1
|
Chandurkar MK, Mittal N, Royer-Weeden SP, Lehmann SD, Michels EB, Haarman SE, Severance SA, Rho Y, Han SJ. Transient low shear-stress preconditioning influences long-term endothelial traction and alignment under high shear flow. Am J Physiol Heart Circ Physiol 2024; 326:H1180-H1192. [PMID: 38457352 DOI: 10.1152/ajpheart.00067.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/01/2024] [Accepted: 03/03/2024] [Indexed: 03/10/2024]
Abstract
Endothelial cells (ECs) within the vascular system encounter fluid shear stress (FSS). High, laminar FSS promotes vasodilation and anti-inflammatory responses, whereas low or disturbed FSS induces dysfunction and inflammation. However, the adaptation of endothelial cells (ECs) to dynamically changing FSS patterns remains underexplored. Here, by combining traction force microscopy with a custom flow chamber, we examined human umbilical vein endothelial cells adapting their traction during transitions from short-term low shear to long-term high shear stress. We discovered that the initial low FSS elevates the traction by only half of the amount in response to direct high FSS even after flow changes to high FSS. However, in the long term under high FSS, the flow started with low FSS triggers a substantial second rise in traction for over 10 h. In contrast, the flow started directly with high FSS results in a quick traction surge followed by a huge reduction below the baseline traction in <30 min. Importantly, we find that the orientation of traction vectors is steered by initial shear exposure. Using Granger causality analysis, we show that the traction that aligns in the flow direction under direct high FSS functionally causes cell alignment toward the flow direction. However, EC traction that orients perpendicular to the flow that starts with temporary low FSS functionally causes cell orientation perpendicular to the flow. Taken together, our findings elucidate the significant influence of initial short-term low FSS on lasting changes in endothelial traction that induces EC alignment.NEW & NOTEWORTHY In our study, we uncover that preconditioning with low shear stress yields enduring impacts on endothelial cell traction and orientation, persisting even after transitioning to high-shear conditions. Using Granger causality analysis, we demonstrate a functional link between the direction of cell traction and subsequent cellular alignment across varying shear environments.
Collapse
Affiliation(s)
- Mohanish K Chandurkar
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan, United States
| | - Nikhil Mittal
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan, United States
| | - Shaina P Royer-Weeden
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan, United States
| | - Steven D Lehmann
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
| | - Etienne B Michels
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
| | - Samuel E Haarman
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan, United States
| | - Scott A Severance
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
| | - Yeonwoo Rho
- Department of Mathematical Sciences, Michigan Technological University, Houghton, Michigan, United States
| | - Sangyoon J Han
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan, United States
- Department of Mechanical Engineering and Engineering Mechanics, Michigan Technological University, Houghton, Michigan, United States
| |
Collapse
|
2
|
Sheng X, Gao S, Sheng Y, Xie X, Wang J, He Y. Vangl2 participates in the primary ciliary assembly under low fluid shear stress in hUVECs. Cell Tissue Res 2021; 387:95-109. [PMID: 34738156 DOI: 10.1007/s00441-021-03546-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 10/13/2021] [Indexed: 11/30/2022]
Abstract
The pattern of blood fluid shear stress (FSS) is considered the main factor that affects ciliogenesis in human umbilical vein endothelial cells (hUVECs), the underlying mechanism is unclear. Microfluidic chamber experiments were carried out to load hUVECs with low fluid shear stress (LSS, 0.1 dynes/cm2) or high fluid shear stress (HSS, 15 dynes/cm2). Van Gogh2 (Vangl2), a core protein in the planar cell polarity (PCP) pathway, was silenced and overexpressed in hUVECs. Immunofluorescence analysis showed that primary cilia assemble under LSS while disassembling under HSS. Vangl2 expression was consistent with cilia assembly, and its localization showed a polar distribution under LSS. Furthermore, the average number of ciliated cells and primary cilia length were increased in the Vangl2 overexpressing cell lines (the OE group) but decreased in the Vangl2 silenced cell lines (the SH group). When these cells were loaded with different FSS, more ciliated cells with longest primary cilia were observed in the LSS loaded OE group compared with those in the other groups. Immunoprecipitation showed that the interaction between Bardet-Biedl syndrome 8 (BBS8) and Vangl2 was enhanced following LSS loading compared to that under HSS. However, the interactions between phosphorylated dishevelled segment polarity protein 2 (pDvl2), kinesin family member 2a (Kif2a), and polo-like kinase 1 (Plk1) and Vangl2 were restrained following LSS loading. Overall, the results indicated that Vangl2 played a significant role during LSS-induced primary cilia assembly by recruiting BBS to promote the apical docking of basal bodies and by restraining Dvl2 phosphorylation from reducing primary cilia disassembly.
Collapse
Affiliation(s)
- Xin Sheng
- Department of Biochemistry, Zunyi Medical University, Zunyi, 563000, People's Republic of China.
| | - Shuanglin Gao
- Department of Biochemistry, Zunyi Medical University, Zunyi, 563000, People's Republic of China
| | - Yan Sheng
- Laboratory of Basic Medical Morphology, Zunyi Medical University, Zunyi, 563000, People's Republic of China
| | - Xiadan Xie
- Department of Biochemistry, Zunyi Medical University, Zunyi, 563000, People's Republic of China
| | - Junhua Wang
- Department of Biochemistry, Zunyi Medical University, Zunyi, 563000, People's Republic of China
| | - Yan He
- Department of Biochemistry, Zunyi Medical University, Zunyi, 563000, People's Republic of China
| |
Collapse
|
3
|
Urschel K, Tauchi M, Achenbach S, Dietel B. Investigation of Wall Shear Stress in Cardiovascular Research and in Clinical Practice-From Bench to Bedside. Int J Mol Sci 2021; 22:5635. [PMID: 34073212 PMCID: PMC8198948 DOI: 10.3390/ijms22115635] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/20/2021] [Accepted: 05/22/2021] [Indexed: 12/16/2022] Open
Abstract
In the 1900s, researchers established animal models experimentally to induce atherosclerosis by feeding them with a cholesterol-rich diet. It is now accepted that high circulating cholesterol is one of the main causes of atherosclerosis; however, plaque localization cannot be explained solely by hyperlipidemia. A tremendous amount of studies has demonstrated that hemodynamic forces modify endothelial athero-susceptibility phenotypes. Endothelial cells possess mechanosensors on the apical surface to detect a blood stream-induced force on the vessel wall, known as "wall shear stress (WSS)", and induce cellular and molecular responses. Investigations to elucidate the mechanisms of this process are on-going: on the one hand, hemodynamics in complex vessel systems have been described in detail, owing to the recent progress in imaging and computational techniques. On the other hand, investigations using unique in vitro chamber systems with various flow applications have enhanced the understanding of WSS-induced changes in endothelial cell function and the involvement of the glycocalyx, the apical surface layer of endothelial cells, in this process. In the clinical setting, attempts have been made to measure WSS and/or glycocalyx degradation non-invasively, for the purpose of their diagnostic utilization. An increasing body of evidence shows that WSS, as well as serum glycocalyx components, can serve as a predicting factor for atherosclerosis development and, most importantly, for the rupture of plaques in patients with high risk of coronary heart disease.
