1
|
Qiu B, Pompe S, Xenaki KT, Di Maggio A, Moreno CB, van Bergen En Henegouwen PMP, Mastrobattista E, Oliveira S, Caiazzo M. Receptor-mediated transcytosis of nanobodies targeting the heparin-binding EGF-like growth factor in human blood-brain barrier models. J Control Release 2025; 383:113852. [PMID: 40393531 DOI: 10.1016/j.jconrel.2025.113852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/20/2025] [Accepted: 05/14/2025] [Indexed: 05/22/2025]
Abstract
Transport of molecules into the brain is regulated by the blood-brain barrier (BBB). Receptor-mediated transcytosis (RMT) is a targeted vesicular transport mechanism of brain endothelial cells that can be employed to specifically transport large therapeutic molecules into the brain. ProHB-EGF is the transmembrane precursor of the heparin-binding EGF-like growth factor (HB-EGF) present on the intraluminal side of the brain endothelial cells. This molecule is characterized as an internalizing transport receptor with so far no discovery of endogenous ligands. In this study, we describe the selection and characterization of two nanobodies (named F12 and H7) with high binding affinity for proHB-EGF and their BBB transcytosis potential were tested in vitro. For the human BBB model, we found that a polarized co-culture environment was crucial for the expression and cell surface display of proHB-EGF. The ability of F12 and H7 to pass the BBB via RMT was demonstrated in both a primary human brain microvascular endothelial cell-based BBB model and a human induced pluripotent stem cell (hiPSC)-derived iBBB model. Our studies demonstrate that the proHB-EGF targeting Nbs are promising BBB shuttle molecules for delivery of therapeutic molecules into the brain.
Collapse
Affiliation(s)
- Boning Qiu
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, the Netherlands.
| | - Sara Pompe
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Science Faculty, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Katerina T Xenaki
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, the Netherlands; Cell Biology, Neurobiology and Biophysics, Department of Biology, Science Faculty, Utrecht University, 3584 CH Utrecht, the Netherlands.
| | - Alessia Di Maggio
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Science Faculty, Utrecht University, 3584 CH Utrecht, the Netherlands.
| | - Clara Belinchón Moreno
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, the Netherlands; Cell Biology, Neurobiology and Biophysics, Department of Biology, Science Faculty, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Paul M P van Bergen En Henegouwen
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Science Faculty, Utrecht University, 3584 CH Utrecht, the Netherlands.
| | - Enrico Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, the Netherlands.
| | - Sabrina Oliveira
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, the Netherlands; Cell Biology, Neurobiology and Biophysics, Department of Biology, Science Faculty, Utrecht University, 3584 CH Utrecht, the Netherlands.
| | - Massimiliano Caiazzo
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, the Netherlands; Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy.
| |
Collapse
|
2
|
Chavarria D, Georges KA, O’Grady BJ, Hassan KK, Lippmann ES. Modular cone-and-plate device for mechanofluidic assays in Transwell inserts. Front Bioeng Biotechnol 2025; 13:1494553. [PMID: 39931136 PMCID: PMC11807968 DOI: 10.3389/fbioe.2025.1494553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/06/2025] [Indexed: 02/13/2025] Open
Abstract
In this work, we present a cost effective and open-source modular cone-and-plate (MoCAP) device that incorporates shear stress in the popular Transwell® insert system. This system acts as a lid that incorporates flow into 24-well Transwell® inserts while preserving the ability to conduct molecular profiling assays. Moreover, the MoCAP device can be rapidly reconfigured to test multiple shear stress profiles within a single device. To demonstrate the utility of the MoCAP, we conducted select assays on several different brain microvascular endothelial cell (BMEC) lines that comprise models of the blood-brain barrier (BBB), since shear stress can play an important role in BBB function. Our results characterize how shear stress modulates passive barrier function and GLUT1 expression across the different BMEC lines. Overall, we anticipate this low cost mechanofluidic device will be useful to the mechanobiology community.
Collapse
Affiliation(s)
- Daniel Chavarria
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
| | - Kissamy A. Georges
- Department of Bioengineering, University of Massachusetts Dartmouth, Dartmouth, MA, United States
| | - Brian J. O’Grady
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
| | - Khalid K. Hassan
- School for Science and Math at Vanderbilt, Vanderbilt University, Nashville, TN, United States
| | - Ethan S. Lippmann
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, United States
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, United States
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
3
|
Chebotarev O, Ugodnikov A, Simmons CA. Porous Membrane Electrical Cell-Substrate Impedance Spectroscopy for Versatile Assessment of Biological Barriers In Vitro. ACS APPLIED BIO MATERIALS 2024; 7:2000-2011. [PMID: 38447196 DOI: 10.1021/acsabm.4c00114] [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] [Indexed: 03/08/2024]
Abstract
Cell culture models of endothelial and epithelial barriers typically use porous membrane inserts (e.g., Transwell inserts) as a permeable substrate on which barrier cells are grown, often in coculture with other cell types on the opposite side of the membrane. Current methods to characterize barrier function in porous membrane inserts can disrupt the barrier or provide bulk measurements that cannot isolate barrier cell resistance alone. Electrical cell-substrate impedance sensing (ECIS) addresses these limitations, but its implementation on porous membrane inserts has been limited by costly manufacturing, low sensitivity, and lack of validation for barrier assessment. Here, we present porous membrane ECIS (PM-ECIS), a cost-effective method to adapt ECIS technology to porous substrate-based in vitro models. We demonstrate high fidelity patterning of electrodes on porous membranes that can be incorporated into well plates of a variety of sizes with excellent cell biocompatibility with mono- and coculture set ups. PM-ECIS provided sensitive, real-time measurement of isolated changes in endothelial cell barrier impedance with cell growth and barrier disruption. Barrier function characterized by PM-ECIS resistance correlated well with permeability coefficients obtained from simultaneous molecular tracer permeability assays performed on the same cultures, validating the device. Integration of ECIS into conventional porous cell culture inserts provides a versatile, sensitive, and automated alternative to current methods to measure barrier function in vitro, including molecular tracer assays and transepithelial/endothelial electrical resistance.
Collapse
Affiliation(s)
- Oleg Chebotarev
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Alisa Ugodnikov
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Craig A Simmons
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| |
Collapse
|
4
|
Bolden CT, Skibber MA, Olson SD, Zamorano Rojas M, Milewicz S, Gill BS, Cox CS. Validation and characterization of a novel blood-brain barrier platform for investigating traumatic brain injury. Sci Rep 2023; 13:16150. [PMID: 37752338 PMCID: PMC10522590 DOI: 10.1038/s41598-023-43214-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 09/21/2023] [Indexed: 09/28/2023] Open
Abstract
The Blood-Brain Barrier (BBB) is a highly-selective physiologic barrier responsible for maintaining cerebral homeostasis. Innovative in vitro models of the BBB are needed to provide useful insights into BBB function with CNS disorders like traumatic brain injury (TBI). TBI is a multidimensional and highly complex pathophysiological condition that requires intrinsic models to elucidate its mechanisms. Current models either lack fluidic shear stress, or neglect hemodynamic parameters important in recapitulating the human in vivo BBB phenotype. To address these limitations in the field, we developed a fluid dynamic novel platform which closely mimics these parameters. To validate our platform, Matrigel-coated Transwells were seeded with brain microvascular endothelial cells, both with and without co-cultured primary human astrocytes and bone-marrow mesenchymal stem cells. In this article we characterized BBB functional properties such as TEER and paracellular permeability. Our platform demonstrated physiologic relevant decreases in TEER in response to an ischemic environment, while directly measuring barrier fluid fluctuation. These recordings were followed with recovery, implying stability of the model. We also demonstrate that our dynamic platform is responsive to inflammatory and metabolic cues with resultant permeability coefficients. These results indicate that this novel dynamic platform will be a valuable tool for evaluating the recapitulating BBB function in vitro, screening potential novel therapeutics, and establishing a relevant paradigm to evaluate the pathophysiology of TBI.
