1
|
Jaiswal C, Dey S, Prasad J, Gupta R, Agarwala M, Mandal BB. 3D bioprinted microfluidic based osteosarcoma-on-a chip model as a physiomimetic pre-clinical drug testing platform for anti-cancer drugs. Biomaterials 2025; 320:123267. [PMID: 40138960 DOI: 10.1016/j.biomaterials.2025.123267] [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: 12/06/2024] [Revised: 02/16/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
Standard chemotherapeutic regimen for osteosarcoma (OS) treatment often leads to poor therapeutic outcome, primarily due to lack of an adequate representative model reflecting native OS structural and cellular complexity, posing a translational gap. Three-dimensional bioprinting (3D-BP) represents an efficient and advanced technique for precise recapitulation of the structural and cellular complexity of OS tumor microenvironment (TME). In the present study, we employed a dual extrusion-based 3D-BP method to develop an improved in vitro OS model consisting of both tumor and stromal components. Additionally, a human physiomimetic microfluidic bioreactor is introduced to mimic the dynamic TME and provide physiologically relevant mechanical stimulation to the cells. The model named TC-OS Dynamic model, demonstrated close resemblance to native OS-TME, validated by in vitro studies. Continuous media flow provided mechanical stimulation in the form of shear stress, positively influencing the growth and aggressiveness of OS. Further, drug screening with the model anticancer drugs (doxorubicin, cis-platin, sorafenib) demonstrated enhanced sensitivity in TC-OS Dynamic model as compared to TC-OS Static model, emphasizing enhanced mass transfer, availability and distribution of anticancer drug due to continuous media flow. Overall, TC-OS Dynamic model holds significant potential as a platform in future for high throughput pre-clinical screening of anticancer drugs.
Collapse
Affiliation(s)
- Chitra Jaiswal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781 039, Assam, India
| | - Souradeep Dey
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781 039, Assam, India
| | - Jayant Prasad
- Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati-781039, Assam, India
| | - Raghvendra Gupta
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781 039, Assam, India; Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781 039, Assam, India; Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati-781039, Assam, India
| | - Manoj Agarwala
- GNRC Institute of Medical Sciences, Guwahati 781039, Assam, India
| | - Biman B Mandal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781 039, Assam, India; Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781 039, Assam, India; Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781 039, Assam, India.
| |
Collapse
|
2
|
Deng W, Yang X, Yu J, Omari-Siaw E, Xu X. Recent advances of physiochemical cues on surfaces for directing cell fates. Colloids Surf B Biointerfaces 2025; 250:114550. [PMID: 39929022 DOI: 10.1016/j.colsurfb.2025.114550] [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: 12/23/2024] [Revised: 01/26/2025] [Accepted: 02/01/2025] [Indexed: 02/12/2025]
Abstract
Surface modification plays an essential role in dictating cell behavior and fate, as it creates a microenvironment that profoundly influences cell attachment, migration, proliferation, and differentiation. This review aims to the intricate interplay of culture surface properties, including topography, stiffness, charge, and chemical modifications, demonstrating their profound impact on cell destiny. We explore the nuanced responses of cells to varying surface topographies, from nano- to microscale features, elucidating the influence of geometric patterns and roughness. We also investigate the impact of substrate stiffness, highlighting the way cells perceive and respond to mechanical cues mimicking their native environments. The role of surface charge is examined, revealing how electrostatic interactions influence cell adhesion, signaling, and cell fate decisions. Finally, we delve into the diverse effects of chemical modifications, including the presentation of bioactive molecules, growth factors, and extracellular matrix (ECM) components, demonstrating their ability to guide cell behavior and stimulate specific cellular responses. This review offers comprehensive insights into the important role of surface properties in shaping cell fate, offering promising avenues for developing sophisticated cell culture platforms for applications in drug discovery, regenerative medicine, and fundamental research.
Collapse
Affiliation(s)
- Wenwen Deng
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China
| | - Xiufen Yang
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China
| | - Jiangnan Yu
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China
| | - Emmanuel Omari-Siaw
- Department of Pharmaceutical Science, Kumasi Technical University, PO Box 854, Kumasi, Ashanti, Ghana
| | - Ximing Xu
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China.
| |
Collapse
|
3
|
Alibrandi S, Rinaldi C, Vinci SL, Conti A, Donato L, Scimone C, Sidoti A, D’Angelo R. Mechanotransduction in Development: A Focus on Angiogenesis. BIOLOGY 2025; 14:346. [PMID: 40282211 PMCID: PMC12024848 DOI: 10.3390/biology14040346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025]
Abstract
Cells respond to external mechanical cues and transduce these forces into biological signals. This process is known as mechanotransduction and requires a group of proteins called mechanosensors. This peculiar class of receptors include extracellular matrix proteins, plasma membrane proteins, the cytoskeleton and the nuclear envelope. These cell components are responsive to a wide spectrum of physical cues including stiffness, tensile force, hydrostatic pressure and shear stress. Among mechanotransducers, the Transient Receptor Potential (TRP) and the PIEZO family members are mechanosensitive ion channels, coupling force transduction with intracellular cation transport. Their activity contributes to embryo development, tissue remodeling and repair, and cell homeostasis. In particular, vessel development is driven by hemodynamic cues such as flow direction and shear stress. Perturbed mechanotransduction is involved in several pathological vascular phenotypes including hereditary hemorrhagic telangiectasia. This review is conceived to summarize the most recent findings of mechanotransduction in development. We first collected main features of mechanosensitive proteins. However, we focused on the role of mechanical cues during development. Mechanosensitive ion channels and their function in vascular development are also discussed, with a focus on brain vessel morphogenesis.
