1
|
Munteanu C, Galaction AI, Onose G, Turnea M, Rotariu M. Harnessing Gasotransmitters to Combat Age-Related Oxidative Stress in Smooth Muscle and Endothelial Cells. Pharmaceuticals (Basel) 2025; 18:344. [PMID: 40143122 PMCID: PMC11946800 DOI: 10.3390/ph18030344] [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: 01/25/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Age-related oxidative stress is a critical factor in vascular dysfunction, contributing to hypertension and atherosclerosis. Smooth muscle cells and endothelial cells are particularly susceptible to oxidative damage, which exacerbates vascular aging through cellular senescence, chronic inflammation, and arterial stiffness. Gasotransmitters-hydrogen sulfide (H2S), nitric oxide (NO), and carbon monoxide (CO)-are emerging as promising therapeutic agents for counteracting these processes. This review synthesizes findings from recent studies focusing on the mechanisms by which H2S, NO, and CO influence vascular smooth muscle and endothelial cell function. Therapeutic strategies involving exogenous gasotransmitter delivery systems and combination therapies were analyzed. H2S enhances mitochondrial bioenergetics, scavenges ROS, and activates antioxidant pathways. NO improves endothelial function, promotes vasodilation, and inhibits platelet aggregation. CO exhibits cytoprotective and anti-inflammatory effects by modulating heme oxygenase activity and ROS production. In preclinical studies, gasotransmitter-releasing molecules (e.g., NaHS, SNAP, CORMs) and targeted delivery systems show significant promise. Synergistic effects with lifestyle modifications and antioxidant therapies further enhance their therapeutic potential. In conclusion, gasotransmitters hold significant promise as therapeutic agents to combat age-related oxidative stress in vascular cells. Their multifaceted mechanisms and innovative delivery approaches make them potential candidates for treating vascular dysfunction and promoting healthy vascular aging. Further research is needed to translate these findings into clinical applications.
Collapse
Affiliation(s)
- Constantin Munteanu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
| | - Anca Irina Galaction
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| | - Gelu Onose
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Marius Turnea
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| | - Mariana Rotariu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| |
Collapse
|
2
|
Liu S, Yan J, Gao M, Yang H. Research progress in the regulation of endothelial cells and smooth muscle cells using a micro-nanostructure. Biomed Eng Online 2025; 24:6. [PMID: 39849451 PMCID: PMC11760742 DOI: 10.1186/s12938-025-01337-0] [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: 12/09/2024] [Accepted: 01/09/2025] [Indexed: 01/25/2025] Open
Abstract
Recently, the incidence rate and mortality of various acute or chronic vascular occlusive diseases have increased yearly. As one of the most effective measures to treat them, vascular stents have been widely studied by researchers, and presently, the most commonly used is a drug-eluting stent, which reduces the process of rapid endothelialization because the drug is not selective. Fortunately, with the discovery and exploration of micro-nanostructures that can regulate cells selectively, reducing the incidence of "intravascular restenosis" and achieving rapid endothelialization simultaneously are possible through a special structure that cannot only improve endothelial cells (ECs), but also inhibit smooth muscle cells (SMCs). Therefore, this paper mainly introduces the preparation methods of micro-nanostructures used in the past, as well as the detection methods of EC and SMC. Then, the various functions of different dimensional structures for different cells are summarized and analyzed. Finally, the application of micro-nanostructure in future stent materials is summarized and proposed.
Collapse
Affiliation(s)
- Songhao Liu
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810008, China
- School of Energy and Electrical Engineering, Qinghai University, Xining, 810016, Qinghai, China
| | - Juan Yan
- Department of Pharmacy, Medical College of Qinghai University, Xining, 810016, China
| | - Mengyu Gao
- School of Energy and Electrical Engineering, Qinghai University, Xining, 810016, Qinghai, China
| | - Hongxia Yang
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810008, China.
| |
Collapse
|
3
|
Sreedasyam R, Wilson BG, Ferrandez PR, Botvinick EL, Venugopalan V. An optical system for cellular mechanostimulation in 3D hydrogels. Acta Biomater 2024; 189:439-448. [PMID: 39368720 DOI: 10.1016/j.actbio.2024.09.050] [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: 01/04/2023] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 10/07/2024]
Abstract
We introduce a method utilizing single laser-generated cavitation bubbles to stimulate cellular mechanotransduction in dermal fibroblasts embedded within 3D hydrogels. We demonstrate that fibroblasts embedded in either amorphous or fibrillar hydrogels engage in Ca2+ signaling following exposure to an impulsive mechanical stimulus provided by a single 250 µm diameter laser-generated cavitation bubble. We find that the spatial extent of the cellular signaling is larger for cells embedded within a fibrous collagen hydrogel as compared to those embedded within an amorphous polyvinyl alcohol polymer (SLO-PVA) hydrogel. Additionally, for fibroblasts embedded in collagen, we find an increased range of cellular mechanosensitivity for cells that are polarized relative to the radial axis as compared to the circumferential axis. By contrast, fibroblasts embedded within SLO-PVA did not display orientation-dependent mechanosensitivity. Fibroblasts embedded in hydrogels and cultured in calcium-free media did not show cavitation-induced mechanotransduction; implicating calcium signaling based on transmembrane Ca2+ transport. This study demonstrates the utility of single laser-generated cavitation bubbles to provide local non-invasive impulsive mechanical stimuli within 3D hydrogel tissue models with concurrent imaging using optical microscopy. STATEMENT OF SIGNIFICANCE: Currently, there are limited methods for the non-invasive real-time assessment of cellular sensitivity to mechanical stimuli within 3D tissue scaffolds. We describe an original approach that utilizes a pulsed laser microbeam within a standard laser scanning microscope system to generate single cavitation bubbles to provide impulsive mechanostimulation to cells within 3D fibrillar and amorphous hydrogels. Using this technique, we measure the cellular mechanosensitivity of primary human dermal fibroblasts embedded in amorphous and fibrillar hydrogels, thereby providing a useful method to examine cellular mechanotransduction in 3D biomaterials. Moreover, the implementation of our method within a standard optical microscope makes it suitable for broad adoption by cellular mechanotransduction researchers and opens the possibility of high-throughput evaluation of biomaterials with respect to cellular mechanosignaling.
Collapse
Affiliation(s)
- Rahul Sreedasyam
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697-2715, United States
| | - Bryce G Wilson
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA 92697-2580, United States
| | - Patricia R Ferrandez
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697-2715, United States
| | - Elliot L Botvinick
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697-2715, United States; Beckman Laser Institute, University of California Irvine, Irvine, CA 92697-3010, United States.
| | - Vasan Venugopalan
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697-2715, United States; Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA 92697-2580, United States; Beckman Laser Institute, University of California Irvine, Irvine, CA 92697-3010, United States.
| |
Collapse
|
4
|
Liu H, Ren J, Mao L, Xiong C, Zhang X, Wang S, Huang WH, Chen MM. Flexible and Stretchable Photoelectrochemical Sensing toward True-to-Life Monitoring of Hydrogen Peroxide Regulation in Endothelial Mechanotransduction. Anal Chem 2024; 96:16825-16833. [PMID: 39382083 DOI: 10.1021/acs.analchem.4c03550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Hydrogen peroxide (H2O2) levels play a vital role in redox regulation and maintaining the physiological balance of living cells, especially in cell mechanotransduction. Despite the achievements on strain-induced cellular H2O2 monitoring, the applied voltage for H2O2 electrooxidation possibly gave rise to an abnormal expression and inadequate accuracy, which was still an inescapable concern. Hence, we decorated an interlaced CuO@TiO2 nanowires (NWs) semiconductor meshwork onto a polydimethylsiloxane film-supported gold nanotubes substrate (Au NTs/PDMS) to construct a flexible photoelectrochemical (PEC) sensing platform. Under white light irradiation, CuO@TiO2 NWs synergistically exhibited great stretchability and the PEC platform enabled stable photocurrent responses from the reduction of H2O2 even during mechanical deformation. Moreover, the admirable biocompatibility and an almost negligible open circuit voltage of +0.18 V for the CuO@TiO2 NWs/Au NTs/PDMS sensor guaranteed human umbilical vein endothelial cells (HUVECs) adhesion tightly thereon even under continuous illumination for 30 min. Finally, the as-proposed stretchable PEC sensor achieved sensitive and true-to-life monitoring of transient H2O2 release during HUVECs deformation, in which H2O2 release was positively correlated to mechanical strains. This investigation opens a new shade path on in situ cellular sensing and meanwhile greatly expands the application mode of the PEC approach.
Collapse
Affiliation(s)
- Hao Liu
- Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Jiang Ren
- Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Lebao Mao
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Chengyi Xiong
- Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Xiuhua Zhang
- Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Shengfu Wang
- Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Wei-Hua Huang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Miao-Miao Chen
- Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| |
Collapse
|
5
|
Lim J, Fang HW, Bupphathong S, Sung PC, Yeh CE, Huang W, Lin CH. The Edifice of Vasculature-On-Chips: A Focused Review on the Key Elements and Assembly of Angiogenesis Models. ACS Biomater Sci Eng 2024; 10:3548-3567. [PMID: 38712543 PMCID: PMC11167599 DOI: 10.1021/acsbiomaterials.3c01978] [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: 12/29/2023] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/08/2024]
Abstract
The conception of vascularized organ-on-a-chip models provides researchers with the ability to supply controlled biological and physical cues that simulate the in vivo dynamic microphysiological environment of native blood vessels. The intention of this niche research area is to improve our understanding of the role of the vasculature in health or disease progression in vitro by allowing researchers to monitor angiogenic responses and cell-cell or cell-matrix interactions in real time. This review offers a comprehensive overview of the essential elements, including cells, biomaterials, microenvironmental factors, microfluidic chip design, and standard validation procedures that currently govern angiogenesis-on-a-chip assemblies. In addition, we emphasize the importance of incorporating a microvasculature component into organ-on-chip devices in critical biomedical research areas, such as tissue engineering, drug discovery, and disease modeling. Ultimately, advances in this area of research could provide innovative solutions and a personalized approach to ongoing medical challenges.
Collapse
Affiliation(s)
- Joshua Lim
- Graduate
Institute of Nanomedicine and Medical Engineering, College of Biomedical
Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsu-Wei Fang
- High-value
Biomaterials Research and Commercialization Center, National Taipei University of Technology, Taipei 10608, Taiwan
- Department
of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei 10608, Taiwan
- Institute
of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Sasinan Bupphathong
- Graduate
Institute of Nanomedicine and Medical Engineering, College of Biomedical
Engineering, Taipei Medical University, Taipei 11031, Taiwan
- High-value
Biomaterials Research and Commercialization Center, National Taipei University of Technology, Taipei 10608, Taiwan
| | - Po-Chan Sung
- School
of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Chen-En Yeh
- School
of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Wei Huang
- Department
of Orthodontics, Rutgers School of Dental
Medicine, Newark, New Jersey 07103, United States
| | - Chih-Hsin Lin
- Graduate
Institute of Nanomedicine and Medical Engineering, College of Biomedical
Engineering, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
6
|
Sharma S, Kishen A. Bioarchitectural Design of Bioactive Biopolymers: Structure-Function Paradigm for Diabetic Wound Healing. Biomimetics (Basel) 2024; 9:275. [PMID: 38786486 PMCID: PMC11117869 DOI: 10.3390/biomimetics9050275] [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: 04/04/2024] [Revised: 04/26/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
Chronic wounds such as diabetic ulcers are a major complication in diabetes caused by hyperglycemia, prolonged inflammation, high oxidative stress, and bacterial bioburden. Bioactive biopolymers have been found to have a biological response in wound tissue microenvironments and are used for developing advanced tissue engineering strategies to enhance wound healing. These biopolymers possess innate bioactivity and are biodegradable, with favourable mechanical properties. However, their bioactivity is highly dependent on their structural properties, which need to be carefully considered while developing wound healing strategies. Biopolymers such as alginate, chitosan, hyaluronic acid, and collagen have previously been used in wound healing solutions but the modulation of structural/physico-chemical properties for differential bioactivity have not been the prime focus. Factors such as molecular weight, degree of polymerization, amino acid sequences, and hierarchical structures can have a spectrum of immunomodulatory, anti-bacterial, and anti-oxidant properties that could determine the fate of the wound. The current narrative review addresses the structure-function relationship in bioactive biopolymers for promoting healing in chronic wounds with emphasis on diabetic ulcers. This review highlights the need for characterization of the biopolymers under research while designing biomaterials to maximize the inherent bioactive potency for better tissue regeneration outcomes, especially in the context of diabetic ulcers.