Collapse
Affiliation(s)
- Christopher T Bolden
- Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
- Center for Translational Injury Research, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
| | - Max A Skibber
- Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Scott D Olson
- Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Miriam Zamorano Rojas
- Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Samantha Milewicz
- Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Brijesh S Gill
- Department of Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Charles S Cox
- Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
- Center for Translational Injury Research, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
| |
Collapse
|
5
|
Arshad M, Cheng S, van Reeuwijk M, Sherwin SJ, Weinberg PD. Modification of the swirling well cell culture model to alter shear stress metrics. Biotechnol Bioeng 2023; 120:1254-1268. [PMID: 36633017 PMCID: PMC10952219 DOI: 10.1002/bit.28331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 12/07/2022] [Accepted: 01/09/2023] [Indexed: 01/13/2023]
Abstract
Effects of hemodynamic shear stress on endothelial cells have been extensively investigated using the "swirling well" method, in which cells are cultured in dishes or multiwell plates placed on an orbital shaker. A wave rotates around the well, producing complex patterns of shear. The method allows chronic exposure to flow with high throughput at low cost but has two disadvantages: a number of shear stress characteristics change in a broadly similar way from the center to the edge of the well, and cells at one location in the well may release mediators into the medium that affect the behavior of cells at other locations, exposed to different shears. These properties make it challenging to correlate cell properties with shear. The present study investigated simple alterations to ameliorate these issues. Flows were obtained by numerical simulation. Increasing the volume of fluid in the well-altered dimensional but not dimensionless shear metrics. Adding a central cylinder to the base of the well-forced fluid to flow in a square toroidal channel and reduced multidirectionality. Conversely, suspending a cylinder above the base of the well made the flow highly multidirectional. Increasing viscosity in the latter model increased the magnitude of dimensional but not dimensionless metrics. Finally, tilting the well changed the patterns of different wall shear stress metrics in different ways. Collectively, these methods allow similar flows over most of the cells cultured and/or allow the separation of different shear metrics. A combination of the methods overcomes the limitations of the baseline model.
Collapse
Affiliation(s)
- Mehwish Arshad
- Department of BioengineeringImperial College LondonLondonUK
- Department of AeronauticsImperial College LondonLondonUK
| | - Shuyu Cheng
- Department of BioengineeringImperial College LondonLondonUK
| | - Maarten van Reeuwijk
- Department of Civil and Environmental EngineeringImperial College LondonLondonUK
| | | | | |
Collapse
|
6
|
Abdelkarim M, Perez-Davalos L, Abdelkader Y, Abostait A, Labouta HI. Critical design parameters to develop biomimetic organ-on-a-chip models for the evaluation of the safety and efficacy of nanoparticles. Expert Opin Drug Deliv 2023; 20:13-30. [PMID: 36440475 DOI: 10.1080/17425247.2023.2152000] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Organ-on-a-chip (OOC) models are based on microfluidics and can recapitulate the healthy and diseased microstructure of organs1 and tissues and the dynamic microenvironment inside the human body. However, the use of OOC models to evaluate the safety and efficacy of nanoparticles (NPs) is still in the early stages. AREAS COVERED The different design parameters of the microfluidic chip and the mechanical forces generated by fluid flow play a pivotal role in simulating the human environment. This review discusses the role of different key parameters on the performance of OOC models. These include the flow pattern, flow rate, shear stress (magnitude, rate, and distribution), viscosity of the media, and the microchannel dimensions and shape. We also discuss how the shear stress and other mechanical forces affect the transport of NPs across biological barriers, cell uptake, and their biocompatibility. EXPERT OPINION We describe several good practices and design parameters to consider for future OOC research. We submit that following these recommendations will help realize the full potential of the OOC models in the preclinical evaluation of novel therapies, including NPs.
Collapse
Affiliation(s)
- Mahmoud Abdelkarim
- Biomedical Engineering, University of Manitoba, R3T 5V6, Winnipeg, Manitoba, Canada.,College of Pharmacy, University of Manitoba, R3E 0T5, Winnipeg, Manitoba, Canada
| | - Luis Perez-Davalos
- College of Pharmacy, University of Manitoba, R3E 0T5, Winnipeg, Manitoba, Canada
| | - Yasmin Abdelkader
- College of Pharmacy, University of Manitoba, R3E 0T5, Winnipeg, Manitoba, Canada.,Department of Cell Biology, Biotechnology Research Institute, National Research Centre, 12622, Cairo, Egypt
| | - Amr Abostait
- College of Pharmacy, University of Manitoba, R3E 0T5, Winnipeg, Manitoba, Canada
| | - Hagar I Labouta
- Biomedical Engineering, University of Manitoba, R3T 5V6, Winnipeg, Manitoba, Canada.,College of Pharmacy, University of Manitoba, R3E 0T5, Winnipeg, Manitoba, Canada.,Children's Hospital Research Institute of Manitoba, R3E 3P4, Winnipeg, Manitoba, Canada.,Faculty of Pharmacy, Alexandria University, 21521, Alexandria, Egypt
| |
Collapse
|
7
|
Tian Y, Seeto WJ, Páez-Arias MA, Hahn MS, Lipke EA. Endothelial colony forming cell rolling and adhesion supported by peptide-grafted hydrogels. Acta Biomater 2022; 152:74-85. [PMID: 36031035 DOI: 10.1016/j.actbio.2022.08.047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 07/28/2022] [Accepted: 08/22/2022] [Indexed: 01/13/2023]
Abstract
The aim of this study was to investigate the ability of peptides and peptide combinations to support circulating endothelial colony forming cell (ECFC) rolling and adhesion under shear flow, informing biomaterial design in moving toward rapid cardiovascular device endothelialization. ECFCs have high proliferative capability and can differentiate into endothelial cells, making them a promising cell source for endothelialization. Both single peptides and peptide combinations designed to target integrins α4β1 and α5β1 were coupled to poly(ethylene glycol) hydrogels, and their performance was evaluated by monitoring velocity patterns during the ECFC rolling process, in addition to firm adhesion (capture). Tether percentage and velocity fluctuation, a parameter newly defined here, were found to be valuable in assessing cell rolling velocity patterns and when used in combination were able to predict cell capture. REDV-containing peptides binding integrin α4β1 have been previously shown to reduce ECFC rolling velocity but not to support firm adhesion. This study finds that the performance of REDV-containing peptides in facilitating ECFC dynamic adhesion and capture can be improved by combination with α5β1 integrin-binding peptides, which support ECFC static adhesion. Moreover, when similar in length, the peptide combinations may have synergistic effects in capturing ECFCs. With matching lengths, the peptide combinations including CRRETAWAC(cyclic)+REDV, P_RGDS+KSSP_REDV, and P_RGDS+P_REDV showed high values in both tether percentage and velocity fluctuation and improvement in ECFC capture compared to the single peptides at the shear rate of 20 s-1. These newly identified peptide combinations have the potential to be used as vascular device coatings to recruit ECFCs. STATEMENT OF SIGNIFICANCE: Restoration of functional endothelium following placement of stents and vascular grafts is critical for maintaining long-term patency. Endothelial colony forming cells (ECFCs) circulating in blood flow are a valuable cell source for rapid endothelialization. Here we identify and test novel peptides and peptide combinations that can potentially be used as coatings for vascular devices to support rolling and capture of ECFCs from flow. In addition to the widely used assessment of final ECFC adhesion, we also recorded the rolling process to quantitatively evaluate the interaction between ECFCs and the peptides, obtaining detailed performance of the peptides and gaining insight into effective capture molecule design. Peptide combinations targeting both integrin α4β1 and integrin α5β1 showed the highest percentages of ECFC capture.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Chemical Engineering, Auburn University, 212 Ross Hall, Auburn, AL, 36849, USA
| | - Wen J Seeto
- Department of Chemical Engineering, Auburn University, 212 Ross Hall, Auburn, AL, 36849, USA
| | - Mayra A Páez-Arias
- Department of Chemical Engineering, Auburn University, 212 Ross Hall, Auburn, AL, 36849, USA
| | - Mariah S Hahn
- Biomedical Engineering Department, Rensselaer Polytechnic Institute, Troy, NY, 12180-3590, USA
| | - Elizabeth A Lipke
- Department of Chemical Engineering, Auburn University, 212 Ross Hall, Auburn, AL, 36849, USA.
