1
|
Chisolm SJ, Guo E, Subramaniam V, Schulze KD, Angelini TE. Elastic modulus versus cell packing density in MDCK epithelial monolayers. J Biomech 2025; 184:112648. [PMID: 40157056 DOI: 10.1016/j.jbiomech.2025.112648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/18/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
The elastic moduli of tissues are connected to their states of health and function. The epithelial monolayer is a simple, minimal, tissue model that is often used to gain understanding of mechanical behavior at the cellular or multi-cellular scale. Here we investigate how the elastic modulus of Madin Darby Canine Kidney (MDCK) cells depends on their packing density. Rather than measuring elasticity at the sub-cellular scale with local probes, we characterize the monolayer at the multi-cellular scale, as one would a thin slab of elastic material. We use a micro-indentation system to apply gentle forces to the apical side of MDCK monolayers, applying a normal force to approximately 100 cells in each experiment. In low-density confluent monolayers, we find that the elastic modulus decreases with increasing cell density. At high densities, the modulus appears to plateau. This finding will help guide our understanding of known collective behaviors in epithelial monolayers and other tissues where variations in cell packing density are correlated with cell motion.
Collapse
Affiliation(s)
- Steven J Chisolm
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32605, United States.
| | - Emily Guo
- Department of Mechanical Engineering, Auburn University, Auburn, AL 36849, United States.
| | - Vignesh Subramaniam
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32605, United States.
| | - Kyle D Schulze
- Department of Mechanical Engineering, Auburn University, Auburn, AL 36849, United States.
| | - Thomas E Angelini
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32605, United States; Department of Materials Science and Engineering, University of Florida, Gainesville, FL 32605, United States; J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32605, United States.
| |
Collapse
|
2
|
Han J, Halwachs K, West T, Larsen B, Sacks MS, Rosales AM, Zoldan J. Matrix Stiffness Regulates Mechanotransduction and Vascular Network Formation of hiPSC-Derived Endothelial Progenitors Encapsulated in 3D Hydrogels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.11.648340. [PMID: 40291699 PMCID: PMC12027365 DOI: 10.1101/2025.04.11.648340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The mechanical properties of the extracellular matrix (ECM), particularly stiffness, regulate endothelial progenitor responses during vascular development, yet their behavior in physiologically compliant matrices (<1 kPa) remains underexplored. Using norbornene-modified hyaluronic acid (NorHA) hydrogels with tunable stiffness (190-884 Pa), we investigated how hydrogel stiffness influences cell morphology, endothelial maturation, mechanotransduction, and microvascular network formation in human induced pluripotent stem cell-derived endothelial progenitors (hiPSC-EPs). Our findings reveal a stiffness-dependent tradeoff between mechanotransduction and vascular network formation. At intermediate stiffness (551 Pa), cells exhibited the greatest increase in endothelial marker CD31 expression and Yes-associated protein (YAP)/ transcriptional coactivator with PDZ-binding motif (TAZ) nuclear translocation, indicating enhanced mechanotransduction and endothelial maturation. However, this did not translate to superior plexus formation. Instead, the most compliant matrix (190 Pa) supported greater vascular connectivity, characterized by longer branches (∼0.03/volume vs. 0.015 at 551 Pa) and enhanced actin remodeling. 3D cell contraction measurements revealed a 15.6-fold higher basal displacement in compliant hydrogels, suggesting that cell-generated forces and matrix deformability collectively drive vascular morphogenesis. Unlike prior studies focusing on pathological stiffness ranges (>10 kPa), our results emphasize that vascularization is not solely driven by the most mechanotransductive environment but rather by a balance of compliance, contractility, and cell-induced remodeling. These findings underscore the need to design hydrogels that provide sufficient mechanotransduction for endothelial maturation while maintaining compliance to support dynamic vascular morphogenesis. This work provides a mechanically tuned framework for optimizing microenvironments to balance endothelial differentiation and vascular network formation in tissue engineering and regenerative medicine.
Collapse
|
3
|
Zhu C, Bishop T, Gregorich ZR, Guo W. Titin is a new factor regulating arterial stiffness through vascular smooth muscle cell tone in male rats. Physiol Rep 2025; 13:e70270. [PMID: 40119572 PMCID: PMC11928681 DOI: 10.14814/phy2.70270] [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: 11/12/2024] [Revised: 02/03/2025] [Accepted: 02/28/2025] [Indexed: 03/24/2025] Open
Abstract
Arterial stiffness is a robust predictor of cardiovascular disease and mortality. As such, there is substantial interest in uncovering its causal factors for the development of targeted treatments to regulate arterial stiffness. The elastic protein titin is a key determinant of myocardial stiffness, yet whether it plays a role in regulating arterial stiffness is unknown. In this study, we aimed to investigate the role of titin in vascular smooth muscle cell (VSMC) and overall arterial stiffness. To do this, we took advantage of rats lacking RNA binding motif 20 (RBM20), the primary splicing regulator of titin, in striated muscles. Using this model, we demonstrate that RBM20 regulates titin isoform expression in smooth muscle, with loss of the protein leading to the expression of larger titin isoforms. We show that the expression of larger titin reduces the stiffness of VSMCs. While decreased titin-based VSMC stiffness did not affect baseline arterial stiffness, we found that arterial stiffness was reduced in response to a challenge with the potent vasoconstrictor angiotensin II (Ang II). The observed reduction in arterial stiffness following Ang II treatment was not the result of changes in either the extracellular matrix or myofilaments. We further show that the expression of a larger titin isoform ameliorates cardiac remodeling caused by Ang II-associated hypertension. In summary, our study provides the first evidence that titin regulates VSMC stiffness, which is relevant for arterial stiffness in the context of elevated blood pressure. Furthermore, our data provide proof-of-concept evidence that targeting RBM20 to reduce arterial stiffness through titin isoform switching may benefit aging- or hypertension-associated arterial stiffness and vascular diseases.
