1
|
Yazdanpanah Moghadam E, Sonenberg N, Packirisamy M. Microfluidic Wound-Healing Assay for Comparative Study on Fluid Dynamic, Chemical and Mechanical Wounding on Microglia BV2 Migration. MICROMACHINES 2024; 15:1004. [PMID: 39203655 PMCID: PMC11356282 DOI: 10.3390/mi15081004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/19/2024] [Accepted: 07/26/2024] [Indexed: 09/03/2024]
Abstract
Microglial cells, or brain immune cells, are highly dynamic and continuously migrate in pathophysiological conditions. Their adhesion, as a physical characteristic, plays a key role in migration. In this study, we presented a microfluidic chip combination of two assays: a microglial BV2 adhesion assay and a wound-healing migration assay. The chip could create the cell-free area (wound) under chemical stimuli with trypsin (chemical assay) and also mechanical stimuli with the PBS flow (mechanical assay). The microfluidic chip functioned as the cell adhesion assay during wounding, when the cell adhesion of microglia BV2 cells was characterized by the cell removal time under various shear stress ranges. The cell detachment pattern on the glass substrate was found under physiological conditions. After wounding, the chip operated as a migration assay; it was shown that cell migration in the cell-free area generated chemically with trypsin was highly improved compared to mechanical cell-free area creations with PBS flow and the scratch assay. Our findings indicated that the increase in inlet flow rate in the mechanical assay led to a reduced experiment time and mechanical force on the cells, which could improve cell migration. Furthermore, the study on the effect of the device geometry showed that the increased channel width had an inhibitory effect on cell migration. The bi-functional chip offers an opportunity for the development of new models for a better understanding of cellular adhesion and migration in in vitro microenvironments.
Collapse
Affiliation(s)
- Ehsan Yazdanpanah Moghadam
- Optical-Bio Microsystems Laboratory, Micro-Nano-Bio Integration Center, Department of Mechanical and Industrial Engineering, Concordia University, Montreal, QC H3G 1M8, Canada;
- Department of Biochemistry, Goodman Cancer Research Center, McGill University, Montreal, QC H3G 1Y62, Canada;
| | - Nahum Sonenberg
- Department of Biochemistry, Goodman Cancer Research Center, McGill University, Montreal, QC H3G 1Y62, Canada;
| | - Muthukumaran Packirisamy
- Optical-Bio Microsystems Laboratory, Micro-Nano-Bio Integration Center, Department of Mechanical and Industrial Engineering, Concordia University, Montreal, QC H3G 1M8, Canada;
| |
Collapse
|
2
|
Cho K, Lee J, Kim J. Integrated high-throughput drug screening microfluidic system for comprehensive ocular toxicity assessment. Toxicol In Vitro 2024; 98:105843. [PMID: 38735502 DOI: 10.1016/j.tiv.2024.105843] [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: 02/28/2024] [Revised: 04/29/2024] [Accepted: 05/08/2024] [Indexed: 05/14/2024]
Abstract
Traditional experimental methodologies suffer from a few limitations in the toxicological evaluation of the preservatives added to eye drops. In this study, we overcame these limitations by using a microfluidic device. We developed a microfluidic system featuring a gradient concentration generator for preservative dosage control with microvalves and micropumps, automatically regulated by a programmable Arduino board. This system facilitated the simultaneous toxicological evaluation of human corneal epithelial cells against eight different concentrations of preservatives, allowing for quadruplicate experiments in a single run. In our study, the IC50 values for healthy eyes and those affected with dry eyes syndrome showed an approximately twofold difference. This variation is likely attributable to the duration for which the preservative remained in contact with corneal cells before being washed off by the medium, suggesting the significance of exposure time in the cytotoxic effect of preservatives. Our microfluidic system, automated by Arduino, simulated healthy and dry eye environments to study benzalkonium chloride toxicity and revealed significant differences in cell viability, with IC50 values of 0.0033% for healthy eyes and 0.0017% for dry eyes. In summary, we implemented the pinch-to-zoom feature of an electronic tablet in our microfluidic system, offering innovative alternatives for eye research.
Collapse
Affiliation(s)
- Kyongjin Cho
- Dept. of Ophthalmology, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Jinho Lee
- Research Institute of Natural Science and Department of Physics Education, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Jeongyun Kim
- Dept. of Physics, College of Natural Science, Dankook University, Cheonan 31116, Republic of Korea.
| |
Collapse
|
3
|
A Cataño J, Farthing S, Mascarenhas Z, Lake N, Yarlagadda PKDV, Li Z, Toh YC. A User-Centric 3D-Printed Modular Peristaltic Pump for Microfluidic Perfusion Applications. MICROMACHINES 2023; 14:mi14050930. [PMID: 37241553 DOI: 10.3390/mi14050930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/17/2023] [Accepted: 04/23/2023] [Indexed: 05/28/2023]
Abstract
Microfluidic organ-on-a-chip (OoC) technology has enabled studies on dynamic physiological conditions as well as being deployed in drug testing applications. A microfluidic pump is an essential component to perform perfusion cell culture in OoC devices. However, it is challenging to have a single pump that can fulfil both the customization function needed to mimic a myriad of physiological flow rates and profiles found in vivo and multiplexing requirements (i.e., low cost, small footprint) for drug testing operations. The advent of 3D printing technology and open-source programmable electronic controllers presents an opportunity to democratize the fabrication of mini-peristaltic pumps suitable for microfluidic applications at a fraction of the cost of commercial microfluidic pumps. However, existing 3D-printed peristaltic pumps have mainly focused on demonstrating the feasibility of using 3D printing to fabricate the structural components of the pump and neglected user experience and customization capability. Here, we present a user-centric programmable 3D-printed mini-peristaltic pump with a compact design and low manufacturing cost (~USD 175) suitable for perfusion OoC culture applications. The pump consists of a user-friendly, wired electronic module that controls the operation of a peristaltic pump module. The peristaltic pump module comprises an air-sealed stepper motor connected to a 3D-printed peristaltic assembly, which can withstand the high-humidity environment of a cell culture incubator. We demonstrated that this pump allows users to either program the electronic module or use different-sized tubing to deliver a wide range of flow rates and flow profiles. The pump also has multiplexing capability as it can accommodate multiple tubing. The performance and user-friendliness of this low-cost, compact pump can be easily deployed for various OoC applications.