| |
Collapse
|
8
|
Nandan S, Schiavi-Tritz J, Hellmuth R, Dunlop C, Vaughan TJ, Dolan EB. Design and Verification of a Novel Perfusion Bioreactor to Evaluate the Performance of a Self-Expanding Stent for Peripheral Artery Applications. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 4:886458. [PMID: 35800467 PMCID: PMC9253816 DOI: 10.3389/fmedt.2022.886458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Endovascular stenting presents a promising approach to treat peripheral artery stenosis. However, a significant proportion of patients require secondary interventions due to complications such as in-stent restenosis and late stent thrombosis. Clinical failure of stents is not only attributed to patient factors but also on endothelial cell (EC) injury response, stent deployment techniques, and stent design. Three-dimensional in vitro bioreactor systems provide a valuable testbed for endovascular device assessment in a controlled environment replicating hemodynamic flow conditions found in vivo. To date, very few studies have verified the design of bioreactors based on applied flow conditions and their impact on wall shear stress, which plays a key role in the development of vascular pathologies. In this study, we develop a computationally informed bioreactor capable of capturing responses of human umbilical vein endothelial cells seeded on silicone tubes subjected to hemodynamic flow conditions and deployment of a self-expanding nitinol stents. Verification of bioreactor design through computational fluid dynamics analysis confirmed the application of pulsatile flow with minimum oscillations. EC responses based on morphology, nitric oxide (NO) release, metabolic activity, and cell count on day 1 and day 4 verified the presence of hemodynamic flow conditions. For the first time, it is also demonstrated that the designed bioreactor is capable of capturing EC responses to stent deployment beyond a 24-hour period with this testbed. A temporal investigation of EC responses to stent implantation from day 1 to day 4 showed significantly lower metabolic activity, EC proliferation, no significant changes to NO levels and EC's aligning locally to edges of stent struts, and random orientation in between the struts. These EC responses were indicative of stent-induced disturbances to local hemodynamics and sustained EC injury response contributing to neointimal growth and development of in-stent restenosis. This study presents a novel computationally informed 3D in vitro testbed to evaluate stent performance in presence of hemodynamic flow conditions found in native peripheral arteries and could help to bridge the gap between the current capabilities of 2D in vitro cell culture models and expensive pre-clinical in vivo models.
Collapse
Affiliation(s)
- Swati Nandan
- Biomedical Engineering and Biomechanics Research Centre (BioMEC), School of Engineering, College of Science and Engineering, National University of Ireland Galway, Galway, Ireland
- Vascular Flow Technology, Dundee, United Kingdom
| | - Jessica Schiavi-Tritz
- Biomedical Engineering and Biomechanics Research Centre (BioMEC), School of Engineering, College of Science and Engineering, National University of Ireland Galway, Galway, Ireland
| | | | - Craig Dunlop
- Vascular Flow Technology, Dundee, United Kingdom
| | - Ted J. Vaughan
- Biomedical Engineering and Biomechanics Research Centre (BioMEC), School of Engineering, College of Science and Engineering, National University of Ireland Galway, Galway, Ireland
- *Correspondence: Ted J. Vaughan
| | - Eimear B. Dolan
- Biomedical Engineering and Biomechanics Research Centre (BioMEC), School of Engineering, College of Science and Engineering, National University of Ireland Galway, Galway, Ireland
- Eimear B. Dolan
| |
Collapse
|
9
|
Guerra DB, Oliveira EMN, Sonntag AR, Sbaraine P, Fay AP, Morrone FB, Papaléo RM. Intercomparison of radiosensitization induced by gold and iron oxide nanoparticles in human glioblastoma cells irradiated by 6 MV photons. Sci Rep 2022; 12:9602. [PMID: 35688846 PMCID: PMC9187689 DOI: 10.1038/s41598-022-13368-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/22/2022] [Indexed: 12/04/2022] Open
Abstract
In this work, an intercomparison of sensitization effects produced by gold (GNP) and dextran-coated iron oxide (SPION-DX) nanoparticles in M059J and U87 human glioblastoma cells was performed using 6 MV-photons. Three variables were mapped: the nanoparticle material, treatment concentration, and cell radiosensitivity. For U87, GNP treatments resulted in high sensitization enhancement ratios (SER[Formula: see text] up to 2.04). More modest effects were induced by SPION-DX, but still significant reductions in survival were achieved (maximum SER[Formula: see text] ). For the radiosensitive M059J, sensitization by both NPs was poor. SER[Formula: see text] increased with the degree of elemental uptake in the cells, but not necessarily with treatment concentration. For GNP, where exposure concentration and elemental uptake were found to be proportional, SER[Formula: see text] increased linearly with concentration in both cell lines. For SPION-DX, saturation of sensitization enhancement and metal uptake occurred at high exposures. Fold change in the [Formula: see text] ratios extracted from survival curves are reduced by the presence of SPION-DX but strongly increased by GNPs , suggesting that sensitization by GNPs occurs mainly via promotion of lethal damage, while for SPION-DX repairable damage dominates. The NPs were more effective in eliminating the radioresistant glioblastoma cells, an interesting finding, as resistant cells are key targets to improve treatment outcome.
Collapse
Affiliation(s)
- Danieli B Guerra
- Interdisciplinary Center of Nanoscience and Micro-Nanotechnology, School of Technology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, 90619-900, Brazil.
| | - Elisa M N Oliveira
- Interdisciplinary Center of Nanoscience and Micro-Nanotechnology, School of Technology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, 90619-900, Brazil
| | - Amanda R Sonntag
- Interdisciplinary Center of Nanoscience and Micro-Nanotechnology, School of Technology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, 90619-900, Brazil
| | - Patricia Sbaraine
- Division of Radiotherapy, São Lucas Hospital of PUCRS, Porto Alegre, 90610-000, Brazil
| | - Andre P Fay
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, 90619-900, Brazil
| | - Fernanda B Morrone
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, 90619-900, Brazil
| | - Ricardo M Papaléo
- Interdisciplinary Center of Nanoscience and Micro-Nanotechnology, School of Technology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, 90619-900, Brazil
| |
Collapse
|
10
|
Camasão DB, Li L, Drouin B, Lau C, Reinhardt DP, Mantovani D. Physiologically relevant platform for an advanced in vitro model of the vascular wall: focus on in situ fabrication and mechanical maturation. IN VITRO MODELS 2022; 1:179-195. [PMID: 39872805 PMCID: PMC11756475 DOI: 10.1007/s44164-022-00012-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/27/2022] [Accepted: 02/28/2022] [Indexed: 01/30/2025]
Abstract
The mechanical stimulation applied on engineered vascular constructs in perfusion bioreactors has been shown to be beneficial for their maturation. The level of mechanical stimulation applied on these constructs depends on the flow parameters of the circuit (e.g., fluid viscosity, flow rate, frequency, and pressure). As a group, these parameters are often overlooked in the literature, and they rarely meet the physiological values of the blood flow. For this reason, the level of circumferential stretching and shear stress that blood vessels experience in the human body are rarely reproduced. In this work, we reported the development of a physiologically relevant platform for (1) the in situ fabrication of vascular wall models based on collagen gel, and (2) their maturation under physiological levels of mechanical stimulation in a perfusion bioreactor (pulsatile flow rate of 100 mL/min, frequency of 1 Hz, pressure of 80-120 mmHg, and viscosity of 4 cP). One week of dynamic maturation oriented the seeded cells into the circumferential direction, increased the deposition of collagen and key elastin fiber-related proteins, and improved the mechanical properties in terms of tensile equilibrium elastic modulus (by 110%) and strength at break (by 63%) when compared to the static condition. In addition to the maturation study under selected physiologically relevant mechanical stimulation (such as adult, fetal, child, and hypertension conditions), the platform might also be used as a relevant in vitro testing system for new drugs or pro-active coating to medical devices (such as stents, endografts, and vascular prostheses) expected to trigger specific mechanisms or activities in vascular cells composing the arterial wall.