Collapse
Affiliation(s)
- Simona Alibrandi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
- Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Istituto Euro-Mediterraneo di Scienza e Tecnologia (I.E.ME.S.T.), Street Michele Miraglia 20, 90139 Palermo, Italy
| | - Carmela Rinaldi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
| | - Sergio Lucio Vinci
- Neuroradiology Unit, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
| | - Alfredo Conti
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, Street Altura 3, 40123 Bologna, Italy
- Department of Biomedical and NeuroMotor Sciences (DiBiNeM), Alma Mater Studiorum—University of Bologna, 40127 Bologna, Italy
| | - Luigi Donato
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
- Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Istituto Euro-Mediterraneo di Scienza e Tecnologia (I.E.ME.S.T.), Street Michele Miraglia 20, 90139 Palermo, Italy
| | - Concetta Scimone
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
| | - Antonina Sidoti
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
- Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Istituto Euro-Mediterraneo di Scienza e Tecnologia (I.E.ME.S.T.), Street Michele Miraglia 20, 90139 Palermo, Italy
| | - Rosalia D’Angelo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
| |
Collapse
|
4
|
Ma L, Fang F, Wang H, Zhao P, Yu H, Liu X. Macrophage co-culture promotes cell reprogramming and prevents ferroptosis in aging fibroblasts for neurodegeneration therapy. J Mol Med (Berl) 2025; 103:301-310. [PMID: 39893308 DOI: 10.1007/s00109-025-02518-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/24/2024] [Accepted: 01/15/2025] [Indexed: 02/04/2025]
Abstract
Ferroptosis, a form of programmed cell death associated with lipid peroxidation and iron dependency, plays a critical role in affecting neuronal function in the aging-related neurodegenerative diseases. Macrophages, influenced by these changes, contribute significantly to the progression of aging nerve diseases. Induced neuronal reprogramming is an advanced technology, which can direct convert somatic cells, such as fibroblasts, into neurons, and offers a promising approach for drug screening aimed at correcting ferroptosis and combating aging-related nerve diseases. However, the efficiency of this reprogramming process remains a significant challenge. In this study, we aimed to manipulate macrophage phenotypes to enhance the direct conversion of fibroblasts into neurons. Specifically, we sought to correct ferroptosis through screening natural compounds using aged fibroblasts and utilizing macrophages to promote induced neuronal (iN) reprogramming. Our findings demonstrate that M2 macrophages effectively promote the direct reprogramming of fibroblasts into iNs. In a novel macrophage-fibroblast co-culture system, M2 macrophages facilitate iN reprogramming by reducing fibroblast adhesion forces and promoting asymmetric cell division. Furthermore, we discovered that manipulating matrix stiffness can induce polarization of macrophages towards the M2 phenotype, thereby enhancing fibroblast reprogramming into iNs. To facilitate these findings, we developed a mechano-cue-based drug screening chip, where soft hydrogels induced and maintained the phenotype of M2 macrophages and effectively promoted cell reprogramming. Using a combinatorial approach with 36 such chips, we screened natural compounds for their anti-aging properties, focusing on reversing fibroblast aging and inducing their conversion into neuronal cells. Notably, Vitexin, an apigenin flavone glycoside with a role as a platelet aggregation inhibitor, emerged as a promising candidate to achieve our therapeutic goals. This study highlights the potential of macrophage-mediated modulation of fibroblast reprogramming as a strategy to address ferroptosis-induced neuronal dysfunction in aging-related nerve diseases. KEY MESSAGE: This study highlights the potential of macrophage-mediated modulation of fibroblast reprogramming as a strategy to address ferroptosis-induced neuronal dysfunction in aging-related nerve diseases.
Collapse
Affiliation(s)
- Lunjie Ma
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Fei Fang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Haonan Wang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Ping Zhao
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Hongchi Yu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China.