Collapse
Affiliation(s)
- Jorge A Cataño
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane 4000, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Kelvin Grove 4059, Australia
| | - Steven Farthing
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane 4000, Australia
| | - Zeus Mascarenhas
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane 4000, Australia
| | - Nathaniel Lake
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane 4000, Australia
| | - Prasad K D V Yarlagadda
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane 4000, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Kelvin Grove 4059, Australia
- School of Engineering, University of Southern Queensland, Springfield Central 4300, Australia
| | - Zhiyong Li
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane 4000, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Kelvin Grove 4059, Australia
| | - Yi-Chin Toh
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane 4000, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Kelvin Grove 4059, Australia
- Max Planck Queensland Centre (MPQC) for the Materials Science of Extracellular Matrices, Queensland University of Technology, Kelvin Grove 4059, Australia
- Centre for Microbiome Research, Queensland University of Technology, Woolloongabba 4102, Australia
| |
Collapse
|
4
|
Yazdanpanah Moghadam E, Sonenberg N, Packirisamy M. Microfluidic Wound-Healing Assay for ECM and Microenvironment Properties on Microglia BV2 Cells Migration. BIOSENSORS 2023; 13:290. [PMID: 36832056 PMCID: PMC9954450 DOI: 10.3390/bios13020290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/12/2023] [Accepted: 02/14/2023] [Indexed: 06/18/2023]
Abstract
Microglia cells, as the resident immune cells of the central nervous system (CNS), are highly motile and migratory in development and pathophysiological conditions. During their migration, microglia cells interact with their surroundings based on the various physical and chemical properties in the brain. Herein, a microfluidic wound-healing chip is developed to investigate microglial BV2 cell migration on the substrates coated with extracellular matrixes (ECMs) and substrates usually used for bio-applications on cell migration. In order to generate the cell-free space (wound), gravity was utilized as a driving force to flow the trypsin with the device. It was shown that, despite the scratch assay, the cell-free area was created without removing the extracellular matrix coating (fibronectin) using the microfluidic assay. It was found that the substrates coated with Poly-L-Lysine (PLL) and gelatin stimulated microglial BV2 migration, while collagen and fibronectin coatings had an inhibitory effect compared to the control conditions (uncoated glass substrate). In addition, the results showed that the polystyrene substrate induced higher cell migration than the PDMS and glass substrates. The microfluidic migration assay provides an in vitro microenvironment closer to in vivo conditions for further understanding the microglia migration mechanism in the brain, where the environment properties change under homeostatic and pathological conditions.
Collapse
Affiliation(s)
- Ehsan Yazdanpanah Moghadam
- Optical-Bio Microsystems Laboratory, Micro-Nano-Bio Integration Center, Department of Mechanical and Industrial Engineering, Concordia University, Montreal, QC H3G 1M8, Canada
- Department of Biochemistry, Goodman Cancer Research Center, McGill University, Montreal, QC H3A 1A3, Canada
| | - Nahum Sonenberg
- Department of Biochemistry, Goodman Cancer Research Center, McGill University, Montreal, QC H3A 1A3, Canada
| | - Muthukumaran Packirisamy
- Optical-Bio Microsystems Laboratory, Micro-Nano-Bio Integration Center, Department of Mechanical and Industrial Engineering, Concordia University, Montreal, QC H3G 1M8, Canada
| |
Collapse
|
5
|
Ahmad N. In Vitro and In Vivo Characterization Methods for Evaluation of Modern Wound Dressings. Pharmaceutics 2022; 15:42. [PMID: 36678671 PMCID: PMC9864730 DOI: 10.3390/pharmaceutics15010042] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/10/2022] [Accepted: 12/17/2022] [Indexed: 12/25/2022] Open
Abstract
Chronic wound management represents a major challenge in the healthcare sector owing to its delayed wound-healing process progression and huge financial burden. In this regard, wound dressings provide an appropriate platform for facilitating wound healing for several decades. However, adherent traditional wound dressings do not provide effective wound healing for highly exudating chronic wounds and need the development of newer and innovative wound dressings to facilitate accelerated wound healing. In addition, these dressings need frequent changing, resulting in more pain and discomfort. In order to overcome these issues, a wide range of affordable and innovative modern wound dressings have been developed and explored recently to accelerate and improve the wound healing process. However, a comprehensive understanding of various in vitro and in vivo characterization methods being utilized for the evaluation of different modern wound dressings is lacking. In this context, an overview of modern dressings and their complete in vitro and in vivo characterization methods for wound healing assessment is provided in this review. Herein, various emerging modern wound dressings with advantages and challenges have also been reviewed. Furthermore, different in vitro wound healing assays and in vivo wound models being utilized for the evaluation of wound healing progression and wound healing rate using wound dressings are discussed in detail. Finally, a summary of modern wound dressings with challenges and the future outlook is highlighted.