Collapse
Affiliation(s)
- Dimitria B. Camasão
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Department of Min-Met-Materials Engineering, Research Center of CHU de Québec, Division of Regenerative Medicine, Laval University, Québec, QC G1V 0A6 Canada
| | - Ling Li
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, Faculty of Dentistry, McGill University, Montreal, QC H3A 0C7 Canada
| | - Bernard Drouin
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Department of Min-Met-Materials Engineering, Research Center of CHU de Québec, Division of Regenerative Medicine, Laval University, Québec, QC G1V 0A6 Canada
| | - Cori Lau
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, Faculty of Dentistry, McGill University, Montreal, QC H3A 0C7 Canada
| | - Dieter P. Reinhardt
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, Faculty of Dentistry, McGill University, Montreal, QC H3A 0C7 Canada
| | - Diego Mantovani
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Department of Min-Met-Materials Engineering, Research Center of CHU de Québec, Division of Regenerative Medicine, Laval University, Québec, QC G1V 0A6 Canada
| |
Collapse
|
11
|
Liu B, Wang X, Jiang L, Xu J, Zohar Y, Yao G. Extracellular Fluid Flow Induces Shallow Quiescence Through Physical and Biochemical Cues. Front Cell Dev Biol 2022; 10:792719. [PMID: 35281101 PMCID: PMC8912726 DOI: 10.3389/fcell.2022.792719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
The balance between cell quiescence and proliferation is fundamental to tissue physiology and homeostasis. Recent studies have shown that quiescence is not a passive and homogeneous state but actively maintained and heterogeneous. These cellular characteristics associated with quiescence were observed primarily in cultured cells under a static medium. However, cells in vivo face different microenvironmental conditions, particularly, under interstitial fluid flows distributed through extracellular matrices. Interstitial fluid flow exerts shear stress on cells and matrix strain, and results in continuous replacement of extracellular factors. In this study, we analyzed individual cells under varying fluid flow rates in microfluidic devices. We found quiescence characteristics previously identified under conventional static medium, including serum signal-dependant quiescence entry and exit and time-dependant quiescence deepening, are also present under continuous fluid flow. Furthermore, increasing the flow rate drives cells to shallower quiescence and become more likely to reenter the cell cycle upon growth stimulation. This effect is due to flow-induced physical and biochemical cues. Specifically, increasing shear stress or extracellular factor replacement individually, without altering other parameters, results in shallow quiescence. We show our experimental results can be quantitatively explained by a mathematical model connecting extracellular fluid flow to an Rb-E2f bistable switch that regulates the quiescence-to-proliferation transition. Our findings uncover a previously unappreciated mechanism that likely underlies the heterogeneous responses of quiescent cells for tissue repair and regeneration in different physiological tissue microenvironments.
Collapse
Affiliation(s)
- Bi Liu
- School of Pharmacy, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, China
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, United States
| | - Xia Wang
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, United States
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Linan Jiang
- Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ, United States
- *Correspondence: Linan Jiang, ; Guang Yao,
| | - Jianhua Xu
- School of Pharmacy, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, China
| | - Yitshak Zohar
- Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ, United States
| | - Guang Yao
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, United States
- *Correspondence: Linan Jiang, ; Guang Yao,
| |
Collapse
|
12
|
Siren EMJ, Luo HD, Bajaj S, MacKenzie J, Daneshi M, Martinez DM, Conway EM, Cheung KC, Kizhakkedathu JN. An improved in vitro model for studying the structural and functional properties of the endothelial glycocalyx in arteries, capillaries and veins. FASEB J 2021; 35:e21643. [PMID: 33977574 DOI: 10.1096/fj.201802376rrrr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 12/30/2022]
Abstract
The endothelial glycocalyx is a dynamic structure integral to blood vessel hemodynamics and capable of tightly regulating a range of biological processes (ie, innate immunity, inflammation, and coagulation) through dynamic changes in its composition of the brush structure. Evaluating the specific roles of the endothelial glycocalyx under a range of pathophysiologic conditions has been a challenge in vitro as it is difficult to generate functional glycocalyces using commonly employed 2D cell culture models. We present a new multi-height microfluidic platform that promotes the growth of functional glycocalyces by eliciting unique shear stress forces over a continuous human umbilical vein endothelial cell monolayer at magnitudes that recapitulate the physical environment in arterial, capillary and venous regions of the vasculature. Following 72 hours of shear stress, unique glycocalyx structures formed within each region that were distinct from that observed in short (3 days) and long-term (21 days) static cell culture. The model demonstrated glycocalyx-specific properties that match the characteristics of the endothelium in arteries, capillaries and veins, with respect to surface protein expression, platelet adhesion, lymphocyte binding and nanoparticle uptake. With artery-to-capillary-to-vein transition on a continuous endothelial monolayer, this in vitro platform is an improved system over static cell culture for more effectively studying the role of the glycocalyx in endothelial biology and disease.
Collapse
Affiliation(s)
- Erika M J Siren
- Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.,Department of Chemistry, University of British Columbia, Vancouver, BC, Canada
| | - Haiming D Luo
- Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.,Department of Chemistry, University of British Columbia, Vancouver, BC, Canada
| | - Sargun Bajaj
- Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Jordan MacKenzie
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC, Canada.,Department of Mathematics, University of British Columbia, Vancouver, BC, Canada
| | - Masoud Daneshi
- Department of Mathematics, University of British Columbia, Vancouver, BC, Canada
| | - D Mark Martinez
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC, Canada.,Department of Mathematics, University of British Columbia, Vancouver, BC, Canada
| | - Edward M Conway
- Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.,Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Karen C Cheung
- Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.,Department of Electrical and Computer Engineering, University of British Columbia, Vancouver, BC, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Jayachandran N Kizhakkedathu
- Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.,Department of Chemistry, University of British Columbia, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
13
|
Gabr MT, Balupuri A, Kang NS. High-Throughput Platform for Real-Time Monitoring of ATP-Generating Enzymes in Living Cells Based on a Lanthanide Probe. ACS Sens 2020; 5:1872-1876. [PMID: 32610895 DOI: 10.1021/acssensors.0c00897] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Remarkable variation between cell-free and cellular measurements of enzyme activity triggered the unmet need to develop tools for monitoring enzyme activity in living cells. Such tools will advance our understanding of the biological functions of enzymes and their potential impact on drug discovery. We report in this study a universal assay for monitoring ATP-generating enzymes in living cells using a self-assembled Tb3+ complex probe. Modulation of the rheological properties of cell culture media enabled shifting the lifetime of the Tb3+ complex in the presence of ATP from micro-to-millisecond range. Based on the response of the Tb3+ complex to ATP, cellular assays for 5 ATP-generating enzymes were developed. Remarkably, assessment of the activity of these enzymes in living cells is made possible for the first time. The pyruvate kinase M2 (PKM2) assay has been optimized for high-throughput screening (HTS) and further implemented in the identification of novel scaffolds as PKM2 inhibitors.
Collapse
Affiliation(s)
- Moustafa T. Gabr
- Department of Radiology, Stanford University, Stanford, California 94305, United States
| | - Anand Balupuri
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 305-764, Republic of Korea
| | - Nam Sook Kang
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 305-764, Republic of Korea
| |
Collapse
|
14
|
Ruano-Salguero JS, Lee KH. Antibody transcytosis across brain endothelial-like cells occurs nonspecifically and independent of FcRn. Sci Rep 2020; 10:3685. [PMID: 32111886 PMCID: PMC7048754 DOI: 10.1038/s41598-020-60438-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/04/2020] [Indexed: 11/09/2022] Open
Abstract
The blood-brain barrier (BBB) hinders the brain delivery of therapeutic immunoglobulin γ (IgG) antibodies. Evidence suggests that IgG-specific processing occurs within the endothelium of the BBB, but any influence on transcytosis remains unclear. Here, involvement of the neonatal Fc receptor (FcRn), which mediates IgG recycling and transcytosis in peripheral endothelium, was investigated by evaluating the transcytosis of IgGs with native or reduced FcRn engagement across human induced pluripotent stem cell-derived brain endothelial-like cells. Despite differential trafficking, the permeability of all tested IgGs were comparable and remained constant irrespective of concentration or competition with excess IgG, suggesting IgG transcytosis occurs nonspecifically and originates from fluid-phase endocytosis. Comparison with the receptor-enhanced permeability of transferrin indicates that the phenomena observed for IgG is ubiquitous for most macromolecules. However, increased permeability was observed for macromolecules with biophysical properties known to engage alternative endocytosis mechanisms, highlighting the importance of biophysical characterizations in assessing transcytosis mechanisms.
Collapse
Affiliation(s)
- John S Ruano-Salguero
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, 19716, USA
- Delaware Biotechnology Institute, University of Delaware, Newark, DE, 19711, USA
| | - Kelvin H Lee
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, 19716, USA.