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
5
|
Ono R, Maeda K, Tanioka T, Isozaki T. Monocyte-derived Langerhans cells express Delta-like 4 induced by peptidoglycan and interleukin-4 mediated suppression. Front Immunol 2025; 16:1532620. [PMID: 40018044 PMCID: PMC11865044 DOI: 10.3389/fimmu.2025.1532620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/30/2025] [Indexed: 03/01/2025] Open
Abstract
T cells contribute to immunotherapy and autoimmune pathogenesis and Langerhans cells (LCs) have a substantial ability to activate T cells. In vitro-generated monocyte-derived LCs (Mo-LCs) are useful models to study LC function in autoimmune diseases and to test future LC-based immunotherapies. Although dendritic cells (DCs) expressing high levels of Delta-like 4 (DLL4+ DCs), which is a member of the Notch ligand family, have greater ability than DLL4- DCs to activate T cells, the induction method of human DLL4+ DCs has yet to be determined. The aim of this study is to establish whether Mo-LCs express DLL4 and establish the induction method of antigen presenting cells, which most potently activate T cells, similar to our previously established induction method of human Mo-LCs. We compared the ratios of DLL4 expression and T cell activation via flow cytometry among monocyte-derived cells, which have a greater ability than the resident cells to activate T cells. Here, we discovered that Mo-LCs expressed DLL4, which most potently activated T cells among monocyte-derived cells, and that Mo-LCs and DLL4 expression were induced by DLL4, granulocyte macrophage colony-stimulating factor, and transforming growth factor-β1. Additionally, peptidoglycan was required for DLL4 expression, whereas interleukin-4 repressed it. These findings provide insights into the roles of DLL4-expressing cells such as DLL4+ Mo-LCs in human diseases, which will assist with the development of more effective therapeutic strategies in the future.
Collapse
Affiliation(s)
- Rei Ono
- Department of Pathogenesis and Translational Medicine, Showa University Graduate School of Pharmacy, Tokyo, Japan
| | - Kohei Maeda
- Department of Pathogenesis and Translational Medicine, Showa University Graduate School of Pharmacy, Tokyo, Japan
| | - Toshihiro Tanioka
- Department of Pathogenesis and Translational Medicine, Showa University Graduate School of Pharmacy, Tokyo, Japan
| | - Takeo Isozaki
- Department of Pathogenesis and Translational Medicine, Showa University Graduate School of Pharmacy, Tokyo, Japan
- Department of Rheumatology, Showa University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
6
|
Hansen CE, Hollaus D, Kamermans A, de Vries HE. Tension at the gate: sensing mechanical forces at the blood-brain barrier in health and disease. J Neuroinflammation 2024; 21:325. [PMID: 39696463 PMCID: PMC11657007 DOI: 10.1186/s12974-024-03321-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024] Open
Abstract
Microvascular brain endothelial cells tightly limit the entry of blood components and peripheral cells into the brain by forming the blood-brain barrier (BBB). The BBB is regulated by a cascade of mechanical and chemical signals including shear stress and elasticity of the adjacent endothelial basement membrane (BM). During physiological aging, but especially in neurological diseases including multiple sclerosis (MS), stroke, small vessel disease, and Alzheimer's disease (AD), the BBB is exposed to inflammation, rigidity changes of the BM, and disturbed cerebral blood flow (CBF). These altered forces lead to increased vascular permeability, reduced endothelial reactivity to vasoactive mediators, and promote leukocyte transmigration. Whereas the molecular players involved in leukocyte infiltration have been described in detail, the importance of mechanical signalling throughout this process has only recently been recognized. Here, we review relevant features of mechanical forces acting on the BBB under healthy and pathological conditions, as well as the endothelial mechanosensory elements detecting and responding to altered forces. We demonstrate the underlying complexity by focussing on the family of transient receptor potential (TRP) ion channels. A better understanding of these processes will provide insights into the pathogenesis of several neurological disorders and new potential leads for treatment.
Collapse
Affiliation(s)
- Cathrin E Hansen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - David Hollaus
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Alwin Kamermans
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands.
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
7
|
Santorelli M, Bhamidipati PS, Courte J, Swedlund B, Jain N, Poon K, Schildknecht D, Kavanagh A, MacKrell VA, Sondkar T, Malaguti M, Quadrato G, Lowell S, Thomson M, Morsut L. Control of spatio-temporal patterning via cell growth in a multicellular synthetic gene circuit. Nat Commun 2024; 15:9867. [PMID: 39562554 DOI: 10.1038/s41467-024-53078-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 10/01/2024] [Indexed: 11/21/2024] Open
Abstract
A major goal in synthetic development is to build gene regulatory circuits that control patterning. In natural development, an interplay between mechanical and chemical communication shapes the dynamics of multicellular gene regulatory circuits. For synthetic circuits, how non-genetic properties of the growth environment impact circuit behavior remains poorly explored. Here, we first describe an occurrence of mechano-chemical coupling in synthetic Notch (synNotch) patterning circuits: high cell density decreases synNotch-gated gene expression in different cellular systems in vitro. We then construct, both in vitro and in silico, a synNotch-based signal propagation circuit whose outcome can be regulated by cell density. Spatial and temporal patterning outcomes of this circuit can be predicted and controlled via modulation of cell proliferation, initial cell density, and/or spatial distribution of cell density. Our work demonstrates that synthetic patterning circuit outcome can be controlled via cellular growth, providing a means for programming multicellular circuit patterning outcomes.