Collapse
Affiliation(s)
- Shivam Sharma
- The Kishen Lab, Dental Research Institute, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Faculty of Dentistry, University of Toronto, 124 Edward Street, Toronto, ON M5G 1G6, Canada
| | - Anil Kishen
- The Kishen Lab, Dental Research Institute, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Faculty of Dentistry, University of Toronto, 124 Edward Street, Toronto, ON M5G 1G6, Canada
- Department of Dentistry, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| |
Collapse
|
7
|
Laowpanitchakorn P, Zeng J, Piantino M, Uchida K, Katsuyama M, Matsusaki M. Biofabrication of engineered blood vessels for biomedical applications. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2024; 25:2330339. [PMID: 38633881 PMCID: PMC11022926 DOI: 10.1080/14686996.2024.2330339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/10/2024] [Indexed: 04/19/2024]
Abstract
To successfully engineer large-sized tissues, establishing vascular structures is essential for providing oxygen, nutrients, growth factors and cells to prevent necrosis at the core of the tissue. The diameter scale of the biofabricated vasculatures should range from 100 to 1,000 µm to support the mm-size tissue while being controllably aligned and spaced within the diffusion limit of oxygen. In this review, insights regarding biofabrication considerations and techniques for engineered blood vessels will be presented. Initially, polymers of natural and synthetic origins can be selected, modified, and combined with each other to support maturation of vascular tissue while also being biocompatible. After they are shaped into scaffold structures by different fabrication techniques, surface properties such as physical topography, stiffness, and surface chemistry play a major role in the endothelialization process after transplantation. Furthermore, biological cues such as growth factors (GFs) and endothelial cells (ECs) can be incorporated into the fabricated structures. As variously reported, fabrication techniques, especially 3D printing by extrusion and 3D printing by photopolymerization, allow the construction of vessels at a high resolution with diameters in the desired range. Strategies to fabricate of stable tubular structures with defined channels will also be discussed. This paper provides an overview of the many advances in blood vessel engineering and combinations of different fabrication techniques up to the present time.
Collapse
Affiliation(s)
| | - Jinfeng Zeng
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| | - Marie Piantino
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
- The Consortium for Future Innovation by Cultured Meat, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| | - Kentaro Uchida
- Materials Solution Department, Product Analysis Center, Panasonic Holdings Corporation, Kadoma, Osaka, Japan
| | - Misa Katsuyama
- Materials Solution Department, Product Analysis Center, Panasonic Holdings Corporation, Kadoma, Osaka, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
- The Consortium for Future Innovation by Cultured Meat, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Chen TY, Cheng KC, Yang PS, Shrestha LK, Ariga K, Hsu SH. Interaction of vascular endothelial cells with hydrophilic fullerene nanoarchitectured structures in 2D and 3D environments. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2024; 25:2315014. [PMID: 38419801 PMCID: PMC10901190 DOI: 10.1080/14686996.2024.2315014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/01/2024] [Indexed: 03/02/2024]
Abstract
The interaction between diverse nanoarchitectured fullerenes and cells is crucial for biomedical applications. Here, we detailed the preparation of hydrophilic self-assembled fullerenes by the liquid-liquid interfacial precipitation (LLIP) method and hydrophilic coating of the materials as a possible vascularization strategy. The interactions of vascular endothelial cells (ECs) with hydrophilic fullerene nanotubes (FNT-P) and hydrophilic fullerene nanowhiskers (FNW-P) were investigated. The average length and diameter of FNT-P were 16 ± 2 μm and 3.4 ± 0.4 μm (i.e. aspect ratios of 4.6), respectively. The average length and diameter of FNW-P were 65 ± 8 μm and 1.2 ± 0.2 μm (i.e. aspect ratios of 53.9), respectively. For two-dimensional (2D) culture after 7 days, the ECs remained viable and proliferated up to ~ 420% and ~ 400% with FNT-P and FNW-P of 50 μg/mL, respectively. Furthermore, an optimized chitosan-based self-healing hydrogel with a modulus of ~400 Pa was developed and used to incorporate self-assembled fullerenes as in vitro three-dimensional (3D) platforms to investigate the impact of FNT-P and FNW-P on ECs within a 3D environment. The addition of FNW-P or FNT-P (50 μg/mL) in the hydrogel system led to proliferation rates of ECs up to ~323% and ~280%, respectively, after 7 days of culture. The ECs in FNW-P hydrogel displayed an elongated shape with aligned morphology, while those in FNT-P hydrogel exhibited a rounded and clustered distribution. Vascular-related gene expressions of ECs were significantly upregulated through interactions with these fullerenes. Thus, the combined use of different nanoarchitectured self-assembled fullerenes and self-healing hydrogels may offer environmental cues influencing EC development in a 3D biomimetic microenvironment, holding promise for advancing vascularization strategy in tissue engineering.
Collapse
Affiliation(s)
- Tsai-Yu Chen
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan, R.O.C
| | - Kun-Chih Cheng
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan, R.O.C
| | - Pei-Syuan Yang
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan, R.O.C
| | - Lok Kumar Shrestha
- Supermolecules Group, Research Center for Materials Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), Tsukuba, Japan
- Department of Materials Science, Faculty of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Japan
| | - Katsuhiko Ariga
- Supermolecules Group, Research Center for Materials Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), Tsukuba, Japan
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Shan-hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan, R.O.C
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan, R.O.C
| |
Collapse
|
11
|
Lakin R, Polidovitch N, Yang S, Parikh M, Liu X, Debi R, Gao X, Chen W, Guzman C, Yakobov S, Izaddoustdar F, Wauchop M, Lei Q, Xu W, Nedospasov SA, Christoffels VM, Backx PH. Cardiomyocyte and endothelial cells play distinct roles in the tumour necrosis factor (TNF)-dependent atrial responses and increased atrial fibrillation vulnerability induced by endurance exercise training in mice. Cardiovasc Res 2023; 119:2607-2622. [PMID: 37713664 PMCID: PMC10730243 DOI: 10.1093/cvr/cvad144] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 06/22/2023] [Accepted: 07/18/2023] [Indexed: 09/17/2023] Open
Abstract
AIMS Endurance exercise is associated with an increased risk of atrial fibrillation (AF). We previously established that adverse atrial remodelling and AF susceptibility induced by intense exercise in mice require the mechanosensitive and pro-inflammatory cytokine tumour necrosis factor (TNF). The cellular and mechanistic basis for these TNF-mediated effects is unknown. METHODS AND RESULTS We studied the impact of Tnf excision, in either atrial cardiomyocytes or endothelial cells (using Cre-recombinase expression controlled by Nppa or Tie2 promoters, respectively), on the cardiac responses to six weeks of intense swim exercise training. TNF ablation, in either cell type, had no impact on the changes in heart rate, autonomic tone, or left ventricular structure and function induced by exercise training. Tnf excision in atrial cardiomyocytes did, however, prevent atrial hypertrophy, fibrosis, and macrophage infiltration as well as conduction slowing and increased AF susceptibility arising from exercise training. In contrast, endothelial-specific excision only reduced the training-induced atrial hypertrophy. Consistent with these cell-specific effects of Tnf excision, inducing TNF loss from atrial cardiomyocytes prevented activation of p38MAPKinase, a strain-dependent downstream mediator of TNF signalling, without affecting the atrial stretch as assessed by atrial pressures induced by exercise. Despite TNF's established role in innate immune responses and inflammation, neither acute nor chronic exercise training caused measurable NLRP3 inflammasome activation. CONCLUSIONS Our findings demonstrate that adverse atrial remodelling and AF vulnerability induced by intense exercise require TNF in atrial cardiomyocytes whereas the impact of endothelial-derived TNF is limited to hypertrophy modulation. The implications of the cell autonomous effects of TNF and crosstalk between cells in the atria are discussed.
Collapse
Affiliation(s)
- Robert Lakin
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Nazari Polidovitch
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Sibao Yang
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130022, China
| | - Mihir Parikh
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Xueyan Liu
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130022, China
| | - Ryan Debi
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Xiaodong Gao
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Wenliang Chen
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Camilo Guzman
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Simona Yakobov
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Farzad Izaddoustdar
- Department of Physiology, University of Toronto, Toronto, ON M5S 3E2, Canada
| | - Marianne Wauchop
- Department of Physiology, University of Toronto, Toronto, ON M5S 3E2, Canada
| | - Qian Lei
- Department of Anesthesiology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Weimin Xu
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130022, China
| | - Sergei A Nedospasov
- Laboratory of Molecular Mechanisms of Immunity, Engelhardt Institute of Molecular Biology, Moscow 119991, Russia
- Division of Immunobiology and Biomedicine, Sirius University of Science and Technology, Sirius 354349, Russia
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, The Netherlands
| | - Peter H Backx
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| |
Collapse
|
12
|
Katoh K. Effects of Mechanical Stress on Endothelial Cells In Situ and In Vitro. Int J Mol Sci 2023; 24:16518. [PMID: 38003708 PMCID: PMC10671803 DOI: 10.3390/ijms242216518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
Endothelial cells lining blood vessels are essential for maintaining vascular homeostasis and mediate several pathological and physiological processes. Mechanical stresses generated by blood flow and other biomechanical factors significantly affect endothelial cell activity. Here, we review how mechanical stresses, both in situ and in vitro, affect endothelial cells. We review the basic principles underlying the cellular response to mechanical stresses. We also consider the implications of these findings for understanding the mechanisms of mechanotransducer and mechano-signal transduction systems by cytoskeletal components.
Collapse
Affiliation(s)
- Kazuo Katoh
- Laboratory of Human Anatomy and Cell Biology, Faculty of Health Sciences, Tsukuba University of Technology, Tsukuba 305-8521, Japan
| |
Collapse
|
13
|
Hamrangsekachaee M, Wen K, Yazdani N, Willits RK, Bencherif SA, Ebong EE. Endothelial glycocalyx sensitivity to chemical and mechanical sub-endothelial substrate properties. Front Bioeng Biotechnol 2023; 11:1250348. [PMID: 38026846 PMCID: PMC10643223 DOI: 10.3389/fbioe.2023.1250348] [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: 06/30/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
Glycocalyx (GCX) is a carbohydrate-rich structure that coats the surface of endothelial cells (ECs) and lines the blood vessel lumen. Mechanical perturbations in the vascular environment, such as blood vessel stiffness, can be transduced and sent to ECs through mechanosensors such as GCX. Adverse stiffness alters GCX-mediated mechanotransduction and leads to EC dysfunction and eventually atherosclerotic cardiovascular diseases. To understand GCX-regulated mechanotransduction events, an in vitro model emulating in vivo vessel conditions is needed. To this end, we investigated the impact of matrix chemical and mechanical properties on GCX expression via fabricating a tunable non-swelling matrix based on the collagen-derived polypeptide, gelatin. To study the effect of matrix composition, we conducted a comparative analysis of GCX expression using different concentrations (60-25,000 μg/mL) of gelatin and gelatin methacrylate (GelMA) in comparison to fibronectin (60 μg/mL), a standard coating material for GCX-related studies. Using immunocytochemistry analysis, we showed for the first time that different substrate compositions and concentrations altered the overall GCX expression on human umbilical vein ECs (HUVECs). Subsequently, GelMA hydrogels were fabricated with stiffnesses of 2.5 and 5 kPa, representing healthy vessel tissues, and 10 kPa, corresponding to diseased vessel tissues. Immunocytochemistry analysis showed that on hydrogels with different levels of stiffness, the GCX expression in HUVECs remained unchanged, while its major polysaccharide components exhibited dysregulation in distinct patterns. For example, there was a significant decrease in heparan sulfate expression on pathological substrates (10 kPa), while sialic acid expression increased with increased matrix stiffness. This study suggests the specific mechanisms through which GCX may influence ECs in modulating barrier function, immune cell adhesion, and mechanotransduction function under distinct chemical and mechanical conditions of both healthy and diseased substrates.