Collapse
Affiliation(s)
| | | | | | - Barbara Dietel
- Department of Medicine 2—Cardiology and Angiology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Universitätsklinikum, 91054 Erlangen, Germany; (K.U.); (M.T.); (S.A.)
| |
Collapse
|
4
|
Yang T, De La Franier B, Thompson M. Anti-Thrombogenicity Study of a Covalently-Attached Monolayer on Stent-Grade Stainless Steel. MATERIALS 2021; 14:ma14092342. [PMID: 33946387 PMCID: PMC8125229 DOI: 10.3390/ma14092342] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/21/2021] [Accepted: 04/26/2021] [Indexed: 12/16/2022]
Abstract
Implantable devices fabricated from austenitic type 316L stainless steel have been employed significantly in medicine, principally because the material displays excellent mechanical characteristics and corrosion resistance. It is well known, however, that interaction of exposure of such a material to blood can initiate platelet adhesion and blood coagulation, leading to a harmful medical condition. In order to prevent undesirable surface platelet adhesion on biomaterials employed in procedures such as renal dialysis, we developed an ultrathin anti-thrombogenic covalently attached monolayer based on monoethylene glycol silane chemistry. This functions by forming an interstitial hydration layer which displays restricted mobility in the prevention of surface fouling. In the present work, the promising anti-thrombogenic properties of this film are examined with respect to platelet aggregation on 316L austenitic stainless steel exposed to whole human blood. Prior to exposure with blood, all major surface modification steps were examined by X-ray photoelectron spectroscopic analysis and surface free-angle measurement by contact angle goniometry. End-stage anti-thrombogenicity detection after 20 min of blood exposure at 100 s-1, 300 s-1, 600 s-1, 750 s-1, and 900 s-1 shear rates revealed that a significant reduction (>90%) of platelet adhesion and aggregation was achieved for surface-modified steel, compared with untreated material. This result is confirmed by experiments conducted in real time for 60-minute exposure to blood at 100 s-1, 600 s-1, and 900 s-1 shear rates.
Collapse
|
5
|
Abutaleb NO, Truskey GA. Differentiation and characterization of human iPSC-derived vascular endothelial cells under physiological shear stress. STAR Protoc 2021; 2:100394. [PMID: 33796871 PMCID: PMC7995664 DOI: 10.1016/j.xpro.2021.100394] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) offer a potentially unlimited source to generate endothelial cells (ECs) for numerous applications. Here, we describe a 7-day protocol to differentiate up to 55 million vascular endothelial cells (viECs) from 3.5 million human iPSCs using small molecules to regulate specific transcription factors. We also describe a parallel-plate flow chamber system to study EC behavior under physiological shear stress. For complete details on the use and execution of this protocol, please refer to Atchison et al. (2020). A protocol for differentiating vascular endothelial cells (viECs) from human iPSCs Generation of up to 55 million viECs from 3.5 million iPSCs within 7 days Design and use of parallel-plate flow chamber to study EC behavior under flow viECs express EC markers, upregulate flow-sensitive genes, and align to flow direction
Collapse
Affiliation(s)
- Nadia O Abutaleb
- Department of Biomedical Engineering, Duke University, Durham, NC 27713, USA
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC 27713, USA
| |
Collapse
|
6
|
Pantazi P, Carollo E, Carter DRF, Brooks SA. A practical toolkit to study aspects of the metastatic cascade in vitro. Acta Histochem 2020; 122:151654. [PMID: 33157489 DOI: 10.1016/j.acthis.2020.151654] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/23/2020] [Accepted: 10/28/2020] [Indexed: 12/30/2022]
Abstract
While metastasis - the spread of cancer from the primary location to distant sites in the body - remains the principle cause of cancer death, it is incompletely understood. It is a complex process, requiring the metastatically successful cancer cell to negotiate a formidable series of interconnected steps, which are described in this paper. For each step, we review the range of in vitro assays that may be used to study them. We also provide a range of detailed, step-by-step protocols that can be undertaken in most modestly-equipped laboratories, including methods for converting qualitative observations into quantitative data for analysis. Assays include: (1) a gelatin degradation assay to study the ability of endothelial cells to degrade extracellular matrix during tumour angiogenesis; (2) the morphological characterisation of cells undergoing epithelial-mesenchymal transition (EMT) as they acquire motility; (3) a 'scratch' or 'wound-healing' assay to study cancer cell migration; (4) a transwell assay to study cancer cell invasion through extracellular matrix; and (5) a static adhesion assay to examine cancer cell interactions with, and adhesion to, endothelial monolayers. This toolkit of protocols will enable researchers who are interested in metastasis to begin to focus on defined aspects of the process. It is only by further understanding this complex, fascinating and clinically relevant series of events that we may ultimately devise ways of better treating, or even preventing, cancer metastasis. The assays may also be of more broad interest to researchers interested in studying aspects of cellular behaviour in relation to other developmental and disease processes.
Collapse
|
7
|
Shurbaji S, G. Anlar G, A. Hussein E, Elzatahry A, C. Yalcin H. Effect of Flow-Induced Shear Stress in Nanomaterial Uptake by Cells: Focus on Targeted Anti-Cancer Therapy. Cancers (Basel) 2020; 12:E1916. [PMID: 32708521 PMCID: PMC7409087 DOI: 10.3390/cancers12071916] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/20/2019] [Accepted: 12/24/2019] [Indexed: 02/02/2023] Open
Abstract
Recently, nanomedicines have gained a great deal of attention in diverse biomedical applications, including anti-cancer therapy. Being different from normal tissue, the biophysical microenvironment of tumor cells and cancer cell mechanics should be considered for the development of nanostructures as anti-cancer agents. Throughout the last decades, many efforts devoted to investigating the distinct cancer environment and understanding the interactions between tumor cells and have been applied bio-nanomaterials. This review highlights the microenvironment of cancer cells and how it is different from that of healthy tissue. We gave special emphasis to the physiological shear stresses existing in the cancerous surroundings, since these stresses have a profound effect on cancer cell/nanoparticle interaction. Finally, this study reviews relevant examples of investigations aimed at clarifying the cellular nanoparticle uptake behavior under both static and dynamic conditions.