Collapse
MESH Headings
- Animals
- Connectin/metabolism
- Connectin/genetics
- Vascular Stiffness/physiology
- Male
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/cytology
- Rats
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/physiology
- Rats, Sprague-Dawley
- Angiotensin II/pharmacology
- Protein Isoforms/metabolism
Collapse
Affiliation(s)
- Chaoqun Zhu
- Department of Animal SciencesUniversity of WyomingLaramieWyomingUSA
| | | | - Zachery R. Gregorich
- Department of Animal and Dairy SciencesUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Cardiovascular Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Wei Guo
- Department of Animal SciencesUniversity of WyomingLaramieWyomingUSA
- Department of Animal and Dairy SciencesUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Cardiovascular Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| |
Collapse
|
4
|
Alfaro-García JP, Orozco-Castaño CA, Sánchez-Rendón JA, Casanova-Yépes HF, Vicente-Manzanares M, Gallego-Gómez JC. Characterization of the Temporal Dynamics of the Endothelial-Mesenchymal-like Transition Induced by Soluble Factors from Dengue Virus Infection in Microvascular Endothelial Cells. Int J Mol Sci 2025; 26:2139. [PMID: 40076764 PMCID: PMC11900998 DOI: 10.3390/ijms26052139] [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: 01/21/2025] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Dengue virus (DV) infection poses a severe life-threatening risk in certain cases. This is mainly due to endothelial dysregulation, which causes plasma leakage and hemorrhage. However, the etiology of DV-induced endothelial dysregulation remains incompletely understood. To identify the potential mechanisms of endothelial dysregulation caused by DV, the effects of conditioned media from Dengue virus (CMDV) on the mechanics and transcriptional profile of the endothelial cells were examined using permeability assays, atomic force microscopy, In-Cell Western blot and in silico transcriptomics. Exposure of HMEC-1 cells to the CMDV increased endothelial permeability and cellular stiffness. It also induced the expression of the key proteins associated with endothelial-to-mesenchymal transition (EndMT). These data support the notion that the DV promotes endothelial dysfunction by triggering transcriptional programs that compromise the endothelial barrier function. Understanding the molecular mechanisms underlying DV-induced endothelial dysregulation is crucial for developing targeted therapeutic strategies to mitigate the severe outcomes associated with dengue infection.
Collapse
Affiliation(s)
- Jenny Paola Alfaro-García
- Grupo Medicina de Translación—Facultad de Medicina, Universidad de Antioquia, Medellín 050010, Colombia;
| | | | - Julián Andrés Sánchez-Rendón
- Grupo de Coloides—Facultad de Ciencias Exactas y Naturales, Universidad de Antioquia, Medellín 050010, Colombia; (J.A.S.-R.); (H.F.C.-Y.)
| | - Herley Fernando Casanova-Yépes
- Grupo de Coloides—Facultad de Ciencias Exactas y Naturales, Universidad de Antioquia, Medellín 050010, Colombia; (J.A.S.-R.); (H.F.C.-Y.)
| | - Miguel Vicente-Manzanares
- Molecular Mechanisms Program, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)—University of Salamanca, 37007 Salamanca, Spain
| | - Juan Carlos Gallego-Gómez
- Grupo Medicina de Translación—Facultad de Medicina, Universidad de Antioquia, Medellín 050010, Colombia;
| |
Collapse
|
5
|
Kanuparthy M, Manthana R, Kaushik H, Xiang K, Hamze J, Marimekala D, Feng J, Sellke FW. Microvascular Dysfunction Following Cardioplegic Arrest and Cardiopulmonary Bypass: Impacts of Diabetes and Hypertension. Biomedicines 2025; 13:409. [PMID: 40002822 PMCID: PMC11853148 DOI: 10.3390/biomedicines13020409] [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: 12/31/2024] [Revised: 01/27/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Cardioplegic arrest and cardiopulmonary bypass (CP/CPB) are known to engender microvascular dysfunction in patients undergoing cardiac surgery. These effects are significantly varied by patient comorbidities including diabetes and hypertension. Both diabetes and hypertension are associated with worse outcomes after cardiac surgery, partly related to increased microvascular complications. In this review, we examine several key facets of microvascular dysfunction after CP/CPB: microvascular endothelial and vasomotor dysfunction, altered gene and protein expression, endothelial adherens junction dysfunction, and programmed cell death as they relate to diabetes and hypertension. This review examines both classical techniques, including microvessel reactivity assays, and modern multiomic approaches to characterizing these microvascular changes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Frank W. Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (M.K.); (R.M.); (H.K.); (K.X.); (J.H.); (D.M.); (J.F.)
| |
Collapse
|
6
|
Vu V, Sullivan B, Hebner E, Rahil Z, Zou Y, Leckband D. Cadherins and growth factor receptors - ligand-selective mechano-switches at cadherin junctions. J Cell Sci 2025; 138:JCS262279. [PMID: 39817537 PMCID: PMC11883276 DOI: 10.1242/jcs.262279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 01/06/2025] [Indexed: 01/18/2025] Open
Abstract
This study investigated possible mechanisms underlying differences between heterophilic and homophilic cadherin adhesions that influence intercellular mechanics and multicellular organization. Results suggest that homophilic cadherin ligation selectively activates force transduction, such that resulting signaling and mechano-transduction amplitudes are independent of cadherin-binding affinities. Epithelial (E-) and neural (N-)cadherin cooperate with distinct growth factors to mechanically activate force transduction cascades. Prior results have demonstrated that E-cadherin and epidermal growth factor receptor form force-sensitive complexes at intercellular junctions. Here, we show that the reconstitution of N-cadherin force transduction requires the co-expression of N-cadherin and fibroblast growth factor receptor. Mechanical measurements further demonstrated that homophilic ligation initiates receptor tyrosine kinase-dependent force transduction cascades, but heterophilic cadherin ligands fail to activate signaling or generate stereotypical mechano-transduction signatures. The all-or-nothing contrast between mechano-transduction by heterophilic versus homophilic cadherin adhesions supersedes differences in cadherin adhesion strength. This mechano-selectivity impacts cell spreading and traction generation on cadherin substrates. Homophilic ligation appears to be a key that selectively unlocks cadherin mechano-transduction. These findings might reconcile the roles of cadherin recognition and cell mechanics in the organization of multicellular assemblies.