Collapse
Affiliation(s)
| | - Ke Wen
- Chemical Engineering Department, Northeastern University, Boston, MA, United States
| | - Narges Yazdani
- Bioengineering Department, Northeastern University, Boston, MA, United States
| | - Rebecca K. Willits
- Chemical Engineering Department, Northeastern University, Boston, MA, United States
- Bioengineering Department, Northeastern University, Boston, MA, United States
| | - Sidi A. Bencherif
- Chemical Engineering Department, Northeastern University, Boston, MA, United States
- Bioengineering Department, Northeastern University, Boston, MA, United States
- Laboratoire de BioMécanique et BioIngénierie (BMBI), UMR CNRS, Sorbonne Universités, Université de Technologie of Compiègne (UTC), Compiègne, France
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States
| | - Eno E. Ebong
- Chemical Engineering Department, Northeastern University, Boston, MA, United States
- Bioengineering Department, Northeastern University, Boston, MA, United States
- Neuroscience Department, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
14
|
Mirza I, Haloul M, Hassan C, Masrur M, Mostafa A, Bianco FM, Ali MM, Minshall RD, Mahmoud AM. Adiposomes from Obese-Diabetic Individuals Promote Endothelial Dysfunction and Loss of Surface Caveolae. Cells 2023; 12:2453. [PMID: 37887297 PMCID: PMC10605845 DOI: 10.3390/cells12202453] [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: 09/22/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
Glycosphingolipids (GSLs) are products of lipid glycosylation that have been implicated in the development of cardiovascular diseases. In diabetes, the adipocyte microenvironment is characterized by hyperglycemia and inflammation, resulting in high levels of GSLs. Therefore, we sought to assess the GSL content in extracellular vesicles derived from the adipose tissues (adiposomes) of obese-diabetic (OB-T2D) subjects and their impact on endothelial cell function. To this end, endothelial cells were exposed to adiposomes isolated from OB-T2D versus healthy subjects. Cells were assessed for caveolar integrity and related signaling, such as Src-kinase and caveolin-1 (cav-1) phosphorylation, and functional pathways, such as endothelial nitric oxide synthase (eNOS) activity. Compared with adiposomes from healthy subjects, OB-T2D adiposomes had higher levels of GSLs, especially LacCer and GM3; they promoted cav-1 phosphorylation coupled to an obvious loss of endothelial surface caveolae and induced eNOS-uncoupling, peroxynitrite generation, and cav-1 nitrosylation. These effects were abolished by Src kinase inhibition and were not observed in GSL-depleted adiposomes. At the functional levels, OB-T2D adiposomes reduced nitric oxide production, shear response, and albumin intake in endothelial cells and impaired flow-induced dilation in healthy arterioles. In conclusion, OB-T2D adiposomes carried a detrimental GSL cargo that disturbed endothelial caveolae and the associated signaling.
Collapse
Affiliation(s)
- Imaduddin Mirza
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (I.M.); (M.H.)
| | - Mohamed Haloul
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (I.M.); (M.H.)
| | - Chandra Hassan
- Department of Surgery, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (C.H.); (M.M.); (F.M.B.); (R.D.M.)
| | - Mario Masrur
- Department of Surgery, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (C.H.); (M.M.); (F.M.B.); (R.D.M.)
| | - Amro Mostafa
- Departments of Anesthesiology and Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA;
| | - Francesco M. Bianco
- Department of Surgery, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (C.H.); (M.M.); (F.M.B.); (R.D.M.)
| | - Mohamed M. Ali
- School of Business and Non-Profit Management, North Park University, Chicago, IL 60625, USA;
| | - Richard D. Minshall
- Department of Surgery, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (C.H.); (M.M.); (F.M.B.); (R.D.M.)
| | - Abeer M. Mahmoud
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (I.M.); (M.H.)
- Department of Kinesiology and Nutrition, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
15
|
Crawshaw JR, Flegg JA, Bernabeu MO, Osborne JM. Mathematical models of developmental vascular remodelling: A review. PLoS Comput Biol 2023; 19:e1011130. [PMID: 37535698 PMCID: PMC10399886 DOI: 10.1371/journal.pcbi.1011130] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023] Open
Abstract
Over the past 40 years, there has been a strong focus on the development of mathematical models of angiogenesis, while developmental remodelling has received little such attention from the mathematical community. Sprouting angiogenesis can be seen as a very crude way of laying out a primitive vessel network (the raw material), while remodelling (understood as pruning of redundant vessels, diameter control, and the establishment of vessel identity and hierarchy) is the key to turning that primitive network into a functional network. This multiscale problem is of prime importance in the development of a functional vasculature. In addition, defective remodelling (either during developmental remodelling or due to a reactivation of the remodelling programme caused by an injury) is associated with a significant number of diseases. In this review, we discuss existing mathematical models of developmental remodelling and explore the important contributions that these models have made to the field of vascular development. These mathematical models are effectively used to investigate and predict vascular development and are able to reproduce experimentally observable results. Moreover, these models provide a useful means of hypothesis generation and can explain the underlying mechanisms driving the observed structural and functional network development. However, developmental vascular remodelling is still a relatively new area in mathematical biology, and many biological questions remain unanswered. In this review, we present the existing modelling paradigms and define the key challenges for the field.
Collapse
Affiliation(s)
- Jessica R. Crawshaw
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
- School of Mathematics and Statistics, The University of Melbourne, Melbourne, Australia
| | - Jennifer A. Flegg
- School of Mathematics and Statistics, The University of Melbourne, Melbourne, Australia
| | - Miguel O. Bernabeu
- Centre for Medical Informatics, The Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
- The Bayes Centre, The University of Edinburgh, Edinburgh, United Kingdom
| | - James M. Osborne
- School of Mathematics and Statistics, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
16
|
Yadav S, Singha P, Nguyen NK, Ooi CH, Kashaninejad N, Nguyen NT. Uniaxial Cyclic Cell Stretching Device for Accelerating Cellular Studies. MICROMACHINES 2023; 14:1537. [PMID: 37630073 PMCID: PMC10456305 DOI: 10.3390/mi14081537] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/24/2023] [Accepted: 07/29/2023] [Indexed: 08/27/2023]
Abstract
Cellular response to mechanical stimuli is a crucial factor for maintaining cell homeostasis. The interaction between the extracellular matrix and mechanical stress plays a significant role in organizing the cytoskeleton and aligning cells. Tools that apply mechanical forces to cells and tissues, as well as those capable of measuring the mechanical properties of biological cells, have greatly contributed to our understanding of fundamental mechanobiology. These tools have been extensively employed to unveil the substantial influence of mechanical cues on the development and progression of various diseases. In this report, we present an economical and high-performance uniaxial cell stretching device. This paper reports the detailed operation concept of the device, experimental design, and characterization. The device was tested with MDA-MB-231 breast cancer cells. The experimental results agree well with previously documented morphological changes resulting from stretching forces on cancer cells. Remarkably, our new device demonstrates comparable cellular changes within 30 min compared with the previous 2 h stretching duration. This third-generation device significantly improved the stretching capabilities compared with its previous counterparts, resulting in a remarkable reduction in stretching time and a substantial increase in overall efficiency. Moreover, the device design incorporates an open-source software interface, facilitating convenient parameter adjustments such as strain, stretching speed, frequency, and duration. Its versatility enables seamless integration with various optical microscopes, thereby yielding novel insights into the realm of mechanobiology.
Collapse
Affiliation(s)
| | | | | | | | | | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan, QLD 4111, Australia; (S.Y.); (P.S.); (N.-K.N.); (C.H.O.); (N.K.)
| |
Collapse
|
17
|
Poznyak AV, Orekhova VA, Sukhorukov VN, Khotina VA, Popov MA, Orekhov AN. Atheroprotective Aspects of Heat Shock Proteins. Int J Mol Sci 2023; 24:11750. [PMID: 37511509 PMCID: PMC10380699 DOI: 10.3390/ijms241411750] [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: 07/06/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Atherosclerosis is a major global health problem. Being a harbinger of a large number of cardiovascular diseases, it ultimately leads to morbidity and mortality. At the same time, effective measures for the prevention and treatment of atherosclerosis have not been developed, to date. All available therapeutic options have a number of limitations. To understand the mechanisms behind the triggering and development of atherosclerosis, a deeper understanding of molecular interactions is needed. Heat shock proteins are important for the normal functioning of cells, actively helping cells adapt to gradual changes in the environment and survive in deadly conditions. Moreover, multiple HSP families play various roles in the progression of cardiovascular disorders. Some heat shock proteins have been shown to have antiatherosclerotic effects, while the role of others remains unclear. In this review, we considered certain aspects of the antiatherosclerotic activity of a number of heat shock proteins.
Collapse
Affiliation(s)
- Anastasia V Poznyak
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
| | - Varvara A Orekhova
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
| | - Vasily N Sukhorukov
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
| | - Victoria A Khotina
- Institute of General Pathology and Pathophysiology, 8, Baltiyskaya St., 125315 Moscow, Russia
| | - Mikhail A Popov
- Department of Cardiac Surgery, Moscow Regional Research and Clinical Institute (MONIKI), 61/2, Shchepkin St., 129110 Moscow, Russia
| | - Alexander N Orekhov
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
| |
Collapse
|
18
|
Galpayage Dona KNU, Ramirez SH, Andrews AM. A Next-Generation 3D Tissue-Engineered Model of the Human Brain Microvasculature to Study the Blood-Brain Barrier. Bioengineering (Basel) 2023; 10:817. [PMID: 37508844 PMCID: PMC10376721 DOI: 10.3390/bioengineering10070817] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/09/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
More than a billion people are affected by neurological disorders, and few have effective therapeutic options. A key challenge that has prevented promising preclinically proven strategies is the translation gap to the clinic. Humanized tissue engineering models that recreate the brain environment may aid in bridging this translational gap. Here, we showcase the methodology that allows for the practical fabrication of a comprehensive microphysicological system (MPS) of the blood-brain barrier (BBB). Compared to other existing 2D and 3D models of the BBB, this model features relevant cytoarchitecture and multicellular arrangement, with branching and network topologies of the vascular bed. This process utilizes 3D bioprinting with digital light processing to generate a vasculature lumen network surrounded by embedded human astrocytes. The lumens are then cellularized with primary human brain microvascular endothelial cells and pericytes. To initiate mechanotransduction pathways and complete maturation, vascular structures are continuously perfused for 7 days. Constructs are validated for complete endothelialization with viability dyes prior to functional assessments that include barrier integrity (permeability) and immune-endothelial interactions. This MPS has applications for the study of novel therapeutics, toxins, and elucidating mechanisms of pathophysiology.
Collapse
Affiliation(s)
- Kalpani N. Udeni Galpayage Dona
- Department of Pathology & Laboratory Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (K.N.U.G.D.)
| | - Servio H. Ramirez
- Department of Pathology & Laboratory Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (K.N.U.G.D.)
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- The Shriners Hospitals Pediatric Research Center, Philadelphia, PA 19140, USA
| | - Allison M. Andrews
- Department of Pathology & Laboratory Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (K.N.U.G.D.)
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
19
|
Wang X, Shen Y, Shang M, Liu X, Munn LL. Endothelial mechanobiology in atherosclerosis. Cardiovasc Res 2023; 119:1656-1675. [PMID: 37163659 PMCID: PMC10325702 DOI: 10.1093/cvr/cvad076] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 02/11/2023] [Accepted: 02/21/2023] [Indexed: 05/12/2023] Open
Abstract
Cardiovascular disease (CVD) is a serious health challenge, causing more deaths worldwide than cancer. The vascular endothelium, which forms the inner lining of blood vessels, plays a central role in maintaining vascular integrity and homeostasis and is in direct contact with the blood flow. Research over the past century has shown that mechanical perturbations of the vascular wall contribute to the formation and progression of atherosclerosis. While the straight part of the artery is exposed to sustained laminar flow and physiological high shear stress, flow near branch points or in curved vessels can exhibit 'disturbed' flow. Clinical studies as well as carefully controlled in vitro analyses have confirmed that these regions of disturbed flow, which can include low shear stress, recirculation, oscillation, or lateral flow, are preferential sites of atherosclerotic lesion formation. Because of their critical role in blood flow homeostasis, vascular endothelial cells (ECs) have mechanosensory mechanisms that allow them to react rapidly to changes in mechanical forces, and to execute context-specific adaptive responses to modulate EC functions. This review summarizes the current understanding of endothelial mechanobiology, which can guide the identification of new therapeutic targets to slow or reverse the progression of atherosclerosis.
