1
|
Midekssa FS, Davidson CD, Wieger ME, Kamen JL, Hanna KM, Jayco DKP, Hu MM, Friend NE, Putnam AJ, Helms AS, Shikanov A, Baker BM. Semi-synthetic fibrous fibrin composites promote 3D microvascular assembly, survival, and host integration of endothelial cells without mesenchymal cell support. Bioact Mater 2025; 49:652-669. [PMID: 40235652 PMCID: PMC11999628 DOI: 10.1016/j.bioactmat.2025.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 01/14/2025] [Accepted: 02/19/2025] [Indexed: 04/17/2025] Open
Abstract
Vasculogenic assembly of 3D capillary networks remains a promising approach to vascularizing tissue-engineered grafts, a significant outstanding challenge in tissue engineering and regenerative medicine. Current approaches for vasculogenic assembly rely on the inclusion of supporting mesenchymal cells alongside endothelial cells, co-encapsulated within vasculo-conducive materials such as low-density fibrin hydrogels. Here, we established a material-based approach to circumvent the need for supporting mesenchymal cells and report that the inclusion of synthetic matrix fibers in dense (>3 mg mL-1) 3D fibrin hydrogels can enhance vasculogenic assembly in endothelial cell monocultures. Surprisingly, we found that the addition of non-cell-adhesive synthetic matrix fibers compared to cell-adhesive synthetic fibers best encouraged vasculogenic assembly, proliferation, lumenogenesis, a vasculogenic transcriptional program, and additionally promoted cell-matrix interactions and intercellular force transmission. Implanting fiber-reinforced prevascularized constructs to assess graft-host vascular integration, we demonstrate additive effects of enhanced vascular network assembly during in vitro pre-culture, fiber-mediated improvements in endothelial cell survival and vascular maintenance post-implantation, and enhanced host cell infiltration that collectively enabled graft vessel integration with host circulation. This work establishes synthetic matrix fibers as an inexpensive alternative to sourcing and expanding secondary supporting cell types for the prevascularization of tissue constructs.
Collapse
Affiliation(s)
- Firaol S. Midekssa
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Christopher D. Davidson
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Megan E. Wieger
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Jordan L. Kamen
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Kaylin M. Hanna
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Danica Kristen P. Jayco
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Michael M. Hu
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Nicole E. Friend
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Andrew J. Putnam
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Adam S. Helms
- Division of Cardiovascular Medicine, University of Michigan Ann Arbor, MI 48109, United States
| | - Ariella Shikanov
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, 48109, United States
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Brendon M. Baker
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| |
Collapse
|
2
|
Yu WL, Deng LW, Li HH, Wang CK, Zuo XY, Wang ZC, Meng L, Wen LX, Zeng WZ, Zhao Y, Wang XH. FBXO38 Regulates Nox1 Stability to Reduce Vascular Endothelial Damage Induced by Low Oscillatory Shear Stress. Cardiovasc Ther 2025; 2025:4506032. [PMID: 40313652 PMCID: PMC12043393 DOI: 10.1155/cdr/4506032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/28/2025] [Indexed: 05/03/2025] Open
Abstract
Oxidative stress and endothelial dysfunction are critical drivers of atherosclerosis, but the mechanisms regulating oxidative stress under disturbed flow conditions remain incompletely understood. The ubiquitin-proteasome system, particularly E3 ubiquitin ligases, may play a pivotal role in modulating these processes. FBXO38, an E3 ligase involved in proteasomal degradation, has been implicated in various physiological pathways, but its role in regulating oxidative stress in endothelial cells is unknown. We hypothesized that FBXO38 mitigates endothelial damage induced by low oscillatory shear stress (LOSS) by promoting the ubiquitin-proteasome-dependent degradation of Nox1, a major source of reactive oxygen species (ROS). Using an in vitro LOSS model in human umbilical vein endothelial cells (HUVECs) and an in vivo mouse partial carotid ligation model, we assessed the expression of FBXO38 and Nox1 through quantitative PCR, western blotting, immunofluorescence, and immunohistochemistry. LOSS significantly reduced FBXO38 protein expression (by ~60%, p < 0.0001 at 24 h), leading to increased Nox1 protein levels (approximately two-fold, p < 0.001) and apoptosis. FBXO38 overexpression markedly attenuated Nox1 accumulation (~50% reduction, p < 0.05), reduced ROS production, and improved cell viability under LOSS conditions, whereas FBXO38 knockdown exacerbated these effects. Moreover, FBXO38 directly interacted with Nox1, suggesting a ubiquitin-dependent degradation mechanism. Our results reveal that FBXO38 regulates endothelial oxidative stress by controlling Nox1 stability under disturbed shear stress conditions. Although FBXO38 emerges as a promising candidate for therapeutic targeting, further studies are necessary to validate its potential in preclinical and clinical settings.
Collapse
Affiliation(s)
- Wan-li Yu
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li-wen Deng
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huan-huan Li
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chun-kai Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiang-yi Zuo
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zi-chang Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Meng
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lan-xin Wen
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wan-zhi Zeng
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Zhao
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xue-hu Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Grego A, Fernandes C, Fonseca I, Dias-Neto M, Costa R, Leite-Moreira A, Oliveira SM, Trindade F, Nogueira-Ferreira R. Endothelial dysfunction in cardiovascular diseases: mechanisms and in vitro models. Mol Cell Biochem 2025:10.1007/s11010-025-05289-w. [PMID: 40259179 DOI: 10.1007/s11010-025-05289-w] [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: 02/09/2025] [Accepted: 04/08/2025] [Indexed: 04/23/2025]
Abstract
Endothelial cells (ECs) are arranged side-by-side to create a semi-permeable monolayer, forming the inner lining of every blood vessel (micro and macrocirculation). Serving as the first barrier for circulating molecules and cells, ECs represent the main regulators of vascular homeostasis being able to respond to environmental changes, either physical or chemical signals, by producing several factors that regulate vascular tone and cellular adhesion. Healthy endothelium has anticoagulant properties that prevent the adhesion of leukocytes and platelets to the vessel walls, contributing to resistance to thrombus formation, and regulating inflammation, and vascular smooth muscle cell proliferation. Many risk factors of cardiovascular diseases (CVDs) promote the endothelial expression of chemokines, cytokines, and adhesion molecules. The resultant endothelial activation can lead to endothelial cell dysfunction (ECD). In vitro models of ECD allow the study of cellular and molecular mechanisms of disease and provide a research platform for screening potential therapeutic agents. Even though alternative models are available, such as animal models or ex vivo models, in vitro models offer higher experimental flexibility and reproducibility, making them a valuable tool for the understanding of pathophysiological mechanisms of several diseases, such as CVDs. Therefore, this review aims to synthesize the currently available in vitro models regarding ECD, emphasizing CVDs. This work will focus on 2D cell culture models (endothelial cell lines and primary ECs), 3D cell culture systems (scaffold-free and scaffold-based), and 3D cell culture models (such as organ-on-a-chip). We will dissect the role of external stimuli-chemical and mechanical-in triggering ECD.
Collapse
Affiliation(s)
- Ana Grego
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Cristiana Fernandes
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Ivo Fonseca
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Marina Dias-Neto
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- Department of Angiology and Vascular Surgery, Unidade Local de Saúde de São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Raquel Costa
- Universidade Católica Portuguesa, CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005, Porto, Portugal
| | - Adelino Leite-Moreira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- Department of Cardiothoracic Surgery, Unidade Local de Saúde de São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Sandra Marisa Oliveira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Fábio Trindade
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Rita Nogueira-Ferreira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.
| |
Collapse
|
4
|
Brunmaier LAE, Ozdemir T, Walker TW. Angiogenesis: Biological Mechanisms and In Vitro Models. Ann Biomed Eng 2025:10.1007/s10439-025-03721-2. [PMID: 40210793 DOI: 10.1007/s10439-025-03721-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/25/2025] [Indexed: 04/12/2025]
Abstract
The translation of biomedical devices and drug research is an expensive and long process with a low probability of receiving FDA approval. Developing physiologically relevant in vitro models with human cells offers a solution to not only improving the odds of FDA approval but also to expand our ability to study complex in vivo systems in a simpler fashion. Animal models remain the standard for pre-clinical testing; however, the data from animal models is an unreliable extrapolation when anticipating a human response in clinical trials, thus contributing to the low rates of translation. In this review, we focus on in vitro vascular or angiogenic models because of the incremental role that the vascular system plays in the translation of biomedical research. The first section of this review discusses the most common angiogenic cytokines that are used in vitro to initiate angiogenesis, followed by angiogenic inhibitors where both initiators and inhibitors work to maintain vascular homeostasis. Next, we evaluate previously published in vitro models, where we evaluate capturing the physical environment for biomimetic in vitro modeling. These topics provide a foundation of parameters that must be considered to improve and achieve vascular biomimicry. Finally, we summarize these topics to suggest a path forward with the goal of engineering human in vitro models that emulate the in vivo environment and provide a platform for biomedical device and drug screening that produces data to support clinical translation.
Collapse
Affiliation(s)
- Laura A E Brunmaier
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA
| | - Tugba Ozdemir
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA
| | - Travis W Walker
- Karen M. Swindler Department of Chemical and Biological Engineering, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA.
| |
Collapse
|
5
|
Zhang Y, Sun H, Gandhi A, Du Y, Ebrahimi S, Jiang Y, Xu S, Uwase H, Seidel A, Bingaman SS, Arnold AC, Nguyen C, Ding W, Woolard MD, Hobbs R, Bagchi P, He P. Role of shear stress-induced red blood cell released ATP in atherosclerosis. Am J Physiol Heart Circ Physiol 2025; 328:H774-H791. [PMID: 39982440 DOI: 10.1152/ajpheart.00875.2024] [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: 12/16/2024] [Revised: 12/31/2024] [Accepted: 02/13/2025] [Indexed: 02/22/2025]
Abstract
Altered hemodynamics is a key factor for atherosclerosis. For decades, endothelial cell (EC) responses to fluid-generated wall shear stress have been the central focus for atherogenesis. However, circulating blood is not a cell-free fluid, it contains mechanosensitive red blood cells (RBCs) that are also subjected to altered hemodynamics and release a large amount of ATP, but their impact on atherosclerosis has been overlooked. The focus of this study is the role of shear stress (SS)-induced RBC-released ATP in atherosclerosis. Hypercholesterolemic mouse models with and without RBC-Pannexin 1 deletion were used for the study. Results showed that SS-induced release of ATP from RBCs was at µM concentrations, three-orders of magnitude higher than that from other cell types. Suppression of RBC-released ATP via deletion of Pannexin 1, a mechanosensitive ATP-permeable channel, reduced high-fat diet-induced aortic plaque burden by 40%-60%. Importantly, the location and the extent of aortic atherosclerotic lesions spatially matched with the ATP deposition profile at aortic wall predicted by a computational fluid dynamic (CFD) model. Furthermore, hypercholesterolemia increases EC susceptibility to ATP with potentiated increase in [Ca2+]i, an initial signaling for aortic EC barrier dysfunction, and an essential cause for lipid accumulation and inflammatory cell infiltration. The computational prediction also provides a physics-based explanation for RBC-released ATP-induced sex disparities in atherosclerosis. Our study reveals an important role of RBC-released ATP in the initiation and progression of atherosclerosis. These novel findings provide a more comprehensive view of how altered hemodynamics and systemic risk factors synergistically contribute to atherosclerosis.NEW & NOTEWORTHY This study reveals that, in addition to fluid-derived wall shear stress, the disturbed blood flow-induced release of ATP from mechanosensitive red blood cells (RBCs), the major cellular components of blood, along with hypercholesterolemia-induced increases in endothelial cell susceptibility to ATP contribute significantly to the initiation and progression of atherosclerosis. These novel findings advance our current understanding of how altered hemodynamics and hypercholesterolemia synergistically contribute to atherosclerosis for the first time with the inclusion of RBCs.
Collapse
Affiliation(s)
- Yunpei Zhang
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Haoyu Sun
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Aayush Gandhi
- Department of Mechanical & Aerospace Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States
| | - Yong Du
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Saman Ebrahimi
- Department of Mechanical & Aerospace Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States
| | - Yanyan Jiang
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Sulei Xu
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Hope Uwase
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Alane Seidel
- Transgenic core, Department of Comparative Medicine, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Sarah S Bingaman
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Amy C Arnold
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Christian Nguyen
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Wei Ding
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Matthew D Woolard
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States
| | - Ryan Hobbs
- Department of Dermatology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Prosenjit Bagchi
- Department of Mechanical & Aerospace Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States
| | - Pingnian He
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| |
Collapse
|
6
|
Grotberg JB, Romanò F, Grotberg JC. Flow mechanisms of the air-blood barrier. PLoS Comput Biol 2025; 21:e1012917. [PMID: 40208907 PMCID: PMC12052194 DOI: 10.1371/journal.pcbi.1012917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 05/05/2025] [Accepted: 02/25/2025] [Indexed: 04/12/2025] Open
Abstract
The air-blood barrier protects the lung from blood/serum entering the air spaces, i.e., from "drowning in your own fluids". Failure leads to pulmonary edema, a regularly fatal complication during the Covid-19 pandemic which claimed 7 million lives worldwide. Finding no mathematical models for the underlying fluid mechanics, we created the first. Governing flow equations for alveolar capillary, interstitium, and alveolus are coupled by crossflows at the capillary and epithelial membranes and end-exit flows to the lymphatics. Case examples include normal/recovery, cardiogenic pulmonary edema, acute respiratory distress syndrome, effects of positive end expiratory pressure, and a wide range of parameter values for permeability of the membranes and interstitial matrix. Previously unknown membrane fluid shear stresses calculate to values that affect cell function in many systems. We add active epithelial reabsorption which has two effects: shifting streamlines to favor alveolar-lymphatic clearance and adding to the direct alveolar-capillary clearance. Simple algebraic equations are derived for the interstitial fluid pressure, pi, membrane crossflow velocities and the critical capillary pressure, pcrit, above which edema occurs. For validation, the pcrit predictions fit clinical definitions and flow calculations of lymphatic vs capillary clearance match animal experimental data. For decades the value of pi has been imposed as an input, whereas we calculate the value as an output. They don't agree. Since the space is too small for measurements, the ability to calculate pi and pcrit offers new insights, questions long-held beliefs, and opens applications from physiological studies to personalized clinical care.