Collapse
Affiliation(s)
- Vinh Vu
- Department of Biochemistry, University of Illinois, 600 South Mathews Ave, Urbana, IL 61801, USA
| | - Brendan Sullivan
- Department of Biochemistry, University of Illinois, 600 South Mathews Ave, Urbana, IL 61801, USA
| | - Evan Hebner
- Department of Biochemistry, University of Illinois, 600 South Mathews Ave, Urbana, IL 61801, USA
| | - Zainab Rahil
- Department of Bioengineering, University of Illinois, 1402 W Green St., Urbana, IL 61801, USA
| | - Yubo Zou
- Department of Biochemistry, University of Illinois, 600 South Mathews Ave, Urbana, IL 61801, USA
| | - Deborah Leckband
- Department of Biochemistry, University of Illinois, 600 South Mathews Ave, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois, 1402 W Green St., Urbana, IL 61801, USA
- Department of Chemical and Biomolecular Engineering, University of Illinois, 600 South Mathews Ave, Urbana, IL 61801, USA
- Department of Chemistry, University of Illinois, 600 South Mathews Ave, Urbana, IL 61801, USA
- Carl W. Woese Institute for Genomic Biology, 1206 West Gregory Drive, Urbana, IL 61801, USA
| |
Collapse
|
7
|
Simões-Faria R, Daems M, Peacock HM, Declercq M, Willems A, Jones EAV, Ghesquière B. Wall shear stress modulates metabolic pathways in endothelial cells. Metabolomics 2025; 21:16. [PMID: 39832080 PMCID: PMC11753319 DOI: 10.1007/s11306-024-02214-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025]
Abstract
INTRODUCTION Hemodynamic forces play a crucial role in modulating endothelial cell (EC) behavior, significantly influencing blood vessel responses. While traditional in vitro studies often explore ECs under static conditions, ECs are exposed to various hemodynamic forces in vivo. This study investigates how wall shear stress (WSS) influences EC metabolism, focusing on the interplay between WSS and key metabolic pathways. OBJECTIVES The aim of this study is to examine the effects of WSS on EC metabolism, specifically evaluating its impact on central carbon metabolism and glycolysis using transcriptomics and tracer metabolomics approaches. METHODS ECs were exposed to WSS, and transcriptomic analysis was performed to assess gene expression changes related to metabolic pathways. Tracer metabolomics was used to track metabolic fluxes, focusing on glutamine and glycolytic metabolism. Additionally, chemical inhibition of glutamate dehydrogenase was conducted to evaluate its role in EC fitness under WSS. RESULTS Transcriptomic data revealed upregulation of glutamine and glutamate pathways, alongside downregulation of glycolytic activity in ECs exposed to WSS. Tracer metabolomics confirmed that WSS promotes glutamine anaplerosis into the Krebs cycle, while decreasing glycolytic metabolism. Suppression of glutamate dehydrogenase impaired EC fitness under WSS conditions. CONCLUSION Our findings illuminate that ECs subjected to WSS exhibit a preference for glutamine as a key nutrient source for central carbon metabolism pathways, indicating diminished reliance on glycolysis. This study elucidates the nutritional predilections and regulatory mechanisms governing EC metabolism under WSS in vitro, underscoring the pivotal role of physical stimuli in shaping EC metabolic responses.
Collapse
Affiliation(s)
- Rita Simões-Faria
- Laboratory of Applied Mass Spectrometry, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- Metabolomics Core Facility Leuven, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Margo Daems
- Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Hanna M Peacock
- Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Mathias Declercq
- Department of Development and Regeneration, CF Centre, Woman and Child, KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Anton Willems
- Laboratory of Applied Mass Spectrometry, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- Metabolomics Core Facility Leuven, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Elizabeth A V Jones
- Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Bart Ghesquière
- Laboratory of Applied Mass Spectrometry, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.
- Metabolomics Core Facility Leuven, Center for Cancer Biology, VIB, Leuven, Belgium.
| |
Collapse
|
8
|
Cronin NM, Dawson LW, DeMali KA. Shear stress-stimulated AMPK couples endothelial cell mechanics, metabolism and vasodilation. J Cell Sci 2024; 137:jcs262232. [PMID: 39513477 PMCID: PMC11795286 DOI: 10.1242/jcs.262232] [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: 04/29/2024] [Accepted: 11/02/2024] [Indexed: 11/15/2024] Open
Abstract
Endothelial cells respond to mechanical force by stimulating cellular signaling, but how these pathways are linked to elevations in cell metabolism and whether metabolism supports the mechanical response remains poorly understood. Here, we show that the application of force to endothelial cells stimulates VE-cadherin to activate liver kinase B1 (LKB1; also known as STK11) and AMP-activated protein kinase (AMPK), a master regulator of energy homeostasis. VE-cadherin-stimulated AMPK increases eNOS (also known as NOS3) activity and localization to the plasma membrane, reinforcement of the actin cytoskeleton and cadherin adhesion complex, and glucose uptake. We present evidence for the increase in metabolism being necessary to fortify the adhesion complex, actin cytoskeleton and cellular alignment. Together, these data extend the paradigm for how mechanotransduction and metabolism are linked to include a connection to vasodilation, thereby providing new insight into how diseases involving contractile, metabolic and vasodilatory disturbances arise.