Collapse
Affiliation(s)
- Naveed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka 72388, Aljouf, Saudi Arabia
| |
Collapse
|
6
|
CircLDLR Modulates the Proliferation and Apoptosis of Vascular Smooth Muscle Cells in Coronary Artery Disease Through miR-26-5p/KDM6A Axis. J Cardiovasc Pharmacol 2022; 80:132-139. [PMID: 35384910 DOI: 10.1097/fjc.0000000000001275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 03/17/2022] [Indexed: 11/25/2022]
Abstract
ABSTRACT The purpose of this study was to investigate the effect of circLDLR on the proliferation and apoptosis of vascular smooth muscle cells (VSMCs) in coronary artery disease and its regulatory mechanism. The expression of KDM6A was detected by qRT-PCR or Western blot. VSMCs were transfected with miR-26-5p mimic/inhibitor or OE KDM6A. Cell proliferation and apoptosis were assessed. Luciferase reporter gene assays were used to examine interactions between miR-26-5p and KDM6A in VSMCs. Downregulation of circLDLR was associated with increased miR-26-5p in coronary artery disease tissues. In addition, circLDLR could inhibit cell proliferation and promote cell apoptosis by regulating miR-26-5p. Moreover, the overexpression of KDM6A reduced VSMCs proliferation and increased apoptosis in an miR-26-5p/circLDLR axis-dependent manner. CircLDLR modulates the proliferation and apoptosis of VSMCs through miR-26-5p/KDM6A axis.
Collapse
|
7
|
Improved mechanical, degradation, and biological performances of Zn–Fe alloys as bioresorbable implants. Bioact Mater 2021; 17:334-343. [PMID: 35386444 PMCID: PMC8965087 DOI: 10.1016/j.bioactmat.2021.12.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/07/2021] [Accepted: 12/24/2021] [Indexed: 01/07/2023] Open
Abstract
Zinc (Zn) is a promising bioresorbable implant material with more moderate degradation rate compared to magnesium (Mg) and iron (Fe). However, the low mechanical strength and localized degradation behavior of pure Zn limit its clinical applications. Alloying is one of the most effective ways to overcome these limitations. After screening the alloying element candidates regarding their potentials for improvement on the degradation and biocompatibility, we proposed Fe as the alloying element for Zn, and investigated the in vitro and in vivo performances of these alloys in both subcutaneous and femoral tissues. Results showed that the uniformly distributed secondary phase in Zn–Fe alloys significantly improved the mechanical property and facilitated uniform degradation, which thus enhanced their biocompatibility, especially the Zn-0.4Fe alloy. Moreover, these Zn–Fe alloys showed outstanding antibacterial property. Taken together, Zn–Fe alloys could be promising candidates as bioresorbable medical implants for various cardiovascular, wound closure, and orthopedic applications. Zn-0.4Fe alloy significantly improved the mechanical strength and ductility. Zn-0.4Fe alloy showed a uniform degradation behavior. Subcutaneous and femoral implantations to compare in vivo performances. Zn-0.4Fe alloy showed superior hemocompatibility and in vivo osteointegration.
Collapse
|
8
|
In vitro methods used for discovering plant derived products as wound healing agents - An update on the cell types and rationale. Fitoterapia 2021; 154:105026. [PMID: 34480992 DOI: 10.1016/j.fitote.2021.105026] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/27/2021] [Accepted: 08/29/2021] [Indexed: 01/13/2023]
Abstract
Wounds still pose a huge burden on human health and healthcare systems in many parts of the world. Phytomedicines are being used to heal the wounds since ancient times. Now-a-days also many researchers are exploring the wound healing activity of phytomedicines. Wound healing is a complex process thus, it is always a question mark regarding the best test model (in vivo, ex vivo and in vitro) model to assess the wound healing activity of phytomedicines. In general, the researchers would opt for in vivo model - probably because of closer physiological relevance to human wounds. However, in vivo experimental models are not suitable for high throughput screening and not ethical in terms of initial screening of the phytomedicines. The in vivo models are associated with difficulties in obtaining the ethical approvals, requires huge budget, and resources. We argue that judicious selection of cell types would serve the purpose of developing a physiologically relevant in vitro experimental model. A lot of progress has been made in molecular biology techniques to bridge the gap between in vitro models and their physiological relevance. The in vitro models are the best suited for high throughput screening and to elucidate the molecular mechanisms. The main aim of this review is to provide insights on selection of the cell types for developing physiologically relevant in vitro wound healing assays, which can be used to improve the value of phytomedicines further.
Collapse
|
9
|
Höving AL, Schmitz J, Schmidt KE, Greiner JFW, Knabbe C, Kaltschmidt B, Grünberger A, Kaltschmidt C. Human Blood Serum Induces p38-MAPK- and Hsp27-Dependent Migration Dynamics of Adult Human Cardiac Stem Cells: Single-Cell Analysis via a Microfluidic-Based Cultivation Platform. BIOLOGY 2021; 10:biology10080708. [PMID: 34439941 PMCID: PMC8389316 DOI: 10.3390/biology10080708] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/16/2021] [Accepted: 07/21/2021] [Indexed: 12/14/2022]
Abstract
Simple Summary Adult human stem cells possess the ability to contribute to endogenous regeneration processes of injured tissue by migrating to specific locations. For stem cell-based clinical applications it is highly important to gain knowledge about the migration behavior of adult human stem cells and the underlying molecular mechanisms of this ability. Human blood serum has been shown to have beneficial effects on other regenerative capacities of adult human stem cells. Within this study we tested the effect of human blood serum on the migration behavior of stem cells from the human heart. We used a microfluidic cultivation device, which allowed us to monitor the living cells and their movement behavior in real time. After addition of human blood serum, the heart stem cells increased their speed of movement and covered distance. Further, we observed that this effect could be diminished by inhibition of a specific kinase, p38-MAPK. Thus, our data suggest beneficial effects of human blood serum on adult human heart stem cells dependent on p38-MAPK. Our study contributes to a deeper understanding of the dynamics of stem cell migration and introduces a new platform to monitor stem cell movement in real time. Abstract Migratory capabilities of adult human stem cells are vital for assuring endogenous tissue regeneration and stem cell-based clinical applications. Although human blood serum has been shown to be beneficial for cell migration and proliferation, little is known about its impact on the migratory behavior of cardiac stem cells and underlying signaling pathways. Within this study, we investigated the effects of human blood serum on primary human cardiac stem cells (hCSCs) from the adult heart auricle. On a technical level, we took advantage of a microfluidic cultivation platform, which allowed us to characterize cell morphologies and track migration of single hCSCs via live cell imaging over a period of up to 48 h. Our findings showed a significantly increased migration distance and speed of hCSCs after treatment with human serum compared to control. Exposure of blood serum-stimulated hCSCs to the p38 mitogen-activated protein kinase (p38-MAPK) inhibitor SB239063 resulted in significantly decreased migration. Moreover, we revealed increased phosphorylation of heat shock protein 27 (Hsp27) upon serum treatment, which was diminished by p38-MAPK-inhibition. In summary, we demonstrate human blood serum as a strong inducer of adult human cardiac stem cell migration dependent on p38-MAPK/Hsp27-signalling. Our findings further emphasize the great potential of microfluidic cultivation devices for assessing spatio-temporal migration dynamics of adult human stem cells on a single-cell level.