Collapse
Affiliation(s)
- James B. Grotberg
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Francesco Romanò
- Univ. Lille, CNRS, ONERA, Arts et Métiers Institute of Technology, Centrale Lille, UMR, LMFL - Laboratoire de Mécanique des Fluides de Lille - Kampé de Fériet, Lille, France
| | - John C. Grotberg
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| |
Collapse
|
7
|
Ching T, van Steen ACI, Gray-Scherr D, Teo JL, Vasan A, Jeon J, Shah J, Patel A, Stoddard AE, Bays JL, Eyckmans J, Chen CS. TapeTech microfluidic connectors: adhesive tape-enabled solution for organ-on-a-chip system integration. LAB ON A CHIP 2025; 25:1474-1488. [PMID: 39907088 PMCID: PMC11795533 DOI: 10.1039/d4lc00970c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/18/2025] [Indexed: 02/06/2025]
Abstract
A longstanding challenge in microfluidics has been the efficient delivery of fluids from macro-scale pumping systems into microfluidic devices, known as the "world-to-chip" problem. Thus far, the entire industry has accepted the use of imperfect, rigid tubing and connectors as the ecosystem within which to operate, which, while functional, are often cumbersome, labor-intensive, prone to errors, and ill-suited for high-throughput experimentation. In this paper, we introduce TapeTech microfluidics, a flexible and scalable solution designed to address the persistent "world-to-chip" problem in microfluidics, particularly in organ-on-a-chip (OoC) applications. TapeTech offers a streamlined alternative, utilizing adhesive tape and thin-film polymers to create adaptable, integrated multi-channel ribbon connectors that simplify fluidic integration with pumps and reservoirs. Key features of TapeTech include reduced pressure surges, easy priming, rapid setup, easy multiplexing, and broad compatibility with existing devices and components, which are essential for maintaining stable fluid dynamics and protecting sensitive cell cultures. Furthermore, TapeTech is designed to flex around the lids of Petri dishes, enhancing sterility and transportability by enabling easy transfer between incubators, biosafety cabinets (BSCs), and microscopes. The rapid design-to-prototype iteration enabled by TapeTech allows users to quickly develop connectors for a wide range of microfluidic devices. Importantly, we showcase the utility of TapeTech in OoC cultures requiring fluid flow. We also highlight other utilities, such as real-time microscopy and a well-plate medium exchanger. The accessibility of this technology should enable more laboratories to simplify design and setup of microfluidic experiments, and increase technology adoption.
Collapse
Affiliation(s)
- Terry Ching
- Biological Design Center, Boston University, Boston, MA 02215, USA.
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Abraham C I van Steen
- Biological Design Center, Boston University, Boston, MA 02215, USA.
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Delaney Gray-Scherr
- Biological Design Center, Boston University, Boston, MA 02215, USA.
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Jessica L Teo
- Biological Design Center, Boston University, Boston, MA 02215, USA.
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Anish Vasan
- Biological Design Center, Boston University, Boston, MA 02215, USA.
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Joshua Jeon
- Biological Design Center, Boston University, Boston, MA 02215, USA.
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Jessica Shah
- Harvard-MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Aayush Patel
- Biological Design Center, Boston University, Boston, MA 02215, USA.
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Amy E Stoddard
- Biological Design Center, Boston University, Boston, MA 02215, USA.
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jennifer L Bays
- Biological Design Center, Boston University, Boston, MA 02215, USA.
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Jeroen Eyckmans
- Biological Design Center, Boston University, Boston, MA 02215, USA.
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Christopher S Chen
- Biological Design Center, Boston University, Boston, MA 02215, USA.
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| |
Collapse
|
8
|
Wu Y, Sun SX. Fluid transport comes to the fore. eLife 2025; 14:e106304. [PMID: 40014042 PMCID: PMC11867611 DOI: 10.7554/elife.106304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025] Open
Abstract
Proteins that allow water to move in and out of cells help shape the development of new blood vessels.
Collapse
Affiliation(s)
- Yufei Wu
- Institute for NanoBioTechnology and Department of Mechanical Engineering, Johns Hopkins UniversityBaltimoreUnited States
| | - Sean X Sun
- Institute for NanoBioTechnology and Department of Mechanical Engineering, Johns Hopkins UniversityBaltimoreUnited States
| |
Collapse
|
9
|
Chandurkar MK, Yang M, Rostami M, Han SJ. TRPV4 Dominates High Shear-Induced Initial Traction Response and Long-Term Relaxation Over Piezo1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637570. [PMID: 39990362 PMCID: PMC11844455 DOI: 10.1101/2025.02.10.637570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Modulation of endothelial traction is critical for the responses of endothelial cells to fluid shear stress (FSS), which has profound implications for vascular health and atherosclerosis. Previously, we demonstrated that under high FSS, endothelial cells rapidly increase traction forces, followed by relaxation, with traction aligning in the flow direction. In contrast, low shear preconditioning induces a modest short-term increase in traction (<30 min), followed by a secondary long-term (>14 hr) rise, with traction/cells aligning perpendicular to the flow. The upstream mechanosensors driving these responses, however, remain unknown. Here, we sought the roles of Piezo1 and TRPV4 ion channels in shear-induced traction modulation. We report that HUVECs with Piezo1 silencing reduced the initial traction rise in half under high FSS compared to those by WT cells, while not affecting the traction modulation in response to low FSS or traction/cell alignment to the flow direction. Conversely, cells with siTRPV4 fully abrogated the initial traction rise, as well as alignment of traction and cells, in response to both high and low FSS conditions. Dual inhibition of Piezo1 and TRPV4 further impaired both initial and long-term traction under high FSS. Interestingly, dual-inhibited cells displayed larger initial traction responses to low FSS compared to control cells, suggesting the involvement of alternative calcium-independent pathways that become dominant when both ion channels are nonfunctional. Additionally, either ion channel inhibition led to secondary long-term traction increase even under high FSS condition. These findings suggest that while both Piezo1 and TRPV4 channels contribute to shear mechanotransduction, TRPV4 plays more dominant role than Piezo1 in mediating the initial traction rise and sustaining long-term relaxation under high or low shear stress, highlighting their critical and distinct contributions to endothelial mechanotransduction and remodeling.
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
| | - Manli Yang
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan, United States
| | - Majid Rostami
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, United States
- Health Research Institute, 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 Biological Sciences, Michigan Technological University, Houghton, Michigan, United States
- Department of Mechanical Engineering and Engineering Mechanics, Michigan Technological University, Houghton, Michigan, United States
| |
Collapse
|
10
|
Ansarizadeh M, Nguyen HT, Lazovic B, Kettunen J, De Silva L, Sivakumar R, Junttila P, Rissanen SL, Hicks R, Singh P, Eklund L. Microfluidic vessel-on-chip platform for investigation of cellular defects in venous malformations and responses to various shear stress and flow conditions. LAB ON A CHIP 2025; 25:613-630. [PMID: 39847008 DOI: 10.1039/d4lc00824c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
A novel microfluidic platform was designed to study the cellular architecture of endothelial cells (ECs) in an environment replicating the 3D organization and flow of blood vessels. In particular, the platform was constructed to investigate EC defects in slow-flow venous malformations (VMs) under varying shear stress and flow conditions. The platform featured a standard microtiter plate footprint containing 32 microfluidic units capable of replicating wall shear stress (WSS) in normal veins and enabling precise control of shear stress and flow directionality without the need for complex pumping systems. Using genetically engineered human umbilical vein endothelial cells (HUVECs) and induced pluripotent stem cell (iPSC)-derived ECs (iECs) to express the recurrent TIE2L914F VM mutation we assessed responses on EC orientation and area, actin organization, and Golgi polarization to uni- and bidirectional flow and varying WSS. Comparison of control and TIE2L914F expressing ECs showed differential cellular responses to flow and WSS in terms of cell shape elongation, orientation of F-actin, and Golgi polarization, indicating altered mechanosensory or mechanotransduction signaling pathways in the presence of the VM causative mutation. The data also revealed significant differences in how the primary and iPSC-derived iECs responded to flow. As a conclusion, the developed microfluidic platform allowed simulation of multiple flow conditions in a scalable and pumpless format. The design made it a desirable tool for studying different EC types as well as cellular changes in vascular disease. The platform should offer new opportunities for biomechanical research by providing a controlled environment to analyze the flow-dependent mechanosensory pathways in ECs.
Collapse
Affiliation(s)
- Mohammadhassan Ansarizadeh
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5000, FI-90014 Oulu, Finland.
| | - Hoang-Tuan Nguyen
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5000, FI-90014 Oulu, Finland.
- Finnadvance Ltd., Oulu, Finland
| | - Bojana Lazovic
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5000, FI-90014 Oulu, Finland.
- BioPharmaceuticals R&D Cell Therapy Department, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Translational Genomics, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Laknee De Silva
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5000, FI-90014 Oulu, Finland.
| | | | | | | | - Ryan Hicks
- BioPharmaceuticals R&D Cell Therapy Department, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Translational Genomics, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, UK
| | | | - Lauri Eklund
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5000, FI-90014 Oulu, Finland.
| |
Collapse
|
11
|
Viana-Mattioli S, Fonseca-Alaniz MH, Pinheiro-de-Sousa I, Junior RR, Mastella MH, de Carvalho Cavalli R, Sandrim VC. Plasma from hypertensive pregnancy patients induce endothelial dysfunction even under atheroprotective shear stress. Sci Rep 2025; 15:4675. [PMID: 39920219 PMCID: PMC11805971 DOI: 10.1038/s41598-025-88902-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 01/31/2025] [Indexed: 02/09/2025] Open
Abstract
Preeclampsia (PE) is a challenge in maternal healthcare due to its complex nature, characterized by high blood pressure, protein in the urine, and damage to various organs. There is evidence linking PE to endothelial dysfunction (ED), triggered by substances released from an oxygen-deprived placenta. Previous in vitro studies have not considered the impact of in vivo elements, such as the different patterns of blood flow, and laminar (LSS) vs. oscillatory (OSS) shear stress, on the development of ED. We investigated the impact of plasma from healthy pregnant women (HP), subjects with gestational hypertension (GH), and PE patients on global gene expression of human coronary endothelial cells (HCAECs) under LSS and OSS. Our findings revealed a unique transcriptional profile of endothelial cells induced by plasma incubation in LSS. Notably, OSS resulted in similar transcriptomes irrespective of plasma treatment. Under LSS, GH plasma resulted in a proliferative profile, whereas PE plasma was linked to pro-inflammatory and antioxidant profiles compared to HP plasma. Our findings demonstrate that shear stress levels influence the endothelial cell transcriptome in response to plasma from hypertensive pregnancy patients. Both PE and GH can induce endothelial dysfunction under atheroprotective LSS, with a more significant effect observed with PE-derived plasma.
Collapse
Affiliation(s)
- Sarah Viana-Mattioli
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu, Universidade Estadual Paulista (UNESP), Distrito Rubião Júnior, Botucatu, São Paulo, SP, Brazil
- Laboratorio de Genetica e Cardiologia Molecular, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Miriam Helena Fonseca-Alaniz
- Laboratorio de Genetica e Cardiologia Molecular, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Iguaracy Pinheiro-de-Sousa
- Laboratorio de Genetica e Cardiologia Molecular, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Ricardo Rosa Junior
- Laboratorio de Genetica e Cardiologia Molecular, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Moises Henrique Mastella
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu, Universidade Estadual Paulista (UNESP), Distrito Rubião Júnior, Botucatu, São Paulo, SP, Brazil
| | - Ricardo de Carvalho Cavalli
- Department of Gynecology and Obstetrics, Hospital das Clínicas, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, SP, Brazil
| | - Valeria Cristina Sandrim
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu, Universidade Estadual Paulista (UNESP), Distrito Rubião Júnior, Botucatu, São Paulo, SP, Brazil.
| |
Collapse
|
12
|
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
|
13
|
Linares I, Chen K, Saffren A, Mansouri M, Abhyankar VV, Miller BL, Begolo S, Awad HA, McGrath JL. Fluid flow impacts endothelial-monocyte interactions in a model of vascular inflammatory fibrosis. Sci Rep 2025; 15:3227. [PMID: 39863621 PMCID: PMC11763004 DOI: 10.1038/s41598-025-85987-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
The aberrant vascular response associated with tendon injury results in circulating immune cell infiltration and a chronic inflammatory feedback loop leading to poor healing outcomes. Studying this dysregulated tendon repair response in human pathophysiology has been historically challenging due to the reliance on animal models. To address this, our group developed the human tendon-on-a-chip (hToC) to model cellular interactions in the injured tendon microenvironment; however, this model lacked the key element of physiological flow in the vascular compartment. Here, we leveraged the modularity of our platform to create a fluidic hToC that enables the study of circulating immune cell and vascular crosstalk in a tendon injury model. Under physiological shear stress consistent with postcapillary venules, we found a significant increase in the endothelial leukocyte activation marker intercellular adhesion molecule 1 (ICAM-1), as well as enhanced adhesion and transmigration of circulating monocytes across the endothelial barrier. The addition of tissue macrophages to the tendon compartment further increased the degree of circulating monocyte infiltration into the tissue matrix. Our findings demonstrate the importance of adding physiological flow to the human tendon-on-a-chip, and more generally, the significance of flow for modeling immune cell interactions in tissue inflammation and disease.
Collapse
Affiliation(s)
- Isabelle Linares
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Kaihua Chen
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Ava Saffren
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Mehran Mansouri
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, USA
| | - Vinay V Abhyankar
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, USA
| | - Benjamin L Miller
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Dermatology, University of Rochester Medical Center, Rochester, NY, USA
- Institute of Optics, University of Rochester, Rochester, NY, USA
| | | | - Hani A Awad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - James L McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA.
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
14
|
Simões-Faria R, Daems M, Peacock HM, Declercq M, Willems A, Jones EAV, Ghesquière B. Wall shear stress modulates metabolic pathways in endothelial cells. Metabolomics 2025; 21:16. [PMID: 39832080 PMCID: PMC11753319 DOI: 10.1007/s11306-024-02214-y] [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: 10/17/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025]
Abstract
INTRODUCTION Hemodynamic forces play a crucial role in modulating endothelial cell (EC) behavior, significantly influencing blood vessel responses. While traditional in vitro studies often explore ECs under static conditions, ECs are exposed to various hemodynamic forces in vivo. This study investigates how wall shear stress (WSS) influences EC metabolism, focusing on the interplay between WSS and key metabolic pathways. OBJECTIVES The aim of this study is to examine the effects of WSS on EC metabolism, specifically evaluating its impact on central carbon metabolism and glycolysis using transcriptomics and tracer metabolomics approaches. METHODS ECs were exposed to WSS, and transcriptomic analysis was performed to assess gene expression changes related to metabolic pathways. Tracer metabolomics was used to track metabolic fluxes, focusing on glutamine and glycolytic metabolism. Additionally, chemical inhibition of glutamate dehydrogenase was conducted to evaluate its role in EC fitness under WSS. RESULTS Transcriptomic data revealed upregulation of glutamine and glutamate pathways, alongside downregulation of glycolytic activity in ECs exposed to WSS. Tracer metabolomics confirmed that WSS promotes glutamine anaplerosis into the Krebs cycle, while decreasing glycolytic metabolism. Suppression of glutamate dehydrogenase impaired EC fitness under WSS conditions. CONCLUSION Our findings illuminate that ECs subjected to WSS exhibit a preference for glutamine as a key nutrient source for central carbon metabolism pathways, indicating diminished reliance on glycolysis. This study elucidates the nutritional predilections and regulatory mechanisms governing EC metabolism under WSS in vitro, underscoring the pivotal role of physical stimuli in shaping EC metabolic responses.