Collapse
Affiliation(s)
- Xiaoli Wang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310020, China
| | - Yang Shen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Min Shang
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310020, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lance L Munn
- Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
20
|
Zhang Y, Zhang Y, Hutterer E, Hultin S, Bergman O, Kolbeinsdottir S, Jin H, Forteza MJ, Ketelhuth DFJ, Roy J, Hedin U, Enge M, Matic L, Eriksson P, Holmgren L. The VE-cadherin/AmotL2 mechanosensory pathway suppresses aortic inflammation and the formation of abdominal aortic aneurysms. NATURE CARDIOVASCULAR RESEARCH 2023; 2:629-644. [PMID: 39195920 PMCID: PMC11358041 DOI: 10.1038/s44161-023-00298-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 06/01/2023] [Indexed: 08/29/2024]
Abstract
Endothelial cells respond to mechanical forces exerted by blood flow. Endothelial cell-cell junctions and the sites of endothelial adhesion to the matrix sense and transmit mechanical forces to the cellular cytoskeleton. Here we show that the scaffold protein AmotL2 connects junctional VE-cadherin and actin filaments to the nuclear lamina. AmotL2 is essential for the formation of radial actin filaments and the alignment of endothelial cells, and, in its absence, nuclear integrity and positioning are altered. Molecular analysis demonstrated that VE-cadherin binds to AmotL2 and actin, resulting in a cascade that transmits extracellular mechanical signals to the nuclear membrane. Furthermore, the endothelial deficit of AmotL2 in mice fed normal diet provoked a pro-inflammatory response and abdominal aortic aneurysms (AAAs). Transcriptome analysis of human AAA samples revealed a negative correlation between AmotL2 and inflammation of the aortic intima. These findings offer insight into the link between junctional mechanotransduction and vascular disease.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Department of Oncology-Pathology, BioClinicum, Karolinska Institutet, Stockholm, Sweden
| | - Yumeng Zhang
- Department of Oncology-Pathology, BioClinicum, Karolinska Institutet, Stockholm, Sweden
| | - Evelyn Hutterer
- Department of Oncology-Pathology, BioClinicum, Karolinska Institutet, Stockholm, Sweden
| | - Sara Hultin
- Department of Oncology-Pathology, BioClinicum, Karolinska Institutet, Stockholm, Sweden
| | - Otto Bergman
- Department of Medicine Solna, BioClinicum, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Solrun Kolbeinsdottir
- Department of Oncology-Pathology, BioClinicum, Karolinska Institutet, Stockholm, Sweden
| | - Hong Jin
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Maria J Forteza
- Department of Medicine Solna, BioClinicum, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Daniel F J Ketelhuth
- Department of Medicine Solna, BioClinicum, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Cardiovascular and Renal Research, Institutet of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Joy Roy
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Martin Enge
- Department of Oncology-Pathology, BioClinicum, Karolinska Institutet, Stockholm, Sweden
| | - Ljubica Matic
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Per Eriksson
- Department of Medicine Solna, BioClinicum, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Lars Holmgren
- Department of Oncology-Pathology, BioClinicum, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
21
|
Shayan M, Huang MS, Navarro R, Chiang G, Hu C, Oropeza BP, Johansson PK, Suhar RA, Foster AA, LeSavage BL, Zamani M, Enejder A, Roth JG, Heilshorn SC, Huang NF. Elastin-like protein hydrogels with controllable stress relaxation rate and stiffness modulate endothelial cell function. J Biomed Mater Res A 2023; 111:896-909. [PMID: 36861665 PMCID: PMC10159914 DOI: 10.1002/jbm.a.37520] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 01/26/2023] [Accepted: 02/15/2023] [Indexed: 03/03/2023]
Abstract
Mechanical cues from the extracellular matrix (ECM) regulate vascular endothelial cell (EC) morphology and function. Since naturally derived ECMs are viscoelastic, cells respond to viscoelastic matrices that exhibit stress relaxation, in which a cell-applied force results in matrix remodeling. To decouple the effects of stress relaxation rate from substrate stiffness on EC behavior, we engineered elastin-like protein (ELP) hydrogels in which dynamic covalent chemistry (DCC) was used to crosslink hydrazine-modified ELP (ELP-HYD) and aldehyde/benzaldehyde-modified polyethylene glycol (PEG-ALD/PEG-BZA). The reversible DCC crosslinks in ELP-PEG hydrogels create a matrix with independently tunable stiffness and stress relaxation rate. By formulating fast-relaxing or slow-relaxing hydrogels with a range of stiffness (500-3300 Pa), we examined the effect of these mechanical properties on EC spreading, proliferation, vascular sprouting, and vascularization. The results show that both stress relaxation rate and stiffness modulate endothelial spreading on two-dimensional substrates, on which ECs exhibited greater cell spreading on fast-relaxing hydrogels up through 3 days, compared with slow-relaxing hydrogels at the same stiffness. In three-dimensional hydrogels encapsulating ECs and fibroblasts in coculture, the fast-relaxing, low-stiffness hydrogels produced the widest vascular sprouts, a measure of vessel maturity. This finding was validated in a murine subcutaneous implantation model, in which the fast-relaxing, low-stiffness hydrogel produced significantly more vascularization compared with the slow-relaxing, low-stiffness hydrogel. Together, these results suggest that both stress relaxation rate and stiffness modulate endothelial behavior, and that the fast-relaxing, low-stiffness hydrogels supported the highest capillary density in vivo.
Collapse
Affiliation(s)
- Mahdis Shayan
- Department of Cardiothoracic Surgery, Stanford University, Palo Alto, CA, USA
- The Stanford Cardiovascular Institute, Stanford University, Palo Alto, CA, USA
| | - Michelle S. Huang
- Department of Chemical Engineering, Stanford University, Palo Alto, CA, USA
| | - Renato Navarro
- Department of Materials Science & Engineering, Stanford University, Palo Alto, CA, USA
| | - Gladys Chiang
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Caroline Hu
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Beu P. Oropeza
- Department of Cardiothoracic Surgery, Stanford University, Palo Alto, CA, USA
- The Stanford Cardiovascular Institute, Stanford University, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Patrik K. Johansson
- Geballe Laboratory for Advanced Materials, Stanford University, Palo Alto, CA, USA
| | - Riley A. Suhar
- Department of Materials Science & Engineering, Stanford University, Palo Alto, CA, USA
| | - Abbygail A. Foster
- Department of Materials Science & Engineering, Stanford University, Palo Alto, CA, USA
| | - Bauer L. LeSavage
- Department of Bioengineering, Stanford University, Palo Alto, CA, USA
| | - Maedeh Zamani
- Department of Cardiothoracic Surgery, Stanford University, Palo Alto, CA, USA
- The Stanford Cardiovascular Institute, Stanford University, Palo Alto, CA, USA
| | - Annika Enejder
- Geballe Laboratory for Advanced Materials, Stanford University, Palo Alto, CA, USA
| | - Julien G. Roth
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Sarah C. Heilshorn
- The Stanford Cardiovascular Institute, Stanford University, Palo Alto, CA, USA
- Department of Chemical Engineering, Stanford University, Palo Alto, CA, USA
- Department of Materials Science & Engineering, Stanford University, Palo Alto, CA, USA
- Geballe Laboratory for Advanced Materials, Stanford University, Palo Alto, CA, USA
| | - Ngan F. Huang
- Department of Cardiothoracic Surgery, Stanford University, Palo Alto, CA, USA
- The Stanford Cardiovascular Institute, Stanford University, Palo Alto, CA, USA
- Department of Chemical Engineering, Stanford University, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| |
Collapse
|
22
|
Isosaari L, Vuorenpää H, Yrjänäinen A, Kapucu FE, Kelloniemi M, Pakarinen TK, Miettinen S, Narkilahti S. Simultaneous induction of vasculature and neuronal network formation on a chip reveals a dynamic interrelationship between cell types. Cell Commun Signal 2023; 21:132. [PMID: 37316873 DOI: 10.1186/s12964-023-01159-4] [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: 03/14/2023] [Accepted: 05/06/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND Neuronal networks receive and deliver information to regulate bodily functions while the vascular network provides oxygen, nutrients, and signaling molecules to tissues. Neurovascular interactions are vital for both tissue development and maintaining homeostasis in adulthood; these two network systems align and reciprocally communicate with one another. Although communication between network systems has been acknowledged, the lack of relevant in vitro models has hindered research at the mechanistic level. For example, the current used in vitro neurovascular models are typically established to be short-term (≤ 7 days) culture models, and they miss the supporting vascular mural cells. METHODS In this study, we utilized human induced pluripotent stem cell (hiPSC) -derived neurons, fluorescence tagged human umbilical vein endothelial cells (HUVECs), and either human bone marrow or adipose stem/stromal cells (BMSCs or ASCs) as the mural cell types to create a novel 3D neurovascular network-on-a-chip model. Collagen 1-fibrin matrix was used to establish long-term (≥ 14 days) 3D cell culture in a perfusable microphysiological environment. RESULTS Aprotinin-supplemented endothelial cell growth medium-2 (EGM-2) supported the simultaneous formation of neuronal networks, vascular structures, mural cell differentiation, and the stability of the 3D matrix. The formed neuronal and vascular networks were morphologically and functionally characterized. Neuronal networks supported vasculature formation based on direct cell contacts and by dramatically increasing the secretion of angiogenesis-related factors in multicultures in contrast to cocultures without neurons. Both utilized mural cell types supported the formation of neurovascular networks; however, the BMSCs seemed to boost neurovascular networks to greater extent. CONCLUSIONS Overall, our study provides a novel human neurovascular network model that is applicable for creating in vivo-like tissue models with intrinsic neurovascular interactions. The 3D neurovascular network model on chip forms an initial platform for the development of vascularized and innervated organ-on-chip and further body-on-chip concepts and offers the possibility for mechanistic studies on neurovascular communication both under healthy and in disease conditions. Video Abstract.
Collapse
Affiliation(s)
- Lotta Isosaari
- NeuroGroup, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Hanna Vuorenpää
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Alma Yrjänäinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Fikret Emre Kapucu
- NeuroGroup, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Minna Kelloniemi
- Department of Plastic and Reconstructive Surgery, Tampere University Hospital, Tampere, Finland
| | | | - Susanna Miettinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Susanna Narkilahti
- NeuroGroup, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| |
Collapse
|
23
|
Tian F, Yin L, Lin P, Liu Y, Wang W, Chen Y, Tang Y. Aligned Nanofibrous Net Deposited Perpendicularly on Microridges Supports Endothelium Formation and Promotes the Structural Maturation of hiPSC-Derived Cardiomyocytes. ACS APPLIED MATERIALS & INTERFACES 2023; 15:17518-17531. [PMID: 36992621 DOI: 10.1021/acsami.2c22551] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Cell alignment widely exists in various in vivo tissues and also plays an essential role in the construction of in vitro models, such as vascular endothelial and myocardial models. Recently, microscale and nanoscale hierarchical topographical structures have been drawing increasing attention for engineering in vitro cell alignment. In the present study, we fabricated a micro-/nanohierarchical substrate based on soft lithography and electrospinning to assess the synergetic effect of both the aligned nanofibrous topographical guidance and the off-ground culture environment provided by the substrate on the endothelium formation and the maturation of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). The morphology, proliferation, and barrier formation of human umbilical vein endothelial cells (HUVECs) as well as the alignment, cardiac-specific proteins, and maturity-related gene expression of hiPSC-CMs on the aligned-nanofiber/microridge (AN-MR) substrate were studied. Compared with the glass slide and the single-aligned nanofiber substrate, the AN-MR substrate enhanced the proliferation, alignment, and cell-cell interaction of HUVECs and improved the length of the sarcomere and maturation-related gene expression of hiPSC-CMs. Finally, the response of hiPSC-CMs on different substrates to two typical cardiac drugs (isoproterenol and E-4031) was tested and analyzed, showing that the hiPSC-CMs on AN-MR substrates were more resistant to drugs than those in other groups, which was related to the higher maturity of the cells. Overall, the proposed micro-/nanohierarchical substrate supports the in vitro endothelium formation and enhances the maturation of hiPSC-CMs, which show great potential to be applied in the construction of in vitro models and tissue engineering.