Collapse
Affiliation(s)
- Samar Shurbaji
- Materials Science and Technology Department, College of Arts and Sciences, Qatar University, Doha 2713, Qatar; (S.S.); (E.A.H.)
| | - Gulsen G. Anlar
- College of Medicine, Department of Medical Sciences, Qatar University, Doha 2713, Qatar;
| | - Essraa A. Hussein
- Materials Science and Technology Department, College of Arts and Sciences, Qatar University, Doha 2713, Qatar; (S.S.); (E.A.H.)
| | - Ahmed Elzatahry
- Materials Science and Technology Department, College of Arts and Sciences, Qatar University, Doha 2713, Qatar; (S.S.); (E.A.H.)
| | - Huseyin C. Yalcin
- Biomedical Research Center, Qatar University, Doha 2713, Qatar
- Department of Biomedical Sciences, College of Health Science-QU Health, Qatar University, Doha 2713, Qatar
| |
Collapse
|
8
|
James BD, Montoya N, Allen J. MechanoBioTester: A Decoupled Multistimulus Cell Culture Device for Studying Complex Microenvironments In Vitro. ACS Biomater Sci Eng 2020; 6:3673-3689. [PMID: 32704528 PMCID: PMC7377433 DOI: 10.1021/acsbiomaterials.0c00498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Increasingly being recognized is the role of the complex microenvironment to regulate cell phenotype; however, the cell culture systems used to study these effects in vitro are lagging. The complex microenvironment is host to a combination of biological interactions, chemical factors, and mechanical stimuli. Many devices have been designed to probe the effects of one mechanical stimulus, but few are capable of systematically interrogating all combinations of mechanical stimuli with independent control. To address this gap, we have developed the MechanoBioTester platform, a decoupled, multi-stimulus cell culture model for studying the cellular response to complex microenvironments in vitro. The system uses an engineered elastomeric chamber with a specially defined region for incorporating different target materials to act as the cell culture substrate. We have tested the system with several target materials including: polydimethylsiloxane elastomer, polyacrylamide gel, poly(1,8-octanediol citrate) elastomer, and type I collagen gel for both 2D and 3D co-culture. Additionally, when the chamber is connected to a flow circuit and our stretching device, stimuli in the form of fluid flow, cyclic stretch, and hydrostatic pressure are able to be imparted with independent control. We validated the device using experimental and computational methods to define a range of capabilities relevant to physiological microenvironments. The MechanoBioTester platform promises to function as a model system for mechanobiology, biomaterial design, and drug discovery applications that focus on probing the impact of a complex microenvironment in an in vitro setting. The protocol described within provides the details characterizing the MechanoBioTester system, the steps for fabricating the MechanoBioTester chamber, and the procedure for operating the MechanoBioTester system to stimulate cells.
Collapse
Affiliation(s)
- Bryan D. James
- Department of Materials Science & Engineering, University of Florida, 100 Rhines Hall, PO Box 116400, Gainesville, Florida 32611, United States
- Institute for Computational Engineering, University of Florida, 300 Weil Hall, PO Box 116550, Gainesville, Florida 32611, United States
| | - Nicolas Montoya
- Department of Electrical & Computer Engineering, University of Florida, 216 Larsen Hall, Gainesville, Florida 32611, United States
| | - Josephine Allen
- Department of Materials Science & Engineering, University of Florida, 100 Rhines Hall, PO Box 116400, Gainesville, Florida 32611, United States
| |
Collapse
|
9
|
Hulander M, Valen-Rukke H, Sundell G, Andersson M. Influence of Fibrinogen on Staphylococcus epidermidis Adhesion Can Be Reversed by Tuning Surface Nanotopography. ACS Biomater Sci Eng 2019; 5:4323-4330. [DOI: 10.1021/acsbiomaterials.9b00450] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Mats Hulander
- Chalmers University of Technology, Chalmersplatsen 4, 412 96 Göteborg, Sweden
- Nordic Institute of Dental Materials, Sognsveien 70 A, 0855 Oslo, Norway
| | - Håkon Valen-Rukke
- Nordic Institute of Dental Materials, Sognsveien 70 A, 0855 Oslo, Norway
| | - Gustav Sundell
- Chalmers University of Technology, Chalmersplatsen 4, 412 96 Göteborg, Sweden
| | - Martin Andersson
- Chalmers University of Technology, Chalmersplatsen 4, 412 96 Göteborg, Sweden
| |
Collapse
|
10
|
Kwiecinski JM, Crosby HA, Valotteau C, Hippensteel JA, Nayak MK, Chauhan AK, Schmidt EP, Dufrêne YF, Horswill AR. Staphylococcus aureus adhesion in endovascular infections is controlled by the ArlRS-MgrA signaling cascade. PLoS Pathog 2019; 15:e1007800. [PMID: 31116795 PMCID: PMC6548404 DOI: 10.1371/journal.ppat.1007800] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 06/04/2019] [Accepted: 04/30/2019] [Indexed: 12/31/2022] Open
Abstract
Staphylococcus aureus is a leading cause of endovascular infections. This bacterial pathogen uses a diverse array of surface adhesins to clump in blood and adhere to vessel walls, leading to endothelial damage, development of intravascular vegetations and secondary infectious foci, and overall disease progression. In this work, we describe a novel strategy used by S. aureus to control adhesion and clumping through activity of the ArlRS two-component regulatory system, and its downstream effector MgrA. Utilizing a combination of in vitro cellular assays, and single-cell atomic force microscopy, we demonstrated that inactivation of this ArlRS—MgrA cascade inhibits S. aureus adhesion to a vast array of relevant host molecules (fibrinogen, fibronectin, von Willebrand factor, collagen), its clumping with fibrinogen, and its attachment to human endothelial cells and vascular structures. This impact on S. aureus adhesion was apparent in low shear environments, and in physiological levels of shear stress, as well as in vivo in mouse models. These effects were likely mediated by the de-repression of giant surface proteins Ebh, SraP, and SasG, caused by inactivation of the ArlRS—MgrA cascade. In our in vitro assays, these giant proteins collectively shielded the function of other surface adhesins and impaired their binding to cognate ligands. Finally, we demonstrated that the ArlRS—MgrA regulatory cascade is a druggable target through the identification of a small-molecule inhibitor of ArlRS signaling. Our findings suggest a novel approach for the pharmacological treatment and prevention of S. aureus endovascular infections through targeting the ArlRS—MgrA regulatory system. Adhesion is central to the success of Staphylococcus aureus as a bacterial pathogen. We describe a novel mechanism through which S. aureus alters adhesion to ligands by regulating expression of giant inhibitory surface proteins. These giant proteins shield normal surface adhesins, preventing binding to ligands commonly found in the bloodstream and vessel walls. Using this unique regulatory scheme, S. aureus can bypass the need for individualized regulation of numerous adhesins to control overall adhesive properties. Our study establishes the importance of these giant proteins for S. aureus pathogenesis and demonstrates that a single regulatory cascade can be targeted for treating infections.