Collapse
Affiliation(s)
- Rita Simões-Faria
- Laboratory of Applied Mass Spectrometry, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- Metabolomics Core Facility Leuven, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Margo Daems
- Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Hanna M Peacock
- Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Mathias Declercq
- Department of Development and Regeneration, CF Centre, Woman and Child, KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Anton Willems
- Laboratory of Applied Mass Spectrometry, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- Metabolomics Core Facility Leuven, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Elizabeth A V Jones
- Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Bart Ghesquière
- Laboratory of Applied Mass Spectrometry, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.
- Metabolomics Core Facility Leuven, Center for Cancer Biology, VIB, Leuven, Belgium.
| |
Collapse
|
15
|
Sun WT, Du JY, Wang J, Wang YL, Dong ED. Potential preservative mechanisms of cardiac rehabilitation pathways on endothelial function in coronary heart disease. SCIENCE CHINA. LIFE SCIENCES 2025; 68:158-175. [PMID: 39395086 DOI: 10.1007/s11427-024-2656-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/17/2024] [Indexed: 10/14/2024]
Abstract
Cardiac rehabilitation, a comprehensive exercise-based lifestyle and medical management, is effective in decreasing morbidity and improving life quality in patients with coronary heart disease. Endothelial function, an irreplaceable indicator in coronary heart disease progression, is measured by various methods in traditional cardiac rehabilitation pathways, including medicinal treatment, aerobic training, and smoking cessation. Nevertheless, studies on the effect of some emerging cardiac rehabilitation programs on endothelial function are limited. This article briefly reviewed the endothelium-beneficial effects of different cardiac rehabilitation pathways, including exercise training, lifestyle modification and psychological intervention in patients with coronary heart disease, and related experimental models, and summarized both uncovered and potential cellular and molecular mechanisms of the beneficial roles of various cardiac rehabilitation pathways on endothelial function. In exercise training and some lifestyle interventions, the enhanced bioavailability of nitric oxide, increased circulating endothelial progenitor cells (EPCs), and decreased oxidative stress are major contributors to preventing endothelial dysfunction in coronary heart disease. Moreover, the preservation of endothelial-dependent hyperpolarizing factors and inflammatory suppression play roles. On the one hand, to develop more endothelium-protective rehabilitation methods in coronary heart disease, adequately designed and sized randomized multicenter clinical trials should be advanced using standardized cardiac rehabilitation programs and existing assessment methods. On the other hand, additional studies using suitable experimental models are warranted to elucidate the relationship between some new interventions and endothelial protection in both macro- and microvasculature.
Collapse
Affiliation(s)
- Wen-Tao Sun
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, 266071, China.
| | - Jian-Yong Du
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, 266071, China
| | - Jia Wang
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, 266071, China
| | - Yi-Long Wang
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, 266071, China
| | - Er-Dan Dong
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, 266071, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, 100191, China.
- The Institute of Cardiovascular Sciences, Peking University, Beijing, 100191, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China.
| |
Collapse
|
16
|
Hansen CE, Hollaus D, Kamermans A, de Vries HE. Tension at the gate: sensing mechanical forces at the blood-brain barrier in health and disease. J Neuroinflammation 2024; 21:325. [PMID: 39696463 PMCID: PMC11657007 DOI: 10.1186/s12974-024-03321-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024] Open
Abstract
Microvascular brain endothelial cells tightly limit the entry of blood components and peripheral cells into the brain by forming the blood-brain barrier (BBB). The BBB is regulated by a cascade of mechanical and chemical signals including shear stress and elasticity of the adjacent endothelial basement membrane (BM). During physiological aging, but especially in neurological diseases including multiple sclerosis (MS), stroke, small vessel disease, and Alzheimer's disease (AD), the BBB is exposed to inflammation, rigidity changes of the BM, and disturbed cerebral blood flow (CBF). These altered forces lead to increased vascular permeability, reduced endothelial reactivity to vasoactive mediators, and promote leukocyte transmigration. Whereas the molecular players involved in leukocyte infiltration have been described in detail, the importance of mechanical signalling throughout this process has only recently been recognized. Here, we review relevant features of mechanical forces acting on the BBB under healthy and pathological conditions, as well as the endothelial mechanosensory elements detecting and responding to altered forces. We demonstrate the underlying complexity by focussing on the family of transient receptor potential (TRP) ion channels. A better understanding of these processes will provide insights into the pathogenesis of several neurological disorders and new potential leads for treatment.
Collapse
Affiliation(s)
- Cathrin E Hansen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - David Hollaus
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Alwin Kamermans
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands.
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
17
|
Chakraborty S, Mantripragada VT, Chakravarty A, Goswami D, Poddar A. Unraveling the complex interplay between abnormal hemorheology and shape asymmetry in flow through stenotic arteries. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 257:108437. [PMID: 39357092 DOI: 10.1016/j.cmpb.2024.108437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND AND OBJECTIVE Stenosis or narrowing of arteries due to the buildup of plaque is a common occurrence in atherosclerosis and coronary artery disease (CAD), limiting blood flow to the heart and posing substantial cardiovascular risk. While the role of geometric irregularities in arterial stenosis is well-documented, the complex interplay between the abnormal hemorheology and asymmetric shape in flow characteristics remains unexplored. METHODS This study investigates the influence of varying hematocrit (Hct) levels, often caused by conditions such as diabetes and anemia, on flow patterns in an idealized eccentric stenotic artery using computational fluid dynamics simulations. We consider three physiological levels of Hct, 25%, 45%, and 65%, representing anemia, healthy, and diabetic conditions, respectively. The numerical simulations are performed for different combinations of shape eccentricity and blood rheological parameters, and hemodynamic indicators such as wall shear stress (WSS), oscillatory shear index (OSI), are relative residence time (RRT) are calculated to assess the arterial health. RESULTS Our results reveal the significant influence of Hct level on stenosis progression. CAD patients with anemia are exposed to lower WSS and higher OSI, which may increase the propensity for plaque progression and rupture. However, for CAD patients with high Hct level - as is often the case in diabetes - the WSS at the minimal lumen area increases rapidly, which may also lead to plaque rupture and cause adverse events such as heart attacks. These disturbances promote endothelial dysfunction, inflammation, and thrombus formation, thereby intensifying cardiovascular risk. CONCLUSIONS Our findings underscore the significance of incorporating hemorheological parameters, such as Hct, into computational models for accurate assessment of flow dynamics. We envision that insights gained from this study will inform the development of tailored treatment strategies and interventions in CAD patients with common comorbidities such as diabetes and anemia, thus mitigating the adverse effects of abnormal hemorheology and reducing the ever-growing burden of cardiovascular diseases.
Collapse
Affiliation(s)
- Soumen Chakraborty
- Department of Mechanical Engineering, Indian Institute of Technology (Indian School of Mines), Dhanbad, Jharkhand 826004, India
| | - Vishnu Teja Mantripragada
- Department of Fuel, Minerals and Metallurgical Engineering, Indian Institute of Technology (Indian School of Mines), Dhanbad, Jharkhand 826004, India
| | - Aranyak Chakravarty
- School of Nuclear Studies and Application, Jadavpur University, Kolkata, West Bengal 700106, India
| | - Debkalpa Goswami
- Department of Cardiovascular Medicine, Heart, Vascular & Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Antarip Poddar
- Department of Mechanical Engineering, Indian Institute of Technology (Indian School of Mines), Dhanbad, Jharkhand 826004, India.
| |
Collapse
|
18
|
Li W, Li J, Pan C, Lee JS, Kim BS, Gao G. Light-based 3D bioprinting techniques for illuminating the advances of vascular tissue engineering. Mater Today Bio 2024; 29:101286. [PMID: 39435375 PMCID: PMC11492625 DOI: 10.1016/j.mtbio.2024.101286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/21/2024] [Accepted: 10/01/2024] [Indexed: 10/23/2024] Open
Abstract
Vascular tissue engineering faces significant challenges in creating in vitro vascular disease models, implantable vascular grafts, and vascularized tissue/organ constructs due to limitations in manufacturing precision, structural complexity, replicating the composited architecture, and mimicking the mechanical properties of natural vessels. Light-based 3D bioprinting, leveraging the unique advantages of light including high resolution, rapid curing, multi-material adaptability, and tunable photochemistry, offers transformative solutions to these obstacles. With the emergence of diverse light-based 3D bioprinting techniques and innovative strategies, the advances in vascular tissue engineering have been significantly accelerated. This review provides an overview of the human vascular system and its physiological functions, followed by an in-depth discussion of advancements in light-based 3D bioprinting, including light-dominated and light-assisted techniques. We explore the application of these technologies in vascular tissue engineering for creating in vitro vascular disease models recapitulating key pathological features, implantable blood vessel grafts, and tissue analogs with the integration of capillary-like vasculatures. Finally, we provide readers with insights into the future perspectives of light-based 3D bioprinting to revolutionize vascular tissue engineering.
Collapse
Affiliation(s)
- Wei Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Jinhua Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
- Beijing Institute of Technology, Zhuhai, Beijing Institute of Technology (BIT), Zhuhai 519088, China
| | - Chen Pan
- School of Mechanical Engineering, Beijing Institute of Technology, Beijing 100081, China
- School of Mechanical and Equipment Engineering, Hebei University of Engineering, Handan, 050024, China
| | - Jae-Seong Lee
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Information Convergence Engineering, Pusan National University, Busan 50612, Republic of Korea
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Information Convergence Engineering, Pusan National University, Busan 50612, Republic of Korea
| | - Ge Gao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
| |
Collapse
|
19
|
Wang Z, Zou Q, Magermans L, Amselem G, Dessalles CA, Louis B, Filoche M, Gacoin T, Kim J. Shearmetry of Fluids with Tunable Rheology by Polarized Luminescence of Rare Earth-Doped Nanorods. ACS NANO 2024; 18:30650-30657. [PMID: 39404453 DOI: 10.1021/acsnano.4c09493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Shear stress plays a critical role in regulating physiological processes within microcirculatory systems. While particle imaging velocimetry is a standard technique for quantifying shear flow, uncertainty near boundaries and low resolution remain severe restrictions. Additionally, shear stress determination is particularly challenging in biofluids due to their significant non-Newtonian behaviors. The present study develops a shearmetry technique in physiological settings using a biomimetic fluid containing rare earth-doped luminescent nanorods acting in two roles. First, they are used as colloidal additives adjusting rheological properties in physiological media. Their anisotropic morphology and interparticle interaction synergistically induce a non-Newtonian shear-thinning effect emulating real biofluids. Second, they can probe shear stress due to the shear-induced alignment. The polarized luminescence of the nanorods allows for quantifying their orientational order parameter and thus correlated shear stress. Using scanning confocal microscopy, we demonstrate the tomographic mapping of the shear stress distribution in microfluidics. High shear stress is evident near the constriction and the cellular periphery, in which non-Newtonian effects can have a significant impact. This emerging shearmetry technique is promising for implementation in physiological and rheological environments of biofluids.
Collapse
Affiliation(s)
- Zijun Wang
- Laboratoire de Physique de la Matière Condensée, Ecole Polytechnique, CNRS, IP Paris, 91128 Palaiseau, France
- L'Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, INSERM U955, CNRS, 94010 Créteil, France
| | - Qilin Zou
- Laboratoire de Physique de la Matière Condensée, Ecole Polytechnique, CNRS, IP Paris, 91128 Palaiseau, France
| | - Lilian Magermans
- Laboratoire de Physique de la Matière Condensée, Ecole Polytechnique, CNRS, IP Paris, 91128 Palaiseau, France
| | - Gabriel Amselem
- Laboratoire d'Hydrodynamique, Ecole Polytechnique, CNRS, IP Paris, 91128 Palaiseau, France
| | - Claire A Dessalles
- Laboratoire d'Hydrodynamique, Ecole Polytechnique, CNRS, IP Paris, 91128 Palaiseau, France
| | - Bruno Louis
- L'Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, INSERM U955, CNRS, 94010 Créteil, France
| | - Marcel Filoche
- L'Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, INSERM U955, CNRS, 94010 Créteil, France
- Institut Langevin, ESPCI Paris, PSL University, CNRS, 75005 Paris, France
| | - Thierry Gacoin
- Laboratoire de Physique de la Matière Condensée, Ecole Polytechnique, CNRS, IP Paris, 91128 Palaiseau, France
| | - Jongwook Kim
- Laboratoire de Physique de la Matière Condensée, Ecole Polytechnique, CNRS, IP Paris, 91128 Palaiseau, France
| |
Collapse
|
20
|
Zhang J. Non-coding RNAs and angiogenesis in cardiovascular diseases: a comprehensive review. Mol Cell Biochem 2024; 479:2921-2953. [PMID: 38306012 DOI: 10.1007/s11010-023-04919-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/18/2023] [Indexed: 02/03/2024]
Abstract
Non-coding RNAs (ncRNAs) have key roles in the etiology of many illnesses, including heart failure, myocardial infarction, stroke, and in physiological processes like angiogenesis. In transcriptional regulatory circuits that control heart growth, signaling, and stress response, as well as remodeling in cardiac disease, ncRNAs have become important players. Studies on ncRNAs and cardiovascular disease have made great progress recently. Here, we go through the functions of non-coding RNAs (ncRNAs) like circular RNAs (circRNAs), and microRNAs (miRNAs) as well as long non-coding RNAs (lncRNAs) in modulating cardiovascular disorders.