- Delaware Biotechnology Institute, University of Delaware, Newark, DE, 19711, USA.
| |
Collapse
|
15
|
Kadam AA, Gersch RP, Rosengart TK, Frame MD. Inflammatory monocyte response due to altered wall shear stress in an isolated femoral artery model. J Biol Methods 2019; 6:e109. [PMID: 31453258 PMCID: PMC6706128 DOI: 10.14440/jbm.2019.274] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 11/12/2018] [Accepted: 11/28/2018] [Indexed: 01/10/2023] Open
Abstract
Arteriogenesis (collateral formation) is accompanied by a pro-inflammatory state that may be related to the wall shear stress (WSS) within the neo-collateral vessels. Examining the pro-inflammatory component in situ or in vivo is complex. In an ex vivo mouse femoral artery perfusion model, we examined the effect of wall shear stress on pro-arteriogenic inflammatory markers and monocyte adhesion. In a femoral artery model with defined pulsatile flow, WSS was controlled (at physiological stress, 1.4×, and 2× physiological stress) during a 24 h perfusion before gene expression levels and monocyte adhesion were assessed. Significant upregulation of expression was found for the cytokine TNFα, adhesion molecule ICAM-1, growth factor TGFβ, and the transcription factor Egr-1 at varying levels of increased WSS compared to physiological control. Further, trends toward upregulation were found for FGF-2, the cytokine MCP-1 and adhesion molecules VCAM-1 and P-selectin with increased WSS. Finally, monocytes adhesion increased in response to increased WSS. We have developed a murine femoral artery model for studying changes in WSS ex vivo and show that the artery responds by upregulating inflammatory cytokines, adhesion molecules and growth factors consistent with previous in vivo findings.
Collapse
Affiliation(s)
- Aparna A Kadam
- Department of Biomedical Engineering, Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794-5281, USA
| | - Robert P Gersch
- Department of Surgery, Stony Brook University, Stony Brook, NY 11794-5281, USA
| | - Todd K Rosengart
- Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mary D Frame
- Department of Biomedical Engineering, Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794-5281, USA
| |
Collapse
|
16
|
Wu Q, Miao T, Feng T, Yang C, Guo Y, Li H. Dextran‑coated superparamagnetic iron oxide nanoparticles activate the MAPK pathway in human primary monocyte cells. Mol Med Rep 2018; 18:564-570. [PMID: 29749448 DOI: 10.3892/mmr.2018.8972] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Accepted: 04/25/2018] [Indexed: 02/05/2023] Open
Abstract
With the increase in applications of superparamagnetic iron oxide nanoparticles (SPIONs) in biomedicine, it is essential to investigate the bio‑security of these nanoparticles, especially with respect to the human immune system. In the present study, the biological effects of dextran‑coated superparamagnetic iron oxide nanoparticles (Dex‑SPIONs) on human primary monocyte cells were evaluated. The results of the present study demonstrated that Dex‑SPIONs can be identified in phagosomes or freed in the cytoplasm and did not affect cell viability or induce apoptosis. Notably, there were certain bulky vacuoles and a number of pseudopodia from the cell membrane, suggesting potential activation of human monocyte cells. In addition, the expression levels of pro‑inflammatory cytokines interleukin (IL)‑1β and tumor necrosis factor (TNF)‑α were also increased following treatment with Dex‑SPIONs. Simultaneously, the phosphorylation levels of mitogen‑activated protein kinase (MAPK) p38, c‑Jun N‑terminal kinase 1 and extracellular signal regulated kinase were markedly enhanced following nanoparticle exposure and MAPK inhibitors could abate the production of IL‑1β and TNF‑α. The results of the present study demonstrated that Dex‑SPIONs could activate human monocyte cells and that activation of MAPK pathway may be involved in these effects.
Collapse
Affiliation(s)
- Qihong Wu
- Key Laboratory of Obstetrics and Gynecology and Pediatric Disease and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Tianyu Miao
- Department of Vascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ting Feng
- Key Laboratory of Obstetrics and Gynecology and Pediatric Disease and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Chuan Yang
- Key Laboratory of Obstetrics and Gynecology and Pediatric Disease and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yingkun Guo
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hong Li
- Key Laboratory of Obstetrics and Gynecology and Pediatric Disease and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
17
|
Wu Q, Jin R, Feng T, Liu L, Yang L, Tao Y, Anderson JM, Ai H, Li H. Iron oxide nanoparticles and induced autophagy in human monocytes. Int J Nanomedicine 2017; 12:3993-4005. [PMID: 28603414 PMCID: PMC5457122 DOI: 10.2147/ijn.s135189] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Superparamagnetic iron oxide nanoparticles have been widely used in biomedical applications, but understanding of their interactions with the biological immune system is relatively limited. This work is focused on dextran-coated iron oxide nanoparticles and their induced autophagy in human monocytes. We found that these nanoparticles can be taken up by human monocytes, followed by localization within vesicles or free in cytoplasm. Autophagosome formation was observed with increased expression of LC3II protein, the specific marker of autophagy. The autophagy substrate p62 was degraded in a dose-dependent manner, and autophagy was blocked by autophagy (or lysosome) inhibitors alone or along with iron oxide nanoparticles, indicating that autophagosome accumulation was mainly due to autophagy induction, rather than blockade of autophagy flux. Interestingly, iron oxide nanoparticles increased the viability of human monocytes, but the mechanism was not clear. We further found that inhibition of autophagy mostly attenuated the survival of cells, with acceleration of the inflammation induced by these nanoparticles. Taken together, autophagic activation in human monocytes may play a protective role against the cytotoxicity of iron oxide nanoparticles.
Collapse
Affiliation(s)
- QiHong Wu
- Key Laboratory of Obstetrics, Gynecology, Pediatric Disease, and Birth Defects, Ministry of Education, West China Second University Hospital
| | - RongRong Jin
- National Engineering Research Center for Biomaterials
| | - Ting Feng
- Key Laboratory of Obstetrics, Gynecology, Pediatric Disease, and Birth Defects, Ministry of Education, West China Second University Hospital
| | - Li Liu
- National Engineering Research Center for Biomaterials
| | - Li Yang
- National Engineering Research Center for Biomaterials
| | - YuHong Tao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - James M Anderson
- Department of Biomedical Engineering.,Department of Pathology, Case Western Reserve University, Cleveland, OH, US
| | - Hua Ai
- National Engineering Research Center for Biomaterials.,Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Li
- Key Laboratory of Obstetrics, Gynecology, Pediatric Disease, and Birth Defects, Ministry of Education, West China Second University Hospital.,Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Glycocalyx Degradation Induces a Proinflammatory Phenotype and Increased Leukocyte Adhesion in Cultured Endothelial Cells under Flow. PLoS One 2016; 11:e0167576. [PMID: 27907146 PMCID: PMC5132265 DOI: 10.1371/journal.pone.0167576] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 11/16/2016] [Indexed: 12/30/2022] Open
Abstract
Leukocyte adhesion to the endothelium is an early step in the pathogenesis of atherosclerosis. Effective adhesion requires the binding of leukocytes to their cognate receptors on the surface of endothelial cells. The glycocalyx covers the surface of endothelial cells and is important in the mechanotransduction of shear stress. This study aimed to identify the molecular mechanisms underlying the role of the glycocalyx in leukocyte adhesion under flow. We performed experiments using 3-D cell culture models, exposing human abdominal aortic endothelial cells to steady laminar shear stress (10 dynes/cm2 for 24 hours). We found that with the enzymatic degradation of the glycocalyx, endothelial cells developed a proinflammatory phenotype when exposed to uniform steady shear stress leading to an increase in leukocyte adhesion. Our results show an up-regulation of ICAM-1 with degradation compared to non-degraded controls (3-fold increase, p<0.05) and we attribute this effect to a de-regulation in NF-κB activity in response to flow. These results suggest that the glycocalyx is not solely a physical barrier to adhesion but rather plays an important role in governing the phenotype of endothelial cells, a key determinant in leukocyte adhesion. We provide evidence for how the destabilization of this structure may be an early and defining feature in the initiation of atherosclerosis.