Collapse
Affiliation(s)
- Jakub M. Kwiecinski
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Heidi A. Crosby
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Claire Valotteau
- Institute of Life Sciences, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Joseph A. Hippensteel
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Manasa K. Nayak
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Anil K. Chauhan
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Eric P. Schmidt
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Yves F. Dufrêne
- Institute of Life Sciences, Université catholique de Louvain, Louvain-la-Neuve, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium
| | - Alexander R. Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- Department of Veterans Affairs Eastern Colorado Healthcare System, Denver, Colorado, United States of America
- * E-mail:
| |
Collapse
|
11
|
Dupuy A, Ju LA, Passam FH. Straight Channel Microfluidic Chips for the Study of Platelet Adhesion under Flow. Bio Protoc 2019; 9:e3195. [PMID: 33654994 PMCID: PMC7854274 DOI: 10.21769/bioprotoc.3195] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/17/2019] [Accepted: 02/28/2019] [Indexed: 01/01/2023] Open
Abstract
Microfluidic devices have become an integral method of cardiovascular research as they enable the study of shear force in biological processes, such as platelet function and thrombus formation. Furthermore, microfluidic chips offer the benefits of ex vivo testing of platelet adhesion using small amounts of blood or purified platelets. Microfluidic chips comprise flow channels of varying dimensions and geometries which are connected to a syringe pump. The pump draws blood or platelet suspensions through the channel(s) allowing for imaging of platelet adhesion and thrombus formation by fluorescence microscopy. The chips can be fabricated from various blood-compatible materials. The current protocol uses commercial plastic or in-house polydimethylsiloxane (PDMS) chips. Commercial biochips offer the advantage of standardization whereas in-house chips offer the advantage of decreased cost and flexibility in design. Microfluidic devices are a powerful tool to study the biorheology of platelets and other cell types with the potential of a diagnostic and monitoring tool for cardiovascular diseases.
Collapse
Affiliation(s)
- Alexander Dupuy
- Heart Research Institute, Newtown, NSW 2042, Australia.,University of Sydney, Camperdown, NSW 2006, Australia
| | - Lining Arnold Ju
- Heart Research Institute, Newtown, NSW 2042, Australia.,University of Sydney, Camperdown, NSW 2006, Australia
| | - Freda H Passam
- Heart Research Institute, Newtown, NSW 2042, Australia.,University of Sydney, Camperdown, NSW 2006, Australia
| |
Collapse
|
12
|
Jiang W, Lin J, Chen AH, Pan J, Liu H. A portable device for studying the effects of fluid flow on degradation properties of biomaterials inside cell incubators. Regen Biomater 2019; 6:39-48. [PMID: 30740241 PMCID: PMC6362820 DOI: 10.1093/rb/rby026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 11/08/2018] [Accepted: 11/10/2018] [Indexed: 12/22/2022] Open
Abstract
A portable device was designed and constructed for studying the properties of biomaterials in physiologically relevant fluids under controllable flow conditions that closely simulate fluid flow inside the body. The device can fit entirely inside a cell incubator; and, thus, it can be used directly under standard cell culture conditions. An impedance-driven pump was built in the sterile flow loop to control the flow rates of fluids, which made the device small and portable for easy deployment in the incubator. To demonstrate the device functions, magnesium (Mg) as a representative biodegradable material was tested in the flow device for immersion degradation under flow versus static conditions, while the flow module was placed inside a standard cell incubator. The flow rate was controlled at 0.17 ± 0.06 ml/s for this study; and, the flow rate is adjustable through the controller module outside of incubators for simulating the flow rates in the ranges of blood flow in human artery (0.05 ∼0.43 ml/s) and vein (0.02 ∼0.08 ml/s). Degradation of Mg under flow versus static conditions was characterized by measuring the changes of sample mass and thickness, and Mg2+ ion concentrations in the immersion media. Surface chemistry and morphology of Mg after immersion under flow versus static conditions were compared. The portable impedance-driven flow device is easy to fit inside an incubator and much smaller than a peristaltic pump, providing a valuable solution for studying biomaterials and implants (e.g. vascular or ureteral stents) in body fluids under flow versus static conditions with or without cells.
Collapse
Affiliation(s)
- Wensen Jiang
- Department of Bioengineering, Materials Science and Engineering Program, Stem Cell Center
| | - Jiajia Lin
- Department of Bioengineering, Materials Science and Engineering Program, Stem Cell Center
| | - Alex H Chen
- Department of Mechanical Engineering, University of California, Riverside, Riverside, CA, USA
- Department of Bioengineering, University of California, Riverside, Riverside, CA, USA
| | - Jianwei Pan
- Department of Neurosurgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Huinan Liu
- Department of Bioengineering, Materials Science and Engineering Program, Stem Cell Center
- Department of Bioengineering, University of California, Riverside, Riverside, CA, USA
| |
Collapse
|
13
|
Man HSJ, Sukumar AN, Ku KH, Dubinsky MK, Subramaniam N, Marsden PA. Gene Expression Analysis of Endothelial Cells Exposed to Shear Stress Using Multiple Parallel-plate Flow Chambers. J Vis Exp 2018. [PMID: 30394398 DOI: 10.3791/58478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
We describe a workflow for the analysis of gene expression from endothelial cells subject to a steady laminar flow using multiple monitored parallel-plate flow chambers. Endothelial cells form the inner cellular lining of blood vessels and are chronically exposed to the frictional force of blood flow called shear stress. Under physiological conditions, endothelial cells function in the presence of various shear stress conditions. Thus, the application of shear stress conditions in in vitro models can provide greater insight into endothelial responses in vivo. The parallel-plate flow chamber previously published by Lane et al.9 is adapted to study endothelial gene regulation in the presence and absence of steady (non-pulsatile) laminar flow. Key adaptations in the set-up for laminar flow as presented here include a large, dedicated environment to house concurrent flow circuits, the monitoring of flow rates in real-time, and the inclusion of an exogenous reference RNA for the normalization of quantitative real-time PCR data. To assess multiple treatments/conditions with the application of shear stress, multiple flow circuits and pumps are used simultaneously within the same heated and humidified incubator. The flow rate of each flow circuit is measured continuously in real-time to standardize shear stress conditions throughout the experiments. Because these experiments have multiple conditions, we also use an exogenous reference RNA that is spiked-in at the time of RNA extraction for the normalization of RNA extraction and first-strand cDNA synthesis efficiencies. These steps minimize the variability between samples. This strategy is employed in our pipeline for the gene expression analysis with shear stress experiments using the parallel-plate flow chamber, but parts of this strategy, such as the exogenous reference RNA spike-in, can easily and cost-effectively be used for other applications.