Collapse
Affiliation(s)
- Jie Zhang
- Medical School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
21
|
Zhang X, Ding H, Ji X, Chen L, Huang P, Lin Z, Zhu J, Zhou S, Liu Z, Zhang M, Xu Q. Predicting vulnerable carotid plaques by detecting wall shear stress based on ultrasonic vector flow imaging. J Vasc Surg 2024; 80:1475-1486.e1. [PMID: 38925348 DOI: 10.1016/j.jvs.2024.06.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVE Carotid plaque vulnerability is a significant factor in the risk of cardiocerebrovascular events, with intraplaque neovascularization (IPN) being a crucial characteristic of plaque vulnerability. This study investigates the value of ultrasound vector flow imaging (V-flow) for measuring carotid plaque wall shear stress (WSS) in predicting the extent of IPN. METHODS We enrolled 140 patients into three groups: 53 in the plaque group (72 plaques), 23 in the stenosis group (27 plaques), and 64 in the control group. V-flow was used to measure WSS parameters, including the average WSS (WSS mean) and the maximum WSS (WSS max), across three plaque locations: mid-upstream, maximum thickness, and mid-downstream. Contrast-enhanced ultrasound examination was used in 76 patients to analyze IPN and its correlation with WSS parameters. RESULTS WSS max in the stenosis group was significantly higher than that in the control and plaque groups at the maximum thickness part (P < .05) and WSS mean in the stenosis group was significantly lower than that in the control group at the mid-upstream and mid-downstream segments (P < .05). WSS mean in the plaque group was significantly lower than that of the control group at all three locations (P < .05). Contrast-enhanced ultrasound examination revealed that plaques with neovascularization enhancement exhibited significantly higher WSS values (P < .05), with a positive correlation between WSS parameters and IPN enhancement grades, particularly WSS max at the thickest part (r = 0.508). Receiver operating characteristic curve analysis of WSS parameters for evaluating IPN showed that the efficacy of WSS max in evaluating IPN was better than that of WSS mean (P < .05), with an area under the curve of 0.7762 and 0.6973 (95% confidence intervals, 0.725-0.822 and 0.642-0.749, respectively). The cut-offs were 4.57 Pa and 1.12 Pa, sensitivities were 74.03% and 63.64%, and specificities were 75.00% and 68.18%. CONCLUSIONS V-flow effectively measures WSS in carotid plaques. WSS max provides a promising metric for assessing IPN, offering potential insights into plaque characteristics and showing some potential in predicting plaque vulnerability.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Ultrasonography, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huanhuan Ding
- Department of Ultrasonography, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoli Ji
- Department of Ultrasonography, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ling Chen
- Department of Ultrasonography, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Peipei Huang
- Department of Ultrasonography, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zengqiao Lin
- Department of Ultrasonography, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianbi Zhu
- Department of Ultrasonography, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shujing Zhou
- Department of Ultrasonography, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zezheng Liu
- Department of Ultrasonography, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Miaomiao Zhang
- Department of Ultrasonography, Lingkun Street Community Health Service Center of Dongtou District, Wenzhou, Zhejiang, China
| | - Qi Xu
- Department of Ultrasonography, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
22
|
Mai-Thi HN, Nguyen DPH, Le P, Tran NQ, Tran CT, Stoldt VR, Huynh K. Low shear-induced fibrillar fibronectin: comparative analyses of morphologies and cellular effects on bovine aortic endothelial cell adhesion and proliferation. Phys Biol 2024; 21:066002. [PMID: 39366409 DOI: 10.1088/1478-3975/ad838c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 10/04/2024] [Indexed: 10/06/2024]
Abstract
Wall shear stress (WSS) is a critical factor in vascular biology, and both high and low WSS are implicated in atherosclerosis. Fibronectin (FN) is a key extracellular matrix protein that plays an important role in cell activities. Under high shear stress, plasma FN undergoes fibrillogenesis; however, its behavior under low shear stress remains unclear. This study aimed to investigate the formation ofin vitrocell-free fibrillar FN (FFN) under low shear rate conditions and its effect on bovine aortic endothelial cell behavior. FN (500µg ml-1) was perfused through slide chambers at three flow rates (0.16 ml h-1, 0.25 ml h-1, and 0.48 ml h-1), corresponding to low shear rates of 0.35 s-1, 0.55 s-1, and 1.05 s-1, respectively, for 4 h at room temperature. The formed FN matrices were observed using fluorescence microscopy and scanning electron microscopy. Under low shear rates, distinct FN matrix structures were observed. FFN0.48 formed immense fibrils with smooth surfaces, FFN0.25 formed a matrix with a rough surface, and FFN16 exhibited nodular structures. FFN0.25 supported cell activities to a greater extent than native FN and other FFN surfaces. Our study suggests that abnormally low shear conditions impact FN structure and function and enhance the understanding of FN fibrillogenesis in vascular biology, particularly in atherosclerosis.
Collapse
Affiliation(s)
- Hoang-Nghi Mai-Thi
- School of Biomedical Engineering, International University, Quarter 6, Linh Trung, Thu Duc District, Ho Chi Minh City, Vietnam
- Vietnam National University, Linh Trung, Thu Duc District, Ho Chi Minh City, Vietnam
- School of Nursing, Eastern International University, Nam Ky Khoi Nghia Street, Hoa Phu Ward, Thu Dau Mot City, Binh Duong Province, Vietnam
| | - Dang Phu-Hai Nguyen
- School of Biomedical Engineering, International University, Quarter 6, Linh Trung, Thu Duc District, Ho Chi Minh City, Vietnam
- School of Biotechnology, International University, Quarter 6, Linh Trung, Thu Duc District, Ho Chi Minh City, Vietnam
- Vietnam National University, Linh Trung, Thu Duc District, Ho Chi Minh City, Vietnam
| | - Phong Le
- School of Biomedical Engineering, International University, Quarter 6, Linh Trung, Thu Duc District, Ho Chi Minh City, Vietnam
- Vietnam National University, Linh Trung, Thu Duc District, Ho Chi Minh City, Vietnam
| | - Ngoc Quyen Tran
- Graduate University of Science and Technology Viet Nam, Vietnam Academy of Science and Technology, Ho Chi Minh City, Viet Nam; and Institute of Applied Materials Science, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
| | - Cam Tu Tran
- Institute of Tropical Biology, Viet Nam Academy of Science and Technology (VAST), Ho Chi Minh City, Vietnam
| | - Volker R Stoldt
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Moorenstr. 5, Düsseldorf 40225, Germany
| | - Khon Huynh
- School of Biomedical Engineering, International University, Quarter 6, Linh Trung, Thu Duc District, Ho Chi Minh City, Vietnam
- Vietnam National University, Linh Trung, Thu Duc District, Ho Chi Minh City, Vietnam
| |
Collapse
|
23
|
Mote N, Kubik S, Polacheck WJ, Baker BM, Trappmann B. A nanoporous hydrogel-based model to study chemokine gradient-driven angiogenesis under luminal flow. LAB ON A CHIP 2024; 24:4892-4906. [PMID: 39308400 DOI: 10.1039/d4lc00460d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
The growth of new blood vessels through angiogenesis is a highly coordinated process, which is initiated by chemokine gradients that activate endothelial cells within a perfused parent vessel to sprout into the surrounding 3D tissue matrix. While both biochemical signals from pro-angiogenic factors, as well as mechanical cues originating from luminal fluid flow that exerts shear stress on the vessel wall, have individually been identified as major regulators of endothelial cell sprouting, it remains unclear whether and how both types of cues synergize. To fill this knowledge gap, here, we created a 3D biomimetic model of chemokine gradient-driven angiogenic sprouting, in which a micromolded tube inside a hydrogel matrix is seeded with endothelial cells and connected to a perfusion system to control fluid flow rates and resulting shear forces on the vessel wall. To allow for the formation of chemokine gradients despite the presence of luminal flow, a nanoporous synthetic hydrogel that supports angiogenesis but limits the interstitial flow proved crucial. Using this system, we find that luminal flow and resulting shear stress is a major regulator of the speed and morphogenesis of angiogenic sprouting, whose action is mediated through changes in vascular permeability.
Collapse
Affiliation(s)
- Nidhi Mote
- Bioactive Materials Laboratory, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Sarah Kubik
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, 27514 USA
| | - William J Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, 27514 USA
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, 2174 Lurie BME Building, 1101 Beal Avenue, Ann Arbor, MI, 48109 USA
| | - Britta Trappmann
- Bioactive Materials Laboratory, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Straße 6, 44227 Dortmund, Germany.
| |
Collapse
|
24
|
Ebrahimi A, Ghorbanpoor H, Apaydın E, Demir Cevizlidere B, Özel C, Tüfekçioğlu E, Koç Y, Topal AE, Tomsuk Ö, Güleç K, Abdullayeva N, Kaya M, Ghorbani A, Şengel T, Benzait Z, Uysal O, Eker Sarıboyacı A, Doğan Güzel F, Singh H, Hassan S, Ankara H, Pat S, Atalay E, Avci H. Convenient rapid prototyping microphysiological niche for mimicking liver native basement membrane: Liver sinusoid on a chip. Colloids Surf B Biointerfaces 2024; 245:114292. [PMID: 39383580 DOI: 10.1016/j.colsurfb.2024.114292] [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: 07/08/2024] [Revised: 09/24/2024] [Accepted: 10/02/2024] [Indexed: 10/11/2024]
Abstract
Liver is responsible for the metabolization processes of up to 90 % of compounds and toxins in the body. Therefore liver-on-a-chip systems, as an in vitro promising cell culture platform, have great importance for fundamental science and drug development. In most of the liver-on-a-chip studies, seeding cells on both sides of a porous membrane, which represents the basement membrane, fail to resemble the native characteristics of biochemical, biophysical, and mechanical properties. In this study, polycarbonate (PC) and polyethylene terephthalate (PET) membranes were coated with gelatin to address this issue by accurately mimicking the native basement membrane present in the space of Disse. Various coating methods were used, including doctor blade, gel micro-injection, electrospinning, and spin coating. Spin coating was demonstrated to be the most effective technique owing to the ability to produce thin gel thickness with desirable surface roughness for cell interactions on both sides of the membrane. HepG2 and EA.HY926 cells were seeded on the upper and bottom sides of the gelatin-coated PET membrane and cultured on-chip for 7 days. Cell viability increased from 90 % to 95 %, while apoptotic index decreased. Albumin secretion notably rose between days 1-7 and 4-7, while GST-α secretion decreased from day 1 to day 7. In conclusion, the optimized spin coating process reported here can effectively modify the membranes to better mimic the native basement membrane niche characteristics.
Collapse
Affiliation(s)
- Aliakbar Ebrahimi
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Hamed Ghorbanpoor
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Biomedical Engineering, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Elif Apaydın
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Biochemistry, Institute of Health Sciences, Anadolu University, Eskisehir, Türkiye
| | - Bahar Demir Cevizlidere
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Ceren Özel
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Emre Tüfekçioğlu
- Department of Industrial Design/Department of Industrial Design, Faculty of Architecture and Design, Eskisehir Technical University, Eskisehir, Türkiye
| | - Yücel Koç
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Ahmet Emin Topal
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Biochemistry, School of Pharmacy, Bahçeşehir University, Istanbul, Türkiye
| | - Özlem Tomsuk
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Mechanical Engineering, Middle East Technical University, Ankara 06800, Türkiye
| | - Kadri Güleç
- Department of Analytical Chemistry, Institute of Health Sciences, Anadolu University, Eskisehir, Türkiye
| | - Nuran Abdullayeva
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Murat Kaya
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Aynaz Ghorbani
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Tayfun Şengel
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye; Central Research Laboratory Research and Application Center (ARUM), Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Zineb Benzait
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Onur Uysal
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Ayla Eker Sarıboyacı
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Fatma Doğan Güzel
- Department of Biomedical Engineering, Ankara Yildirim Beyazit University, Ankara, Türkiye
| | - Hemant Singh
- Department of Biological Sciences, Khalifa University, Main Campus, Abu Dhabi, United Arab Emirates; Center for Biotechnology, Khalifa University, Main Campus, Abu Dhabi, United Arab Emirates; Functional Biomaterials Group, Khalifa University, San Campus, Abu Dhabi, United Arab Emirates
| | - Shabir Hassan
- Department of Biological Sciences, Khalifa University, Main Campus, Abu Dhabi, United Arab Emirates; Center for Biotechnology, Khalifa University, Main Campus, Abu Dhabi, United Arab Emirates; Functional Biomaterials Group, Khalifa University, San Campus, Abu Dhabi, United Arab Emirates
| | - Hüseyin Ankara
- Mining Engineering Department, Engineering-Architecture Faculty, Eskisehir Osmangazi University, Meşelik Campus, Eskisehir 26480, Türkiye
| | - Suat Pat
- Eskisehir Osmangazi University, Faculty of Science, Department of Physics, Eskisehir TR-26040, Türkiye
| | - Eray Atalay
- Department of Ophthalmology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir 26040, Türkiye
| | - Huseyin Avci
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Metallurgical and Materials Engineering, Eskisehir Osmangazi University, Eskisehir, Türkiye; Translational Medicine Research and Clinical Center (TATUM), Eskisehir Osmangazi University, Eskisehir, Türkiye.
| |
Collapse
|
25
|
Alcaide D, Alric B, Cacheux J, Nakano S, Doi K, Shinohara M, Kondo M, Bancaud A, Matsunaga YT. Laminin and hyaluronan supplementation of collagen hydrogels enhances endothelial function and tight junction expression on three-dimensional cylindrical microvessel-on-a-chip. Biochem Biophys Res Commun 2024; 724:150234. [PMID: 38865812 DOI: 10.1016/j.bbrc.2024.150234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 05/30/2024] [Accepted: 06/05/2024] [Indexed: 06/14/2024]
Abstract
Vasculature-on-chip (VoC) models have become a prominent tool in the study of microvasculature functions because of their cost-effective and ethical production process. These models typically use a hydrogel in which the three-dimensional (3D) microvascular structure is embedded. Thus, VoCs are directly impacted by the physical and chemical cues of the supporting hydrogel. Endothelial cell (EC) response in VoCs is critical, especially in organ-specific vasculature models, in which ECs exhibit specific traits and behaviors that vary between organs. Many studies customize the stimuli ECs perceive in different ways; however, customizing the hydrogel composition accordingly to the target organ's extracellular matrix (ECM), which we believe has great potential, has been rarely investigated. We explored this approach to organ-specific VoCs by fabricating microvessels (MVs) with either human umbilical vein ECs or human brain microvascular ECs in a 3D cylindrical VoC using a collagen hydrogel alone or one supplemented with laminin and hyaluronan, components found in the brain ECM. We characterized the physical properties of these hydrogels and analyzed the barrier properties of the MVs. Barrier function and tight junction (ZO-1) expression improved with the addition of laminin and hyaluronan in the composite hydrogel.