Collapse
Affiliation(s)
- Anna L. Höving
- Department of Cell Biology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany; (K.E.S.); (J.F.W.G.); (B.K.); (C.K.)
- Heart and Diabetes Centre NRW, Institute for Laboratory and Transfusion Medicine, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany;
- Correspondence:
| | - Julian Schmitz
- Multiscale Bioengineering, Faculty of Technology, Bielefeld University, 33615 Bielefeld, Germany; (J.S.); (A.G.)
| | - Kazuko E. Schmidt
- Department of Cell Biology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany; (K.E.S.); (J.F.W.G.); (B.K.); (C.K.)
- Heart and Diabetes Centre NRW, Institute for Laboratory and Transfusion Medicine, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany;
| | - Johannes F. W. Greiner
- Department of Cell Biology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany; (K.E.S.); (J.F.W.G.); (B.K.); (C.K.)
| | - Cornelius Knabbe
- Heart and Diabetes Centre NRW, Institute for Laboratory and Transfusion Medicine, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany;
| | - Barbara Kaltschmidt
- Department of Cell Biology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany; (K.E.S.); (J.F.W.G.); (B.K.); (C.K.)
- Molecular Neurobiology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany
| | - Alexander Grünberger
- Multiscale Bioengineering, Faculty of Technology, Bielefeld University, 33615 Bielefeld, Germany; (J.S.); (A.G.)
| | - Christian Kaltschmidt
- Department of Cell Biology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany; (K.E.S.); (J.F.W.G.); (B.K.); (C.K.)
| |
Collapse
|
10
|
Microfluidic and Lab-on-a-Chip Systems for Cutaneous Wound Healing Studies. Pharmaceutics 2021; 13:pharmaceutics13060793. [PMID: 34073346 PMCID: PMC8228894 DOI: 10.3390/pharmaceutics13060793] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/19/2021] [Accepted: 05/24/2021] [Indexed: 12/26/2022] Open
Abstract
Cutaneous wound healing is a complex, multi-stage process involving direct and indirect cell communication events with the aim of efficiently restoring the barrier function of the skin. One key aspect in cutaneous wound healing is associated with cell movement and migration into the physically, chemically, and biologically injured area, resulting in wound closure. Understanding the conditions under which cell migration is impaired and elucidating the cellular and molecular mechanisms that improve healing dynamics are therefore crucial in devising novel therapeutic strategies to elevate patient suffering, reduce scaring, and eliminate chronic wounds. Following the global trend towards the automation, miniaturization, and integration of cell-based assays into microphysiological systems, conventional wound healing assays such as the scratch assay and cell exclusion assay have recently been translated and improved using microfluidics and lab-on-a-chip technologies. These miniaturized cell analysis systems allow for precise spatial and temporal control over a range of dynamic microenvironmental factors including shear stress, biochemical and oxygen gradients to create more reliable in vitro models that resemble the in vivo microenvironment of a wound more closely on a molecular, cellular, and tissue level. The current review provides (a) an overview on the main molecular and cellular processes that take place during wound healing, (b) a brief introduction into conventional in vitro wound healing assays, and (c) a perspective on future cutaneous and vascular wound healing research using microfluidic technology.
Collapse
|
11
|
Pu Y, Gingrich J, Veiga-Lopez A. A 3-dimensional microfluidic platform for modeling human extravillous trophoblast invasion and toxicological screening. LAB ON A CHIP 2021; 21:546-557. [PMID: 33166377 PMCID: PMC8212566 DOI: 10.1039/d0lc01013h] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Placental trophoblast cells invasion into the maternal uterus is an essential and complex event in the formation of the maternal-fetal interface. Commonly used two-dimensional (2D) cell invasion tools do not accurately represent the in vivo cell invasion microenvironment. Three-dimensional (3D) silicone polymer polydimethylsiloxane (PDMS) microfluidic platforms are an emerging technology in developing organ-on-a-chip models. Here, we present a placenta-on-a-chip platform that enables the evaluation of trophoblast invasion with intraluminal flow within an engineered PDMS 3D microfluidic chip. This platform reproduces key elements of the placental microenvironment, including endothelial and trophoblast cells, layered with an extracellular matrix, and incorporates dynamic medium flow while allowing for real-time monitoring, imaging, evaluation of trophoblast cell invasion, and heterocellular cell-to-cell interactions. Coupled with fluorescent cell tagging and flow cytometry, this platform also allows collection of the invasive cells. This will help our understanding of pathways that regulate trophoblast cell invasion and may prove important for toxicological screening of exposures that interfere with invasiveness in a complex organ such as the placenta.