Collapse
|
19
|
Wolf F, Vogt F, Schmitz-Rode T, Jockenhoevel S, Mela P. Bioengineered vascular constructs as living models for in vitro cardiovascular research. Drug Discov Today 2016; 21:1446-1455. [PMID: 27126777 DOI: 10.1016/j.drudis.2016.04.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 04/01/2016] [Accepted: 04/18/2016] [Indexed: 12/20/2022]
Abstract
Cardiovascular diseases represent the most common cause of morbidity and mortality worldwide. In this review, we explore the potential of bioengineered vascular constructs as living models for in vitro cardiovascular research to advance the current knowledge of pathophysiological processes and support the development of clinical therapies. Bioengineered vascular constructs capable of recapitulating the cellular and mechanical environment of native vessels represent a valuable platform to study cellular interactions and signaling cascades, test drugs and medical devices under (patho)physiological conditions, with the additional potential benefit of reducing the number of animals required for preclinical testing.
Collapse
Affiliation(s)
- Frederic Wolf
- Department of Tissue Engineering & Textile Implants, Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Pauwelsstraße 20, 52074 Aachen, Germany
| | - Felix Vogt
- Department of Cardiology, Pulmonology, Intensive Care and Vascular Medicine, Medical Faculty, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Thomas Schmitz-Rode
- Department of Tissue Engineering & Textile Implants, Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Pauwelsstraße 20, 52074 Aachen, Germany; Department of Cardiovascular Engineering, Institute of Applied Medical Engineering, Helmholtz Institute Aachen, RWTH Aachen University, Pauwelsstraße 20, 52074 Aachen, Germany
| | - Stefan Jockenhoevel
- Department of Tissue Engineering & Textile Implants, Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Pauwelsstraße 20, 52074 Aachen, Germany; Institut für Textiltechnik, RWTH Aachen University, Otto-Blumenthal-Str. 1, 52074 Aachen, Germany; Aachen-Maastricht Institute for Biobased Materials, Maastricht University at Chemelot Campus, Urmonderbaan 22, 6167 RD Geleen, The Netherlands.
| | - Petra Mela
- Department of Tissue Engineering & Textile Implants, Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Pauwelsstraße 20, 52074 Aachen, Germany
| |
Collapse
|
20
|
Franzoni M, Cattaneo I, Ene-Iordache B, Oldani A, Righettini P, Remuzzi A. Design of a cone-and-plate device for controlled realistic shear stress stimulation on endothelial cell monolayers. Cytotechnology 2016; 68:1885-96. [PMID: 26754843 DOI: 10.1007/s10616-015-9941-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 12/21/2015] [Indexed: 11/29/2022] Open
Abstract
Endothelial cells are constantly exposed to blood flow and the resulting frictional force, the wall shear stress, varies in magnitude and direction with time, depending on vasculature geometry. Previous studies have shown that the structure and function of endothelial cells, and ultimately of the vessel wall, are deeply affected by the nature of wall shear stress waveforms. To investigate the in vitro effects of these stimuli, we developed a compact, programmable, real-time operated system based on cone-and-plate geometry, that can be used within a standard cell incubator. To verify the capability to replicate realistic shear stress waveforms, we calculated both analytically and numerically to what extent the system is able to correctly deliver the stimuli defined by the user at plate level. Our results indicate that for radii greater than 25 mm, the shear stress is almost uniform and directly proportional to cone rotation velocity. We further established that using a threshold of 10 Hz of wall shear stress waveform frequency components, oscillating flow conditions can be reproduced on cell monolayer surface. Finally, we verified the capability of the system to perform long-term flow exposure experiments ensuring sterility and cell culture viability on human umbilical vein endothelial cells exposed to unidirectional and oscillating shear stress. In conclusion, the system we developed is a highly dynamic, easy to handle, and able to generate pulsatile and unsteady oscillating wall shear stress waveforms. This system can be used to investigate the effects of realistic stimulations on endothelial cells, similar to those exerted in vivo by blood flow.
Collapse
Affiliation(s)
- Marco Franzoni
- Department of Biomedical Engineering, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Via Stezzano, 87, 24126, Bergamo, Italy
| | - Irene Cattaneo
- Department of Biomedical Engineering, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Via Stezzano, 87, 24126, Bergamo, Italy
| | - Bogdan Ene-Iordache
- Department of Biomedical Engineering, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Via Stezzano, 87, 24126, Bergamo, Italy
| | - Alberto Oldani
- Department of Management, Information and Production Engineering, University of Bergamo, Viale Marconi 4, 24144, Dalmine, BG, Italy
| | - Paolo Righettini
- Department of Management, Information and Production Engineering, University of Bergamo, Viale Marconi 4, 24144, Dalmine, BG, Italy
| | - Andrea Remuzzi
- Department of Biomedical Engineering, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Via Stezzano, 87, 24126, Bergamo, Italy. .,Department of Management, Information and Production Engineering, University of Bergamo, Viale Marconi 4, 24144, Dalmine, BG, Italy.
| |
Collapse
|
21
|
Avari H, Savory E, Rogers KA. An In Vitro Hemodynamic Flow System to Study the Effects of Quantified Shear Stresses on Endothelial Cells. Cardiovasc Eng Technol 2015; 7:44-57. [PMID: 26621672 DOI: 10.1007/s13239-015-0250-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 11/20/2015] [Indexed: 11/27/2022]
Abstract
Numerous in vitro systems have previously been developed and employed for studying the effects of hemodynamics on endothelial cell (EC) dysfunction. In the majority of that work, accurate flow quantification (e.g., uniformity of the flow over the ECs) remains elusive and wall shear stress (WSS) quantifications are determined using theoretical relationships (without considering the flow channel aspect ratio effects). In addition, those relationships are not applicable to flows other than steady laminar cases. The present work discusses the development of a novel hemodynamic flow system for studying the effects of various well-quantified flow regimes over ECs. The current work presents a novel hemodynamic flow system applying the concept of a parallel plate flow chamber (PPFC) with live microscopy access for studying the effects of quantified WSS on ECs. A range of steady laminar, pulsatile (carotid wave form) and low-Reynolds number turbulent WSSs were quantified through velocity field measurements by a laser Doppler velocimetry (LDV) system, to validate the functionality of the current hemodynamic flow system. Uniformity of the flow across the channel width can be analyzed with the current system (e.g., the flow was uniform across about 65-75% of the channel width for the steady cases). The WSS obtained from the experiments had higher values in almost all of the cases when compared to the most commonly-used theoretical solution (9% < error < 16%), whereas another relationship, which considers the channel dimensions, had better agreement with the experimental results (1% < error < 8%). Additionally, the latter relationship predicted the uniform flow region in the PPFC with an average difference of <5% when compared to the experimental results. The experimental data also showed that the WSS at various locations (D, E and F) at the test section differed by less than 4% for the laminar cases representing a fully developed flow. WSS was also determined for a low-Re (Re = 2750) turbulent flow using (1) the Reynolds shears stress and (2) the time-averaged velocity profile gradient at the wall, with a good agreement (differences <16%) between the two where the first method returned a higher value than the second. Porcine aortic endothelial cell (PAEC) viability in the system and morphological cell response to laminar WSS of about 11 dyne/cm(2), were observed. These results provide performance validation of this novel in vitro system with many improved features compared to previous similar prototypes for investigation of flow effects on ECs. The integration of the LDV technique in the current study and the comparison of the results with those from theory revealed that great care must be taken when using PPFCs since the commonly used theoretical relation for laminar steady flows is unable to predict the flow uniformity (which may introduce significant statistical bias in biological studies) and the predicted WSS was subjected to greater error when compared to a more comprehensive equation presented in the current work. Moreover, application of the LDV technique in the current system is essential for studies of more complex cases, such as disturbed flows, where the WSS cannot be predicted using theoretical or numerical modelling methods.