Collapse
Affiliation(s)
- Daniel Alcaide
- Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan; Department of Bioengineering, School of Engineering, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan; LIMMS, CNRS-IIS UMI 2820, The University of Tokyo, Tokyo, 153-8505, Japan
| | - Baptiste Alric
- Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan; LIMMS, CNRS-IIS UMI 2820, The University of Tokyo, Tokyo, 153-8505, Japan
| | - Jean Cacheux
- Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan; LIMMS, CNRS-IIS UMI 2820, The University of Tokyo, Tokyo, 153-8505, Japan; Centre de Recherches en Cancérologie de Toulouse, Inserm, CNRS, Université Paul Sabatier, Université de Toulouse, 31037, Toulouse, France
| | - Shizuka Nakano
- Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan; Department of Bioengineering, School of Engineering, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan
| | - Kotaro Doi
- Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan
| | - Marie Shinohara
- Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan; Department of Bioengineering, School of Engineering, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan
| | - Makoto Kondo
- Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan
| | - Aurelien Bancaud
- Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan; LIMMS, CNRS-IIS UMI 2820, The University of Tokyo, Tokyo, 153-8505, Japan; LAAS-CNRS, CNRS UPR8001, 7 Avenue du Colonel Roche, 31400, Toulouse, France.
| | - Yukiko T Matsunaga
- Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan; Department of Bioengineering, School of Engineering, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan; LIMMS, CNRS-IIS UMI 2820, The University of Tokyo, Tokyo, 153-8505, Japan.
| |
Collapse
|
26
|
Hu SY, Ye JR, Chou HW, Chen YS, Chang CI, Chiu IS, Lin CT, Tsai HE, Chen SJ, Huang SC. Impact of the pulmonary venous entry site morphology on postoperative pulmonary vein stenosis in total anomalous pulmonary venous connection patients. J Formos Med Assoc 2024:S0929-6646(24)00410-8. [PMID: 39242224 DOI: 10.1016/j.jfma.2024.09.006] [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: 02/08/2024] [Revised: 07/29/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND To evaluate the association between the pulmonary vein (PV) entry site morphology after total anomalous pulmonary vein repair (TAPVC) and postoperative pulmonary vein stenosis (PVS). METHODS Computed tomography (CT) examination was performed to determine the PV entry site morphology. The width of the PV confluence was divided by the width of the left atrium (LA) to obtain the cPV/LA index. The cPV/LA index was compared between patients with and without postoperative PVS. RESULTS Fifty-one patients who had undergone CT after TAPVC repair were included, with a median cPV/LA index of 0.5 (interquartile range (IQR) = 0.349-0.654). Among them, 27 patients developed postoperative PVS. The median cPV/LA index after primary TAPVC repair was significantly lower in patients with PVS compared to those without PVS (0.367, IQR = 0.308-0.433 vs. 0.657, IQR = 0.571-0.783, P < 0.0001). Additionally, the cPV/LA index after surgical re-intervention for PVS was significantly smaller in patients who developed recurrent stenosis compared to those who remained free-from re-stenosis after surgical relief (0.459, IQR = 0.349-0.556; vs. 0.706, IQR = 0.628-0.810, P = 0.0045). CONCLUSION A small PV confluence width is associated with the development of postoperative PVS and recurrent stenosis after surgical relief of PVS. Our results suggest that adequate bilateral pulmonary vein lateralization during TAPVC surgery is crucial.
Collapse
Affiliation(s)
- Szu-Yen Hu
- Department of Cardiovascular Surgery, National Taiwan University Hospital Hsinchu Branch, Hsinchu, Taiwan; Graduate Institute of Biomedical Electronics and Bioinformatics National Taiwan University Taipei Taiwan
| | - Jing-Ren Ye
- Department of Cardiovascular Surgery, National Taiwan University Hospital Hsinchu Branch, Hsinchu, Taiwan
| | - Heng-Wen Chou
- Department of Surgery, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yih-Sharng Chen
- Department of Surgery, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chung-I Chang
- Department of Surgery, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ing-Sh Chiu
- Department of Surgery, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chih-Ting Lin
- Graduate Institute of Biomedical Electronics and Bioinformatics National Taiwan University Taipei Taiwan
| | - Hsiao-En Tsai
- Department of Cardiovascular Surgery, National Taiwan University Hospital Hsinchu Branch, Hsinchu, Taiwan
| | - Shyh-Jye Chen
- Department of Radiology and Medical Imaging, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Shu-Chien Huang
- Department of Surgery, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
27
|
Allen MF, Park SY, Kwak YS. Oxidative stress and vascular dysfunction: Potential therapeutic targets and therapies in peripheral artery disease. Microvasc Res 2024; 155:104713. [PMID: 38914307 DOI: 10.1016/j.mvr.2024.104713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 06/26/2024]
Abstract
Peripheral artery disease (PAD) is the manifestation of atherosclerosis characterized by the accumulation of plaques in the arteries of the lower limbs. Interestingly, growing evidence suggests that the pathology of PAD is multifaceted and encompasses both vascular and skeletal muscle dysfunctions, which contributes to blunted physical capabilities and diminished quality of life. Importantly, it has been suggested that many of these pathological impairments may stem from blunted reduction-oxidation (redox) handling. Of note, in those with PAD, excessive production of reactive oxygen species (ROS) outweighs antioxidant capabilities resulting in oxidative damage, which may have systemic consequences. It has been suggested that antioxidant supplementation may be able to assist in handling ROS. However, the activation of various ROS production sites makes it difficult to determine the efficacy of these antioxidant supplements. Therefore, this review focuses on the common cellular mechanisms that facilitate ROS production and discusses how excessive ROS may impair vascular and skeletal muscle function in PAD. Furthermore, we provide insight for current and potential antioxidant therapies, specifically highlighting activation of the Kelch-like ECH-associated protein 1 (Keap1) - Nuclear Factor Erythroid 2-related factor 2 (Nrf2) pathway as a potential pharmacological therapy to combat ROS accumulation and aid in vascular function, and physical performance in patients with PAD. Altogether, this review provides a better understanding of excessive ROS in the pathophysiology of PAD and enhances our perception of potential therapeutic targets that may improve vascular function, skeletal muscle function, walking capacity, and quality of life in patients with PAD.
Collapse
Affiliation(s)
- Michael F Allen
- School of Health and Kinesiology, University of Nebraska at Omaha, Omaha, NE, United States of America
| | - Song-Young Park
- School of Health and Kinesiology, University of Nebraska at Omaha, Omaha, NE, United States of America; Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Yi-Sub Kwak
- Department of Physical Education, College of Arts, Design, and Sports Science, Dong-Eui University, Busan, Republic of Korea.
| |
Collapse
|
28
|
Liu Y, Kamran R, Han X, Wang M, Li Q, Lai D, Naruse K, Takahashi K. Human heart-on-a-chip microphysiological system comprising endothelial cells, fibroblasts, and iPSC-derived cardiomyocytes. Sci Rep 2024; 14:18063. [PMID: 39117679 PMCID: PMC11310341 DOI: 10.1038/s41598-024-68275-0] [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: 05/09/2024] [Accepted: 07/22/2024] [Indexed: 08/10/2024] Open
Abstract
In recent years, research on organ-on-a-chip technology has been flourishing, particularly for drug screening and disease model development. Fibroblasts and vascular endothelial cells engage in crosstalk through paracrine signaling and direct cell-cell contact, which is essential for the normal development and function of the heart. Therefore, to faithfully recapitulate cardiac function, it is imperative to incorporate fibroblasts and vascular endothelial cells into a heart-on-a-chip model. Here, we report the development of a human heart-on-a-chip composed of induced pluripotent stem cell (iPSC)-derived cardiomyocytes, fibroblasts, and vascular endothelial cells. Vascular endothelial cells cultured on microfluidic channels responded to the flow of culture medium mimicking blood flow by orienting themselves parallel to the flow direction, akin to in vivo vascular alignment in response to blood flow. Furthermore, the flow of culture medium promoted integrity among vascular endothelial cells, as evidenced by CD31 staining and lower apparent permeability. The tri-culture condition of iPSC-derived cardiomyocytes, fibroblasts, and vascular endothelial cells resulted in higher expression of the ventricular cardiomyocyte marker IRX4 and increased contractility compared to the bi-culture condition with iPSC-derived cardiomyocytes and fibroblasts alone. Such tri-culture-derived cardiac tissues exhibited cardiac responses similar to in vivo hearts, including an increase in heart rate upon noradrenaline administration. In summary, we have achieved the development of a heart-on-a-chip composed of cardiomyocytes, fibroblasts, and vascular endothelial cells that mimics in vivo cardiac behavior.
Collapse
Affiliation(s)
- Yun Liu
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama City, 700-8558, Japan
| | - Rumaisa Kamran
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama City, 700-8558, Japan
| | - Xiaoxia Han
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama City, 700-8558, Japan
| | - Mengxue Wang
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama City, 700-8558, Japan
| | - Qiang Li
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama City, 700-8558, Japan
| | - Daoyue Lai
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama City, 700-8558, Japan
| | - Keiji Naruse
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama City, 700-8558, Japan
| | - Ken Takahashi
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama City, 700-8558, Japan.
| |
Collapse
|
29
|
Corti A, Marradi M, Çelikbudak Orhon C, Boccafoschi F, Büchler P, Rodriguez Matas JF, Chiastra C. Impact of Tissue Damage and Hemodynamics on Restenosis Following Percutaneous Transluminal Angioplasty: A Patient-Specific Multiscale Model. Ann Biomed Eng 2024; 52:2203-2220. [PMID: 38702558 PMCID: PMC11247064 DOI: 10.1007/s10439-024-03520-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 04/17/2024] [Indexed: 05/06/2024]
Abstract
Multiscale agent-based modeling frameworks have recently emerged as promising mechanobiological models to capture the interplay between biomechanical forces, cellular behavior, and molecular pathways underlying restenosis following percutaneous transluminal angioplasty (PTA). However, their applications are mainly limited to idealized scenarios. Herein, a multiscale agent-based modeling framework for investigating restenosis following PTA in a patient-specific superficial femoral artery (SFA) is proposed. The framework replicates the 2-month arterial wall remodeling in response to the PTA-induced injury and altered hemodynamics, by combining three modules: (i) the PTA module, consisting in a finite element structural mechanics simulation of PTA, featuring anisotropic hyperelastic material models coupled with a damage formulation for fibrous soft tissue and the element deletion strategy, providing the arterial wall damage and post-intervention configuration, (ii) the hemodynamics module, quantifying the post-intervention hemodynamics through computational fluid dynamics simulations, and (iii) the tissue remodeling module, based on an agent-based model of cellular dynamics. Two scenarios were explored, considering balloon expansion diameters of 5.2 and 6.2 mm. The framework captured PTA-induced arterial tissue lacerations and the post-PTA arterial wall remodeling. This remodeling process involved rapid cellular migration to the PTA-damaged regions, exacerbated cell proliferation and extracellular matrix production, resulting in lumen area reduction up to 1-month follow-up. After this initial reduction, the growth stabilized, due to the resolution of the inflammatory state and changes in hemodynamics. The similarity of the obtained results to clinical observations in treated SFAs suggests the potential of the framework for capturing patient-specific mechanobiological events occurring after PTA intervention.
Collapse
Affiliation(s)
- Anna Corti
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Via Ponzio 34/5, 20133, Milan, Italy.
| | - Matilde Marradi
- Laboratory of Biological Structure Mechanics (LaBS), Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Cemre Çelikbudak Orhon
- Laboratory of Hemodynamics and Cardiovascular Technology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Francesca Boccafoschi
- Department of Health Sciences, University of Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Philippe Büchler
- ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
| | - Jose F Rodriguez Matas
- Laboratory of Biological Structure Mechanics (LaBS), Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Claudio Chiastra
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| |
Collapse
|
30
|
Snyder Y, Jana S. Influence of Substrate Structure and Associated Properties on Endothelial Cell Behavior in the Context of Behaviors Associated with Laminar Flow Conditions. ACS APPLIED BIO MATERIALS 2024; 7:4664-4678. [PMID: 38939951 DOI: 10.1021/acsabm.4c00504] [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: 06/29/2024]
Abstract
In order to treat most vascular diseases, arterial grafts are commonly employed for replacing small-diameter vessels, yet they often cause thrombosis. The growth of endothelial cells along the interior surfaces of these grafts (substrates) is critical to mitigate thrombosis. Typically, endothelial cells are cultured inside these grafts under laminar flow conditions to emulate the native environment of blood vessels and produce an endothelium. Alternatively, the substrate structure could have a similar influence on endothelial cell behavior as laminar flow conditions. In this study, we investigated whether substrates with aligned fiber structures could induce responses in human umbilical vein endothelial cells (HUVECs) akin to those elicited by laminar flow. Our observations revealed that HUVECs on aligned substrates displayed significant morphological changes, aligning parallel to the fibers, similar to effects reported under laminar flow conditions. Conversely, HUVECs on random substrates maintained their characteristic cobblestone appearance. Notably, cell migration was more significant on aligned substrates. Also, we observed that while vWF expression was similar between both substrates, the HUVECs on aligned substrates showed more expression of platelet/endothelial cell adhesion molecule-1 (PECAM-1/CD31), laminin, and collagen IV. Additionally, these cells exhibited increased gene expression related to critical functions such as proliferation, extracellular matrix production, cytoskeletal reorganization, autophagy, and antithrombotic activity. These findings indicated that aligned substrates enhanced endothelial growth and behavior compared to random substrates. These improvements are similar to the beneficial effects of laminar flow on endothelial cells, which are well-documented compared to static or turbulent flow conditions.