Collapse
Affiliation(s)
- Nicholas M. Cronin
- Roy J. and Lucille A. Carver College of Medicine at the University of Iowa, Department of Biochemistry and Molecular Biology, 51 Newton Rd, Iowa City, IA 52242, USA
| | - Logan W. Dawson
- Roy J. and Lucille A. Carver College of Medicine at the University of Iowa, Department of Biochemistry and Molecular Biology, 51 Newton Rd, Iowa City, IA 52242, USA
| | - Kris A. DeMali
- Roy J. and Lucille A. Carver College of Medicine at the University of Iowa, Department of Biochemistry and Molecular Biology, 51 Newton Rd, Iowa City, IA 52242, USA
| |
Collapse
|
9
|
Mierke CT. Mechanosensory entities and functionality of endothelial cells. Front Cell Dev Biol 2024; 12:1446452. [PMID: 39507419 PMCID: PMC11538060 DOI: 10.3389/fcell.2024.1446452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
The endothelial cells of the blood circulation are exposed to hemodynamic forces, such as cyclic strain, hydrostatic forces, and shear stress caused by the blood fluid's frictional force. Endothelial cells perceive mechanical forces via mechanosensors and thus elicit physiological reactions such as alterations in vessel width. The mechanosensors considered comprise ion channels, structures linked to the plasma membrane, cytoskeletal spectrin scaffold, mechanoreceptors, and junctional proteins. This review focuses on endothelial mechanosensors and how they alter the vascular functions of endothelial cells. The current state of knowledge on the dysregulation of endothelial mechanosensitivity in disease is briefly presented. The interplay in mechanical perception between endothelial cells and vascular smooth muscle cells is briefly outlined. Finally, future research avenues are highlighted, which are necessary to overcome existing limitations.
Collapse
|
10
|
Gráczer É, Pászty K, Harsányi L, Lehoczky C, Fülöp A, Varga A. BRAF Modulates the Interplay Between Cell-Cell and Cell-Extracellular Matrix Adhesions in PECAM-1-Mediated Mechanotransduction. Int J Mol Sci 2024; 25:11234. [PMID: 39457016 PMCID: PMC11508702 DOI: 10.3390/ijms252011234] [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: 09/11/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Mechanotransduction, the process of how cells sense and convert mechanical stimuli into biochemical response, is crucial in the migration of leukocytes or cancer cells through the endothelium during inflammation or metastasis. Migrating cells exert forces on the endothelium through cell surface adhesion molecules, such as platelet endothelial adhesion molecule PECAM-1, and this is essential for a successful transmigration. To study PECAM-1-mediated mechanotransduction, we applied PECAM-1-antibody-coated magnetic beads and exerted about 40 pN force on the endothelial monolayer. We show that force increases cell-ECM adhesion in the cell center and is accompanied by the opening of cell-cell junctions. Upon depletion of the MEK/ERK kinase, BRAF force increases cell-ECM adhesion both at the cell periphery and in the cell center, but this does not result in the opening of cell-cell junctions. Decreasing cell-ECM adhesion in BRAF-depleted cells through FAK inhibition results in the remodeling of cell-cell junctions. Force-induced increase in cell-ECM adhesion in the cell center correlates with the activation of the transcriptional cofactor Yes-associated protein (YAP). Furthermore, the induced activation of YAP through LATS inhibition prevents junctional remodeling in control cells. Thus, the activation of YAP might determine the strength of cell-cell junctions during PECAM-1-mediated mechanotransduction.
Collapse
Affiliation(s)
- Éva Gráczer
- Department of Biophysics and Radiation Biology, Semmelweis University, H-1094 Budapest, Hungary; (É.G.); (K.P.)
| | - Katalin Pászty
- Department of Biophysics and Radiation Biology, Semmelweis University, H-1094 Budapest, Hungary; (É.G.); (K.P.)
| | - Laura Harsányi
- Department of Biophysics and Radiation Biology, Semmelweis University, H-1094 Budapest, Hungary; (É.G.); (K.P.)
| | - Csilla Lehoczky
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, H-1083 Budapest, Hungary
| | - Antónia Fülöp
- Faculty of Electrical Engineering and Informatics, Budapest University of Technology and Economics, H-1111 Budapest, Hungary
| | - Andrea Varga
- Department of Biophysics and Radiation Biology, Semmelweis University, H-1094 Budapest, Hungary; (É.G.); (K.P.)
| |
Collapse
|
11
|
Suhardi VJ, Oktarina A, Hammad M, Niu Y, Li Q, Thomson A, Lopez J, McCormick J, Ayturk UM, Greenblatt MB, Ivashkiv LB, Bostrom MPG, Yang X. Prevention and treatment of peri-implant fibrosis by functionally inhibiting skeletal cells expressing the leptin receptor. Nat Biomed Eng 2024; 8:1285-1307. [PMID: 39085645 PMCID: PMC12016487 DOI: 10.1038/s41551-024-01238-y] [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: 01/18/2023] [Accepted: 06/25/2024] [Indexed: 08/02/2024]
Abstract
The cellular and molecular mediators of peri-implant fibrosis-a most common reason for implant failure and for surgical revision after the replacement of a prosthetic joint-remain unclear. Here we show that peri-implant fibrotic tissue in mice and humans is largely composed of a specific population of skeletal cells expressing the leptin receptor (LEPR) and that these cells are necessary and sufficient to generate and maintain peri-implant fibrotic tissue. In a mouse model of tibial implantation and osseointegration that mimics partial knee arthroplasty, genetic ablation of LEPR+ cells prevented peri-implant fibrosis and the implantation of LEPR+ cells from peri-implant fibrotic tissue was sufficient to induce fibrosis in secondary hosts. Conditional deletion of the adhesion G-protein-coupled receptor F5 (ADGRF5) in LEPR+ cells attenuated peri-implant fibrosis while augmenting peri-implant bone formation, and ADGRF5 inhibition by the intra-articular or systemic administration of neutralizing anti-ADGRF5 in the mice prevented and reversed peri-implant fibrosis. Pharmaceutical agents that inhibit the ADGRF5 pathway in LEPR+ cells may be used to prevent and treat peri-implant fibrosis.