Collapse
Affiliation(s)
- Hamed Avari
- Advanced Fluid Mechanics Research Group, Department of Mechanical and Materials Engineering, University of Western Ontario, London, ON, N6A 5B8, Canada.
| | - Eric Savory
- Advanced Fluid Mechanics Research Group, Department of Mechanical and Materials Engineering, University of Western Ontario, London, ON, N6A 5B8, Canada.
| | - Kem A Rogers
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 5B8, Canada.
| |
Collapse
|
22
|
Ma J, Zhao N, Zhu D. Sirolimus-eluting dextran and polyglutamic acid hybrid coatings on AZ31 for stent applications. J Biomater Appl 2015. [PMID: 26202889 DOI: 10.1177/0885328215596324] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
UNLABELLED Magnesium (Mg)-based cardiovascular stents are promising candidate as the next generation of novel stents. Clinical studies have revealed encouraging outcomes, but late restenosis and thrombogenesis still largely exist. Blood and vascular biocompatible coatings with drug-eluting features could be the solution to such problems. OBJECTIVE This study was to investigate the feasibility of a three-layer hybrid coating on Mg alloy AZ31 with sirolimus-eluting feature for cardiovascular stent application. MATERIALS AND METHODS The first and third layers were low molecular weight dextran loaded with sirolimus, and the second layer was polyglutamic acid (PGA) to control sirolimus release. The hybrid coating was verified by scanning electron microscope (SEM). DC polarization and immersion tests were used to evaluate corrosion rate of the materials. Indirect cell viability and cell proliferation tests were performed by culturing cells with extract solutions of AZ31 samples. Blood compatibility was assessed using hemolysis assay. RESULTS Coated samples had an enhanced corrosion resistance than that of uncoated controls, more PGA slower corrosion. Sirolimus had a burst release for the initial ∼3 days and then a slower release until reached a plateau. The PGA thickness was able to control the sirolimus release, the thicker of PGA the slower release. The overall cell viability was extract concentration-dependent, and improved by the hybrid coatings. Cell proliferation was correlated to coating thickness and was inhibited by sirolimus. In addition, all coated AZ31 samples were non-hemolytic. CONCLUSION Results demonstrated that such a three-layer hybrid coating may be useful to improve the vascular biocompatibility of Mg stent materials.
Collapse
Affiliation(s)
- Jun Ma
- Department of Chemical, Biological and Bio-Engineering, North Carolina Agricultural and Technical State University, Greensboro, NC, USA NSF Engineering Research Center-Revolutionizing Metallic Biomaterials, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| | - Nan Zhao
- Department of Chemical, Biological and Bio-Engineering, North Carolina Agricultural and Technical State University, Greensboro, NC, USA NSF Engineering Research Center-Revolutionizing Metallic Biomaterials, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| | - Donghui Zhu
- Department of Chemical, Biological and Bio-Engineering, North Carolina Agricultural and Technical State University, Greensboro, NC, USA NSF Engineering Research Center-Revolutionizing Metallic Biomaterials, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| |
Collapse
|
23
|
Sylman JL, Artzer DT, Rana K, Neeves KB. A vascular injury model using focal heat-induced activation of endothelial cells. Integr Biol (Camb) 2015; 7:801-14. [PMID: 26087748 PMCID: PMC4494879 DOI: 10.1039/c5ib00108k] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Endothelial cells (EC) both inhibit and promote platelet function depending on their activation state. Quiescent EC inhibit platelet activation by constitutive secretion of platelet inhibitors. Activated EC promote platelet adhesion by secretion of von Willebrand factor (vWF). EC also secrete an extracellular matrix that support platelet adhesion when exposed following vascular injury. Previous studies of EC-platelet interactions under flow activate entire monolayers of cells by chemical activation. In this study, EC cultured in microfluidic channels were focally activated by heat from an underlying microelectrode. Based on finite element modeling, microelectrodes induced peak temperature increases of 10-40 °C above 37 °C after applying 5-9 V for 30 s resulting in three zones: (1) a quiescent zone corresponded to peak temperatures of less than 15 °C characterized by no EC activation or platelet accumulation. (2) An activation zone corresponding to an increase of 16-22 °C yielded EC that were viable, secreted elevated levels of vWF, and were P-selectin positive. Platelets accumulated in the retracted spaces between EC in the activation zone at a wall shear rate of 150 and 1500 s(-1). Experiments with blocking antibodies show that platelets adhere via GPIbα-vWF and α6β1-laminin interactions. (3) A kill zone corresponded to peak temperatures of greater than 23 °C where EC were not viable and did not support platelet adhesion. These data define heating conditions for the activation of EC, causing the secretion of vWF and the exposure of a subendothelial matrix that support platelet adhesion and aggregation. This model provides for spatially defined zones of EC activation that could be a useful tool for measuring the relative roles of anti- and prothrombotic roles of EC at the site of vascular injury.
Collapse
Affiliation(s)
- J L Sylman
- Department of Chemical and Biological Engineering, Colorado School of Mines, 1500 Illinois Street, Golden, CO 80401, USA.
| | | | | | | |
Collapse
|
24
|
Superparamagnetic iron oxide nanoparticles impair endothelial integrity and inhibit nitric oxide production. Acta Biomater 2014; 10:4896-4911. [PMID: 25123083 DOI: 10.1016/j.actbio.2014.07.027] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 06/26/2014] [Accepted: 07/22/2014] [Indexed: 01/08/2023]
Abstract
Superparamagnetic iron oxide nanoparticles (SPION) are widely used both clinically and experimentally for diverse in vivo applications, such as contrast enhancement in magnetic resonance imaging, hyperthermia and drug delivery. Biomedical applications require particles to have defined physical and chemical properties, and to be stable in biological media. Despite a suggested low cytotoxic action, adverse reactions of SPION in concentrations relevant for biomedical use have not yet been studied in sufficient detail. In the present work we employed Endorem®, dextran-stabilized SPION approved as an intravenous contrast agent, and compared its action to a set of other nanoparticles with potential for magnetic resonance imaging applications. SPION in concentrations relevant for in vivo applications were rapidly taken up by endothelial cells and exhibited no direct cytotoxicity. Electric cell impedance sensing measurements demonstrated that SPION, but not BaSO4/Gd nanoparticles, impaired endothelial integrity, as was confirmed by increased intercellular gap formation in endothelial monolayers. These structural changes induced the subcellular translocation and inhibition of the cytoprotective and anti-atherosclerotic enzyme endothelial NO-synthase and reduced NO production. Lipopolysaccharide-induced inflammatory NO production of macrophages was not affected by SPION. In conclusion, our data suggest that SPION might substantially alter endothelial integrity and function at therapeutically relevant doses, which are not cytotoxic.
Collapse
|
25
|
Moreno-Ulloa A, Romero-Perez D, Villarreal F, Ceballos G, Ramirez-Sanchez I. Cell membrane mediated (-)-epicatechin effects on upstream endothelial cell signaling: evidence for a surface receptor. Bioorg Med Chem Lett 2014; 24:2749-52. [PMID: 24794111 DOI: 10.1016/j.bmcl.2014.04.038] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 04/07/2014] [Accepted: 04/09/2014] [Indexed: 10/25/2022]
Abstract
The consumption of cacao-derived products, particularly in the form of dark chocolate is known to provide beneficial cardiovascular effects in normal individuals and in those with vascular dysfunction (reduced nitric oxide [NO] bioavailability and/or synthesis). Upstream mechanisms by which flavonoids exert these effects are poorly understood and may involve the participation of cell membrane receptors. We previously demonstrated that the flavanol (-)-epicatechin (EPI) stimulates NO production via Ca(+2)-independent eNOS activation/phosphorylation. We wished to investigate the plausible participation of a cell surface receptor using a novel cell-membrane impermeable EPI-Dextran conjugate (EPI-Dx). Under Ca(2+)-free conditions, human coronary artery endothelial cells (HCAEC) were treated for 10min with EPI or EPI-Dx at equimolar concentrations (100nM). Results demonstrate that both EPI and EPI-Dx induced the phosphorylation/activation of PI3K, PDK-1, AKT and eNOS. Interestingly, EPI-Dx effects were significantly higher in magnitude than those of EPI alone. The capacity of EPI-Dx to stimulate cell responses supports the existence of an EPI cell membrane receptor mediating eNOS activation.