Collapse
Affiliation(s)
- Feng Tian
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Linlin Yin
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Peiran Lin
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Yurong Liu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Wenlong Wang
- School of Mechanical and Electrical Engineering, Guangzhou University, Guangzhou 510006, China
| | - Yong Chen
- PASTEUR, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 24 Rue Lhomond, Paris 75005, France
| | - Yadong Tang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| |
Collapse
|
24
|
Chen X, Li L, Liu X, Liao Y, Qu C, Sun H, Zhao A, Yang P, Huang N, Chen J. Photofunctionalized TiO 2-TiN micropattern coating with anticoagulant properties and ECs contact-guidance effect. Colloids Surf B Biointerfaces 2023; 223:113150. [PMID: 36731267 DOI: 10.1016/j.colsurfb.2023.113150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/09/2023] [Accepted: 01/15/2023] [Indexed: 01/21/2023]
Abstract
Titanium nitride (TiN) and titanium dioxide (TiO2) are two titanium-based coatings commonly used in cardiovascular stent surface engineering. Generally, TiN has good mechanical properties and endothelial cell (ECs) compatibility but poor anticoagulant properties and cannot modulate cell growth orientation and morphology. TiO2 has excellent corrosion resistance and biosafety. Besides, TiO2 has the photocatalytic anticoagulant property, which can migrate to other materials tens of microns away. Based on the above properties, a striped TiO2-TiN micropattern coating was designed and fabricated in this study, and the coating was photofunctionalized by UV irradiation. The obtained photo-functionalized TiO2-TiN micropattern coating showed anticoagulant properties by the migrating effect of the photocatalytic anticoagulant property of TiO2. Besides, the TiO2-TiN micropattern coatings showed ECs compatibility. Furthermore, the growth orientation and cell shape of ECs on TiO2-TiN samples were effectively regulated by the stripe pattern's contact guidance effect, which was particularly evident on the photo-functionalized TiO2-TiN samples. We envision that this photofunctionalized TiO2-TiN striped micropattern coating has significant potential for the surface engineering of vascular stents.
Collapse
Affiliation(s)
- Xiao Chen
- The Department of Ophthalmology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Institute of Biomaterials and Surface Engineering, Key Laboratory for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Linhua Li
- The Department of Ophthalmology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoqi Liu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, the Department of Medical Genetics, the Institute of Laboratory Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology, Chengdu, China
| | - Yuzhen Liao
- Institute of Biomaterials and Surface Engineering, Key Laboratory for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Chao Qu
- The Department of Ophthalmology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hong Sun
- Institute of Biomaterials and Surface Engineering, Key Laboratory for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Ansha Zhao
- Institute of Biomaterials and Surface Engineering, Key Laboratory for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Ping Yang
- Institute of Biomaterials and Surface Engineering, Key Laboratory for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Nan Huang
- Institute of Biomaterials and Surface Engineering, Key Laboratory for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Jiang Chen
- The Department of Ophthalmology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
25
|
Beltrán-Partida E, Valdez-Salas B, García-López Portillo M, Gutierrez-Perez C, Castillo-Uribe S, Salvador-Carlos J, Alcocer-Cañez J, Cheng N. Atherosclerotic-Derived Endothelial Cell Response Conducted by Titanium Oxide Nanotubes. MATERIALS (BASEL, SWITZERLAND) 2023; 16:794. [PMID: 36676534 PMCID: PMC9865858 DOI: 10.3390/ma16020794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/04/2023] [Accepted: 01/11/2023] [Indexed: 06/17/2023]
Abstract
Atherosclerosis lesions are described as the formation of an occlusive wall-vessel plaque that can exacerbate infarctions, strokes, and even death. Furthermore, atherosclerosis damages the endothelium integrity, avoiding proper regeneration after stent implantation. Therefore, we investigate the beneficial effects of TiO2 nanotubes (NTs) in promoting the initial response of detrimental human atherosclerotic-derived endothelial cells (AThEC). We synthesized and characterized NTs on Ti6Al4V by anodization. We isolated AThEC and tested the adhesion long-lasting proliferation activity, and the modulation of focal adhesions conducted on the materials. Moreover, ultrastructural cell-surface contact at the nanoscale and membrane roughness were evaluated to explain the results. Our findings depicted improved filopodia and focal adhesions stimulated by the NTs. Similarly, the NTs harbored long-lasting proliferative metabolism after 5 days, explained by overcoming cell-contact interactions at the nanoscale. Furthermore, the senescent activity detected in the AThEC could be mitigated by the modified membrane roughness and cellular stretch orchestrated by the NTs. Importantly, the NTs stimulate the initial endothelial anchorage and metabolic recovery required to regenerate the endothelial monolayer. Despite the dysfunctional status of the AThEC, our study brings new evidence for the potential application of nano-configured biomaterials for innovation in stent technologies.
Collapse
Affiliation(s)
- Ernesto Beltrán-Partida
- Laboratorio de Biología Molecular y Cáncer, Instituto de Ingeniería, Universidad Autónoma de Baja California, Blvd. Benito Juárez y Calle de la Normal s/n, Mexicali C.P. 21040, Baja California, Mexico
| | - Benjamín Valdez-Salas
- Laboratorio de Biología Molecular y Cáncer, Instituto de Ingeniería, Universidad Autónoma de Baja California, Blvd. Benito Juárez y Calle de la Normal s/n, Mexicali C.P. 21040, Baja California, Mexico
| | - Martha García-López Portillo
- Laboratorio de Biología Molecular y Cáncer, Instituto de Ingeniería, Universidad Autónoma de Baja California, Blvd. Benito Juárez y Calle de la Normal s/n, Mexicali C.P. 21040, Baja California, Mexico
| | - Claudia Gutierrez-Perez
- Laboratorio de Biología Molecular y Cáncer, Instituto de Ingeniería, Universidad Autónoma de Baja California, Blvd. Benito Juárez y Calle de la Normal s/n, Mexicali C.P. 21040, Baja California, Mexico
| | - Sandra Castillo-Uribe
- Laboratorio de Biología Molecular y Cáncer, Instituto de Ingeniería, Universidad Autónoma de Baja California, Blvd. Benito Juárez y Calle de la Normal s/n, Mexicali C.P. 21040, Baja California, Mexico
| | - Jorge Salvador-Carlos
- Laboratorio de Biología Molecular y Cáncer, Instituto de Ingeniería, Universidad Autónoma de Baja California, Blvd. Benito Juárez y Calle de la Normal s/n, Mexicali C.P. 21040, Baja California, Mexico
| | - José Alcocer-Cañez
- Coordinación Clínica de Cirugía, Hospital General de Zona No. 30, Instituto Mexicano del Seguro Social (IMSS), Av. Lerdo de Tejada s/n, Mexicali C.P. 21100, Baja California, Mexico
| | - Nelson Cheng
- Magna International Pte Ltd., 10 H Enterprise Road, Singapore 629834, Singapore
| |
Collapse
|
26
|
Hydrogel-Based Tissue-Mimics for Vascular Regeneration and Tumor Angiogenesis. Regen Med 2023. [DOI: 10.1007/978-981-19-6008-6_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
27
|
Mieremet A, van der Stoel M, Li S, Coskun E, van Krimpen T, Huveneers S, de Waard V. Endothelial dysfunction in Marfan syndrome mice is restored by resveratrol. Sci Rep 2022; 12:22504. [PMID: 36577770 PMCID: PMC9797556 DOI: 10.1038/s41598-022-26662-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022] Open
Abstract
Patients with Marfan syndrome (MFS) develop thoracic aortic aneurysms as the aorta presents excessive elastin breaks, fibrosis, and vascular smooth muscle cell (vSMC) death due to mutations in the FBN1 gene. Despite elaborate vSMC to aortic endothelial cell (EC) signaling, the contribution of ECs to the development of aortic pathology remains largely unresolved. The aim of this study is to investigate the EC properties in Fbn1C1041G/+ MFS mice. Using en face immunofluorescence confocal microscopy, we showed that EC alignment with blood flow was reduced, EC roundness was increased, individual EC surface area was larger, and EC junctional linearity was decreased in aortae of Fbn1C1041G/+ MFS mice. This modified EC phenotype was most prominent in the ascending aorta and occurred before aortic dilatation. To reverse EC morphology, we performed treatment with resveratrol. This restored EC blood flow alignment, junctional linearity, phospho-eNOS expression, and improved the structural integrity of the internal elastic lamina of Fbn1C1041G/+ mice. In conclusion, these experiments identify the involvement of ECs and underlying internal elastic lamina in MFS aortic pathology, which could act as potential target for future MFS pharmacotherapies.
Collapse
Affiliation(s)
- Arnout Mieremet
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
| | - Miesje van der Stoel
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | - Siyu Li
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
| | - Evrim Coskun
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
| | - Tsveta van Krimpen
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | - Stephan Huveneers
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | - Vivian de Waard
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands.
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands.
| |
Collapse
|
28
|
Lai MW, Chow N, Checco A, Kunar B, Redmond D, Rafii S, Rabbany SY. Systems Biology Analysis of Temporal Dynamics That Govern Endothelial Response to Cyclic Stretch. Biomolecules 2022; 12:1837. [PMID: 36551265 PMCID: PMC9775567 DOI: 10.3390/biom12121837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
Endothelial cells in vivo are subjected to a wide array of mechanical stimuli, such as cyclic stretch. Notably, a 10% stretch is associated with an atheroprotective endothelial phenotype, while a 20% stretch is associated with an atheroprone endothelial phenotype. Here, a systems biology-based approach is used to present a comprehensive overview of the functional responses and molecular regulatory networks that characterize the transition from an atheroprotective to an atheroprone phenotype in response to cyclic stretch. Using primary human umbilical vein endothelial cells (HUVECs), we determined the role of the equibiaxial cyclic stretch in vitro, with changes to the radius of the magnitudes of 10% and 20%, which are representative of physiological and pathological strain, respectively. Following the transcriptome analysis of next-generation sequencing data, we identified four key endothelial responses to pathological cyclic stretch: cell cycle regulation, inflammatory response, fatty acid metabolism, and mTOR signaling, driven by a regulatory network of eight transcription factors. Our study highlights the dynamic regulation of several key stretch-sensitive endothelial functions relevant to the induction of an atheroprone versus an atheroprotective phenotype and lays the foundation for further investigation into the mechanisms governing vascular pathology. This study has significant implications for the development of treatment modalities for vascular disease.
Collapse
Affiliation(s)
- Michael W. Lai
- Bioengineering Program, DeMatteis School of Engineering and Applied Science, Hofstra University, New York, NY 11549, USA
| | - Nathan Chow
- Bioengineering Program, DeMatteis School of Engineering and Applied Science, Hofstra University, New York, NY 11549, USA
| | - Antonio Checco
- Bioengineering Program, DeMatteis School of Engineering and Applied Science, Hofstra University, New York, NY 11549, USA
| | - Balvir Kunar
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine (WCM), New York, NY 10065, USA
| | - David Redmond
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine (WCM), New York, NY 10065, USA
| | - Shahin Rafii
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine (WCM), New York, NY 10065, USA
| | - Sina Y. Rabbany
- Bioengineering Program, DeMatteis School of Engineering and Applied Science, Hofstra University, New York, NY 11549, USA
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine (WCM), New York, NY 10065, USA
| |
Collapse
|
29
|
Barrasa-Ramos S, Dessalles CA, Hautefeuille M, Barakat AI. Mechanical regulation of the early stages of angiogenesis. J R Soc Interface 2022; 19:20220360. [PMID: 36475392 PMCID: PMC9727679 DOI: 10.1098/rsif.2022.0360] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Favouring or thwarting the development of a vascular network is essential in fields as diverse as oncology, cardiovascular disease or tissue engineering. As a result, understanding and controlling angiogenesis has become a major scientific challenge. Mechanical factors play a fundamental role in angiogenesis and can potentially be exploited for optimizing the architecture of the resulting vascular network. Largely focusing on in vitro systems but also supported by some in vivo evidence, the aim of this Highlight Review is dual. First, we describe the current knowledge with particular focus on the effects of fluid and solid mechanical stimuli on the early stages of the angiogenic process, most notably the destabilization of existing vessels and the initiation and elongation of new vessels. Second, we explore inherent difficulties in the field and propose future perspectives on the use of in vitro and physics-based modelling to overcome these difficulties.