Collapse
Affiliation(s)
- H S Jeffrey Man
- Institute of Medical Science, University of Toronto; Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital
| | - Aravin N Sukumar
- Institute of Medical Science, University of Toronto; Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital
| | - Kyung Ha Ku
- Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital; Department of Laboratory Medicine and Pathobiology, University of Toronto
| | - Michelle K Dubinsky
- Institute of Medical Science, University of Toronto; Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital
| | - Noeline Subramaniam
- Institute of Medical Science, University of Toronto; Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital
| | - Philip A Marsden
- Institute of Medical Science, University of Toronto; Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital; Department of Laboratory Medicine and Pathobiology, University of Toronto; Department of Medicine, University of Toronto;
| |
Collapse
|
14
|
McCall AD, Edgerton M. Real-time Imaging and Quantification of Fungal Biofilm Development Using a Two-Phase Recirculating Flow System. J Vis Exp 2018:58457. [PMID: 30394387 PMCID: PMC6235572 DOI: 10.3791/58457] [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] [Indexed: 10/31/2022] Open
Abstract
In oropharyngeal candidiasis, members of the genus Candida must adhere to and grow on the oral mucosal surface while under the effects of salivary flow. While models for the growth under flow have been developed, many of these systems are expensive, or do not allow imaging while the cells are under flow. We have developed a novel apparatus that allows us to image the growth and development of Candida albicans cells under flow and in real-time. Here, we detail the protocol for the assembly and use of this flow apparatus, as well as the quantification of data that are generated. We are able to quantify the rates that the cells attach to and detach from the slide, as well as to determine a measure of the biomass on the slide over time. This system is both economical and versatile, working with many types of light microscopes, including inexpensive benchtop microscopes, and is capable of extended imaging times compared to other flow systems. Overall, this is a low-throughput system that can provide highly detailed real-time information on the biofilm growth of fungal species under flow.
Collapse
|
15
|
Li SS, Ip CKM, Tang MYH, Sy SKH, Yung S, Chan TM, Yang M, Shum HC, Wong AST. Modeling Ovarian Cancer Multicellular Spheroid Behavior in a Dynamic 3D Peritoneal Microdevice. J Vis Exp 2017. [PMID: 28287578 DOI: 10.3791/55337] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Ovarian cancer is characterized by extensive peritoneal metastasis, with tumor spheres commonly found in the malignant ascites. This is associated with poor clinical outcomes and currently lacks effective treatment. Both the three-dimensional (3D) environment and the dynamic mechanical forces are very important factors in this metastatic cascade. However, traditional cell cultures fail to recapitulate this natural tumor microenvironment. Thus, in vivo-like models that can emulate the intraperitoneal environment are of obvious importance. In this study, a new microfluidic platform of the peritoneum was set up to mimic the situation of ovarian cancer spheroids in the peritoneal cavity during metastasis. Ovarian cancer spheroids generated under a non-adherent condition were cultured in microfluidic channels coated with peritoneal mesothelial cells subjected to physiologically relevant shear stress. In summary, this dynamic 3D ovarian cancer-mesothelium microfluidic platform can provide new knowledge on basic cancer biology and serve as a platform for potential drug screening and development.
Collapse
Affiliation(s)
- Shan-Shan Li
- School of Biological Sciences, University of Hong Kong
| | - Carman K M Ip
- School of Biological Sciences, University of Hong Kong
| | | | - Samuel K H Sy
- Department of Mechanical Engineering, University of Hong Kong
| | - Susan Yung
- Department of Medicine, University of Hong Kong
| | | | - Mengsu Yang
- Department of Biomedical Sciences, Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institutes of City University of Hong Kong
| | - Ho Cheung Shum
- Department of Mechanical Engineering, University of Hong Kong;
| | | |
Collapse
|
16
|
Muhamed I, Chowdhury F, Maruthamuthu V. Biophysical Tools to Study Cellular Mechanotransduction. Bioengineering (Basel) 2017; 4:E12. [PMID: 28952491 PMCID: PMC5590431 DOI: 10.3390/bioengineering4010012] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 01/30/2017] [Accepted: 02/02/2017] [Indexed: 01/25/2023] Open
Abstract
The cell membrane is the interface that volumetrically isolates cellular components from the cell's environment. Proteins embedded within and on the membrane have varied biological functions: reception of external biochemical signals, as membrane channels, amplification and regulation of chemical signals through secondary messenger molecules, controlled exocytosis, endocytosis, phagocytosis, organized recruitment and sequestration of cytosolic complex proteins, cell division processes, organization of the cytoskeleton and more. The membrane's bioelectrical role is enabled by the physiologically controlled release and accumulation of electrochemical potential modulating molecules across the membrane through specialized ion channels (e.g., Na⁺, Ca2+, K⁺ channels). The membrane's biomechanical functions include sensing external forces and/or the rigidity of the external environment through force transmission, specific conformational changes and/or signaling through mechanoreceptors (e.g., platelet endothelial cell adhesion molecule (PECAM), vascular endothelial (VE)-cadherin, epithelial (E)-cadherin, integrin) embedded in the membrane. Certain mechanical stimulations through specific receptor complexes induce electrical and/or chemical impulses in cells and propagate across cells and tissues. These biomechanical sensory and biochemical responses have profound implications in normal physiology and disease. Here, we discuss the tools that facilitate the understanding of mechanosensitive adhesion receptors. This article is structured to provide a broad biochemical and mechanobiology background to introduce a freshman mechano-biologist to the field of mechanotransduction, with deeper study enabled by many of the references cited herein.