Collapse
Affiliation(s)
- Yong Pu
- Department of Pathology, University of Illinois at Chicago, 909 S. Wolcott Ave, Rm 6093, Chicago, IL 60612, USA.
| | - Jeremy Gingrich
- Department of Pharmacology and Toxicology, Michigan State University, USA
| | - Almudena Veiga-Lopez
- Department of Pathology, University of Illinois at Chicago, 909 S. Wolcott Ave, Rm 6093, Chicago, IL 60612, USA.
| |
Collapse
|
12
|
Soitu C, Panea M, Castrejón-Pita AA, Cook PR, Walsh EJ. Creating wounds in cell monolayers using micro-jets. BIOMICROFLUIDICS 2021; 15:014108. [PMID: 33598064 PMCID: PMC7872715 DOI: 10.1063/5.0043312] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 01/26/2021] [Indexed: 06/12/2023]
Abstract
Many wound-healing assays are used in cell biology and biomedicine; they are often labor intensive and/or require specialized and costly equipment. We describe a contactless method to create wounds with any imaginable 2D pattern in cell monolayers using the micro-jets of either media or an immiscible and biocompatible fluorocarbon (i.e., FC40). We also combine this with another method that allows automation and multiplexing using standard Petri dishes. A dish is filled with a thin film of media overlaid with FC40, and the two liquids are reshaped into an array of microchambers within minutes. Each chamber in such a grid is isolated from others by the fluid walls of FC40. Cells are now added, allowed to grow into a monolayer, and wounds are created using the microjets; then, healing is monitored by microscopy. As arrays of chambers can be made using media and Petri dishes familiar to biologists, and as dishes fit seamlessly into their incubators, microscopes, and workflows, we anticipate that this assay will find wide application in wound healing.
Collapse
Affiliation(s)
- Cristian Soitu
- Osney Thermofluids Institute, Department of Engineering Science, University of Oxford, Osney Mead, Oxford OX2 0ES, United Kingdom
| | - Mirela Panea
- Neurosciences Group, Nuffield Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford OX3 9DS, United Kingdom
| | | | - Peter R. Cook
- The Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, United Kingdom
| | - Edmond J. Walsh
- Osney Thermofluids Institute, Department of Engineering Science, University of Oxford, Osney Mead, Oxford OX2 0ES, United Kingdom
| |
Collapse
|
13
|
Deal HE, Brown AC, Daniele MA. Microphysiological systems for the modeling of wound healing and evaluation of pro-healing therapies. J Mater Chem B 2020; 8:7062-7075. [PMID: 32756718 PMCID: PMC7460719 DOI: 10.1039/d0tb00544d] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Wound healing is a multivariate process involving the coordinated response of numerous proteins and cell types. Accordingly, biomedical research has seen an increased adoption of the use of in vitro wound healing assays with complexity beyond that offered by traditional well-plate constructs. These microphysiological systems (MPS) seek to recapitulate one or more physiological features of the in vivo microenvironment, while retaining the analytical capacity of more reductionist assays. Design efforts to achieve relevant wound healing physiology include the use of dynamic perfusion over static culture, the incorporation of multiple cell types, the arrangement of cells in three dimensions, the addition of biomechanically and biochemically relevant hydrogels, and combinations thereof. This review provides a brief overview of the wound healing process and in vivo assays, and we critically review the current state of MPS and supporting technologies for modelling and studying wound healing. We distinguish between MPS that seek to inform a particular phase of wound healing, and constructs that have the potential to inform multiple phases of wound healing. This distinction is a product of whether analysis of a particular process is prioritized, or a particular physiology is prioritized, during design. Material selection is emphasized throughout, and relevant fabrication techniques discussed.
Collapse
Affiliation(s)
- Halston E Deal
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA. and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA
| | - Ashley C Brown
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA. and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA
| | - Michael A Daniele
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA. and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA and Department of Electrical & Computer Engineering, North Carolina State University, 890 Oval Dr., Raleigh, NC 27695, USA
| |
Collapse
|
14
|
Tong A, Pham QL, Shah V, Naik A, Abatemarco P, Voronov R. Automated Addressable Microfluidic Device for Minimally Disruptive Manipulation of Cells and Fluids within Living Cultures. ACS Biomater Sci Eng 2020; 6:1809-1820. [DOI: 10.1021/acsbiomaterials.9b01969] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Anh Tong
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark College of Engineering, 161 Warren Street, Newark, New Jersey 07102, United States
| | - Quang Long Pham
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark College of Engineering, 161 Warren Street, Newark, New Jersey 07102, United States
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Vatsal Shah
- Department of Computer Science, Ying Wu College of Computing Sciences, New Jersey Institute of Technology, Newark College of Engineering, Suite 3500, University Heights, Newark, New Jersey 07102, United States
- Federated Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark College of Engineering, Suite 204, University Heights, Newark, New Jersey 07102, United States
| | - Akshay Naik
- Helen and John C. Hartmann Department of Electrical and Computer Engineering, New Jersey Institute of Technology, Newark College of Engineering, Suite 200, University Heights, Newark, New Jersey 07102, United States
| | - Paul Abatemarco
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark College of Engineering, 161 Warren Street, Newark, New Jersey 07102, United States
| | - Roman Voronov
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark College of Engineering, 161 Warren Street, Newark, New Jersey 07102, United States
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark College of Engineering, 323 Dr. Martin Luther King Jr. Boulevard, Newark, New Jersey 07103, United States
| |
Collapse
|
15
|
Fu J, Su Y, Qin YX, Zheng Y, Wang Y, Zhu D. Evolution of metallic cardiovascular stent materials: A comparative study among stainless steel, magnesium and zinc. Biomaterials 2020; 230:119641. [PMID: 31806406 PMCID: PMC6934082 DOI: 10.1016/j.biomaterials.2019.119641] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 11/16/2019] [Accepted: 11/19/2019] [Indexed: 12/22/2022]
Abstract
A cardiovascular stent is a small mesh tube that expands a narrowed or blocked coronary artery. Unfortunately, current stents, regardless metallic or polymeric, still largely fall short to the ideal clinical needs due to late restenosis, thrombosis and other clinical complications. Nonetheless, metallic stents are preferred clinically thanks to their superior mechanical property and radiopacity to their polymeric counterparts. The emergence of bioresorbable metals opens a window for better stent materials as they may have the potential to reduce or eliminate late restenosis and thrombosis. In fact, some bioresorbable magnesium (Mg)-based stents have obtained regulatory approval or under trials with mixed clinical outcomes. Some major issues with Mg include the too rapid degradation rate and late restenosis. To mitigate these problems, bioresorbable zinc (Zn)-based stent materials are being developed lately with the more suitable degradation rate and better biocompatibility. The past decades have witnessed the unprecedented evolution of metallic stent materials from first generation represented by stainless steel (SS), to second generation represented by Mg, and to third generation represented by Zn. To further elucidate their pros and cons as metallic stent materials, we systematically evaluated their performances in vitro and in vivo through direct side-by-side comparisons. Our results demonstrated that tailored Zn-based material with proper configurations could be a promising candidate for a better stent material in the future.