Collapse
Affiliation(s)
- Sara Barrasa-Ramos
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Claire A. Dessalles
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Mathieu Hautefeuille
- Laboratoire de Biologie du Développement (UMR7622), Institut de Biologie Paris Seine, Sorbonne Université, Paris, France,Facultad de Ciencias, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Abdul I. Barakat
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| |
Collapse
|
30
|
Zamir A, Li G, Chase K, Moskovitch R, Sun B, Zaritsky A. Emergence of synchronized multicellular mechanosensing from spatiotemporal integration of heterogeneous single-cell information transfer. Cell Syst 2022; 13:711-723.e7. [PMID: 35921844 PMCID: PMC9509451 DOI: 10.1016/j.cels.2022.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/14/2021] [Accepted: 07/07/2022] [Indexed: 01/26/2023]
Abstract
Multicellular synchronization is a ubiquitous phenomenon in living systems. However, how noisy and heterogeneous behaviors of individual cells are integrated across a population toward multicellular synchronization is unclear. Here, we study the process of multicellular calcium synchronization of the endothelial cell monolayer in response to mechanical stimuli. We applied information theory to quantify the asymmetric information transfer between pairs of cells and defined quantitative measures to how single cells receive or transmit information within a multicellular network. Our analysis revealed that multicellular synchronization was established by gradual enhancement of information spread from the single cell to the multicellular scale. Synchronization was associated with heterogeneity in the cells' communication properties, reinforcement of the cells' state, and information flow. Altogether, we suggest a phenomenological model where cells gradually learn their local environment, adjust, and reinforce their internal state to stabilize the multicellular network architecture to support information flow from local to global scales toward multicellular synchronization.
Collapse
Affiliation(s)
- Amos Zamir
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Guanyu Li
- Department of Physics, Oregon State University, Corvallis, OR 97331, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Katelyn Chase
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Robert Moskovitch
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Bo Sun
- Department of Physics, Oregon State University, Corvallis, OR 97331, USA.
| | - Assaf Zaritsky
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.
| |
Collapse
|
31
|
Wang S, Zhao Q, Li J, Du X. Morphing-to-Adhesion Polysaccharide Hydrogel for Adaptive Biointerfaces. ACS APPLIED MATERIALS & INTERFACES 2022; 14:42420-42429. [PMID: 36083279 DOI: 10.1021/acsami.2c10117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Reliable functions of medical implants highly depend on biocompatible, conformal, and stable biointerfaces for seamless biointegration with biological tissues. Though flexible biointerfaces based on synthetic hydrogels have shown promise in optimizing implant biointegration via surgical suturing, physical attachment, or manual preshaping, they still suffer from poor adaptability, such as tissue damage by surgical suturing, low bioactivity, and difficulties in conformal contact and stable fixation, especially for specific tissues of large surface curvatures. Here, we report a bilayer hydrogel-based adaptive biointerface (HAB) made of two polysaccharide derivates, N-hydroxysuccinimide (NHS) ester-activated alginate and chitosan, harnessing dual advantages of their different swelling and active groups. Leveraging on the differential swelling between the two hydrogel layers and covalent linkages with active groups at hydrogel interfaces, HABs can be programmed into sealed tubes with tunable diameters via water-induced compliable shape morphing and instant interfacial adhesion. We further demonstrate that the polysaccharide-based morphing-to-adhesion HAB possesses outstanding bioactivity in directing cellular focal adhesion and intercellular junction, versatile geometrical adaptability to diverse tubular tissues with a wide range of surface curvatures (2.8 × 102-1.3 × 103 m-1), and excellent mechanical stability in high load-/shear-bearing physiological environments (blood flow volume: 85 mm·s-1). HABs overcome the limitations of existing biointerfaces in terms of poor bioactivity and difficult biointegration with biological tissues of large surface curvatures, holding promise to open new avenues for adaptive biointerfaces and reliable medical implants.
Collapse
Affiliation(s)
- Shanshan Wang
- Institute of Biomedical & Health Engineering, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen 518035, China
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Science and Technology, China University of Geosciences, Beijing 100083, China
| | - Qilong Zhao
- Institute of Biomedical & Health Engineering, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen 518035, China
| | - Jinhong Li
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Science and Technology, China University of Geosciences, Beijing 100083, China
| | - Xuemin Du
- Institute of Biomedical & Health Engineering, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen 518035, China
| |
Collapse
|
32
|
Lopina OD, Fedorov DA, Sidorenko SV, Bukach OV, Klimanova EA. Sodium Ions as Regulators of Transcription in Mammalian Cells. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:789-799. [PMID: 36171659 DOI: 10.1134/s0006297922080107] [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: 04/18/2022] [Revised: 06/13/2022] [Accepted: 06/13/2022] [Indexed: 06/16/2023]
Abstract
The maintenance of an uneven distribution of Na+ and K+ ions between the cytoplasm and extracellular medium is the basis for the functioning of any animal cell. Changes in the intracellular ratio of these cations occur in response to numerous stimuli and are important for the cell activity regulation. Numerous experimental data have shown that gene transcription in mammalian cells can be regulated by changes in the intracellular [Na+]i/[K+]i ratio. Here, we discuss possible mechanisms of such regulation in various cell types, with special attention to the [Ca2+]-independent signaling pathways that suggest the presence of an intracellular sensor of monovalent cations. As such sensor, we propose the secondary structures of nucleic acids called G-quadruplexes. They are widely represented in mammalian genomes and are often found in the promoters of genes encoding transcription factors.
Collapse
Affiliation(s)
- Olga D Lopina
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119234, Russia.
| | - Dmitrii A Fedorov
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119234, Russia
| | | | - Olesya V Bukach
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119234, Russia
| | | |
Collapse
|
33
|
Jiao Y, Zhang Y, Xiao Y, Xing Y, Cai Z, Wang C, Zhou Z, Feng Z, Gu Y. The crescendo pulse frequency of shear stress stimulates the endothelialization of bone marrow mesenchymal stem cells on the luminal surface of decellularized scaffold in the bioreactor. Bioengineered 2022; 13:7925-7938. [PMID: 35358008 PMCID: PMC9278976 DOI: 10.1080/21655979.2022.2039502] [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] [Indexed: 11/24/2022] Open
Abstract
A completely confluent endothelial cell (EC) monolayer is required to maintain proper vascular function in small diameter tissue-engineered vascular graft (TEVG). However, the most effective method for EC attachment to the luminal surface and formation of an entire endothelium layer that works in vitro remains a complicated challenge that requires urgent resolution. Although pulsatile flow has been shown to be better suited for the generation of functional endothelium, the optimal frequency setting is unknown. Several pulsatile flow frequencies were used to implant rat bone mesenchymal stem cells (MSC) into the lumen of decellularized porcine carotid arteries. The endothelium's integrity and cell activity were investigated in order to determine the best pulse frequency settings. The results showed that MSC were maximally preserved and exhibited maximal morphological changes with improved endothelialization performance in response to increased pulse stimulation frequency. Increased pulse frequency stimulation stimulates the expression of mechanoreceptor markers, cytoskeleton reorganization in the direction of blood flow, denser skeletal proteins fibronectin, and stronger intercellular connections when compared to constant pulse frequency stimulation. MSC eventually develops an intact endothelial layer with anti-thrombotic properties on the inner wall of the decellularized tubular lumen. Conclusion: The decellularized vessels retain the three-dimensional structure of the vasculature, have a surface topography suitable for MSC growth, and have good mechanical properties. By increasing the frequency of pulsed stimulation, MSC endothelialize the lumen of the decellularized vasculature. It is expected to have anti-thrombotic and anti-neointimal hyperplasia properties after implantation, ultimately improving the patency of TEVG.
Collapse
Affiliation(s)
- Yuhao Jiao
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuanguo Zhang
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yonghao Xiao
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, China
| | - Yuehao Xing
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhiwen Cai
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Cong Wang
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhengtong Zhou
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zengguo Feng
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, China
| | - Yongquan Gu
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
34
|
Karakaya C, van Asten JGM, Ristori T, Sahlgren CM, Loerakker S. Mechano-regulated cell-cell signaling in the context of cardiovascular tissue engineering. Biomech Model Mechanobiol 2022; 21:5-54. [PMID: 34613528 PMCID: PMC8807458 DOI: 10.1007/s10237-021-01521-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 09/15/2021] [Indexed: 01/18/2023]
Abstract
Cardiovascular tissue engineering (CVTE) aims to create living tissues, with the ability to grow and remodel, as replacements for diseased blood vessels and heart valves. Despite promising results, the (long-term) functionality of these engineered tissues still needs improvement to reach broad clinical application. The functionality of native tissues is ensured by their specific mechanical properties directly arising from tissue organization. We therefore hypothesize that establishing a native-like tissue organization is vital to overcome the limitations of current CVTE approaches. To achieve this aim, a better understanding of the growth and remodeling (G&R) mechanisms of cardiovascular tissues is necessary. Cells are the main mediators of tissue G&R, and their behavior is strongly influenced by both mechanical stimuli and cell-cell signaling. An increasing number of signaling pathways has also been identified as mechanosensitive. As such, they may have a key underlying role in regulating the G&R of tissues in response to mechanical stimuli. A more detailed understanding of mechano-regulated cell-cell signaling may thus be crucial to advance CVTE, as it could inspire new methods to control tissue G&R and improve the organization and functionality of engineered tissues, thereby accelerating clinical translation. In this review, we discuss the organization and biomechanics of native cardiovascular tissues; recent CVTE studies emphasizing the obtained engineered tissue organization; and the interplay between mechanical stimuli, cell behavior, and cell-cell signaling. In addition, we review past contributions of computational models in understanding and predicting mechano-regulated tissue G&R and cell-cell signaling to highlight their potential role in future CVTE strategies.
Collapse
Affiliation(s)
- Cansu Karakaya
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Jordy G M van Asten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Tommaso Ristori
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Cecilia M Sahlgren
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
- Faculty of Science and Engineering, Biosciences, Åbo Akademi, Turku, Finland
| | - Sandra Loerakker
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
35
|
Current Progress in Vascular Engineering and Its Clinical Applications. Cells 2022; 11:cells11030493. [PMID: 35159302 PMCID: PMC8834640 DOI: 10.3390/cells11030493] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 02/04/2023] Open
Abstract
Coronary heart disease (CHD) is caused by narrowing or blockage of coronary arteries due to atherosclerosis. Coronary artery bypass grafting (CABG) is widely used for the treatment of severe CHD cases. Although autologous vessels are a preferred choice, healthy autologous vessels are not always available; hence there is a demand for tissue engineered vascular grafts (TEVGs) to be used as alternatives. However, producing clinical grade implantable TEVGs that could healthily survive in the host with long-term patency is still a great challenge. There are additional difficulties in producing small diameter (<6 mm) vascular conduits. As a result, there have not been TEVGs that are commercially available. Properties of vascular scaffolds such as tensile strength, thrombogenicity and immunogenicity are key factors that determine the biocompatibility of TEVGs. The source of vascular cells employed to produce TEVGs is a limiting factor for large-scale productions. Advanced technologies including the combined use of natural and biodegradable synthetic materials for scaffolds in conjunction with the use of mesenchyme stem cells or induced pluripotent stem cells (iPSCs) provide promising solutions for vascular tissue engineering. The aim of this review is to provide an update on various aspects in this field and the current status of TEVG clinical applications.