Collapse
Affiliation(s)
- Ismaeel Muhamed
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA.
| | - Farhan Chowdhury
- Department of Mechanical Engineering and Energy Processes, Southern Illinois University Carbondale, Carbondale, IL 62901, USA.
| | - Venkat Maruthamuthu
- Department of Mechanical and Aerospace Engineering, Old Dominion University, Norfolk, VA 23529, USA.
| |
Collapse
|
17
|
Wang YX, Xiang C, Liu B, Zhu Y, Luan Y, Liu ST, Qin KR. A multi-component parallel-plate flow chamber system for studying the effect of exercise-induced wall shear stress on endothelial cells. Biomed Eng Online 2016; 15:154. [PMID: 28155716 PMCID: PMC5259904 DOI: 10.1186/s12938-016-0273-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND In vivo studies have demonstrated that reasonable exercise training can improve endothelial function. To confirm the key role of wall shear stress induced by exercise on endothelial cells, and to understand how wall shear stress affects the structure and the function of endothelial cells, it is crucial to design and fabricate an in vitro multi-component parallel-plate flow chamber system which can closely replicate exercise-induced wall shear stress waveforms in artery. METHODS The in vivo wall shear stress waveforms from the common carotid artery of a healthy volunteer in resting and immediately after 30 min acute aerobic cycling exercise were first calculated by measuring the inner diameter and the center-line blood flow velocity with a color Doppler ultrasound. According to the above in vivo wall shear stress waveforms, we designed and fabricated a parallel-plate flow chamber system with appropriate components based on a lumped parameter hemodynamics model. To validate the feasibility of this system, human umbilical vein endothelial cells (HUVECs) line were cultured within the parallel-plate flow chamber under abovementioned two types of wall shear stress waveforms and the intracellular actin microfilaments and nitric oxide (NO) production level were evaluated using fluorescence microscope. RESULTS Our results show that the trends of resting and exercise-induced wall shear stress waveforms, especially the maximal, minimal and mean wall shear stress as well as oscillatory shear index, generated by the parallel-plate flow chamber system are similar to those acquired from the common carotid artery. In addition, the cellular experiments demonstrate that the actin microfilaments and the production of NO within cells exposed to the two different wall shear stress waveforms exhibit different dynamic behaviors; there are larger numbers of actin microfilaments and higher level NO in cells exposed in exercise-induced wall shear stress condition than resting wall shear stress condition. CONCLUSION The parallel-plate flow chamber system can well reproduce wall shear stress waveforms acquired from the common carotid artery in resting and immediately after exercise states. Furthermore, it can be used for studying the endothelial cells responses under resting and exercise-induced wall shear stress environments in vitro.
Collapse
Affiliation(s)
- Yan-Xia Wang
- State Key Laboratory of Structural Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
| | - Cheng Xiang
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore, Singapore
| | - Bo Liu
- Department of Biomedical Engineering, Dalian University of Technology, Dalian, China
| | - Yong Zhu
- Department of Biomedical Engineering, Dalian University of Technology, Dalian, China
| | - Yong Luan
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shu-Tian Liu
- State Key Laboratory of Structural Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
| | - Kai-Rong Qin
- Department of Biomedical Engineering, Dalian University of Technology, Dalian, China.
| |
Collapse
|
18
|
White LA, Stevenson EV, Yun JW, Eshaq R, Harris NR, Mills DK, Minagar A, Couraud PO, Alexander JS. The Assembly and Application of 'Shear Rings': A Novel Endothelial Model for Orbital, Unidirectional and Periodic Fluid Flow and Shear Stress. J Vis Exp 2016. [PMID: 27842359 DOI: 10.3791/54632] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Deviations from normal levels and patterns of vascular fluid shear play important roles in vascular physiology and pathophysiology by inducing adaptive as well as pathological changes in endothelial phenotype and gene expression. In particular, maladaptive effects of periodic, unidirectional flow induced shear stress can trigger a variety of effects on several vascular cell types, particularly endothelial cells. While by now endothelial cells from diverse anatomic origins have been cultured, in-depth analyses of their responses to fluid shear have been hampered by the relative complexity of shear models (e.g., parallel plate flow chamber, cone and plate flow model). While these all represent excellent approaches, such models are technically complicated and suffer from drawbacks including relatively lengthy and complex setup time, low surface areas, requirements for pumps and pressurization often requiring sealants and gaskets, creating challenges to both maintenance of sterility and an inability to run multiple experiments. However, if higher throughput models of flow and shear were available, greater progress on vascular endothelial shear responses, particularly periodic shear research at the molecular level, might be more rapidly advanced. Here, we describe the construction and use of shear rings: a novel, simple-to-assemble, and inexpensive tissue culture model with a relatively large surface area that easily allows for a high number of experimental replicates in unidirectional, periodic shear stress studies on endothelial cells.
Collapse
Affiliation(s)
- Luke A White
- Molecular and Cellular Physiology, Louisiana State University Health Sciences Center in Shreveport
| | - Emily V Stevenson
- Molecular and Cellular Physiology, Louisiana State University Health Sciences Center in Shreveport
| | - J Winny Yun
- Molecular and Cellular Physiology, Louisiana State University Health Sciences Center in Shreveport
| | - Randa Eshaq
- Molecular and Cellular Physiology, Louisiana State University Health Sciences Center in Shreveport
| | - Norman R Harris
- Molecular and Cellular Physiology, Louisiana State University Health Sciences Center in Shreveport
| | | | - Alireza Minagar
- Neurology, Louisiana State University Health Sciences Center in Shreveport
| | | | - J Steven Alexander
- Molecular and Cellular Physiology, Louisiana State University Health Sciences Center in Shreveport;
| |
Collapse
|
19
|
Stemness and chemoresistance in epithelial ovarian carcinoma cells under shear stress. Sci Rep 2016; 6:26788. [PMID: 27245437 PMCID: PMC4887794 DOI: 10.1038/srep26788] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 05/09/2016] [Indexed: 01/06/2023] Open
Abstract
One of greatest challenges to the successful treatment of cancer is drug resistance. An exciting approach is the eradication of cancer stem cells (CSCs). However, little is known about key signals regulating the formation and expansion of CSCs. Moreover, lack of a reliable predictive preclinical model has been a major obstacle to discover new cancer drugs and predict their clinical activity. Here, in ovarian cancer, a highly chemoresistant tumor that is rapidly fatal, we provide the first evidence demonstrating the causal involvement of mechanical stimulus in the CSC phenotype using a customizable microfluidic platform and three-dimensional spheroids, which most closely mimic tumor behavior. We found that ovarian cancer cells significantly acquired the expression of epithelial-to-mesenchymal transition and CSC markers and a remarkable chemoresistance to clinically relevant doses of frontline chemotherapeutic drugs cisplatin and paclitaxel when grown under fluid shear stress, which corroborates with the physiological attainable levels in the malignant ascites, but not under static condition. Furthermore, we uncovered a new link of microRNA-199a-3p, phosphatidylinositol 3-kinase/Akt, and multidrug transporter activation in shear stress-induced CSC enrichment. Our findings shed new light on the significance of hydrodynamics in cancer progression, emphasizing the need of a flow-informed framework in the development of therapeutics.