Collapse
Affiliation(s)
- Jiayin Fu
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Yingchao Su
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Yi-Xian Qin
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Yufeng Zheng
- Department of Materials Science and Engineering, College of Engineering, Peking University, Beijing, China
| | - Yadong Wang
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
| | - Donghui Zhu
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
16
|
A microfluidics-based wound-healing assay for studying the effects of shear stresses, wound widths, and chemicals on the wound-healing process. Sci Rep 2019; 9:20016. [PMID: 31882962 PMCID: PMC6934480 DOI: 10.1038/s41598-019-56753-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/30/2019] [Indexed: 01/22/2023] Open
Abstract
Collective cell migration plays important roles in various physiological processes. To investigate this collective cellular movement, various wound-healing assays have been developed. In these assays, a “wound” is created mechanically, chemically, optically, or electrically out of a cellular monolayer. Most of these assays are subject to drawbacks of run-to-run variations in wound size/shape and damages to cells/substrate. Moreover, in all these assays, cells are cultured in open, static (non-circulating) environments. In this study, we reported a microfluidics-based wound-healing assay by using the trypsin flow-focusing technique. Fibroblasts were first cultured inside this chip to a cellular monolayer. Then three parallel fluidic flows (containing normal medium and trypsin solution) were introduced into the channels, and cells exposed to protease trypsin were enzymatically detached from the surface. Wounds of three different widths were generated, and subsequent wound-healing processes were observed. This assay is capable of creating three or more wounds of different widths for investigating the effects of various physical and chemical stimuli on wound-healing speeds. The effects of shear stresses, wound widths, and β-lapachone (a wound healing-promoting chemical) on wound-healing speeds were studied. It was found that the wound-healing speed (total area healed per unit time) increased with increasing shear stress and wound width, but under a shear stress of 0.174 mPa the linear healing speed (percent area healed per unit time) was independent of the wound width. Also, the addition of β-lapachone up to 0.5 μM did not accelerate wound healing. This microfluidics-based assay can definitely help in understanding the mechanisms of the wound-healing process and developing new wound-healing therapies.
Collapse
|
17
|
Soitu C, Feuerborn A, Deroy C, Castrejón-Pita AA, Cook PR, Walsh EJ. Raising fluid walls around living cells. SCIENCE ADVANCES 2019; 5:eaav8002. [PMID: 31183401 PMCID: PMC6551168 DOI: 10.1126/sciadv.aav8002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 04/29/2019] [Indexed: 05/04/2023]
Abstract
An effective transformation of the cell culture dishes that biologists use every day into microfluidic devices would open many avenues for miniaturizing cell-based workflows. In this article, we report a simple method for creating microfluidic arrangements around cells already growing on the surface of standard petri dishes, using the interface between immiscible fluids as a "building material." Conventional dishes are repurposed into sophisticated microfluidic devices by reshaping, on demand, the fluid structures around living cells. Moreover, these microfluidic arrangements can be further reconfigured during experiments, which is impossible with most existing microfluidic platforms. The method is demonstrated using workflows involving cell cloning, the selection of a particular clone from among others in a dish, drug treatments, and wound healing. The versatility of the approach and its biologically friendly aspects may hasten uptake by biologists of microfluidics, so the technology finally fulfills its potential.
Collapse
Affiliation(s)
- Cristian Soitu
- Oxford Thermofluids Institute, Department of Engineering Science, University of Oxford, Osney Mead, Oxford OX2 0ES, UK
| | - Alexander Feuerborn
- The Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
- Iota Sciences Ltd., Begbroke Science Park, Begbroke, Oxfordshire OX5 1PF, UK
| | - Cyril Deroy
- Oxford Thermofluids Institute, Department of Engineering Science, University of Oxford, Osney Mead, Oxford OX2 0ES, UK
| | | | - Peter R. Cook
- The Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Edmond J. Walsh
- Oxford Thermofluids Institute, Department of Engineering Science, University of Oxford, Osney Mead, Oxford OX2 0ES, UK
- Iota Sciences Ltd., Begbroke Science Park, Begbroke, Oxfordshire OX5 1PF, UK
- Corresponding author.
| |
Collapse
|
18
|
Ishida T, Shimamoto T, Kaminaga M, Kuchimaru T, Kizaka-Kondoh S, Omata T. Microfluidic High-Migratory Cell Collector Suppressing Artifacts Caused by Microstructures. MICROMACHINES 2019; 10:E116. [PMID: 30754704 PMCID: PMC6412487 DOI: 10.3390/mi10020116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/01/2019] [Accepted: 02/07/2019] [Indexed: 12/22/2022]
Abstract
The small number of high-migratory cancer cells in a cell population make studies on high-migratory cancer cells difficult. For the development of migration assays for such cancer cells, several microfluidic devices have been developed. However, they measure migration that is influenced by microstructures and they collect not only high-migratory cells, but also surrounding cells. In order to find high-migratory cells in cell populations while suppressing artifacts and to collect these cells while minimizing damages, we developed a microfluidic high-migratory cell collector with the ability to sort cancer cells according to cellular migration and mechanical detachment. High-migratory cancer cells travel further from the starting line when all of the cells are seeded on the same starting line. The high-migratory cells are detached using a stretch of cell adhesive surface using a water-driven balloon actuator. Using this cell collector, we selected high-migratory HeLa cells that migrated about 100m in 12 h and collected the cells.