Collapse
|
36
|
Lee GH, Huang SA, Aw WY, Rathod M, Cho C, Ligler FS, Polacheck WJ. Multilayer microfluidic platform for the study of luminal, transmural, and interstitial flow. Biofabrication 2022; 14:10.1088/1758-5090/ac48e5. [PMID: 34991082 PMCID: PMC8867496 DOI: 10.1088/1758-5090/ac48e5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 01/06/2022] [Indexed: 01/27/2023]
Abstract
Efficient delivery of oxygen and nutrients to tissues requires an intricate balance of blood, lymphatic, and interstitial fluid pressures (IFPs), and gradients in fluid pressure drive the flow of blood, lymph, and interstitial fluid through tissues. While specific fluid mechanical stimuli, such as wall shear stress, have been shown to modulate cellular signaling pathways along with gene and protein expression patterns, an understanding of the key signals imparted by flowing fluid and how these signals are integrated across multiple cells and cell types in native tissues is incomplete due to limitations with current assays. Here, we introduce a multi-layer microfluidic platform (MμLTI-Flow) that enables the culture of engineered blood and lymphatic microvessels and independent control of blood, lymphatic, and IFPs. Using optical microscopy methods to measure fluid velocity for applied input pressures, we demonstrate varying rates of interstitial fluid flow as a function of blood, lymphatic, and interstitial pressure, consistent with computational fluid dynamics (CFD) models. The resulting microfluidic and computational platforms will provide for analysis of key fluid mechanical parameters and cellular mechanisms that contribute to diseases in which fluid imbalances play a role in progression, including lymphedema and solid cancer.
Collapse
Affiliation(s)
- Gi-hun Lee
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University
| | - Stephanie A. Huang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University
| | - Wen Y. Aw
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University
| | - Mitesh Rathod
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University
| | - Crescentia Cho
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University
| | - Frances S. Ligler
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University
| | - William J. Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University,Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill,McAllister Heart Institute, University of North Carolina at Chapel Hill
| |
Collapse
|
37
|
Fang Y, Sun W, Zhang T, Xiong Z. Recent advances on bioengineering approaches for fabrication of functional engineered cardiac pumps: A review. Biomaterials 2021; 280:121298. [PMID: 34864451 DOI: 10.1016/j.biomaterials.2021.121298] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 12/18/2022]
Abstract
The field of cardiac tissue engineering has advanced over the past decades; however, most research progress has been limited to engineered cardiac tissues (ECTs) at the microscale with minimal geometrical complexities such as 3D strips and patches. Although microscale ECTs are advantageous for drug screening applications because of their high-throughput and standardization characteristics, they have limited translational applications in heart repair and the in vitro modeling of cardiac function and diseases. Recently, researchers have made various attempts to construct engineered cardiac pumps (ECPs) such as chambered ventricles, recapitulating the geometrical complexity of the native heart. The transition from microscale ECTs to ECPs at a translatable scale would greatly accelerate their translational applications; however, researchers are confronted with several major hurdles, including geometrical reconstruction, vascularization, and functional maturation. Therefore, the objective of this paper is to review the recent advances on bioengineering approaches for fabrication of functional engineered cardiac pumps. We first review the bioengineering approaches to fabricate ECPs, and then emphasize the unmatched potential of 3D bioprinting techniques. We highlight key advances in bioprinting strategies with high cell density as researchers have begun to realize the critical role that the cell density of non-proliferative cardiomyocytes plays in the cell-cell interaction and functional contracting performance. We summarize the current approaches to engineering vasculatures both at micro- and meso-scales, crucial for the survival of thick cardiac tissues and ECPs. We showcase a variety of strategies developed to enable the functional maturation of cardiac tissues, mimicking the in vivo environment during cardiac development. By highlighting state-of-the-art research, this review offers personal perspectives on future opportunities and trends that may bring us closer to the promise of functional ECPs.
Collapse
Affiliation(s)
- Yongcong Fang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China; "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, PR China
| | - Wei Sun
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China; "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, PR China; Department of Mechanical Engineering, Drexel University, Philadelphia, PA, 19104, USA
| | - Ting Zhang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China; "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, PR China.
| | - Zhuo Xiong
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China; "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, PR China.
| |
Collapse
|
38
|
Novak C, Ballinger MN, Ghadiali S. Mechanobiology of Pulmonary Diseases: A Review of Engineering Tools to Understand Lung Mechanotransduction. J Biomech Eng 2021; 143:110801. [PMID: 33973005 PMCID: PMC8299813 DOI: 10.1115/1.4051118] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 05/01/2021] [Indexed: 12/17/2022]
Abstract
Cells within the lung micro-environment are continuously subjected to dynamic mechanical stimuli which are converted into biochemical signaling events in a process known as mechanotransduction. In pulmonary diseases, the abrogated mechanical conditions modify the homeostatic signaling which influences cellular phenotype and disease progression. The use of in vitro models has significantly expanded our understanding of lung mechanotransduction mechanisms. However, our ability to match complex facets of the lung including three-dimensionality, multicellular interactions, and multiple simultaneous forces is limited and it has proven difficult to replicate and control these factors in vitro. The goal of this review is to (a) outline the anatomy of the pulmonary system and the mechanical stimuli that reside therein, (b) describe how disease impacts the mechanical micro-environment of the lung, and (c) summarize how existing in vitro models have contributed to our current understanding of pulmonary mechanotransduction. We also highlight critical needs in the pulmonary mechanotransduction field with an emphasis on next-generation devices that can simulate the complex mechanical and cellular environment of the lung. This review provides a comprehensive basis for understanding the current state of knowledge in pulmonary mechanotransduction and identifying the areas for future research.
Collapse
Affiliation(s)
- Caymen Novak
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Davis Heart and Lung Research Institute, The Ohio State University, Wexner Medical Center, 473 West 12th Avenue, Columbus, OH 43210
| | - Megan N. Ballinger
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Davis Heart and Lung Research Institute, The Ohio State University, Wexner Medical Center, 473 West 12th Avenue, Columbus, OH 43210
| | - Samir Ghadiali
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Davis Heart and Lung Research Institute, The Ohio State University, Wexner Medical Center, 473 West 12th Avenue, Columbus, OH 43210; Department of Biomedical Engineering, The Ohio State University, 2124N Fontana Labs, 140 West 19th Avenue, Columbus, OH 43210
| |
Collapse
|
39
|
Dessalles CA, Ramón-Lozano C, Babataheri A, Barakat AI. Luminal flow actuation generates coupled shear and strain in a microvessel-on-chip. Biofabrication 2021; 14. [PMID: 34592728 DOI: 10.1088/1758-5090/ac2baa] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022]
Abstract
In the microvasculature, blood flow-derived forces are key regulators of vascular structure and function. Consequently, the development of hydrogel-based microvessel-on-chip systems that strive to mimic thein vivocellular organization and mechanical environment has received great attention in recent years. However, despite intensive efforts, current microvessel-on-chip systems suffer from several limitations, most notably failure to produce physiologically relevant wall strain levels. In this study, a novel microvessel-on-chip based on the templating technique and using luminal flow actuation to generate physiologically relevant levels of wall shear stress and circumferential stretch is presented. Normal forces induced by the luminal pressure compress the surrounding soft collagen hydrogel, dilate the channel, and create large circumferential strain. The fluid pressure gradient in the system drives flow forward and generates realistic pulsatile wall shear stresses. Rigorous characterization of the system reveals the crucial role played by the poroelastic behavior of the hydrogel in determining the magnitudes of the wall shear stress and strain. The experimental measurements are combined with an analytical model of flow in both the lumen and the porous hydrogel to provide an exceptionally versatile user manual for an application-based choice of parameters in microvessels-on-chip. This unique strategy of flow actuation adds a dimension to the capabilities of microvessel-on-chip systems and provides a more general framework for improving hydrogel-basedin vitroengineered platforms.
Collapse
Affiliation(s)
- Claire A Dessalles
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, 91120 Palaiseau, France
| | - Clara Ramón-Lozano
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, 91120 Palaiseau, France
| | - Avin Babataheri
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, 91120 Palaiseau, France
| | - Abdul I Barakat
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, 91120 Palaiseau, France
| |
Collapse
|
40
|
Na + i/K + i imbalance contributes to gene expression in endothelial cells exposed to elevated NaCl. Heliyon 2021; 7:e08088. [PMID: 34632152 PMCID: PMC8488490 DOI: 10.1016/j.heliyon.2021.e08088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/27/2021] [Accepted: 09/26/2021] [Indexed: 12/16/2022] Open
Abstract
High-salt consumption contributes to the development of hypertension and is considered an independent risk factor for vascular remodelling, cardiac hypertrophy and stroke incidence. Alterations in NO production, inflammation and endothelial cell stiffening are considered now as plausible mediators of cardiovascular dysfunction. We studied early responses of endothelial cells (HUVEC) caused by a moderate increase in extracellular sodium concentration. Exposure of HUVEC to elevated sodium within the physiological range up to 24 h is accompanied by changes in monovalent cations fluxes and Na,K-ATPase activation, and, in turn, results in a significant decrease in the content of PTGS2, IL6 and IL1LR1 mRNAs. The expression of NOS3 and FOS genes, as well as the abundance of cytosolic and nuclear NFAT5 protein, remained unchanged. We assessed the mechanical properties of endothelial cells by estimating Young's modulus and equivalent elastic constant using atomic force and interference microscopy, respectively. These parameters were unaffected by elevated-salt exposure for 24 h. The data obtained suggest that even small and short-term elevations of extracellular sodium concentration affect the expression of genes involved in the control of endothelial function through the Na+i/K+i-dependent mechanism(s).
Collapse
|
41
|
Islam S, Boström KI, Di Carlo D, Simmons CA, Tintut Y, Yao Y, Hsu JJ. The Mechanobiology of Endothelial-to-Mesenchymal Transition in Cardiovascular Disease. Front Physiol 2021; 12:734215. [PMID: 34566697 PMCID: PMC8458763 DOI: 10.3389/fphys.2021.734215] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/09/2021] [Indexed: 12/31/2022] Open
Abstract
Endothelial cells (ECs) lining the cardiovascular system are subjected to a highly dynamic microenvironment resulting from pulsatile pressure and circulating blood flow. Endothelial cells are remarkably sensitive to these forces, which are transduced to activate signaling pathways to maintain endothelial homeostasis and respond to changes in the environment. Aberrations in these biomechanical stresses, however, can trigger changes in endothelial cell phenotype and function. One process involved in this cellular plasticity is endothelial-to-mesenchymal transition (EndMT). As a result of EndMT, ECs lose cell-cell adhesion, alter their cytoskeletal organization, and gain increased migratory and invasive capabilities. EndMT has long been known to occur during cardiovascular development, but there is now a growing body of evidence also implicating it in many cardiovascular diseases (CVD), often associated with alterations in the cellular mechanical environment. In this review, we highlight the emerging role of shear stress, cyclic strain, matrix stiffness, and composition associated with EndMT in CVD. We first provide an overview of EndMT and context for how ECs sense, transduce, and respond to certain mechanical stimuli. We then describe the biomechanical features of EndMT and the role of mechanically driven EndMT in CVD. Finally, we indicate areas of open investigation to further elucidate the complexity of EndMT in the cardiovascular system. Understanding the mechanistic underpinnings of the mechanobiology of EndMT in CVD can provide insight into new opportunities for identification of novel diagnostic markers and therapeutic interventions.
Collapse
Affiliation(s)
- Shahrin Islam
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Kristina I Boström
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States.,UCLA Molecular Biology Institute, Los Angeles, CA, United States.,Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Dino Di Carlo
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Electrical and Computer Engineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Craig A Simmons
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada.,Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada.,Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Yin Tintut
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States.,Department of Physiology, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Orthopedic Surgery, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yucheng Yao
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Jeffrey J Hsu
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States.,Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| |
Collapse
|
42
|
Lv H, Ai D. Hippo/yes-associated protein signaling functions as a mechanotransducer in regulating vascular homeostasis. J Mol Cell Cardiol 2021; 162:158-165. [PMID: 34547259 DOI: 10.1016/j.yjmcc.2021.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/25/2021] [Accepted: 09/13/2021] [Indexed: 10/20/2022]
Abstract
Cells are constantly exposed to various mechanical forces, including hydrostatic pressure, cyclic stretch, fluid shear stress, and extracellular matrix stiffness. Mechanical cues can be translated into the cell-specific transcriptional process by a cellular mechanic-transducer. Evidence suggests that mechanical signals assist activated intracellular signal transduction pathways and the relative phenotypic adaptation to coordinate cell behavior and disease appropriately. The Hippo/yes-associated protein (YAP) signaling pathway is regulated in response to numerous mechanical stimuli. It plays an important role in the mechanotransduction mechanism, which converts mechanical forces to cascades of molecular signaling to modulate gene expression. This review summarizes the recent findings relevant to the Hippo/YAP pathway-based mechanotransduction in cell behavior and maintaining blood vessels, as well as cardiovascular disease.