Collapse
Affiliation(s)
- Vincentius Jeremy Suhardi
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, NY, USA
- Research Institute, Hospital for Special Surgery, New York, NY, USA
| | | | - Mohammed Hammad
- Research Institute, Hospital for Special Surgery, New York, NY, USA
| | - Yingzhen Niu
- Research Institute, Hospital for Special Surgery, New York, NY, USA
- Department of Joint Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| | - Qingdian Li
- Research Institute, Hospital for Special Surgery, New York, NY, USA
- Department of Orthopedics, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Andrew Thomson
- Research Institute, Hospital for Special Surgery, New York, NY, USA
| | - Juan Lopez
- Research Institute, Hospital for Special Surgery, New York, NY, USA
| | - Jason McCormick
- Flow Cytometry Core Facility, Weill Cornell Medicine, New York, NY, USA
| | - Ugur M Ayturk
- Research Institute, Hospital for Special Surgery, New York, NY, USA
- Department of Orthopedic Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Matthew B Greenblatt
- Research Institute, Hospital for Special Surgery, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | - Mathias P G Bostrom
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, NY, USA
- Research Institute, Hospital for Special Surgery, New York, NY, USA
- Department of Orthopedic Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Xu Yang
- Research Institute, Hospital for Special Surgery, New York, NY, USA.
- Department of Orthopedic Surgery, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
12
|
Kimura Y, Nakai Y, Ino Y, Akiyama T, Moriyama K, Ohira T, Saito T, Inaba Y, Kumagai K, Ryo A, Hirano H. Identification of gravity-responsive serum proteins in spaceflight mice using a quantitative proteomic approach with data-independent acquisition mass spectrometry. Proteomics 2024; 24:e2300214. [PMID: 38475964 DOI: 10.1002/pmic.202300214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 03/05/2024] [Accepted: 03/05/2024] [Indexed: 03/14/2024]
Abstract
Physical inactivity associated with gravity unloading, such as microgravity during spaceflight and hindlimb unloading (HU), can cause various physiological changes. In this study, we attempted to identify serum proteins whose levels fluctuated in response to gravity unloading. First, we quantitatively assessed changes in the serum proteome profiles of spaceflight mice using mass spectrometry with data-independent acquisition. The serum levels of several proteins involved in the responses to estrogen and glucocorticoid, blood vessel maturation, osteoblast differentiation, and ossification were changed by microgravity exposure. Furthermore, a collective evaluation of serum proteomic data from spaceflight and HU mice identified 30 serum proteins, including Mmp2, Igfbp2, Tnc, Cdh5, and Pmel, whose levels varied to a similar extent in both gravity unloading models. These changes in serum levels could be involved in the physiological changes induced by gravity unloading. A collective evaluation of serum, femur, and soleus muscle proteome data of spaceflight mice also showed 24 serum proteins, including Igfbp5, Igfbp3, and Postn, whose levels could be associated with biological changes induced by microgravity. This study examined serum proteome profiles in response to gravity unloading, and may help deepen our understanding of microgravity adaptation mechanisms during prolonged spaceflight missions.
Collapse
Affiliation(s)
- Yayoi Kimura
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Yusuke Nakai
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Yoko Ino
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Tomoko Akiyama
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Kayano Moriyama
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Takashi Ohira
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| | | | - Yutaka Inaba
- Department of Orthopaedic Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Ken Kumagai
- Department of Orthopaedic Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Akihide Ryo
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Hisashi Hirano
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| |
Collapse
|
13
|
Yu XH, Wu JB, Fan HY, Dai L, Xian HC, Chen BJ, Liao P, Huang MC, Pang X, Zhang M, Liang XH, Tang YL. Artemisinin suppressed tumour growth and induced vascular normalisation in oral squamous cell carcinoma via inhibition of macrophage migration inhibitory factor. Oral Dis 2024; 30:363-375. [PMID: 36321394 DOI: 10.1111/odi.14418] [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: 06/14/2022] [Revised: 09/05/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Tumour vascular normalisation therapy advocates a balance between pro-angiogenic factors and anti-angiogenic factors in tumours. Artemisinin (ART), which is derived from traditional Chinese medicine, has been shown to inhibit tumour growth; however, the relationship between ART and tumour vascular normalisation in oral squamous cell carcinoma (OSCC) has not been previously reported. METHODS Different concentrations(0 mg/kg, 25 mg/kg, 50 mg/kg, 100 mg/kg)of ART were used to treat the xenograft nude mice model of OSCC. The effects of ART on migration and proliferation of OSCC and human umbilical vein endothelial cells (HUVEC) cells were detected by scratch assay and CCK-8 assay. OSCC cells with macrophage migration inhibitory factor (MIF) silenced were constructed to explore the effect of MIF. RESULTS Treatment with ART inhibited the growth and angiogenesis of OSCC xenografts in nude mice and downregulated vascular endothelial growth factor (VEGF), IL-8, and MIF expression levels. ART reduced the proliferation, migration, and tube formation of HUVEC, as well as the expression of VEGFR1 and VEGFR2. When the dose of ART was 50 mg/kg, vascular normalisation of OSCC xenografts was induced. Moreover, VEGF and IL-8 were needed in rhMIF restoring tumour growth and inhibit vascular normalisation after the addition of rhMIF to ART-treated cells. CONCLUSION Artemisinin might induce vascular normalisation and inhibit tumour growth in OSCC through the MIF-signalling pathway.