Collapse
Affiliation(s)
- Marco Santorelli
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Pranav S Bhamidipati
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Josquin Courte
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Benjamin Swedlund
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Naisargee Jain
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kyle Poon
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Dominik Schildknecht
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Andriu Kavanagh
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Biology, California State University Northridge, Northridge, CA, USA
| | - Victoria A MacKrell
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Trusha Sondkar
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Mattias Malaguti
- Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh, UK
- Centre for Engineering Biology, Institute of Quantitative Biology, Biochemistry and Biotechnology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Giorgia Quadrato
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Sally Lowell
- Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh, UK
| | - Matt Thomson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
- Department of Computing and Mathematical Sciences, California Institute of Technology, Pasadena, CA, USA.
- Beckman Center for Single-Cell Profiling and Engineering, Pasadena, CA, USA.
| | - Leonardo Morsut
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
8
|
Passier M, Bentley K, Loerakker S, Ristori T. YAP/TAZ drives Notch and angiogenesis mechanoregulation in silico. NPJ Syst Biol Appl 2024; 10:116. [PMID: 39368976 PMCID: PMC11455968 DOI: 10.1038/s41540-024-00444-3] [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: 04/30/2024] [Accepted: 09/22/2024] [Indexed: 10/07/2024] Open
Abstract
Endothelial cells are key players in the cardiovascular system. Among other things, they are responsible for sprouting angiogenesis, the process of new blood vessel formation essential for both health and disease. Endothelial cells are strongly regulated by the juxtacrine signaling pathway Notch. Recent studies have shown that both Notch and angiogenesis are influenced by extracellular matrix stiffness; however, the underlying mechanisms are poorly understood. Here, we addressed this challenge by combining computational models of Notch signaling and YAP/TAZ, stiffness- and cytoskeleton-regulated mechanotransducers whose activity inhibits both Dll4 (Notch ligand) and LFng (Notch-Dll4 binding modulator). Our simulations successfully mimicked previous experiments, indicating that this YAP/TAZ-Notch crosstalk elucidates the Notch and angiogenesis mechanoresponse to stiffness. Additional simulations also identified possible strategies to control Notch activity and sprouting angiogenesis via cytoskeletal manipulations or spatial patterns of alternating stiffnesses. Our study thus inspires new experimental avenues and provides a promising modeling framework for further investigations into the role of Notch, YAP/TAZ, and mechanics in determining endothelial cell behavior during angiogenesis and similar processes.
Collapse
Affiliation(s)
- Margot Passier
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Katie Bentley
- The Francis Crick Institute, London, UK
- Department of Informatics, King's College London, London, UK
| | - Sandra Loerakker
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems (ICMS), 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 (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
9
|
Al-Nuaimi DA, Rütsche D, Abukar A, Hiebert P, Zanetti D, Cesarovic N, Falk V, Werner S, Mazza E, Giampietro C. Hydrostatic pressure drives sprouting angiogenesis via adherens junction remodelling and YAP signalling. Commun Biol 2024; 7:940. [PMID: 39097636 PMCID: PMC11297954 DOI: 10.1038/s42003-024-06604-9] [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: 11/08/2023] [Accepted: 07/17/2024] [Indexed: 08/05/2024] Open
Abstract
Endothelial cell physiology is governed by its unique microenvironment at the interface between blood and tissue. A major contributor to the endothelial biophysical environment is blood hydrostatic pressure, which in mechanical terms applies isotropic compressive stress on the cells. While other mechanical factors, such as shear stress and circumferential stretch, have been extensively studied, little is known about the role of hydrostatic pressure in the regulation of endothelial cell behavior. Here we show that hydrostatic pressure triggers partial and transient endothelial-to-mesenchymal transition in endothelial monolayers of different vascular beds. Values mimicking microvascular pressure environments promote proliferative and migratory behavior and impair barrier properties that are characteristic of a mesenchymal transition, resulting in increased sprouting angiogenesis in 3D organotypic model systems ex vivo and in vitro. Mechanistically, this response is linked to differential cadherin expression at the adherens junctions, and to an increased YAP expression, nuclear localization, and transcriptional activity. Inhibition of YAP transcriptional activity prevents pressure-induced sprouting angiogenesis. Together, this work establishes hydrostatic pressure as a key modulator of endothelial homeostasis and as a crucial component of the endothelial mechanical niche.
Collapse
Affiliation(s)
| | - Dominic Rütsche
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Experimental Continuum Mechanics, Dübendorf, 8600, Switzerland
| | - Asra Abukar
- ETH Zürich, DMAVT, Experimental Continuum Mechanics, Zürich, 8092, Switzerland
| | - Paul Hiebert
- Department of Biology, ETH Zürich, Institute of Molecular Health Sciences, 8093, Zürich, Switzerland
- Centre for Biomedicine, Hull York Medical School, The University of Hull, Hull, HU6 7RX, UK
| | - Dominik Zanetti
- Department of Biology, ETH Zürich, Institute of Molecular Health Sciences, 8093, Zürich, Switzerland
| | - Nikola Cesarovic
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, 13353, Berlin, Germany
- Department of Health Sciences and Technology, ETH Zürich, 8093, Zürich, Switzerland
| | - Volkmar Falk
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, 13353, Berlin, Germany
- Department of Health Sciences and Technology, ETH Zürich, 8093, Zürich, Switzerland
| | - Sabine Werner
- Department of Biology, ETH Zürich, Institute of Molecular Health Sciences, 8093, Zürich, Switzerland
| | - Edoardo Mazza
- ETH Zürich, DMAVT, Experimental Continuum Mechanics, Zürich, 8092, Switzerland.