Collapse
Affiliation(s)
- Yuriy Snyder
- Department of Bioengineering, University of Missouri, Columbia, Missouri 65211, United States
| | - Soumen Jana
- Department of Bioengineering, University of Missouri, Columbia, Missouri 65211, United States
| |
Collapse
|
31
|
Mzimela N, Dimba N, Sosibo A, Khathi A. Evaluating the impact of type 2 diabetes mellitus on pulmonary vascular function and the development of pulmonary fibrosis. Front Endocrinol (Lausanne) 2024; 15:1431405. [PMID: 39050565 PMCID: PMC11266053 DOI: 10.3389/fendo.2024.1431405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
The increasing prevalence of type 2 diabetes mellitus (T2DM) is a significant worldwide health concern caused by sedentary lifestyles and unhealthy diets. Beyond glycemic control, T2DM impacts multiple organ systems, leading to various complications. While traditionally associated with cardiovascular and microvascular complications, emerging evidence indicates significant effects on pulmonary health. Pulmonary vascular dysfunction and fibrosis, characterized by alterations in vascular tone and excessive extracellular matrix deposition, are increasingly recognized in individuals with T2DM. The onset of T2DM is often preceded by prediabetes, an intermediate hyperglycemic state that is associated with increased diabetes and cardiovascular disease risk. This review explores the relationship between T2DM, pulmonary vascular dysfunction and pulmonary fibrosis, with a focus on potential links with prediabetes. Pulmonary vascular function, including the roles of nitric oxide (NO), prostacyclin (PGI2), endothelin-1 (ET-1), thromboxane A2 (TxA2) and thrombospondin-1 (THBS1), is discussed in the context of T2DM and prediabetes. Mechanisms linking T2DM to pulmonary fibrosis, such as oxidative stress, dysregulated fibrotic signaling, and chronic inflammation, are explained. The impact of prediabetes on pulmonary health, including endothelial dysfunction, oxidative stress, and dysregulated vasoactive mediators, is highlighted. Early detection and intervention during the prediabetic stage may reduce respiratory complications associated with T2DM, emphasizing the importance of management strategies targeting blood glucose regulation and vascular health. More research that looks into the mechanisms underlying pulmonary complications in T2DM and prediabetes is needed.
Collapse
Affiliation(s)
- Nhlakanipho Mzimela
- Department of Human Physiology, Faculty of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | | | | | | |
Collapse
|
32
|
Das A, Smith RJ, Andreadis ST. Harnessing the potential of monocytes/macrophages to regenerate tissue-engineered vascular grafts. Cardiovasc Res 2024; 120:839-854. [PMID: 38742656 PMCID: PMC11218695 DOI: 10.1093/cvr/cvae106] [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: 09/22/2023] [Revised: 02/19/2024] [Accepted: 04/02/2024] [Indexed: 05/16/2024] Open
Abstract
Cell-free tissue-engineered vascular grafts provide a promising alternative to treat cardiovascular disease, but timely endothelialization is essential for ensuring patency and proper functioning post-implantation. Recent studies from our lab showed that blood cells like monocytes (MCs) and macrophages (Mϕ) may contribute directly to cellularization and regeneration of bioengineered arteries in small and large animal models. While MCs and Mϕ are leucocytes that are part of the innate immune response, they share common developmental origins with endothelial cells (ECs) and are known to play crucial roles during vessel formation (angiogenesis) and vessel repair after inflammation/injury. They are highly plastic cells that polarize into pro-inflammatory and anti-inflammatory phenotypes upon exposure to cytokines and differentiate into other cell types, including EC-like cells, in the presence of appropriate chemical and mechanical stimuli. This review focuses on the developmental origins of MCs and ECs; the role of MCs and Mϕ in vessel repair/regeneration during inflammation/injury; and the role of chemical signalling and mechanical forces in Mϕ inflammation that mediates vascular graft regeneration. We postulate that comprehensive understanding of these mechanisms will better inform the development of strategies to coax MCs/Mϕ into endothelializing the lumen and regenerate the smooth muscle layers of cell-free bioengineered arteries and veins that are designed to treat cardiovascular diseases and perhaps the native vasculature as well.
Collapse
Affiliation(s)
- Arundhati Das
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, 908 Furnas Hall, Buffalo, NY 14260-4200, USA
| | - Randall J Smith
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, 332 Bonner Hall, Buffalo, NY 14260-1920, USA
| | - Stelios T Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, 908 Furnas Hall, Buffalo, NY 14260-4200, USA
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, 332 Bonner Hall, Buffalo, NY 14260-1920, USA
- Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, The State University of New York, 701 Ellicott St, Buffalo, NY 14203, USA
- Cell, Gene and Tissue Engineering (CGTE) Center, University at Buffalo, The State University of New York, 813 Furnas Hall, Buffalo, NY 14260-4200, USA
| |
Collapse
|
33
|
Cevik Y, Onan HB, Erdem H, Kizilkanat ED, Yucel SP, Oguz O. Investigation of the morphometric characteristics of internal carotid artery between sexes and in patients with intracranial aneurysms. Surg Radiol Anat 2024; 46:859-869. [PMID: 38630269 DOI: 10.1007/s00276-024-03351-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/16/2024] [Indexed: 06/09/2024]
Abstract
PURPOSE The purpose of this study is to investigate the morphometric properties of the internal carotid artery (ICA) by measuring the diameters and angles of its segments and exploring variations related to sex and the presence of aneurysms. METHODS Digital subtraction angiography (DSA) images were utilized from 130 aneurysm patients and 75 non-aneurysm individuals to create 3D ICA models using 3D Slicer software. Segment diameters were measured via Autodesk Meshmixer 3.5.474 and angles were evaluated using ImageJ software. RESULTS In total, DSA images of 130 aneurysm patients and 75 individuals with normally reported carotid systems were evaluated. It was found that the intracranial aneurysms (IAs) were predominantly formed on the anterior cerebral artery (ACA) in males (%43), whereas in females IAs were frequently localized in the C6 segment (31.7%) and middle cerebral artery (MCA) (30.2%). In the control group, the evaluation of gender differences in segment diameters and angles revealed that males had significantly larger C4 and C5 segment diameters (4.62 vs. 4.32 mm and 4.41 vs. 4.09 mm, respectively) and a greater C6 angle (146.9° vs. 139.7°) compared to females. Comparisons between patients with an aneurysm at the anterior cerebral artery (ACA) and the control group revealed that the ACA group had wider diameters in the C1 (4.88 vs. 4.53 mm), C3 (4.65 vs. 4.4 mm), C5 (4.51 vs. 4.25 mm), and ACA (2.36 vs. 2.06 mm) segments. Additionally, the ACA group had wider angles in the ACA (104.1° vs. 94.1°) and C6 segments (147.7° vs. 143.3°), whereas the control group exhibited wider angles in the middle cerebral artery (MCA) segment (141.5° vs. 135.5°) compared to the ACA aneurysm group. Patients with anterior cerebral artery (ACA) aneurysms exhibited larger diameters in C1, C3, C5, C6, and ACA segments compared to the control group. Additionally, while the control group had larger MCA angle, patients with ACA aneurysms had larger angles in C6 segment and ACA. CONCLUSION Our results demonstrated that formation of aneurysms is affected by anatomical configuration of the ICA as well as sex characteristics, particularly regarding the ACA and MCA bifurcation angles, which showed associations with aneurysms in the respective branches.
Collapse
Affiliation(s)
- Yigit Cevik
- Department of Anatomy, Faculty of Medicine, Cukurova University, Adana, 01330, Turkey.
| | - Hasan Bilen Onan
- Department of Radiology, Faculty of Medicine, Cukurova University, Adana, 01330, Turkey
| | - Huseyin Erdem
- Department of Anatomy, Faculty of Medicine, Cukurova University, Adana, 01330, Turkey
| | | | - Sevinc Puren Yucel
- Department of Biostatistics, Faculty of Medicine, Cukurova University, Adana, 01330, Turkey
| | - Ozkan Oguz
- Department of Anatomy, Faculty of Medicine, Cukurova University, Adana, 01330, Turkey
| |
Collapse
|
34
|
Pinto TS, Feltran GDS, Fernandes CJDC, de Camargo Andrade AF, Coque ADC, Silva SL, Abuderman AA, Zambuzzi WF, Foganholi da Silva RA. Epigenetic changes in shear-stressed endothelial cells. Cell Biol Int 2024; 48:665-681. [PMID: 38420868 DOI: 10.1002/cbin.12138] [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: 03/19/2023] [Revised: 01/18/2024] [Accepted: 01/28/2024] [Indexed: 03/02/2024]
Abstract
Epigenetic changes, particularly histone compaction modifications, have emerged as critical regulators in the epigenetic pathway driving endothelial cell phenotype under constant exposure to laminar forces induced by blood flow. However, the underlying epigenetic mechanisms governing endothelial cell behavior in this context remain poorly understood. To address this knowledge gap, we conducted in vitro experiments using human umbilical vein endothelial cells subjected to various tensional forces simulating pathophysiological blood flow shear stress conditions, ranging from normotensive to hypertensive forces. Our study uncovers a noteworthy observation wherein endothelial cells exposed to high shear stress demonstrate a decrease in the epigenetic marks H3K4ac and H3K27ac, accompanied by significant alterations in the levels of HDAC (histone deacetylase) proteins. Moreover, we demonstrate a negative regulatory effect of increased shear stress on HOXA13 gene expression and a concomitant increase in the expression of the long noncoding RNA, HOTTIP, suggesting a direct association with the suppression of HOXA13. Collectively, these findings represent the first evidence of the role of histone-related epigenetic modifications in modulating chromatin compaction during mechanosignaling of endothelial cells in response to elevated shear stress forces. Additionally, our results highlight the importance of understanding the physiological role of HOXA13 in vascular biology and hypertensive patients, emphasizing the potential for developing small molecules to modulate its activity. These findings warrant further preclinical investigations and open new avenues for therapeutic interventions targeting epigenetic mechanisms in hypertensive conditions.
Collapse
Affiliation(s)
- Thaís Silva Pinto
- Lab. of Bioassays and Cellular Dynamics, Department of Chemical and Biological Sciences, Institute of Biosciences, Paulista State University-UNESP, Botucatu, São Paulo, Brazil
| | - Geórgia da Silva Feltran
- Lab. of Bioassays and Cellular Dynamics, Department of Chemical and Biological Sciences, Institute of Biosciences, Paulista State University-UNESP, Botucatu, São Paulo, Brazil
| | - Célio Júnior da C Fernandes
- Lab. of Bioassays and Cellular Dynamics, Department of Chemical and Biological Sciences, Institute of Biosciences, Paulista State University-UNESP, Botucatu, São Paulo, Brazil
| | - Amanda Fantini de Camargo Andrade
- Lab. of Bioassays and Cellular Dynamics, Department of Chemical and Biological Sciences, Institute of Biosciences, Paulista State University-UNESP, Botucatu, São Paulo, Brazil
| | - Alex de Camargo Coque
- Epigenetic Study Center and Gene Regulation-CEEpiRG, Program in Environmental and Experimental Pathology, Paulista University, São Paulo, São Paulo, Brazil
| | - Simone L Silva
- School of Dentistry, University of Taubaté, Taubaté, São Paulo, Brazil
| | - Abdulwahab A Abuderman
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam bin Abdulaziz University, Riyadh, Saudi Arabia
| | - Willian F Zambuzzi
- Lab. of Bioassays and Cellular Dynamics, Department of Chemical and Biological Sciences, Institute of Biosciences, Paulista State University-UNESP, Botucatu, São Paulo, Brazil
| | - Rodrigo A Foganholi da Silva
- Epigenetic Study Center and Gene Regulation-CEEpiRG, Program in Environmental and Experimental Pathology, Paulista University, São Paulo, São Paulo, Brazil
- School of Dentistry, University of Taubaté, Taubaté, São Paulo, Brazil
| |
Collapse
|
35
|
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 PMCID: PMC11649189 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
|
36
|
Scheidecker B, Poulain S, Sugimoto M, Arakawa H, Kim SH, Kawanishi T, Kato Y, Danoy M, Nishikawa M, Sakai Y. Mechanobiological stimulation in organ-on-a-chip systems reduces hepatic drug metabolic capacity in favor of regenerative specialization. Biotechnol Bioeng 2024; 121:1435-1452. [PMID: 38184801 DOI: 10.1002/bit.28653] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/08/2024]
Abstract
Hepatic physiology depends on the liver's complex structural composition which among others, provides high oxygen supply rates, locally differential oxygen tension, endothelial paracrine signaling, as well as residual hemodynamic shear stress to resident hepatocytes. While functional improvements were shown by implementing these factors into hepatic culture systems, direct cause-effect relationships are often not well characterized-obfuscating their individual contribution in more complex microphysiological systems. By comparing increasingly complex hepatic in vitro culture systems that gradually implement these parameters, we investigate the influence of the cellular microenvironment to overall hepatic functionality in pharmacological applications. Here, hepatocytes were modulated in terms of oxygen tension and supplementation, endothelial coculture, and exposure to fluid shear stress delineated from oxygen influx. Results from transcriptomic and metabolomic evaluation indicate that particularly oxygen supply rates are critical to enhance cellular functionality-with cellular drug metabolism remaining comparable to physiological conditions after prolonged static culture. Endothelial signaling was found to be a major contributor to differential phenotype formation known as metabolic zonation, indicated by WNT pathway activity. Lastly, oxygen-delineated shear stress was identified to direct cellular fate towards increased hepatic plasticity and regenerative phenotypes at the cost of drug metabolic functionality - in line with regenerative effects observed in vivo. With these results, we provide a systematic evaluation of critical parameters and their impact in hepatic systems. Given their adherence to physiological effects in vivo, this highlights the importance of their implementation in biomimetic devices, such as organ-on-a-chip systems. Considering recent advances in basic liver biology, direct translation of physiological structures into in vitro models is a promising strategy to expand the capabilities of pharmacological models.
Collapse
Affiliation(s)
| | - Stéphane Poulain
- Institute of Industrial Science, University of Tokyo, Tokyo, Japan
| | - Masahiro Sugimoto
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
- Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Hiroshi Arakawa
- Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Soo H Kim
- Institute of Industrial Science, University of Tokyo, Tokyo, Japan
| | - Takumi Kawanishi
- Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Yukio Kato
- Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Mathieu Danoy
- Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| | - Masaki Nishikawa
- Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| |
Collapse
|
37
|
Liu S, Han Y, Kong L, Wang G, Ye Z. Atomic force microscopy in disease-related studies: Exploring tissue and cell mechanics. Microsc Res Tech 2024; 87:660-684. [PMID: 38063315 DOI: 10.1002/jemt.24471] [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: 07/30/2023] [Revised: 10/22/2023] [Accepted: 11/26/2023] [Indexed: 03/02/2024]
Abstract
Despite significant progress in human medicine, certain diseases remain challenging to promptly diagnose and treat. Hence, the imperative lies in the development of more exhaustive criteria and tools. Tissue and cellular mechanics exhibit distinctive traits in both normal and pathological states, suggesting that "force" represents a promising and distinctive target for disease diagnosis and treatment. Atomic force microscopy (AFM) holds great promise as a prospective clinical medical device due to its capability to concurrently assess surface morphology and mechanical characteristics of biological specimens within a physiological setting. This review presents a comprehensive examination of the operational principles of AFM and diverse mechanical models, focusing on its applications in investigating tissue and cellular mechanics associated with prevalent diseases. The findings from these studies lay a solid groundwork for potential clinical implementations of AFM. RESEARCH HIGHLIGHTS: By examining the surface morphology and assessing tissue and cellular mechanics of biological specimens in a physiological setting, AFM shows promise as a clinical device to diagnose and treat challenging diseases.