Collapse
Affiliation(s)
- Xiang-Hua Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral Pathology, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Jing-Biao Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), Chengdu, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, China
| | - Hua-Yang Fan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Li Dai
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Hong-Chun Xian
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral Pathology, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Bing-Jun Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Peng Liao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Mei-Chang Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral Pathology, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Xin Pang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Mei Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral Pathology, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| |
Collapse
|
14
|
Valdivia A, Avalos AM, Leyton L. Thy-1 (CD90)-regulated cell adhesion and migration of mesenchymal cells: insights into adhesomes, mechanical forces, and signaling pathways. Front Cell Dev Biol 2023; 11:1221306. [PMID: 38099295 PMCID: PMC10720913 DOI: 10.3389/fcell.2023.1221306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 09/25/2023] [Indexed: 12/17/2023] Open
Abstract
Cell adhesion and migration depend on the assembly and disassembly of adhesive structures known as focal adhesions. Cells adhere to the extracellular matrix (ECM) and form these structures via receptors, such as integrins and syndecans, which initiate signal transduction pathways that bridge the ECM to the cytoskeleton, thus governing adhesion and migration processes. Integrins bind to the ECM and soluble or cell surface ligands to form integrin adhesion complexes (IAC), whose composition depends on the cellular context and cell type. Proteomic analyses of these IACs led to the curation of the term adhesome, which is a complex molecular network containing hundreds of proteins involved in signaling, adhesion, and cell movement. One of the hallmarks of these IACs is to sense mechanical cues that arise due to ECM rigidity, as well as the tension exerted by cell-cell interactions, and transduce this force by modifying the actin cytoskeleton to regulate cell migration. Among the integrin/syndecan cell surface ligands, we have described Thy-1 (CD90), a GPI-anchored protein that possesses binding domains for each of these receptors and, upon engaging them, stimulates cell adhesion and migration. In this review, we examine what is currently known about adhesomes, revise how mechanical forces have changed our view on the regulation of cell migration, and, in this context, discuss how we have contributed to the understanding of signaling mechanisms that control cell adhesion and migration.
Collapse
Affiliation(s)
- Alejandra Valdivia
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Ana María Avalos
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
15
|
Maltabe VA, Melidoni AN, Beis D, Kokkinopoulos I, Paschalidis N, Kouklis P. VE-CADHERIN is expressed transiently in early ISL1 + cardiovascular progenitor cells and facilitates cardiac differentiation. Stem Cell Reports 2023; 18:1827-1840. [PMID: 37541259 PMCID: PMC10545488 DOI: 10.1016/j.stemcr.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/07/2023] [Accepted: 07/09/2023] [Indexed: 08/06/2023] Open
Abstract
Adherens junctions (AJs) provide adhesive properties through cadherins and associated cytoplasmic catenins and participate in morphogenetic processes. We examined AJs formed between ISL1+ cardiovascular progenitor cells during differentiation of embryonic stem cells (ESCs) in vitro and in mouse embryogenesis in vivo. We found that, in addition to N-CADHERIN, a percentage of ISL1+ cells transiently formed vascular endothelial (VE)-CADHERIN-mediated AJs during in vitro differentiation on days 4 and 5, and the same pattern was observed in vivo. Fluorescence-activated cell sorting (FACS) analysis extended morphological data showing that VE-CADHERIN+/ISL1+ cells constitute a significant percentage of cardiac progenitors on days 4 and 5. The VE-CADHERIN+/ISL1+ cell population represented one-third of the emerging FLK1+/PDGFRa+ cardiac progenitor cells (CPCs) for a restricted time window (days 4-6). Ablation of VE-CADHERIN during ESC differentiation results in severe inhibition of cardiac differentiation. Disruption of all classic cadherins in the VE-CADHERIN+ population via a cadherin dominant-negative mutant's expression resulted in a dramatic decrease in the ISL1+ population and inhibition of cardiac differentiation.
Collapse
Affiliation(s)
- Violetta A Maltabe
- Laboratory of Biology, Department of Medicine, University of Ioannina, Ioannina, Greece; Division of Biomedical Research, Foundation for Research and Technology, Institute of Molecular Biology and Biotechnology, Ioannina, Greece
| | - Anna N Melidoni
- Laboratory of Biology, Department of Medicine, University of Ioannina, Ioannina, Greece
| | - Dimitris Beis
- Developmental Biology, Center for Experimental Surgery Clinical and Translational Research, Biomedical Research Foundation Academy of Athens (BRFAA), 11527 Athens, Greece; Laboratory of Biochemistry, Department of Medicine, University of Ioannina, Ioannina, Greece
| | - Ioannis Kokkinopoulos
- Developmental Biology and Immunobiology Laboratories, Center for Clinical, Experimental Surgery, and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Nikolaos Paschalidis
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 115 27 Athens, Greece
| | - Panos Kouklis
- Laboratory of Biology, Department of Medicine, University of Ioannina, Ioannina, Greece; Division of Biomedical Research, Foundation for Research and Technology, Institute of Molecular Biology and Biotechnology, Ioannina, Greece.
| |
Collapse
|
16
|
Nan W, He Y, Wang S, Zhang Y. Molecular mechanism of VE-cadherin in regulating endothelial cell behaviour during angiogenesis. Front Physiol 2023; 14:1234104. [PMID: 37601629 PMCID: PMC10433914 DOI: 10.3389/fphys.2023.1234104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 07/18/2023] [Indexed: 08/22/2023] Open
Abstract
Vascular endothelial (VE)-cadherin, an endothelium-specific adhesion protein, is found in the junctions between endothelial cells (ECs). It's crucial to maintain the homogeneity of ECs. Keeping and controlling the contact between ECs is essential. In addition to its adhesive function, VE-cadherin plays important roles in vascular development, permeability, and tumour angiogenesis. Signal transfer, cytoskeletal reconstruction, and contractile integrating, which are crucial for constructing and maintaining monolayer integrity as well as for repair and regeneration, are the foundation of endothelial cell (EC) junctional dynamics. The molecular basis of adhesion junctions (AJs), which are closely related and work with actin filaments, is provided by the VE-cadherin-catenin complex. They can activate intracellular signals that drive ECs to react or communicate structural changes to junctions. An increasing number of molecules, including the vascular endothelial growth factor receptor 2 (VEGFR2) and vascular endothelial protein tyrosine phosphatase (VE-PTP), have been connected to VE-cadherin in addition to the conventional VE-cadherin-catenin complex. This review demonstrates significant progress in our understanding of the molecular mechanisms that affect VE-cadherin's function in the regulation of EC behaviour during angiogenesis. The knowledge of the molecular processes that control VE-cadherin's role in the regulation of EC behaviour during angiogenesis has recently advanced, as shown in this review.