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Experimental Continuum Mechanics, Dübendorf, 8600, Switzerland.
| | - Costanza Giampietro
- ETH Zürich, DMAVT, Experimental Continuum Mechanics, Zürich, 8092, Switzerland.
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Experimental Continuum Mechanics, Dübendorf, 8600, Switzerland.
| |
Collapse
|
10
|
Yu Y, Leng Y, Song X, Mu J, Ma L, Yin L, Zheng Y, Lu Y, Li Y, Qiu X, Zhu H, Li J, Wang D. Extracellular Matrix Stiffness Regulates Microvascular Stability by Controlling Endothelial Paracrine Signaling to Determine Pericyte Fate. Arterioscler Thromb Vasc Biol 2023; 43:1887-1899. [PMID: 37650330 DOI: 10.1161/atvbaha.123.319119] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 08/15/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND The differentiation of pericytes into myofibroblasts causes microvascular degeneration, ECM (extracellular matrix) accumulation, and tissue stiffening, characteristics of fibrotic diseases. It is unclear how pericyte-myofibroblast differentiation is regulated in the microvascular environment. Our previous study established a novel 2-dimensional platform for coculturing microvascular endothelial cells (ECs) and pericytes derived from the same tissue. This study investigated how ECM stiffness regulated microvascular ECs, pericytes, and their interactions. METHODS Primary microvessels were cultured in the TGM2D medium (tubular microvascular growth medium on 2-dimensional substrates). Stiff ECM was prepared by incubating ECM solution in regular culture dishes for 1 hour followed by PBS wash. Soft ECM with Young modulus of ≈6 kPa was used unless otherwise noted. Bone grafts were prepared from the rat skull. Immunostaining, RNA sequencing, RT-qPCR (real-time quantitative polymerase chain reaction), Western blotting, and knockdown experiments were performed on the cells. RESULTS Primary microvascular pericytes differentiated into myofibroblasts (NG2+αSMA+) on stiff ECM, even with the TGFβ (transforming growth factor beta) signaling inhibitor A83-01. Soft ECM and A83-01 cooperatively maintained microvascular stability while inhibiting pericyte-myofibroblast differentiation (NG2+αSMA-/low). We thus defined 2 pericyte subpopulations: primary (NG2+αSMA-/low) and activated (NG2+αSMA+) pericytes. Soft ECM promoted microvascular regeneration and inhibited fibrosis in bone graft transplantation in vivo. As integrins are the major mechanosensor, we performed RT-qPCR screening of integrin family members and found Itgb1 (integrin β1) was the major subunit downregulated by soft ECM and A83-01 treatment. Knocking down Itgb1 suppressed myofibroblast differentiation on stiff ECM. Interestingly, ITGB1 phosphorylation (Y783) was mainly located on microvascular ECs on stiff ECM, which promoted EC secretion of paracrine factors, including CTGF (connective tissue growth factor), to induce pericyte-myofibroblast differentiation. CTGF knockdown or monoclonal antibody treatment partially reduced myofibroblast differentiation, implying the participation of multiple pathways in fibrosis formation. CONCLUSIONS ECM stiffness and TGFβ signaling cooperatively regulate microvascular stability and pericyte-myofibroblast differentiation. Stiff ECM promotes EC ITGB1 phosphorylation (Y783) and CTGF secretion, which induces pericyte-myofibroblast differentiation.
Collapse
Affiliation(s)
- Yali Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, China (Y.Y., Y. Leng, X.S., J.M., L.M., L.Y., Y.Z., J.L., D.W.)
- School of Basic Medicine, Qingdao University, China (Y.Y., Y. Leng, X.S., L.M., L.Y., Y.Z.)
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao University, Jinan, China (Y.Y., L.M., D.W.)
| | - Yu Leng
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, China (Y.Y., Y. Leng, X.S., J.M., L.M., L.Y., Y.Z., J.L., D.W.)
- School of Basic Medicine, Qingdao University, China (Y.Y., Y. Leng, X.S., L.M., L.Y., Y.Z.)
| | - Xiuyue Song
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, China (Y.Y., Y. Leng, X.S., J.M., L.M., L.Y., Y.Z., J.L., D.W.)
- School of Basic Medicine, Qingdao University, China (Y.Y., Y. Leng, X.S., L.M., L.Y., Y.Z.)
| | - Jie Mu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, China (Y.Y., Y. Leng, X.S., J.M., L.M., L.Y., Y.Z., J.L., D.W.)