Collapse
Affiliation(s)
- Shuaiyuan Liu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Yibo Han
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Lingwen Kong
- Department of Cardiothoracic Surgery, Central Hospital of Chongqing University, Chongqing Emergency Medical Center, Chongqing, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
- JinFeng Laboratory, Chongqing, China
| | - Zhiyi Ye
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
- JinFeng Laboratory, Chongqing, China
| |
Collapse
|
38
|
Zhang H, Rahman T, Lu S, Adam AP, Wan LQ. Helical vasculogenesis driven by cell chirality. SCIENCE ADVANCES 2024; 10:eadj3582. [PMID: 38381835 PMCID: PMC10881055 DOI: 10.1126/sciadv.adj3582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 01/19/2024] [Indexed: 02/23/2024]
Abstract
The cellular helical structure is well known for its crucial role in development and disease. Nevertheless, the underlying mechanism governing this phenomenon remains largely unexplored, particularly in recapitulating it in well-controlled engineering systems. Leveraging advanced microfluidics, we present compelling evidence of the spontaneous emergence of helical endothelial tubes exhibiting robust right-handedness governed by inherent cell chirality. To strengthen our findings, we identify a consistent bias toward the same chirality in mouse vascular tissues. Manipulating endothelial cell chirality using small-molecule drugs produces a dose-dependent reversal of the handedness in engineered vessels, accompanied by non-monotonic changes in vascular permeability. Moreover, our three-dimensional cell vertex model provides biomechanical insights into the chiral morphogenesis process, highlighting the role of cellular torque and tissue fluidity in its regulation. Our study unravels an intriguing mechanism underlying vascular chiral morphogenesis, shedding light on the broader implications and distinctive perspectives of tubulogenesis within biological systems.
Collapse
Affiliation(s)
- Haokang Zhang
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Tasnif Rahman
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Shuhan Lu
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Alejandro Pablo Adam
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
- Department of Ophthalmology, Albany Medical College, Albany, NY 12208, USA
| | - Leo Q. Wan
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
- Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Modeling, Simulation and Imaging in Medicine, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| |
Collapse
|
39
|
Zhang Y, O'Mahony A, He Y, Barber T. Hydrodynamic shear stress' impact on mammalian cell properties and its applications in 3D bioprinting. Biofabrication 2024; 16:022003. [PMID: 38277669 DOI: 10.1088/1758-5090/ad22ee] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 01/26/2024] [Indexed: 01/28/2024]
Abstract
As an effective cell assembly method, three-dimensional bioprinting has been widely used in building organ models and tissue repair over the past decade. However, different shear stresses induced throughout the entire printing process can cause complex impacts on cell integrity, including reducing cell viability, provoking morphological changes and altering cellular functionalities. The potential effects that may occur and the conditions under which these effects manifest are not clearly understood. Here, we review systematically how different mammalian cells respond under shear stress. We enumerate available experimental apparatus, and we categorise properties that can be affected under disparate stress patterns. We also summarise cell damaging mathematical models as a predicting reference for the design of bioprinting systems. We concluded that it is essential to quantify specific cell resistance to shear stress for the optimisation of bioprinting systems. Besides, as substantial positive impacts, including inducing cell alignment and promoting cell motility, can be generated by shear stress, we suggest that we find the proper range of shear stress and actively utilise its positive influences in the development of future systems.
Collapse
Affiliation(s)
- Yani Zhang
- School of Mechanical Engineering, UNSW, Sydney, NSW 2052, Australia
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, People's Republic of China
| | - Aidan O'Mahony
- Inventia Life Science Pty Ltd, Alexandria, Sydney, NSW 2015, Australia
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, People's Republic of China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Tracie Barber
- School of Mechanical Engineering, UNSW, Sydney, NSW 2052, Australia
| |
Collapse
|
40
|
Chandurkar MK, Mittal N, Royer-Weeden SP, Lehmann SD, Rho Y, Han SJ. Low Shear in Short-Term Impacts Endothelial Cell Traction and Alignment in Long-Term. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.20.558732. [PMID: 37790318 PMCID: PMC10542130 DOI: 10.1101/2023.09.20.558732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Within the vascular system, endothelial cells (ECs) are exposed to fluid shear stress (FSS), a mechanical force exerted by blood flow that is critical for regulating cellular tension and maintaining vascular homeostasis. The way ECs react to FSS varies significantly; while high, laminar FSS supports vasodilation and suppresses inflammation, low or disturbed FSS can lead to endothelial dysfunction and increase the risk of cardiovascular diseases. Yet, the adaptation of ECs to dynamically varying FSS remains poorly understood. This study focuses on the dynamic responses of ECs to brief periods of low FSS, examining its impact on endothelial traction-a measure of cellular tension that plays a crucial role in how endothelial cells respond to mechanical stimuli. By integrating traction force microscopy (TFM) with a custom-built flow chamber, we analyzed how human umbilical vein endothelial cells (HUVECs) adjust their traction in response to shifts from low to high shear stress. We discovered that initial exposure to low FSS prompts a marked increase in traction force, which continues to rise over 10 hours before slowly decreasing. In contrast, immediate exposure to high FSS causes a quick spike in traction followed by a swift reduction, revealing distinct patterns of traction behavior under different shear conditions. Importantly, the direction of traction forces and the resulting cellular alignment under these conditions indicate that the initial shear experience dictates long-term endothelial behavior. Our findings shed light on the critical influence of short-lived low-shear stress experiences in shaping endothelial function, indicating that early exposure to low FSS results in enduring changes in endothelial contractility and alignment, with significant consequences for vascular health and the development of cardiovascular diseases.
Collapse
Affiliation(s)
- Mohanish K. Chandurkar
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
- Health Research Institute, Michigan Technological University, Houghton, MI 49931
| | - Nikhil Mittal
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
- Health Research Institute, Michigan Technological University, Houghton, MI 49931
| | - Shaina P. Royer-Weeden
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
- Health Research Institute, Michigan Technological University, Houghton, MI 49931
| | - Steven D. Lehmann
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
| | - Yeonwoo Rho
- Department of Mathematical Sciences, Michigan Technological University, Houghton, MI 49931
| | - Sangyoon J. Han
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
- Health Research Institute, Michigan Technological University, Houghton, MI 49931
- Department of Mechanical Engineering and Engineering Mechanics, Michigan Technological University, Houghton, MI 49931
| |
Collapse
|
41
|
Fish A, Kulkarni A. Flow-Induced Shear Stress Primes NLRP3 Inflammasome Activation in Macrophages via Piezo1. ACS APPLIED MATERIALS & INTERFACES 2024; 16:4505-4518. [PMID: 38240257 DOI: 10.1021/acsami.3c18645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
The NLRP3 inflammasome is a crucial component of the innate immune system, playing a pivotal role in initiating and regulating the body's inflammatory response to various pathogens and cellular damage. Environmental stimuli, such as temperature, pH level, and nutrient availability, can influence the behavior and functions of innate immune cells, including immune cell activity, proliferation, and cytokine production. However, there is limited understanding regarding how mechanical forces, like shear stress, govern the intrinsic inflammatory reaction, particularly the activation of the NLRP3 inflammasome, and how shear stress impacts NLRP3 inflammasome activation through its capacity to induce alterations in gene expression and cytokine secretion. Here, we investigated how shear stress can act as a priming signal in NLRP3 inflammasome activation by exposing immortalized bone marrow-derived macrophages (iBMDMs) to numerous physiologically relevant magnitudes of shear stress before chemically inducing inflammasome activation. We demonstrated that shear stress of large magnitudes was able to prime iBMDMs more effectively for inflammasome activation compared to lower shear stress magnitudes, as quantified by the percentage of cells where ASC-CFP specks formed and IL-1β secretion, the hallmarks of inflammasome activation. Testing this in NLRP3 and caspase-1 knockout iBMDMs showed that the NLRP3 inflammasome was primarily primed for activation due to shear stress exposure. Quantitative polymerase chain reaction (qPCR) and a small-molecule inhibitor study mechanistically determined that shear stress regulates the NLRP3 inflammasome by upregulating Piezo1, IKKβ, and NLRP3. These findings offer insights into the mechanistic relationship among physiological shear stresses, inflammasome activation, and their impact on the progression of inflammatory diseases and their interconnected pathogenesis.
Collapse
Affiliation(s)
- Adam Fish
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Ashish Kulkarni
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
42
|
Benitez-Albiter A, Anderson CP, Jones M, Park SS, Layec G, Park SY. Contributing Factors to Endothelial Dysfunction in Individuals with Spinal Cord Injuries. Pulse (Basel) 2024; 12:49-57. [PMID: 39022560 PMCID: PMC11250044 DOI: 10.1159/000539199] [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: 02/27/2024] [Accepted: 04/27/2024] [Indexed: 07/20/2024] Open
Abstract
Background Patients with spinal cord injuries (SCIs) are at a greater risk for the development of cardiovascular diseases (CVDs) than able-bodied individuals due to the high risk of endothelial dysfunction. Summary For instance, patients with SCIs lose autonomic control of the heart and vasculature, which results in severe fluctuations in blood pressure. These oscillations between hypotension and hypertension have been shown to damage blood vessel endothelial cells and may contribute to the development of atherosclerosis. Furthermore, the loss of skeletal muscle control results in skeletal muscle atrophy and inward remodeling of the conduit arteries. It has been shown that blood vessels in the legs are chronically exposed to high shear, while the aorta experiences chronically low shear. These alterations to shear forces may adversely impact endothelial vasodilatory capacity and promote inflammatory signaling and leukocyte adherence. Additionally, microvascular endothelial vasodilatory capacity is impaired in patients with an SCI, and this may precede changes in conduit artery endothelial function. Finally, due to immobility and a loss of skeletal muscle mass, patients with SCIs have a higher risk of metabolic disorders, inflammation, and oxidative stress. Key Messages Collectively, these factors may impair endothelium-dependent vasodilatory capacity, promote leukocyte adhesion and infiltration, promote the peroxidation of lipids, and ultimately support the development of atherosclerosis. Therefore, future interventions to prevent CVDs in patients with SCIs should focus on the management of endothelial health to prevent endothelial dysfunction and atherosclerosis.
Collapse
Affiliation(s)
| | - Cody P. Anderson
- School of Health and Kinesiology, University of Nebraska at Omaha, Omaha, NE, USA
| | - Matthew Jones
- School of Health and Kinesiology, University of Nebraska at Omaha, Omaha, NE, USA
| | - Sang-Seo Park
- Department of Physiology, Kyung Hee University, Seoul, Republic of Korea
| | - Gwenael Layec
- School of Health and Kinesiology, University of Nebraska at Omaha, Omaha, NE, USA
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Song-Young Park
- School of Health and Kinesiology, University of Nebraska at Omaha, Omaha, NE, USA
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
43
|
Abstract
The deterioration of the brain's microvasculature, particularly in the hippocampus, appears to be a very early event in the development of Alzheimer's disease (AD), preceding even the deposition of amyloid-β. A damaged microvasculature reduces the supply of oxygen and glucose to this region and limits the production of energy, ATP. The damage may be a function of the rise with age in the expression and activity of NADPH oxidase (NOX) in these microvessels. This rise renders these vessels vulnerable to the effects of oxidative stress and inflammation. The rise in NOX activity with age is even more marked in the AD brain where an inverse correlation has been demonstrated between NOX activity and cognitive ability. Apocynin, a putative NOX inhibitor, has been shown to block the damaging effects of NOX activation. Apocynin acts as a strong scavenger of H2O2, and as a weak scavenger of superoxide. Like apocynin, sodium oxybate (SO) has also been shown to block the toxic effects of NOX activation. The application of SO generates NADPH and ATP. SO inhibits oxidative stress and maintains normal cerebral ATP levels under hypoxic conditions. Moreover, it acts epigenetically to attenuate the expression of NOX. SO may delay the onset and slow the progress of AD by suppling energy and maintaining an antioxidative environment in the brain throughout the night. The slow wave activity produced by SO may also activate the glymphatic system and promote the clearance of amyloid-β from the brain.
Collapse
Affiliation(s)
- Mortimer Mamelak
- Department of Psychiatry, Baycrest Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
44
|
Tröndle K, Rizzo L, Pichler R, Zimmermann S, Lienkamp SS. Flow induces common and specific transcriptional changes in renal tubular epithelial cells involving the PI3K pathway. FASEB J 2024; 38:e23329. [PMID: 38050412 DOI: 10.1096/fj.202300834r] [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: 05/12/2023] [Revised: 10/30/2023] [Accepted: 11/10/2023] [Indexed: 12/06/2023]
Abstract
Flow-induced shear stress affects renal epithelial cells in the nephron tubule with potential implications for differential functionalities of the individual segments. Disruptions of cellular mechanosensation or flow conditions are associated with the development and progression of various renal diseases. This study investigates the effects of flow on the transcriptome of various renal tubular epithelial cell types. We analyzed the transcriptome of induced renal epithelial cells (iREC) cultured under physiological flow (0.57 ± 0.05 dyn/cm2 ) or in static conditions for 72 h. RNA sequencing showed 861 differentially expressed genes (DEGs), with 503 up- and 358 downregulated under flow. DEGs were linked to extracellular matrix (ECM) components (e.g. Col1a1, Col4a3, Col4a4, Fn1, Smoc2), junctions (Gja1, Tubb5), channel activities (Abcc4, Aqp1), and transcription factors (Foxq1, Lgr6). Next, we performed a meta-analysis comparing our data with three published datasets that subjected epithelial cell lines from distinct segments to flow, including proximal tubule and collecting duct cells. We found that TGF-ß, p53, MAPK, and PI3K are common flow-regulated pathways. Tfrc expression and thus the capability of iron uptake is commonly upregulated under flow. Many DEGs were related to kidney diseases, such as fibrosis (e.g. Tgfb1-3 and Serpine1). To obtain further mechanistic insights we investigated the role of the PI3K pathway in flow sensing. Applying flow and inhibition of PI3K showed significantly altered expression of transcripts related to ECM remodeling, angiogenesis, and ion transport. This suggests that the PI3K pathway is a critical mediator in flow-dependent cellular processes and gene expression, potentially influencing renal development and tissue remodeling. Finally, we derived a cross-cell-line summary of common as well as segment-specific transcriptomic effects, thus providing insights into the molecular mechanisms underlying flow sensing in the nephron tubule.