Collapse
Affiliation(s)
- Weijin Nan
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Yuxi He
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Shurong Wang
- Department of Ophthalmology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yan Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
17
|
Saygili Demir C, Sabine A, Gong M, Dormond O, Petrova TV. Mechanosensitive mTORC1 signaling maintains lymphatic valves. J Cell Biol 2023; 222:e202207049. [PMID: 37036444 PMCID: PMC10097975 DOI: 10.1083/jcb.202207049] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 01/26/2023] [Accepted: 03/23/2023] [Indexed: 04/11/2023] Open
Abstract
Homeostatic maintenance and repair of lymphatic vessels are essential for health. We investigated the dynamics and the molecular mechanisms of lymphatic endothelial cell (LEC) renewal in adult mesenteric quiescent lymphatic vasculature using label-retention, lineage tracing, and cell ablation strategies. Unlike during development, adult LEC turnover and proliferation was confined to the valve regions of collecting vessels, with valve cells displaying the shortest lifespan. Proliferating valve sinus LECs were the main source for maintenance and repair of lymphatic valves. We identified mechanistic target of rapamycin complex 1 (mTORC1) as a mechanoresponsive pathway activated by fluid shear stress in LECs. Depending on the shear stress level, mTORC1 activity drives division of valve cells or dictates their mechanic resilience through increased protein synthesis. Overactivation of lymphatic mTORC1 in vivo promoted supernumerary valve formation. Our work provides insights into the molecular mechanisms of maintenance of healthy lymphatic vascular system.
Collapse
Affiliation(s)
- Cansaran Saygili Demir
- Department of Oncology, Lausanne University Hospital-University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Amélie Sabine
- Department of Oncology, Lausanne University Hospital-University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Muyun Gong
- Department of Oncology, Lausanne University Hospital-University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Olivier Dormond
- Department of Visceral Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Tatiana V. Petrova
- Department of Oncology, Lausanne University Hospital-University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
- Swiss Institute for Experimental Cancer Research, École polytechnique fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
18
|
Aitken C, Mehta V, Schwartz MA, Tzima E. Mechanisms of endothelial flow sensing. NATURE CARDIOVASCULAR RESEARCH 2023; 2:517-529. [PMID: 39195881 DOI: 10.1038/s44161-023-00276-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 04/14/2023] [Indexed: 08/29/2024]
Abstract
Fluid shear stress plays a key role in sculpting blood vessels during development, in adult vascular homeostasis and in vascular pathologies. During evolution, endothelial cells evolved several mechanosensors that convert physical forces into biochemical signals, a process termed mechanotransduction. This Review discusses our understanding of endothelial flow sensing and suggests important questions for future investigation.
Collapse
Affiliation(s)
- Claire Aitken
- Wellcome Centre for Human Genetics, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Vedanta Mehta
- Wellcome Centre for Human Genetics, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, and Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT, USA.
| | - Ellie Tzima
- Wellcome Centre for Human Genetics, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
19
|
Lin WH, Cooper LM, Anastasiadis PZ. Cadherins and catenins in cancer: connecting cancer pathways and tumor microenvironment. Front Cell Dev Biol 2023; 11:1137013. [PMID: 37255594 PMCID: PMC10225604 DOI: 10.3389/fcell.2023.1137013] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/03/2023] [Indexed: 06/01/2023] Open
Abstract
Cadherin-catenin complexes are integral components of the adherens junctions crucial for cell-cell adhesion and tissue homeostasis. Dysregulation of these complexes is linked to cancer development via alteration of cell-autonomous oncogenic signaling pathways and extrinsic tumor microenvironment. Advances in multiomics have uncovered key signaling events in multiple cancer types, creating a need for a better understanding of the crosstalk between cadherin-catenin complexes and oncogenic pathways. In this review, we focus on the biological functions of classical cadherins and associated catenins, describe how their dysregulation influences major cancer pathways, and discuss feedback regulation mechanisms between cadherin complexes and cellular signaling. We discuss evidence of cross regulation in the following contexts: Hippo-Yap/Taz and receptor tyrosine kinase signaling, key pathways involved in cell proliferation and growth; Wnt, Notch, and hedgehog signaling, key developmental pathways involved in human cancer; as well as TGFβ and the epithelial-to-mesenchymal transition program, an important process for cancer cell plasticity. Moreover, we briefly explore the role of cadherins and catenins in mechanotransduction and the immune tumor microenvironment.
Collapse
|
20
|
Ryltseva GA, Dudaev AE, Menzyanova NG, Volova TG, Alexandrushkina NA, Efimenko AY, Shishatskaya EI. Influence of PHA Substrate Surface Characteristics on the Functional State of Endothelial Cells. J Funct Biomater 2023; 14:jfb14020085. [PMID: 36826884 PMCID: PMC9959859 DOI: 10.3390/jfb14020085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/24/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
The needs of modern regenerative medicine for biodegradable polymers are wide and varied. Restoration of the viability of the vascular tree is one of the most important components of the preservation of the usefulness of organs and tissues. The creation of vascular implants compatible with blood is an important task of vascular bioengineering. The function of the endothelial layer of the vessel, being largely responsible for the development of thrombotic complications, is of great importance for hemocompatibility. The development of surfaces with specific characteristics of biomaterials that are used in vascular technologies is one of the solutions for their correct endothelialization. Linear polyhydroxyalkanoates (PHAs) are biodegradable structural polymeric materials suitable for obtaining various types of implants and tissue engineering, having a wide range of structural and physicomechanical properties. The use of PHA of various monomeric compositions in endothelial cultivation makes it possible to evaluate the influence of material properties, especially surface characteristics, on the functional state of cells. It has been established that PHA samples with the inclusion of 3-hydroxyhexanoate have optimal characteristics for the formation of a human umbilical vein endothelial cell, HUVEC, monolayer in terms of cell morphology as well as the levels of expression of vinculin and VE-cadherin. The obtained results provide a rationale for the use of PHA copolymers as materials for direct contact with the endothelium in vascular implants.