- College of Life Sciences and School of Pharmacy, Medical College, Qingdao University, China (J.M.)
| | - Lei Ma
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, China (Y.Y., Y. Leng, X.S., J.M., L.M., L.Y., Y.Z., J.L., D.W.)
- School of Basic Medicine, Qingdao University, China (Y.Y., Y. Leng, X.S., L.M., L.Y., Y.Z.)
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao University, Jinan, China (Y.Y., L.M., D.W.)
| | - Lin Yin
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, China (Y.Y., Y. Leng, X.S., J.M., L.M., L.Y., Y.Z., J.L., D.W.)
- School of Basic Medicine, Qingdao University, China (Y.Y., Y. Leng, X.S., L.M., L.Y., Y.Z.)
| | - Yu Zheng
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, China (Y.Y., Y. Leng, X.S., J.M., L.M., L.Y., Y.Z., J.L., D.W.)
- School of Basic Medicine, Qingdao University, China (Y.Y., Y. Leng, X.S., L.M., L.Y., Y.Z.)
- Department of Urology, Qingdao Municipal Hospital Affiliated to Qingdao University, China (Y.Z., Y. Lu, H.Z.)
| | - Yi Lu
- Department of Urology, Qingdao Municipal Hospital Affiliated to Qingdao University, China (Y.Z., Y. Lu, H.Z.)
| | - Yuanming Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y. Li, X.Q.)
| | - Xuefeng Qiu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y. Li, X.Q.)
| | - Hai Zhu
- Department of Urology, Qingdao Municipal Hospital Affiliated to Qingdao University, China (Y.Z., Y. Lu, H.Z.)
| | - Jing Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, China (Y.Y., Y. Leng, X.S., J.M., L.M., L.Y., Y.Z., J.L., D.W.)
| | - Dong Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, China (Y.Y., Y. Leng, X.S., J.M., L.M., L.Y., Y.Z., J.L., D.W.)
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao University, Jinan, China (Y.Y., L.M., D.W.)
- Shandong Provincial Institute of Cancer Prevention, Jinan, China (D.W.)
| |
Collapse
|
11
|
Rojas-González DM, Babendreyer A, Ludwig A, Mela P. Analysis of flow-induced transcriptional response and cell alignment of different sources of endothelial cells used in vascular tissue engineering. Sci Rep 2023; 13:14384. [PMID: 37658092 PMCID: PMC10474151 DOI: 10.1038/s41598-023-41247-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 08/23/2023] [Indexed: 09/03/2023] Open
Abstract
Endothelialization of tissue-engineered vascular grafts has proven crucial for implant functionality and thus clinical outcome, however, the choice of endothelial cells (ECs) is often driven by availability rather than by the type of vessel to be replaced. In this work we studied the response to flow of different human ECs with the aim of examining whether their response in vitro is dictated by their original in vivo conditions. Arterial, venous, and microvascular ECs were cultured under shear stress (SS) of 0, 0.3, 3, 1, 10, and 30 dyne/cm2 for 24 h. Regulation of flow-induced marker KLF2 was similar across the different ECs. Upregulation of anti-thrombotic markers, TM and TPA, was mainly seen at higher SS. Cell elongation and alignment was observed for the different ECs at 10 and 30 dyne/cm2 while at lower SS cells maintained a random orientation. Downregulation of pro-inflammatory factors SELE, IL8, and VCAM1 and up-regulation of anti-oxidant markers NQO1 and HO1 was present even at SS for which cell alignment was not observed. Our results evidenced similarities in the response to flow among the different ECs, suggesting that the maintenance of the resting state in vitro is not dictated by the SS typical of the tissue of origin and that absence of flow-induced cell orientation does not necessarily correlate with a pro-inflammatory state of the ECs. These results support the use of ECs from easily accessible sources for in vitro vascular tissue engineering independently from the target vessel.
Collapse
Affiliation(s)
- Diana M Rojas-González
- Department of Biohybrid & Medical Textiles (BioTex) at Center of Biohybrid Medical Systems (CBMS), AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstr. 55, 52074, Aachen, Germany
- Chair of Medical Materials and Implants, Department of Mechanical Engineering, School of Engineering and Design and Munich Institute of Biomedical Engineering, Technical University of Munich, Boltzmannstr 15, 85748, Garching, Germany
| | - Aaron Babendreyer
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany.
| | - Andreas Ludwig
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Petra Mela
- Department of Biohybrid & Medical Textiles (BioTex) at Center of Biohybrid Medical Systems (CBMS), AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstr. 55, 52074, Aachen, Germany.