Collapse
Affiliation(s)
- Kevin Tröndle
- Faculty of Medicine, Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Ludovica Rizzo
- Faculty of Medicine, Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Roman Pichler
- Department of Medicine IV, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Stefan Zimmermann
- Laboratory for MEMS Applications, Department of Microsystems Engineering, IMTEK, University of Freiburg, Freiburg, Germany
| | - Soeren S Lienkamp
- Faculty of Medicine, Institute of Anatomy, University of Zurich, Zurich, Switzerland
| |
Collapse
|
45
|
Toprak K, Balaban İ, Pala S. The effect of successful lower extremity revascularization on aortic stiffness in patients with peripheral arterial disease. Vascular 2023; 31:1253-1261. [PMID: 36796873 DOI: 10.1177/17085381231153223] [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: 02/18/2023]
Abstract
OBJECTIVE Peripheral arterial disease (PAD) is a global health problem and associated with poor outcomes. It causes increased arterial stiffness. The association of PAD with aortic arterial stiffness was investigated in previous studies. However, there is limited data regarding the effect of peripheral revascularization on arterial stiffness. The aim of our study is to investigate the effect of peripheral revascularization on aortic stiffness parameters in patients with symptomatic PAD. METHODS A total of 48 patients with PAD who underwent peripheral revascularization were included in the study. Echocardiography was performed before and after the procedure, and aortic stiffness parameters were obtained by using aortic diameters and arterial blood pressure measurements. RESULTS Post-procedural aortic strain (5.1 [1.3-14] vs. 6.3 [2.8-6.3], p = 0.009) and aortic distensibility (0.2 [0.0-0.9] vs. 0.3 [0.1-1.1], p = 0.001) measurements were significantly increased compared to pre-procedural values. Patients were also compared according to the lesion laterality, site and treatment methods. It was found that the change in aortic strain (p = 0.031) and distensibility (p = 0.043) were significantly higher in unilateral lesion compared to bilateral lesion. Also, the change in aortic strain (p = 0.042) and distensibility (p = 0.033) were significantly higher in iliac site lesion compared to superficial femoral artery (SFA) site lesion. Moreover, the change in aortic strain was significantly higher (p = 0.013) in patients treated with stent compared to only balloon angioplasty. CONCLUSION Our study showed that successful percutaneous revascularization significantly reduced aortic stiffness in PAD. The change in aortic stiffness was significantly higher in unilateral lesions, iliac site lesions and stent-treated lesions.
Collapse
Affiliation(s)
- Kenan Toprak
- Department of Cardiology, Faculty of Medicine, Harran University, Sanliurfa, Turkey
| | - İsmail Balaban
- Clinic of Cardiology, Kartal Koşuyolu High Specialization Training and Research Hospital, İstanbul, Turkey
| | - Selçuk Pala
- Clinic of Cardiology, Kartal Koşuyolu High Specialization Training and Research Hospital, İstanbul, Turkey
| |
Collapse
|
46
|
Dombroski JA, Rowland SJ, Fabiano AR, Knoblauch SV, Hope JM, King MR. Fluid shear stress enhances dendritic cell activation. Immunobiology 2023; 228:152744. [PMID: 37729773 PMCID: PMC10841200 DOI: 10.1016/j.imbio.2023.152744] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/25/2023] [Accepted: 09/11/2023] [Indexed: 09/22/2023]
Abstract
Ex vivo activation of dendritic cells (DCs) has been widely explored for targeted therapies, although these treatments remain expensive. Reducing treatment costs while enhancing cell activation could help to make immunotherapies more accessible. Cells can be activated by both internal and external forces including fluid shear stress (FSS). FSS activates cells via opening of mechanosensitive ion channels. In this study, dendritic cells were activated by sustained exposure to circulatory levels of fluid shear stress using a cone-and-plate flow device and analyzed for activation markers. After 1 h of shear stress exposure, an increase in cytokine release was present in immortalized cells as well as phosphorylation of the proteins NF-κB and cFos in primary DCs. Changes in DC morphology, metabolism and proliferation were also observed. These compelling new findings point to the potential for using FSS to activate DCs for ex vivo therapeutics.
Collapse
Affiliation(s)
- Jenna A Dombroski
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Ave, Nashville, TN 37212, United States
| | - Schyler J Rowland
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Ave, Nashville, TN 37212, United States
| | - Abigail R Fabiano
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Ave, Nashville, TN 37212, United States
| | - Samantha V Knoblauch
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Ave, Nashville, TN 37212, United States
| | - Jacob M Hope
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Ave, Nashville, TN 37212, United States
| | - Michael R King
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Ave, Nashville, TN 37212, United States.
| |
Collapse
|
47
|
Sharma U, Verma S, Adithan S. Morphology of middle cerebral artery using computed tomography angiographic study in a tertiary care hospital. Anat Cell Biol 2023; 56:360-366. [PMID: 37402565 PMCID: PMC10520858 DOI: 10.5115/acb.22.242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/25/2023] [Accepted: 04/27/2023] [Indexed: 07/06/2023] Open
Abstract
Increased tortuosity of vessel is associated with high incidence of plaque formation leading to atherosclerosis. Surgical procedures are done after analyzing morphology of middle cerebral artery (MCA). However, literature describing MCA morphology using computed tomography angiography (CTA) is limited, so this study was planned to determine its incidence in Indian population. Datasets of CTA from 289 patients (180 males and 109 females), average age: 49.29±16.16 years (range: 11 to 85 years), from a tertiary care hospital were systematically reviewed for morphology of MCA. Cases involving aneurysms and infarcts were excluded. Four shapes of MCA were recognized: straight, U, inverted U, and S-shaped. MCA was straight in 44% (254/578), U-shaped in 37% (215/578), S shaped in 15% (89/578) and inverted U-shaped in 3% (20/578) cases. In males, MCA was straight in 46% (166/360), U-shaped in 37% (134/360), S-shaped in 16% (58/360) and inverted U-shaped in 4% (14/360) cases. In females, MCA was straight in 42% cases (92/218), U-shaped in 37% (81/218), S-shaped in 17% (36/218) and inverted U-shaped in 4% (9/218). On comparing shape with various age groups using chi square test, U shaped (P≤0.001) and S-shaped (P=0.003) MCA were found to be statistically significant. The incidence of straight shape was higher in advanced age group (>60 years). Knowledge of MCA shape will be useful for clinicians and surgeons in successful endovascular recanalization. Also, this data would help surgeons during neurointerventional procedures.
Collapse
Affiliation(s)
- Urvi Sharma
- Department of Anatomy, All India Institute of Medical Sciences, Raipur, India
| | - Suman Verma
- Department of Anatomy, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Subathra Adithan
- Department of Radiodiagnosis, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| |
Collapse
|
48
|
Espina JA, Cordeiro MH, Milivojevic M, Pajić-Lijaković I, Barriga EH. Response of cells and tissues to shear stress. J Cell Sci 2023; 136:jcs260985. [PMID: 37747423 PMCID: PMC10560560 DOI: 10.1242/jcs.260985] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023] Open
Abstract
Shear stress is essential for normal physiology and malignancy. Common physiological processes - such as blood flow, particle flow in the gut, or contact between migratory cell clusters and their substrate - produce shear stress that can have an impact on the behavior of different tissues. In addition, shear stress has roles in processes of biomedical interest, such as wound healing, cancer and fibrosis induced by soft implants. Thus, understanding how cells react and adapt to shear stress is important. In this Review, we discuss in vivo and in vitro data obtained from vascular and epithelial models; highlight the insights these have afforded regarding the general mechanisms through which cells sense, transduce and respond to shear stress at the cellular levels; and outline how the changes cells experience in response to shear stress impact tissue organization. Finally, we discuss the role of shear stress in collective cell migration, which is only starting to be appreciated. We review our current understanding of the effects of shear stress in the context of embryo development, cancer and fibrosis, and invite the scientific community to further investigate the role of shear stress in these scenarios.
Collapse
Affiliation(s)
- Jaime A. Espina
- Mechanisms of Morphogenesis Lab, Gulbenkian Institute of Science (IGC), 2780-156 Oeiras, Portugal
| | - Marilia H. Cordeiro
- Mechanisms of Morphogenesis Lab, Gulbenkian Institute of Science (IGC), 2780-156 Oeiras, Portugal
| | - Milan Milivojevic
- Faculty of Technology and Metallurgy, Belgrade University, 11120 Belgrade, Serbia
| | | | - Elias H. Barriga
- Mechanisms of Morphogenesis Lab, Gulbenkian Institute of Science (IGC), 2780-156 Oeiras, Portugal
| |
Collapse
|
49
|
Moris JM, Cardona A, Hinckley B, Mendez A, Blades A, Paidisetty VK, Chang CJ, Curtis R, Allen K, Koh Y. A framework of transient hypercapnia to achieve an increased cerebral blood flow induced by nasal breathing during aerobic exercise. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2023; 5:100183. [PMID: 37745894 PMCID: PMC10514094 DOI: 10.1016/j.cccb.2023.100183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/30/2023] [Accepted: 09/11/2023] [Indexed: 09/26/2023]
Abstract
During exercise, cerebral blood flow (CBF) is expected to only increase to a maximal volume up to a moderate intensity aerobic effort, suggesting that CBF is expected to decline past 70 % of a maximal aerobic effort. Increasing CBF during exercise permits an increased cerebral metabolic activity that stimulates neuroplasticity and other key processes of cerebral adaptations that ultimately improve cognitive health. Recent work has focused on utilizing gas-induced exposure to intermittent hypoxia during aerobic exercise to maximize the improvements in cognitive function compared to those seen under normoxic conditions. However, it is postulated that exercising by isolating breathing only to the nasal route may provide a similar effect by stimulating a transient hypercapnic condition that is non-gas dependent. Because nasal breathing prevents hyperventilation during exercise, it promotes an increase in the partial arterial pressure of CO2. The rise in systemic CO2 stimulates hypercapnia and permits the upregulation of hypoxia-related genes. In addition, the rise in systemic CO2 stimulates cerebral vasodilation, promoting a greater increase in CBF than seen during normoxic conditions. While more research is warranted, nasal breathing might also promote benefits related to improved sleep, greater immunity, and body fat loss. Altogether, this narrative review presents a theoretical framework by which exercise-induced hypercapnia by utilizing nasal breathing during moderate-intensity aerobic exercise may promote greater health adaptations and cognitive improvements than utilizing oronasal breathing.
Collapse
Affiliation(s)
- Jose M. Moris
- Department of Health, Human Performance, and Recreation, Baylor University, One Bear Place #97313, 1312 S. 5th St., Waco, TX 76798, United States
| | - Arturo Cardona
- Department of Health, Human Performance, and Recreation, Baylor University, One Bear Place #97313, 1312 S. 5th St., Waco, TX 76798, United States
| | - Brendan Hinckley
- Department of Health, Human Performance, and Recreation, Baylor University, One Bear Place #97313, 1312 S. 5th St., Waco, TX 76798, United States
| | - Armando Mendez
- Department of Health, Human Performance, and Recreation, Baylor University, One Bear Place #97313, 1312 S. 5th St., Waco, TX 76798, United States
| | - Alexandra Blades
- Department of Health, Human Performance, and Recreation, Baylor University, One Bear Place #97313, 1312 S. 5th St., Waco, TX 76798, United States
| | - Vineet K. Paidisetty
- Department of Health, Human Performance, and Recreation, Baylor University, One Bear Place #97313, 1312 S. 5th St., Waco, TX 76798, United States
| | - Christian J. Chang
- Department of Health, Human Performance, and Recreation, Baylor University, One Bear Place #97313, 1312 S. 5th St., Waco, TX 76798, United States
| | - Ryan Curtis
- Department of Health, Human Performance, and Recreation, Baylor University, One Bear Place #97313, 1312 S. 5th St., Waco, TX 76798, United States
| | - Kylie Allen
- Department of Health, Human Performance, and Recreation, Baylor University, One Bear Place #97313, 1312 S. 5th St., Waco, TX 76798, United States
| | - Yunsuk Koh
- Department of Health, Human Performance, and Recreation, Baylor University, One Bear Place #97313, 1312 S. 5th St., Waco, TX 76798, United States
| |
Collapse
|
50
|
Rojas-González DM, Babendreyer A, Ludwig A, Mela P. Analysis of flow-induced transcriptional response and cell alignment of different sources of endothelial cells used in vascular tissue engineering. Sci Rep 2023; 13:14384. [PMID: 37658092 PMCID: PMC10474151 DOI: 10.1038/s41598-023-41247-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 08/23/2023] [Indexed: 09/03/2023] Open
Abstract
Endothelialization of tissue-engineered vascular grafts has proven crucial for implant functionality and thus clinical outcome, however, the choice of endothelial cells (ECs) is often driven by availability rather than by the type of vessel to be replaced. In this work we studied the response to flow of different human ECs with the aim of examining whether their response in vitro is dictated by their original in vivo conditions. Arterial, venous, and microvascular ECs were cultured under shear stress (SS) of 0, 0.3, 3, 1, 10, and 30 dyne/cm2 for 24 h. Regulation of flow-induced marker KLF2 was similar across the different ECs. Upregulation of anti-thrombotic markers, TM and TPA, was mainly seen at higher SS. Cell elongation and alignment was observed for the different ECs at 10 and 30 dyne/cm2 while at lower SS cells maintained a random orientation. Downregulation of pro-inflammatory factors SELE, IL8, and VCAM1 and up-regulation of anti-oxidant markers NQO1 and HO1 was present even at SS for which cell alignment was not observed. Our results evidenced similarities in the response to flow among the different ECs, suggesting that the maintenance of the resting state in vitro is not dictated by the SS typical of the tissue of origin and that absence of flow-induced cell orientation does not necessarily correlate with a pro-inflammatory state of the ECs. These results support the use of ECs from easily accessible sources for in vitro vascular tissue engineering independently from the target vessel.
Collapse
Affiliation(s)
- Diana M Rojas-González
- Department of Biohybrid & Medical Textiles (BioTex) at Center of Biohybrid Medical Systems (CBMS), AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstr. 55, 52074, Aachen, Germany
- Chair of Medical Materials and Implants, Department of Mechanical Engineering, School of Engineering and Design and Munich Institute of Biomedical Engineering, Technical University of Munich, Boltzmannstr 15, 85748, Garching, Germany
| | - Aaron Babendreyer
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany.
| | - Andreas Ludwig
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Petra Mela
- Department of Biohybrid & Medical Textiles (BioTex) at Center of Biohybrid Medical Systems (CBMS), AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstr. 55, 52074, Aachen, Germany.
- Chair of Medical Materials and Implants, Department of Mechanical Engineering, School of Engineering and Design and Munich Institute of Biomedical Engineering, Technical University of Munich, Boltzmannstr 15, 85748, Garching, Germany.
| |
Collapse
|