Collapse
|
20
|
Jamiolkowski RM, Kang SD, Rodriguez AK, Haseltine JM, Galinat LJ, Jantzen AE, Carlon TA, Darrabie MD, Arciniegas AJ, Mantilla JG, Haley NR, Noviani M, Allen JD, Stabler TV, Frederiksen JW, Alzate O, Keil LG, Liu S, Lin FH, Truskey GA, Achneck HE. Increased yield of endothelial cells from peripheral blood for cell therapies and tissue engineering. Regen Med 2016; 10:447-60. [PMID: 26022764 DOI: 10.2217/rme.15.2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
AIM Peripheral blood-derived endothelial cells (pBD-ECs) are an attractive tool for cell therapies and tissue engineering, but have been limited by their low isolation yield. We increase pBD-EC yield via administration of the chemokine receptor type 4 antagonist AMD3100, as well as via a diluted whole blood incubation (DWBI). MATERIALS & METHODS Porcine pBD-ECs were isolated using AMD3100 and DWBI and tested for EC markers, acetylated LDL uptake, growth kinetics, metabolic activity, flow-mediated nitric oxide production and seeded onto titanium tubes implanted into vessels of pigs. RESULTS DWBI increased the yield of porcine pBD-ECs 6.6-fold, and AMD3100 increased the yield 4.5-fold. AMD3100-mobilized ECs were phenotypically indistinguishable from nonmobilized ECs. In porcine implants, the cells expressed endothelial nitric oxide synthase, reduced thrombin-antithrombin complex systemically and prevented thrombosis. CONCLUSION Administration of AMD3100 and the DWBI method both increase pBD-EC yield.
Collapse
Affiliation(s)
| | - Sa Do Kang
- 1Department of Surgery, Duke University Medical Center, NC, USA
| | | | - Justin M Haseltine
- 1Department of Surgery, Duke University Medical Center, NC, USA.,2Department of Biomedical Engineering, Duke University, NC, USA
| | - Lauren J Galinat
- 1Department of Surgery, Duke University Medical Center, NC, USA.,2Department of Biomedical Engineering, Duke University, NC, USA
| | | | - Tim A Carlon
- 1Department of Surgery, Duke University Medical Center, NC, USA.,2Department of Biomedical Engineering, Duke University, NC, USA
| | | | | | - Jose G Mantilla
- 1Department of Surgery, Duke University Medical Center, NC, USA
| | | | - Maria Noviani
- 1Department of Surgery, Duke University Medical Center, NC, USA.,4Duke-National University of Singapore Graduate Medical School, Singapore
| | - Jason D Allen
- 5Department of Community and Family Medicine, Duke University Medical Center, NC, USA.,6Duke Molecular Physiology Institute, Duke University Medical Center, NC, USA
| | - Thomas V Stabler
- 6Duke Molecular Physiology Institute, Duke University Medical Center, NC, USA
| | | | - Oscar Alzate
- 8University of North Carolina, School of Medicine, NC, USA
| | - Lukas G Keil
- 8University of North Carolina, School of Medicine, NC, USA
| | - Siyao Liu
- 8University of North Carolina, School of Medicine, NC, USA
| | - Fu-Hsiung Lin
- 1Department of Surgery, Duke University Medical Center, NC, USA
| | | | - Hardean E Achneck
- 1Department of Surgery, Duke University Medical Center, NC, USA.,9Department of Pathology, Duke University Medical Center, NC, USA
| |
Collapse
|
21
|
Preclinical models for in vitro mechanical loading of bone-derived cells. BONEKEY REPORTS 2015; 4:728. [PMID: 26331007 DOI: 10.1038/bonekey.2015.97] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 05/29/2015] [Indexed: 02/06/2023]
Abstract
It is well established that bone responds to mechanical stimuli whereby physical forces are translated into chemical signals between cells, via mechanotransduction. It is difficult however to study the precise cellular and molecular responses using in vivo systems. In vitro loading models, which aim to replicate forces found within the bone microenvironment, make the underlying processes of mechanotransduction accessible to the researcher. Direct measurements in vivo and predictive modeling have been used to define these forces in normal physiological and pathological states. The types of mechanical stimuli present in the bone include vibration, fluid shear, substrate deformation and compressive loading, which can all be applied in vitro to monolayer and three-dimensional (3D) cultures. In monolayer, vibration can be readily applied to cultures via a low-magnitude, high-frequency loading rig. Fluid shear can be applied to cultures in multiwell plates via a simple rocking platform to engender gravitational fluid movement or via a pump to cells attached to a slide within a parallel-plate flow chamber, which may be micropatterned for use with osteocytes. Substrate strain can be applied via the vacuum-driven FlexCell system or via a four-point loading jig. 3D cultures better replicate the bone microenvironment and can also be subjected to the same forms of mechanical stimuli as monolayer, including vibration, fluid shear via perfusion flow, strain or compression. 3D cocultures that more closely replicate the bone microenvironment can be used to study the collective response of several cell types to loading. This technical review summarizes the methods for applying mechanical stimuli to bone cells in vitro.
Collapse
|
22
|
A Review of Cell Adhesion Studies for Biomedical and Biological Applications. Int J Mol Sci 2015; 16:18149-84. [PMID: 26251901 PMCID: PMC4581240 DOI: 10.3390/ijms160818149] [Citation(s) in RCA: 526] [Impact Index Per Article: 58.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Revised: 06/21/2015] [Accepted: 06/24/2015] [Indexed: 01/13/2023] Open
Abstract
Cell adhesion is essential in cell communication and regulation, and is of fundamental importance in the development and maintenance of tissues. The mechanical interactions between a cell and its extracellular matrix (ECM) can influence and control cell behavior and function. The essential function of cell adhesion has created tremendous interests in developing methods for measuring and studying cell adhesion properties. The study of cell adhesion could be categorized into cell adhesion attachment and detachment events. The study of cell adhesion has been widely explored via both events for many important purposes in cellular biology, biomedical, and engineering fields. Cell adhesion attachment and detachment events could be further grouped into the cell population and single cell approach. Various techniques to measure cell adhesion have been applied to many fields of study in order to gain understanding of cell signaling pathways, biomaterial studies for implantable sensors, artificial bone and tooth replacement, the development of tissue-on-a-chip and organ-on-a-chip in tissue engineering, the effects of biochemical treatments and environmental stimuli to the cell adhesion, the potential of drug treatments, cancer metastasis study, and the determination of the adhesion properties of normal and cancerous cells. This review discussed the overview of the available methods to study cell adhesion through attachment and detachment events.
Collapse
|
23
|
Hovell CM, Sei YJ, Kim Y. Microengineered vascular systems for drug development. JOURNAL OF LABORATORY AUTOMATION 2015; 20:251-8. [PMID: 25424383 PMCID: PMC5663643 DOI: 10.1177/2211068214560767] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Indexed: 11/15/2022]
Abstract
Recent advances in microfabrication technologies and advanced biomaterials have allowed for the development of in vitro platforms that recapitulate more physiologically relevant cellular components and function. Microengineered vascular systems are of particular importance for the efficient assessment of drug candidates to physiological barriers lining microvessels. This review highlights advances in the development of microengineered vascular structures with an emphasis on the potential impact on drug delivery studies. Specifically, this article examines the development of models for the study of drug delivery to the central nervous system and cardiovascular system. We also discuss current challenges and future prospects of the development of microengineered vascular systems.