- Chair of Medical Materials and Implants, Department of Mechanical Engineering, School of Engineering and Design and Munich Institute of Biomedical Engineering, Technical University of Munich, Boltzmannstr 15, 85748, Garching, Germany.
| |
Collapse
|
12
|
Hamrangsekachaee M, Wen K, Bencherif SA, Ebong EE. Atherosclerosis and endothelial mechanotransduction: current knowledge and models for future research. Am J Physiol Cell Physiol 2023; 324:C488-C504. [PMID: 36440856 PMCID: PMC10069965 DOI: 10.1152/ajpcell.00449.2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/16/2022] [Accepted: 11/20/2022] [Indexed: 11/29/2022]
Abstract
Endothelium health is essential to the regulation of physiological vascular functions. Because of the critical capability of endothelial cells (ECs) to sense and transduce chemical and mechanical signals in the local vascular environment, their dysfunction is associated with a vast variety of vascular diseases and injuries, especially atherosclerosis and subsequent cardiovascular diseases. This review describes the mechanotransduction events that are mediated through ECs, the EC subcellular components involved, and the pathways reported to be potentially involved. Up-to-date research efforts involving in vivo animal models and in vitro biomimetic models are also discussed, including their advantages and drawbacks, with recommendations on future modeling approaches to aid the development of novel therapies targeting atherosclerosis and related cardiovascular diseases.
Collapse
Affiliation(s)
| | - Ke Wen
- Chemical Engineering Department, Northeastern University, Boston, Massachusetts
| | - Sidi A Bencherif
- Chemical Engineering Department, Northeastern University, Boston, Massachusetts
- Bioengineering Department, Northeastern University, Boston, Massachusetts
- Laboratoire de BioMécanique et BioIngénierie, UMR CNRS 7388, Sorbonne Universités, Université de Technologie of Compiègne, Compiègne, France
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
| | - Eno E Ebong
- Chemical Engineering Department, Northeastern University, Boston, Massachusetts
- Bioengineering Department, Northeastern University, Boston, Massachusetts
- Neuroscience Department, Albert Einstein College of Medicine, New York, New York
| |
Collapse
|
13
|
Kretschmer M, Mamistvalov R, Sprinzak D, Vollmar AM, Zahler S. Matrix stiffness regulates Notch signaling activity in endothelial cells. J Cell Sci 2023; 136:286810. [PMID: 36718783 DOI: 10.1242/jcs.260442] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 12/22/2022] [Indexed: 02/01/2023] Open
Abstract
Notch signaling is critical for many developmental and disease-related processes. It is widely accepted that Notch has a mechanotransduction module that regulates receptor cleavage. However, the role of biomechanical properties of the cellular environment in Notch signaling in general is still poorly understood. During angiogenesis, differentiation of endothelial cells into tip and stalk cells is regulated by Notch signaling, and remodeling of the extracellular matrix occurs. We investigated the influence of substrate stiffness on the Notch signaling pathway in endothelial cells. Using stiffness-tuned polydimethylsiloxane (PDMS) substrates, we show that activity of the Notch signaling pathway inversely correlates with a physiologically relevant range of substrate stiffness (i.e. increased Notch signaling activity on softer substrates). Trans-endocytosis of the Notch extracellular domain, but not the overall endocytosis, is regulated by substrate stiffness, and integrin cell-matrix connections are both stiffness dependent and influenced by Notch signaling. We conclude that mechanotransduction of Notch activation is modulated by substrate stiffness, highlighting the role of substrate rigidity as an important cue for signaling. This might have implications in pathological situations associated with stiffening of the extracellular matrix, such as tumor growth.
Collapse
Affiliation(s)
- Maibritt Kretschmer
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-Universität München, Butenandtstraße 5-13, 81377 Munich, Germany
| | - Rose Mamistvalov
- The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - David Sprinzak
- The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Angelika M Vollmar
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-Universität München, Butenandtstraße 5-13, 81377 Munich, Germany
| | - Stefan Zahler
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-Universität München, Butenandtstraße 5-13, 81377 Munich, Germany
| |
Collapse
|
14
|
Dabravolski SA, Markin AM, Andreeva ER, Eremin II, Orekhov AN, Melnichenko AA. Molecular Mechanisms Underlying Pathological and Therapeutic Roles of Pericytes in Atherosclerosis. Int J Mol Sci 2022; 23:11663. [PMID: 36232962 PMCID: PMC9570222 DOI: 10.3390/ijms231911663] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022] Open
Abstract
Pericytes are multipotent mesenchymal stromal cells playing an active role in angiogenesis, vessel stabilisation, maturation, remodelling, blood flow regulation and are able to trans-differentiate into other cells of the mesenchymal lineage. In this review, we summarised recent data demonstrating that pericytes play a key role in the pathogenesis and development of atherosclerosis (AS). Pericytes are involved in lipid accumulation, inflammation, growth, and vascularization of the atherosclerotic plaque. Decreased pericyte coverage, endothelial and pericyte dysfunction is associated with intraplaque angiogenesis and haemorrhage, calcification and cholesterol clefts deposition. At the same time, pericytes can be used as a novel therapeutic target to promote vessel maturity and stability, thus reducing plaque vulnerability. Finally, we discuss recent studies exploring effective AS treatments with pericyte-mediated anti-atherosclerotic, anti-inflammatory and anti-apoptotic effects.
