieve only fetal functionality at best. Moreover, COs are usually avascular, inducing the development of necrotic cores, which can limit growth, development, and maturation. Recent efforts have attempted to vascularize cortical and other organoid types. In this review, we will outline the components of a fully vascularized CO as they relate to neocortical development in vivo. These components address challenges in recapitulating neurovascular tissue patterning, biomechanical properties, and functionality with the goal of mirroring the quality of organoid vascularization only achieved with an in vivo host. We will provide a comprehensive summary of the current progress made in each one of these categories, highlighting advances in vascularization technologies and areas still under investigation.
Collapse
Affiliation(s)
- Erin LaMontagne
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
| | - Alysson R. Muotri
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, United States
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Adam J. Engler
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| |
Collapse
|
49
|
Tan SY, Jing Q, Leung Z, Xu Y, Cheng LKW, Tam SST, Wu AR. Transcriptomic analysis of 3D vasculature-on-a-chip reveals paracrine factors affecting vasculature growth and maturation. LAB ON A CHIP 2022; 22:3885-3897. [PMID: 36093896 DOI: 10.1039/d2lc00570k] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
In vitro models of vasculature are of great importance for modelling vascular physiology and pathology. However, there is usually a lack of proper spatial patterning of interacting heterotypic cells in conventional vasculature dish models, which might confound results between contact and non-contact interactions. We use a microfluidic platform with structurally defined separation between human microvasculature and fibroblasts to probe their dynamic, paracrine interactions. We also develop a novel, versatile technique to retrieve cells embedded in extracellular matrix from the microfluidic device for downstream transcriptomic analysis, and uncover growth factor and cytokine expression profiles associated with improved vasculature growth. Paired receptor-ligand analysis further reveals paracrine signaling molecules that could be supplemented into the medium for vasculatures models where fibroblast coculture is undesirable or infeasible. These findings also provide deeper insights into the molecular cues for more physiologically relevant vascular mimicry and vascularized organoid model for clinical applications such as drug screening and disease modeling.
Collapse
Affiliation(s)
- Sin Yen Tan
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong.
| | - Qiuyu Jing
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Ziuwin Leung
- Department of Electrical and Computer Engineering, Centre for Applied Synthetic Biology, Concordia University, Montréal, Québec H3G1M8, Canada
| | - Ying Xu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Lily Kwan Wai Cheng
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Sindy Sing Ting Tam
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Angela Ruohao Wu
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong.
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
- State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong S.A.R., China
| |
Collapse
|
50
|
Simitian G, Virumbrales-Muñoz M, Sánchez-de-Diego C, Beebe DJ, Kosoff D. Microfluidics in vascular biology research: a critical review for engineers, biologists, and clinicians. LAB ON A CHIP 2022; 22:3618-3636. [PMID: 36047330 PMCID: PMC9530010 DOI: 10.1039/d2lc00352j] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Neovascularization, the formation of new blood vessels, has received much research attention due to its implications for physiological processes and diseases. Most studies using traditional in vitro and in vivo platforms find challenges in recapitulating key cellular and mechanical cues of the neovascularization processes. Microfluidic in vitro models have been presented as an alternative to these limitations due to their capacity to leverage microscale physics to control cell organization and integrate biochemical and mechanical cues, such as shear stress, cell-cell interactions, or nutrient gradients, making them an ideal option for recapitulating organ physiology. Much has been written about the use of microfluidics in vascular biology models from an engineering perspective. However, a review introducing the different models, components and progress for new potential adopters of these technologies was absent in the literature. Therefore, this paper aims to approach the use of microfluidic technologies in vascular biology from a perspective of biological hallmarks to be studied and written for a wide audience ranging from clinicians to engineers. Here we review applications of microfluidics in vascular biology research, starting with design considerations and fabrication techniques. After that, we review the state of the art in recapitulating angiogenesis and vasculogenesis, according to the hallmarks recapitulated and complexity of the models. Finally, we discuss emerging research areas in neovascularization, such as drug discovery, and potential future directions.
Collapse
Affiliation(s)
- Grigor Simitian
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA.
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - María Virumbrales-Muñoz
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Cristina Sánchez-de-Diego
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - David J Beebe
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - David Kosoff
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA.
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|