Collapse
Affiliation(s)
- Aldo Moreno-Ulloa
- University of California, San Diego, Department of Medicine, San Diego, CA, United States; Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Miron, México, DF CP11340, Mexico
| | - Diego Romero-Perez
- Facultad de Ciencias Químicas e Ingeniería, Universidad Autónoma de Baja California, Tijuana, Baja California, Mexico
| | - Francisco Villarreal
- University of California, San Diego, Department of Medicine, San Diego, CA, United States
| | - Guillermo Ceballos
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Miron, México, DF CP11340, Mexico
| | - Israel Ramirez-Sanchez
- University of California, San Diego, Department of Medicine, San Diego, CA, United States; Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Miron, México, DF CP11340, Mexico.
| |
Collapse
|
26
|
Obi S, Masuda H, Akimaru H, Shizuno T, Yamamoto K, Ando J, Asahara T. Dextran induces differentiation of circulating endothelial progenitor cells. Physiol Rep 2014; 2:e00261. [PMID: 24760515 PMCID: PMC4002241 DOI: 10.1002/phy2.261] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Endothelial progenitor cells (EPCs) have been demonstrated to be effective for the treatment of cardiovascular diseases. However, the differentiation process from circulation to adhesion has not been clarified because circulating EPCs rarely attached to dishes in EPC cultures previously. Here we investigated whether immature circulating EPCs differentiate into mature adhesive EPCs in response to dextran. When floating‐circulating EPCs derived from ex vivo expanded human cord blood were cultured with 5% and 10% dextran, they attached to fibronectin‐coated dishes and grew exponentially. The bioactivities of adhesion, proliferation, migration, tube formation, and differentiated type of EPC colony formation increased in EPCs exposed to dextran. The surface protein expression rate of the endothelial markers vascular endothelial growth factor (VEGF)‐R1/2, VE‐cadherin, Tie2, ICAM1, VCAM1, and integrin αv/β3 increased in EPCs exposed to dextran. The mRNA levels of VEGF‐R1/2, VE‐cadherin, Tie2, endothelial nitric oxide synthase, MMP9, and VEGF increased in EPCs treated with dextran. Those of endothelium‐related transcription factors ID1/2, FOXM1, HEY1, SMAD1, FOSL1, NFkB1, NRF2, HIF1A, EPAS1 increased in dextran‐treated EPCs; however, those of hematopoietic‐ and antiangiogenic‐related transcription factors TAL1, RUNX1, c‐MYB, GATA1/2, ERG, FOXH1, HHEX, SMAD2/3 decreased in dextran‐exposed EPCs. Inhibitor analysis showed that PI3K/Akt, ERK1/2, JNK, and p38 signal transduction pathways are involved in the differentiation in response to dextran. In conclusion, dextran induces differentiation of circulating EPCs in terms of adhesion, migration, proliferation, and vasculogenesis. The differentiation mechanism in response to dextran is regulated by multiple signal transductions including PI3K/Akt, ERK1/2, JNK, and p38. These findings indicate that dextran is an effective treatment for EPCs in regenerative medicines.
Collapse
Affiliation(s)
- Syotaro Obi
- Department of Regenerative Medicine Science, Tokai University School of Medicine, Isehara, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Dick M, Jonak P, Leask RL. Statin therapy influences endothelial cell morphology and F-actin cytoskeleton structure when exposed to static and laminar shear stress conditions. Life Sci 2013; 92:859-65. [DOI: 10.1016/j.lfs.2013.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 02/19/2013] [Accepted: 03/02/2013] [Indexed: 01/09/2023]
|
28
|
Kang TY, Hong JM, Kim BJ, Cha HJ, Cho DW. Enhanced endothelialization for developing artificial vascular networks with a natural vessel mimicking the luminal surface in scaffolds. Acta Biomater 2013; 9:4716-25. [PMID: 22947325 DOI: 10.1016/j.actbio.2012.08.042] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 08/22/2012] [Accepted: 08/26/2012] [Indexed: 12/19/2022]
Abstract
Large tissue regeneration remains problematic because of a lack of oxygen and nutrient supply. An attempt to meet the metabolic needs of cells has been made by preforming branched vascular networks within a scaffold to act as channels for mass transport. When constructing functional vascular networks with channel patency, emphasis should be placed on anti-thrombogenic surface issues. The aim of this study was to develop a rapid endothelialization method for creating an anti-thrombogenic surface mimicking the natural vessel wall in the artificial vascular networks. Shear stress preconditioning and scaffold surface modification were investigated as effective approaches for promoting biomaterial endothelialization. We found that a transient increase in shear stress at the appropriate time is key to enhancing endothelialization. Moreover, surface modification with bioactive materials such as collagen and recombinant mussel adhesive protein fused with arginine-glycine-aspartic acid peptide (MAP-RGD) showed a synergetic effect with shear stress preconditioning. Platelet adhesion tests demonstrated the anti-thrombogenic potential of MAP-RGD itself without endothelialization. The rapid endothelialization method established in this study can be easily applied to preformed artificial vascular networks in porous scaffolds. Development of artificial vascular networks with an anti-thrombogenic luminal surface will open up a new chapter in tissue engineering and regenerative medicine.
Collapse
|
29
|
Doiron AL, Clark B, Rinker KD. Endothelial nanoparticle binding kinetics are matrix and size dependent. Biotechnol Bioeng 2011; 108:2988-98. [DOI: 10.1002/bit.23253] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Revised: 06/07/2011] [Accepted: 06/20/2011] [Indexed: 11/07/2022]
|
30
|
Rouleau L, Farcas M, Tardif JC, Mongrain R, Leask RL. Endothelial cell morphologic response to asymmetric stenosis hemodynamics: effects of spatial wall shear stress gradients. J Biomech Eng 2010; 132:081013. [PMID: 20670062 DOI: 10.1115/1.4001891] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Endothelial cells are known to respond to hemodynamic forces. Their phenotype has been suggested to differ between atheroprone and atheroprotective regions of the vasculature, which are characterized by the local hemodynamic environment. Once an atherosclerotic plaque has formed in a vessel, the obstruction creates complex spatial gradients in wall shear stress. Endothelial cell response to wall shear stress may be linked to the stability of coronary plaques. Unfortunately, in vitro studies of the endothelial cell involvement in plaque stability have been limited by unrealistic and simplified geometries, which cannot reproduce accurately the hemodynamics created by a coronary stenosis. Hence, in an attempt to better replicate the spatial wall shear stress gradient patterns in an atherosclerotic region, a three dimensional asymmetric stenosis model was created. Human abdominal aortic endothelial cells were exposed to steady flow (Re=50, 100, and 200 and tau=4.5 dyn/cm(2), 9 dyn/cm(2), and 18 dyn/cm(2)) in idealized 50% asymmetric stenosis and straight/tubular in vitro models. Local morphological changes that occur due to magnitude, duration, and spatial gradients were quantified to identify differences in cell response. In the one dimensional flow regions, where flow is fully developed and uniform wall shear stress is observed, cells aligned in flow direction and had a spindlelike shape when compared with static controls. Morphological changes were progressive and a function of time and magnitude in these regions. Cells were more randomly oriented and had a more cobblestone shape in regions of spatial wall shear stress gradients. These regions were present, both proximal and distal, at the stenosis and on the wall opposite to the stenosis. The response of endothelial cells to spatial wall shear stress gradients both in regions of acceleration and deceleration and without flow recirculation has not been previously reported. This study shows the dependence of endothelial cell morphology on spatial wall shear stress gradients and demonstrates that care must be taken to account for altered phenotype due to geometric features. These results may help explain plaque stability, as cells in shoulder regions near an atherosclerotic plaque had a cobblestone morphology indicating that they may be more permeable to subendothelial transport and express prothrombotic factors, which would increase the risk of atherothrombosis.
Collapse
Affiliation(s)
- Leonie Rouleau
- Department of Chemical Engineering, McGill University, 3610 University, Montreal, QC, H3A 2B2, Canada
| | | | | | | | | |
Collapse
|
31
|
Laminar shear stress prevents simvastatin-induced adhesion molecule expression in cytokine activated endothelial cells. Eur J Pharmacol 2010; 649:268-76. [DOI: 10.1016/j.ejphar.2010.09.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Revised: 08/23/2010] [Accepted: 09/09/2010] [Indexed: 11/17/2022]
|
32
|
Differential Response of Endothelial Cells to Simvastatin When Conditioned with Steady, Non-Reversing Pulsatile or Oscillating Shear Stress. Ann Biomed Eng 2010; 39:402-13. [DOI: 10.1007/s10439-010-0145-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Accepted: 08/12/2010] [Indexed: 10/19/2022]
|
33
|
Rossi J, Rouleau L, Tardif JC, Leask RL. Effect of simvastatin on Kruppel-like factor2, endothelial nitric oxide synthase and thrombomodulin expression in endothelial cells under shear stress. Life Sci 2010; 87:92-9. [DOI: 10.1016/j.lfs.2010.05.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 04/22/2010] [Accepted: 05/14/2010] [Indexed: 10/19/2022]
|