Collapse
Affiliation(s)
- Tadashi Ishida
- Department of Mechanical Engineering, School of Engineering, Tokyo Institute of Technology, Kanagawa 226-8503, Japan.
- Department of Mechano-Micro Engineering, Interdisciplinary Graduate School of Science and Engineering, Tokyo Institute of Technology, Kanagawa 226-8503, Japan.
| | - Takuya Shimamoto
- Department of Mechano-Micro Engineering, Interdisciplinary Graduate School of Science and Engineering, Tokyo Institute of Technology, Kanagawa 226-8503, Japan.
| | - Maho Kaminaga
- Department of Mechanical Engineering, School of Engineering, Tokyo Institute of Technology, Kanagawa 226-8503, Japan.
| | - Takahiro Kuchimaru
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa 226-8503, Japan.
| | - Shinae Kizaka-Kondoh
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa 226-8503, Japan.
| | - Toru Omata
- Department of Mechanical Engineering, School of Engineering, Tokyo Institute of Technology, Kanagawa 226-8503, Japan.
- Department of Mechano-Micro Engineering, Interdisciplinary Graduate School of Science and Engineering, Tokyo Institute of Technology, Kanagawa 226-8503, Japan.
| |
Collapse
|
19
|
Roveimiab Z, Lin F, Anderson JE. Emerging Development of Microfluidics-Based Approaches to Improve Studies of Muscle Cell Migration. TISSUE ENGINEERING PART B-REVIEWS 2018; 25:30-45. [PMID: 30073911 DOI: 10.1089/ten.teb.2018.0181] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
IMPACT STATEMENT The essential interactions between and among cells in the three types of muscle tissue in development, wound healing, and regeneration of tissues, are underpinned by the ability of cardiac, smooth, and skeletal muscle cells to migrate in maintaining functional capacity after pathologies such as myocardial infarction, tissue grafting, and traumatic and postsurgical injury. Microfluidics-based devices now offer significant enhancement over conventional approaches to studying cell chemotaxis and haptotaxis that are inherent in migration. Advances in experimental approaches to muscle cell movement and tissue formation will contribute to innovations in tissue engineering for patching wound repair and muscle tissue replacement.
Collapse
Affiliation(s)
- Ziba Roveimiab
- 1 Department of Biological Sciences and University of Manitoba, Winnipeg, Canada.,2 Department of Physics and Astronomy, University of Manitoba, Winnipeg, Canada
| | - Francis Lin
- 1 Department of Biological Sciences and University of Manitoba, Winnipeg, Canada.,2 Department of Physics and Astronomy, University of Manitoba, Winnipeg, Canada
| | - Judy E Anderson
- 1 Department of Biological Sciences and University of Manitoba, Winnipeg, Canada
| |
Collapse
|
20
|
Qiu H, Shi S, Wang S, Peng H, Ding SJ, Wang L. Proteomic Profiling Exosomes from Vascular Smooth Muscle Cell. Proteomics Clin Appl 2018; 12:e1700097. [PMID: 29687628 PMCID: PMC6298740 DOI: 10.1002/prca.201700097] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 02/26/2018] [Indexed: 01/29/2023]
Abstract
PURPOSE Vascular smooth muscle cells (VSMC) and endothelial cells (EC) communicate mutually to coordinate vascular development and homeostasis. Exosomes are emerging as one type of the mediators involved in this communication. Characterizing proteins in the exosomes is the critical first step in understanding how the VSMC-EC crosstalk is mediated by exosomes. EXPERIMENTAL DESIGN The proteins in the human VSMC-derived exosomes are profiled using nanoLC-MS/MS based proteomics. The identified proteins are subjected to gene ontology analysis. The VSMC-derived exosomes are also assessed for proangiogenic activity in vivo. RESULTS Four hundred and fifty-nine proteins are identified in the VSMC-derived exosomes. Gene ontology analysis revealed that the exosome proteins are involved in 179 cellular components, 120 molecular functions, and 337 biological processes, with cell-cell adhesion and platelet activation/coagulation ranked at the top. VSMC-derived exosomes do not display a proangiogenic activity in the in vivo angiogenesis assay, suggesting that the major function of VSMC-derived exosomes is to maintain vessel homeostasis. CONCLUSION AND CLINICAL RELEVANCE The analyses obtained a systematic view of proteins in the VSMC-derived exosomes, revealed the potential regulatory functions of the exosome in VSMC-EC communication, and suggest that dysregulation of VSMC-derived exosome-mediated functions may disturb vessel homeostasis thereby contributing to vascular diseases.