Collapse
Affiliation(s)
- Candice M Hovell
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Atlanta, GA, USA
| | - Yoshitaka J Sei
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Atlanta, GA, USA
| | - YongTae Kim
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Atlanta, GA, USA George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Atlanta, GA, USA Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, Atlanta, GA, USA Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
24
|
Davidoff S, Au D, Gale B, Brooks B, Brooks A. Maximizing Fibroblast Adhesion on Protein-Coated Surfaces Using Microfluidic Cell Printing. RSC Adv 2015; 5:104101-104109. [PMID: 26989480 PMCID: PMC4792286 DOI: 10.1039/c5ra18673k] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
translation of in vitro cell based assays to in vivo cellular response is imprecise at best. The advent of three-dimensional cell cultures in addition to bioreactor type microfluidics has improved the situation. However, these technical advances cannot be easily combined due to practical limitations. Development of a vertical microfluidic cell printer overcomes this obstacle, providing the ability to more closely recapitulate complex cellular environments and responses. As a proof of concept, we investigated the adhesion of fibroblasts under flow on protein-coated surfaces using a novel vertical microfluidic print head to isolate and manipulate both mechanical and biological factors as a model of fibroblast behavior during the foreign body response following implant insertion. A low flow rate with larger microfluidic channels onto a serum-coated surface has been determined to allow the highest density of viable fibroblasts to attach to the surface. While these insights into fibroblast surface attachment may lead to better material designs, the methods developed herein will certainly be useful as a biomaterials testing platform.
Collapse
Affiliation(s)
| | - D. Au
- Wasatch Microfluidics, Salt Lake City, UT 84103
| | - B.K. Gale
- Department of Mechanical Engineering, University of Utah, Salt Lake, UT 84112
| | - B.D. Brooks
- Wasatch Microfluidics, Salt Lake City, UT 84103
| | - A.E. Brooks
- Department of Pharmaceutics, University of Utah, Salt Lake City, UT 84112
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105
| |
Collapse
|
25
|
Baratchi S, Tovar-Lopez FJ, Khoshmanesh K, Grace MS, Darby W, Almazi J, Mitchell A, McIntyre P. Examination of the role of transient receptor potential vanilloid type 4 in endothelial responses to shear forces. BIOMICROFLUIDICS 2014; 8:044117. [PMID: 25379102 PMCID: PMC4189315 DOI: 10.1063/1.4893272] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 08/05/2014] [Indexed: 05/02/2023]
Abstract
Shear stress is the major mechanical force applied on vascular endothelial cells by blood flow, and is a crucial factor in normal vascular physiology and in the development of some vascular pathologies. The exact mechanisms of cellular mechano-transduction in mammalian cells and tissues have not yet been elucidated, but it is known that mechanically sensitive receptors and ion channels play a crucial role. This paper describes the use of a novel and efficient microfluidic device to study mechanically-sensitive receptors and ion channels in vitro, which has three independent channels from which recordings can be made and has a small surface area such that fewer cells are required than for conventional flow chambers. The contoured channels of the device enabled examination of a range of shear stresses in one field of view, which is not possible with parallel plate flow chambers and other previously used devices, where one level of flow-induced shear stress is produced per fixed flow-rate. We exposed bovine aortic endothelial cells to different levels of shear stress, and measured the resulting change in intracellular calcium levels ([Ca(2+)]i) using the fluorescent calcium sensitive dye Fluo-4AM. Shear stress caused an elevation of [Ca(2+)]i that was proportional to the level of shear experienced. The response was temperature dependant such that at lower temperatures more shear stress was required to elicit a given level of calcium signal and the magnitude of influx was reduced. We demonstrated that shear stress-induced elevations in [Ca(2+)]i are largely due to calcium influx through the transient receptor potential vanilloid type 4 ion channel.
Collapse
Affiliation(s)
| | - Francisco J Tovar-Lopez
- Microplatforms Research Group, School of Electrical and Computer Engineering, RMIT University , Victoria 3001, Australia
| | - Khashayar Khoshmanesh
- Microplatforms Research Group, School of Electrical and Computer Engineering, RMIT University , Victoria 3001, Australia
| | - Megan S Grace
- Health Innovations Research Institute, RMIT University , Victoria 3083, Australia
| | - William Darby
- Health Innovations Research Institute, RMIT University , Victoria 3083, Australia
| | - Juhura Almazi
- Health Innovations Research Institute, RMIT University , Victoria 3083, Australia
| | - Arnan Mitchell
- Microplatforms Research Group, School of Electrical and Computer Engineering, RMIT University , Victoria 3001, Australia
| | - Peter McIntyre
- Health Innovations Research Institute, RMIT University , Victoria 3083, Australia
| |
Collapse
|
26
|
Isolation of functional human endothelial cells from small volumes of umbilical cord blood. Ann Biomed Eng 2013; 41:2181-92. [PMID: 23604849 DOI: 10.1007/s10439-013-0807-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 04/04/2013] [Indexed: 12/11/2022]
Abstract
Endothelial cells (ECs) isolated from endothelial progenitor cells in blood have great potential as a therapeutic tool to promote vasculogenesis and angiogenesis and treat cardiovascular diseases. However, current methods to isolate ECs are limited by a low yield with few colonies appearing during isolation. In order to utilize blood-derived ECs for therapeutic applications, a simple method is needed that can produce a high yield of ECs from small volumes of blood without the addition of animal-derived products. For the first time, we show that human ECs can be isolated without the prior separation of blood components through the technique of diluted whole blood incubation (DWBI) utilizing commercially available human serum. We isolated ECs from small volumes of blood (~10 mL) via DWBI and characterized them with flow cytometry, immunohistochemistry, and uptake of DiI-labeled acetylated low density lipoprotein (DiI-Ac-LDL). These ECs are functional as demonstrated by their ability to form tubular networks in Matrigel, adhere and align with flow under physiological fluid shear stress, and produce increased nitric oxide under fluid flow. An average of 7.0 ± 2.5 EC colonies that passed all functional tests described above were obtained per 10 mL of blood as compared to only 0.3 ± 0.1 colonies with the traditional method based on density centrifugation. The time until first colony appearance was 8.3 ± 1.2 days for ECs isolated with the DWBI method and 12 ± 1.4 days for ECs isolated with the traditional isolation method. A simplified method, such as DWBI, in combination with advances in isolation yield could enable the use of blood-derived ECs in clinical practice.
Collapse
|