Collapse
Affiliation(s)
- Siarhei A. Dabravolski
- Department of Biotechnology Engineering, ORT Braude College, Snunit 51, P.O. Box 78, Karmiel 2161002, Israel
| | - Alexander M. Markin
- Petrovsky National Research Center of Surgery, Abrikosovsky Lane, 2, 119991 Moscow, Russia
| | - Elena R. Andreeva
- Laboratory of Cell Physiology, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye Shosse, 76a, 123007 Moscow, Russia
| | - Ilya I. Eremin
- Petrovsky National Research Center of Surgery, Abrikosovsky Lane, 2, 119991 Moscow, Russia
| | - Alexander N. Orekhov
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
| | | |
Collapse
|
15
|
Zhang W, Li QQ, Gao HY, Wang YC, Cheng M, Wang YX. The regulation of yes-associated protein/transcriptional coactivator with PDZ-binding motif and their roles in vascular endothelium. Front Cardiovasc Med 2022; 9:925254. [PMID: 35935626 PMCID: PMC9354077 DOI: 10.3389/fcvm.2022.925254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/04/2022] [Indexed: 12/14/2022] Open
Abstract
Normal endothelial function plays a pivotal role in maintaining cardiovascular homeostasis, while endothelial dysfunction causes the occurrence and development of cardiovascular diseases. Yes-associated protein (YAP) and its homolog transcriptional co-activator with PDZ-binding motif (TAZ) serve as crucial nuclear effectors in the Hippo signaling pathway, which are regulated by mechanical stress, extracellular matrix stiffness, drugs, and other factors. Increasing evidence supports that YAP/TAZ play an important role in the regulation of endothelial-related functions, including oxidative stress, inflammation, and angiogenesis. Herein, we systematically review the factors affecting YAP/TAZ, downstream target genes regulated by YAP/TAZ and the roles of YAP/TAZ in regulating endothelial functions, in order to provide novel potential targets and effective approaches to prevent and treat cardiovascular diseases.
Collapse
Affiliation(s)
- Wen Zhang
- School of Rehabilitation Medicine, Weifang Medical University, Weifang, China
| | - Qian-qian Li
- School of Rehabilitation Medicine, Weifang Medical University, Weifang, China
| | - Han-yi Gao
- Department of Rehabilitation Medicine, Affiliated Hospital, Weifang Medical University, Weifang, China
| | - Yong-chun Wang
- The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Min Cheng
- School of Basic Medicine, Weifang Medical University, Weifang, China
- *Correspondence: Min Cheng,
| | - Yan-Xia Wang
- School of Rehabilitation Medicine, Weifang Medical University, Weifang, China
- Yan-Xia Wang,
| |
Collapse
|
16
|
Miller B, Sewell-Loftin MK. Mechanoregulation of Vascular Endothelial Growth Factor Receptor 2 in Angiogenesis. Front Cardiovasc Med 2022; 8:804934. [PMID: 35087885 PMCID: PMC8787114 DOI: 10.3389/fcvm.2021.804934] [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: 10/29/2021] [Accepted: 12/10/2021] [Indexed: 12/17/2022] Open
Abstract
The endothelial cells that compose the vascular system in the body display a wide range of mechanotransductive behaviors and responses to biomechanical stimuli, which act in concert to control overall blood vessel structure and function. Such mechanosensitive activities allow blood vessels to constrict, dilate, grow, or remodel as needed during development as well as normal physiological functions, and the same processes can be dysregulated in various disease states. Mechanotransduction represents cellular responses to mechanical forces, translating such factors into chemical or electrical signals which alter the activation of various cell signaling pathways. Understanding how biomechanical forces drive vascular growth in healthy and diseased tissues could create new therapeutic strategies that would either enhance or halt these processes to assist with treatments of different diseases. In the cardiovascular system, new blood vessel formation from preexisting vasculature, in a process known as angiogenesis, is driven by vascular endothelial growth factor (VEGF) binding to VEGF receptor 2 (VEGFR-2) which promotes blood vessel development. However, physical forces such as shear stress, matrix stiffness, and interstitial flow are also major drivers and effectors of angiogenesis, and new research suggests that mechanical forces may regulate VEGFR-2 phosphorylation. In fact, VEGFR-2 activation has been linked to known mechanobiological agents including ERK/MAPK, c-Src, Rho/ROCK, and YAP/TAZ. In vascular disease states, endothelial cells can be subjected to altered mechanical stimuli which affect the pathways that control angiogenesis. Both normalizing and arresting angiogenesis associated with tumor growth have been strategies for anti-cancer treatments. In the field of regenerative medicine, harnessing biomechanical regulation of angiogenesis could enhance vascularization strategies for treating a variety of cardiovascular diseases, including ischemia or permit development of novel tissue engineering scaffolds. This review will focus on the impact of VEGFR-2 mechanosignaling in endothelial cells (ECs) and its interaction with other mechanotransductive pathways, as well as presenting a discussion on the relationship between VEGFR-2 activation and biomechanical forces in the extracellular matrix (ECM) that can help treat diseases with dysfunctional vascular growth.
Collapse
Affiliation(s)
- Bronte Miller
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mary Kathryn Sewell-Loftin
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|