Collapse
Affiliation(s)
- Hong Qiu
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, 30602, USA
| | - Songshan Shi
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, 30602, USA
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P. R. China
| | - Shunchun Wang
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, 30602, USA
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P. R. China
| | - Hong Peng
- Department of Pathology and Microbiology, Mass Spectrometry and Proteomics Core Facility, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Shi-Jian Ding
- Department of Pathology and Microbiology, Mass Spectrometry and Proteomics Core Facility, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Lianchun Wang
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, 30602, USA
| |
Collapse
|
21
|
Luan S, Hao R, Wei Y, Chen D, Fan B, Dong F, Guo W, Wang J, Chen J. A microfabricated 96-well wound-healing assay. Cytometry A 2017; 91:1192-1199. [PMID: 29156109 DOI: 10.1002/cyto.a.23286] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/23/2017] [Accepted: 11/01/2017] [Indexed: 01/09/2023]
Abstract
This article presents a microfabricated 96-well wound-healing assay enabling high-throughput measurement of cellular migration capabilities. Within each well, the middle area is the wound region, made of microfabricated gold surface with self-assembled PEG repellent for cell seeding. After the formation of a cellular confluent monolayer around the wound region, collagen solution was applied to form three-dimensional matrix to cover the PEG surface, initiating the wound-healing process. By interpreting the numbers of migrated cells into the wound regions as a function of specific stimuli with different concentrations, EC50 (half-maximal effective concentration) was obtained. Using H1299 as a model, values of EC50 were quantified as 8% and 160 ng/ml for fetal bovine serum and CXCL12, respectively. In addition, the values of EC50 were demonstrated not to be affected by variations in compositions of extracellular matrix and geometries of wounds, which can thus be regarded as an intrinsic marker. Furthermore, the migration capabilities of a second cell type (HeLa) were characterized by the developed wound-healing assay, producing EC50 of 2% when fetal bovine serum was used as the stimuli. These results validated the proposed high-throughput wound-healing assay, which may function as an enabling tool in studying cellular capabilities of migration and invasion. © 2017 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Shaoliang Luan
- Department of Vascular Surgery, Clinical Division of Surgery, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
| | - Rui Hao
- State Key Laboratory of Transducer Technology, Institute of Electronics of the Chinese Academy of Sciences, Beijing 100190, People's Republic of China
- Univesity of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Yuanchen Wei
- State Key Laboratory of Transducer Technology, Institute of Electronics of the Chinese Academy of Sciences, Beijing 100190, People's Republic of China
| | - Deyong Chen
- State Key Laboratory of Transducer Technology, Institute of Electronics of the Chinese Academy of Sciences, Beijing 100190, People's Republic of China
- Univesity of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Beiyuan Fan
- State Key Laboratory of Transducer Technology, Institute of Electronics of the Chinese Academy of Sciences, Beijing 100190, People's Republic of China
- Univesity of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Fengliang Dong
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
| | - Wei Guo
- Department of Vascular Surgery, Clinical Division of Surgery, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
| | - Junbo Wang
- State Key Laboratory of Transducer Technology, Institute of Electronics of the Chinese Academy of Sciences, Beijing 100190, People's Republic of China
- Univesity of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Jian Chen
- State Key Laboratory of Transducer Technology, Institute of Electronics of the Chinese Academy of Sciences, Beijing 100190, People's Republic of China
- Univesity of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| |
Collapse
|
22
|
Vidmar J, Chingwaru C, Chingwaru W. Mammalian cell models to advance our understanding of wound healing: a review. J Surg Res 2017; 210:269-280. [DOI: 10.1016/j.jss.2016.10.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 07/12/2016] [Accepted: 10/14/2016] [Indexed: 12/30/2022]
|
23
|
Vu TQ, de Castro RMB, Qin L. Bridging the gap: microfluidic devices for short and long distance cell-cell communication. LAB ON A CHIP 2017; 17:1009-1023. [PMID: 28205652 PMCID: PMC5473339 DOI: 10.1039/c6lc01367h] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Cell-cell communication is a crucial component of many biological functions. For example, understanding how immune cells and cancer cells interact, both at the immunological synapse and through cytokine secretion, can help us understand and improve cancer immunotherapy. The study of how cells communicate and form synaptic connections is important in neuroscience, ophthalmology, and cancer research. But in order to increase our understanding of these cellular phenomena, better tools need to be developed that allow us to study cell-cell communication in a highly controlled manner. Some technical requirements for better communication studies include manipulating cells spatiotemporally, high resolution imaging, and integrating sensors. Microfluidics is a powerful platform that has the ability to address these requirements and other current limitations. In this review, we describe some new advances in microfluidic technologies that have provided researchers with novel methods to study intercellular communication. The advantages of microfluidics have allowed for new capabilities in both single cell-cell communication and population-based communication. This review highlights microfluidic communication devices categorized as "short distance", or primarily at the single cell level, and "long distance", which mostly encompasses population level studies. Future directions and translation/commercialization will also be discussed.
Collapse
Affiliation(s)
- Timothy Quang Vu
- Department of Bioengineering, Rice University, Houston, TX 77030, USA and Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA.
| | - Ricardo Miguel Bessa de Castro
- College of Engineering, Swansea University Singleton Park, Swansea, UK and Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA.
| | - Lidong Qin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA. and Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
24
|
Ma Y, Pan JZ, Zhao SP, Lou Q, Zhu Y, Fang Q. Microdroplet chain array for cell migration assays. LAB ON A CHIP 2016; 16:4658-4665. [PMID: 27833945 DOI: 10.1039/c6lc00823b] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Establishing cell migration assays in multiple different microenvironments is important in the study of tissue repair and regeneration, cancer progression, atherosclerosis, and arthritis. In this work, we developed a miniaturized and massive parallel microfluidic platform for multiple cell migration assays combining the traditional membrane-based cell migration technique and the droplet-based microfluidic technique. Nanoliter-scale droplets are flexibly assembled as building blocks based on a porous membrane to form microdroplet chains with diverse configurations for different assay modes. Multiple operations including in-droplet 2D/3D cell culture, cell co-culture and cell migration induced by a chemoattractant concentration gradient in droplet chains could be flexibly performed with reagent consumption in the nanoliter range for each assay and an assay scale-up to 81 assays in parallel in one microchip. We have applied the present platform to multiple modes of cell migration assays including the accurate cell migration assay, competitive cell migration assay, biomimetic chemotaxis assay, and multifactor cell migration assay based on the organ-on-a-chip concept, for demonstrating its versatility, applicability, and potential in cell migration-related research.
Collapse
Affiliation(s)
- Yan Ma
- Institute of Microanalytical Systems, Department of Chemistry and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058, China.
| | - Jian-Zhang Pan
- Institute of Microanalytical Systems, Department of Chemistry and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058, China.
| | - Shi-Ping Zhao
- Institute of Microanalytical Systems, Department of Chemistry and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058, China.
| | - Qi Lou
- Institute of Microanalytical Systems, Department of Chemistry and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058, China.
| | - Ying Zhu
- Institute of Microanalytical Systems, Department of Chemistry and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058, China.
| | - Qun Fang
- Institute of Microanalytical Systems, Department of Chemistry and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|