Collapse
Affiliation(s)
- Huizhen Lv
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Ion and Molecular Function of Cardiovascular Diseases, Tianjin Institute of Cardiology, Tianjin Medical University, Tianjin 300070, China; Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Ding Ai
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Ion and Molecular Function of Cardiovascular Diseases, Tianjin Institute of Cardiology, Tianjin Medical University, Tianjin 300070, China; Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
43
|
Ewald ML, Chen YH, Lee AP, Hughes CCW. The vascular niche in next generation microphysiological systems. LAB ON A CHIP 2021; 21:3244-3262. [PMID: 34396383 PMCID: PMC8635227 DOI: 10.1039/d1lc00530h] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
In recent years, microphysiological system (MPS, also known as, organ-on-a-chip or tissue chip) platforms have emerged with great promise to improve the predictive capacity of preclinical modeling thereby reducing the high attrition rates when drugs move into trials. While their designs can vary quite significantly, in general MPS are bioengineered in vitro microenvironments that recapitulate key functional units of human organs, and that have broad applications in human physiology, pathophysiology, and clinical pharmacology. A critical next step in the evolution of MPS devices is the widespread incorporation of functional vasculature within tissues. The vasculature itself is a major organ that carries nutrients, immune cells, signaling molecules and therapeutics to all other organs. It also plays critical roles in inducing and maintaining tissue identity through expression of angiocrine factors, and in providing tissue-specific milieus (i.e., the vascular niche) that can support the survival and function of stem cells. Thus, organs are patterned, maintained and supported by the vasculature, which in turn receives signals that drive tissue specific gene expression. In this review, we will discuss published vascularized MPS platforms and present considerations for next-generation devices looking to incorporate this critical constituent. Finally, we will highlight the organ-patterning processes governed by the vasculature, and how the incorporation of a vascular niche within MPS platforms will establish a unique opportunity to study stem cell development.
Collapse
Affiliation(s)
- Makena L Ewald
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA 92697, USA.
| | | | | | | |
Collapse
|
44
|
Dessalles CA, Leclech C, Castagnino A, Barakat AI. Integration of substrate- and flow-derived stresses in endothelial cell mechanobiology. Commun Biol 2021; 4:764. [PMID: 34155305 PMCID: PMC8217569 DOI: 10.1038/s42003-021-02285-w] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 06/02/2021] [Indexed: 02/05/2023] Open
Abstract
Endothelial cells (ECs) lining all blood vessels are subjected to large mechanical stresses that regulate their structure and function in health and disease. Here, we review EC responses to substrate-derived biophysical cues, namely topography, curvature, and stiffness, as well as to flow-derived stresses, notably shear stress, pressure, and tensile stresses. Because these mechanical cues in vivo are coupled and are exerted simultaneously on ECs, we also review the effects of multiple cues and describe burgeoning in vitro approaches for elucidating how ECs integrate and interpret various mechanical stimuli. We conclude by highlighting key open questions and upcoming challenges in the field of EC mechanobiology.
Collapse
Affiliation(s)
- Claire A Dessalles
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, Palaiseau, France
| | - Claire Leclech
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, Palaiseau, France
| | - Alessia Castagnino
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, Palaiseau, France
| | - Abdul I Barakat
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, Palaiseau, France.
| |
Collapse
|
45
|
Mohindra P, Desai TA. Micro- and nanoscale biophysical cues for cardiovascular disease therapy. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2021; 34:102365. [PMID: 33571682 PMCID: PMC8217090 DOI: 10.1016/j.nano.2021.102365] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/15/2021] [Indexed: 11/19/2022]
Abstract
After cardiovascular injury, numerous pathological processes adversely impact the homeostatic function of cardiomyocyte, macrophage, fibroblast, endothelial cell, and vascular smooth muscle cell populations. Subsequent malfunctioning of these cells may further contribute to cardiovascular disease onset and progression. By modulating cellular responses after injury, it is possible to create local environments that promote wound healing and tissue repair mechanisms. The extracellular matrix continuously provides these mechanosensitive cell types with physical cues spanning the micro- and nanoscale to influence behaviors such as adhesion, morphology, and phenotype. It is therefore becoming increasingly compelling to harness these cell-substrate interactions to elicit more native cell behaviors that impede cardiovascular disease progression and enhance regenerative potential. This review discusses recent in vitro and preclinical work that have demonstrated the therapeutic implications of micro- and nanoscale biophysical cues on cell types adversely affected in cardiovascular diseases - cardiomyocytes, macrophages, fibroblasts, endothelial cells, and vascular smooth muscle cells.
Collapse
Affiliation(s)
- Priya Mohindra
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA, United States
| | - Tejal A Desai
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA, United States; Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA; Department of Bioengineering, University of California, Berkeley, Berkeley, CA.
| |
Collapse
|
46
|
Inbody SC, Sinquefield BE, Lewis JP, Horton RE. Biomimetic microsystems for cardiovascular studies. Am J Physiol Cell Physiol 2021; 320:C850-C872. [PMID: 33760660 DOI: 10.1152/ajpcell.00026.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Traditional tissue culture platforms have been around for several decades and have enabled key findings in the cardiovascular field. However, these platforms failed to recreate the mechanical and dynamic features found within the body. Organs-on-chips (OOCs) are cellularized microfluidic-based devices that can mimic the basic structure, function, and responses of organs. These systems have been successfully utilized in disease, development, and drug studies. OOCs are designed to recapitulate the mechanical, electrical, chemical, and structural features of the in vivo microenvironment. Here, we review cardiovascular-themed OOC studies, design considerations, and techniques used to generate these cellularized devices. Furthermore, we will highlight the advantages of OOC models over traditional cell culture vessels, discuss implementation challenges, and provide perspectives on the state of the field.
Collapse
Affiliation(s)
- Shelby C Inbody
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| | - Bridgett E Sinquefield
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| | - Joshua P Lewis
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| | - Renita E Horton
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| |
Collapse
|
47
|
Citi V, Martelli A, Gorica E, Brogi S, Testai L, Calderone V. Role of hydrogen sulfide in endothelial dysfunction: Pathophysiology and therapeutic approaches. J Adv Res 2021; 27:99-113. [PMID: 33318870 PMCID: PMC7728589 DOI: 10.1016/j.jare.2020.05.015] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The vascular endothelium represents a fundamental mechanical and biological barrier for the maintenance of vascular homeostasis along the entire vascular tree. Changes in its integrity are associated to several cardiovascular diseases, including hypertension, atherosclerosis, hyperhomocysteinemia, diabetes, all linked to the peculiar condition named endothelial dysfunction, which is referred to the loss of endothelial physiological functions, comprehending the regulation of vascular relaxation and/or cell redox balance, the inhibition of leukocyte infiltration and the production of NO. Among the endothelium-released vasoactive factors, in the last years hydrogen sulfide has been viewed as one of the main characters involved in the regulation of endothelium functionality, and many studies demonstrated that H2S behaves as a vasoprotective gasotransmitter in those cardiovascular diseases where endothelial dysfunction seems to be the central issue. AIM The role of hydrogen sulfide in endothelial dysfunction-related cardiovascular diseases is discussed in this review. KEY SCIENTIFIC CONCEPTS Possible therapeutic approaches using molecules able to release H2S.
Collapse
Affiliation(s)
- Valentina Citi
- Department of Pharmacy, University of Pisa, via Bonanno n.6, 56125 Pisa, Italy
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, via Bonanno n.6, 56125 Pisa, Italy
| | - Era Gorica
- Department of Pharmacy, University of Pisa, via Bonanno n.6, 56125 Pisa, Italy
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, via Bonanno n.6, 56125 Pisa, Italy
| | - Lara Testai
- Department of Pharmacy, University of Pisa, via Bonanno n.6, 56125 Pisa, Italy
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, via Bonanno n.6, 56125 Pisa, Italy
| |
Collapse
|
48
|
Liu YL, Huang WH. Stretchable Electrochemical Sensors for Cell and Tissue Detection. Angew Chem Int Ed Engl 2020; 60:2757-2767. [PMID: 32632992 DOI: 10.1002/anie.202007754] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/04/2020] [Indexed: 12/21/2022]
Abstract
Electrochemical sensing based on conventional rigid electrodes has great restrictions for characterizing biomolecules in deformed cells or soft tissues. The recent emergence of stretchable sensors allows electrodes to conformally contact to curved surfaces and perfectly comply with the deformation of living cells and tissues. This provides a powerful strategy to monitor biomolecules from mechanically deformed cells, tissues, and organisms in real time, and opens up new opportunities to explore the mechanotransduction process. In this minireview, we first summarize the fabrication of stretchable electrodes with emphasis on the nanomaterial-enabled strategies. We then describe representative applications of stretchable sensors in the real-time monitoring of mechanically sensitive cells and tissues. Finally, we present the future possibilities and challenges of stretchable electrochemical sensing in cell, tissue, and in vivo detection.
Collapse
Affiliation(s)
- Yan-Ling Liu
- Sauvage Center for Molecular Sciences, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Wei-Hua Huang
- Sauvage Center for Molecular Sciences, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| |
Collapse
|
49
|
Liu Y, Huang W. Stretchable Electrochemical Sensors for Cell and Tissue Detection. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202007754] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Yan‐Ling Liu
- Sauvage Center for Molecular Sciences College of Chemistry and Molecular Sciences Wuhan University Wuhan 430072 China
| | - Wei‐Hua Huang
- Sauvage Center for Molecular Sciences College of Chemistry and Molecular Sciences Wuhan University Wuhan 430072 China
| |
Collapse
|
50
|
Fan WT, Qin Y, Hu XB, Yan J, Wu WT, Liu YL, Huang WH. Stretchable Electrode Based on Au@Pt Nanotube Networks for Real-Time Monitoring of ROS Signaling in Endothelial Mechanotransduction. Anal Chem 2020; 92:15639-15646. [PMID: 33179904 DOI: 10.1021/acs.analchem.0c04015] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Vascular endothelial cells (ECs) are natively exposed to dynamic cyclic stretch and respond to it by the production of vasoactive molecules. Among them, reactive oxygen species (ROS) are closely implicated to the endothelial function and vascular homeostasis. However, the dynamic monitoring of ROS release during endothelial mechanotransduction remains a steep challenge. Herein, we developed a stretchable electrochemical sensor by decoration of uniform and ultrasmall platinum nanoparticles (Pt NPs) on gold nanotube (Au NT) networks (denoted as Au@Pt NTs). The orchestrated structure exhibited prominent electrocatalytic property toward the oxidation of hydrogen peroxide (H2O2) (as the most stable ROS) while maintaining excellent mechanical compliance of Au NT networks. Moreover, the favorable biocompatibility of Au NTs and Pt NPs promoted the adhesion and proliferation of ECs cultured thereon. These allowed in situ inducing ECs mechanotransduction and synchronously real-time monitoring of H2O2 release. Further investigation revealed that the production of H2O2 was positively correlated with the applied mechanical strains and could be boosted by other coexisting pathogenic factors. This indicates the great prospect of our proposed sensor in exploring ROS-related signaling for the deep understanding of cell mechanotransduction and vascular disorder.
Collapse
Affiliation(s)
- Wen-Ting Fan
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Yu Qin
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Xue-Bo Hu
- College of Chemistry and Chemical Engineering, Xinyang Normal University, Xinyang 464000, China
| | - Jing Yan
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Wen-Tao Wu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Yan-Ling Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Wei-Hua Huang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| |
Collapse
|