Collapse
Affiliation(s)
- Galina A. Ryltseva
- Department of Medical Biology, School of Fundamental Biology and Biotechnology, Siberian Federal University, 79 Svobodnyi Av., 660041 Krasnoyarsk, Russia
- Correspondence: (G.A.R.); (E.I.S.)
| | - Alexey E. Dudaev
- Department of Medical Biology, School of Fundamental Biology and Biotechnology, Siberian Federal University, 79 Svobodnyi Av., 660041 Krasnoyarsk, Russia
- Institute of Biophysics SB RAS, Federal Research Center “Krasnoyarsk Science Center SB RAS”, 50/50 Akademgorodok, 660036 Krasnoyarsk, Russia
| | - Natalia G. Menzyanova
- Department of Medical Biology, School of Fundamental Biology and Biotechnology, Siberian Federal University, 79 Svobodnyi Av., 660041 Krasnoyarsk, Russia
| | - Tatiana G. Volova
- Institute of Biophysics SB RAS, Federal Research Center “Krasnoyarsk Science Center SB RAS”, 50/50 Akademgorodok, 660036 Krasnoyarsk, Russia
- Basic Department of Biotechnology, School of Fundamental Biology and Biotechnology, Siberian Federal University, 79 Svobodnyi Av., 660041 Krasnoyarsk, Russia
| | - Natalia A. Alexandrushkina
- Institute for Regenerative Medicine, Medical Research and Education Center, M.V. Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Anastasia Yu. Efimenko
- Institute for Regenerative Medicine, Medical Research and Education Center, M.V. Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Ekaterina I. Shishatskaya
- Department of Medical Biology, School of Fundamental Biology and Biotechnology, Siberian Federal University, 79 Svobodnyi Av., 660041 Krasnoyarsk, Russia
- Institute of Biophysics SB RAS, Federal Research Center “Krasnoyarsk Science Center SB RAS”, 50/50 Akademgorodok, 660036 Krasnoyarsk, Russia
- Correspondence: (G.A.R.); (E.I.S.)
| |
Collapse
|
21
|
Rosenbohm J, Minnick G, Safa BT, Esfahani AM, Jin X, Zhai H, Lavrik NV, Yang R. A multi-material platform for imaging of single cell-cell junctions under tensile load fabricated with two-photon polymerization. Biomed Microdevices 2022; 24:33. [PMID: 36207557 PMCID: PMC11104271 DOI: 10.1007/s10544-022-00633-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] [Accepted: 09/04/2022] [Indexed: 11/29/2022]
Abstract
We previously reported a single-cell adhesion micro tensile tester (SCAμTT) fabricated from IP-S photoresin with two-photon polymerization (TPP) for investigating the mechanics of a single cell-cell junction under defined tensile loading. A major limitation of the platform is the autofluorescence of IP-S, the photoresin for TPP fabrication, which significantly increases background signal and makes fluorescent imaging of stretched cells difficult. In this study, we report the design and fabrication of a new SCAμTT platform that mitigates autofluorescence and demonstrate its capability in imaging a single cell pair as its mutual junction is stretched. By employing a two-material design using IP-S and IP-Visio, a photoresin with reduced autofluorescence, we show a significant reduction in autofluorescence of the platform. Further, by integrating apertures onto the substrate with a gold coating, the influence of autofluorescence on imaging is almost completely mitigated. With this new platform, we demonstrate the ability to image a pair of epithelial cells as they are stretched up to 250% strain, allowing us to observe junction rupture and F-actin retraction while simultaneously recording the accumulation of over 800 kPa of stress in the junction. The platform and methodology presented here can potentially enable detailed investigation of the mechanics of and mechanotransduction in cell-cell junctions and improve the design of other TPP platforms in mechanobiology applications.
Collapse
Affiliation(s)
- Jordan Rosenbohm
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Grayson Minnick
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Bahareh Tajvidi Safa
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Amir Monemian Esfahani
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Xiaowei Jin
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Haiwei Zhai
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Nickolay V Lavrik
- Center for Nanophase Materials Sciences, Oak Ridge National Laboratory, Oak Ridge, TN, 37831-6054, USA.
| | - Ruiguo Yang
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA.
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA.
| |
Collapse
|
22
|
Kong X, Kapustka A, Sullivan B, Schwarz GJ, Leckband DE. Extracellular matrix regulates force transduction at VE-cadherin junctions. Mol Biol Cell 2022; 33:ar95. [PMID: 35653290 DOI: 10.1091/mbc.e22-03-0075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Increased tension on VE-cadherin (VE-cad) complexes activates adaptive cell stiffening and local cytoskeletal reinforcement--two key signatures of intercellular mechanotransduction. Here we demonstrate that tugging on VE-cad receptors initiates a cascade that results in downstream integrin activation. The formation of new integrin adhesions potentiates vinculin and actin recruitment to mechanically reinforce stressed cadherin adhesions. This cascade differs from documented antagonistic effects of integrins on intercellular junctions. We identify focal adhesion kinase, Abl kinase, and RhoA GTPase as key components of the positive feedback loop. Results further show that a consequence of integrin involvement is the sensitization of intercellular force transduction to the extracellular matrix (ECM) not by regulating junctional tension but by altering signal cascades that reinforce cell-cell adhesions. On type 1 collagen or fibronectin substrates, integrin subtypes α2β1 and α5β1, respectively, differentially control actin remodeling at VE-cad adhesions. Specifically, ECM-dependent differences in VE-cad force transduction mirror differences in the rigidity sensing mechanisms of α2β1 and α5β1 integrins. The findings verify the role of integrins in VE-cad force transduction and uncover a previously unappreciated mechanism by which the ECM impacts the mechanical reinforcement of interendothelial junctions.
Collapse
Affiliation(s)
- Xinyu Kong
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Adrian Kapustka
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Brendan Sullivan
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Gregory J Schwarz
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Deborah E Leckband
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801.,Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801.,Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801.,Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| |
Collapse
|
23
|
Sri-Ranjan K, Sanchez-Alonso JL, Swiatlowska P, Rothery S, Novak P, Gerlach S, Koeninger D, Hoffmann B, Merkel R, Stevens MM, Sun SX, Gorelik J, Braga VMM. Intrinsic cell rheology drives junction maturation. Nat Commun 2022; 13:4832. [PMID: 35977954 PMCID: PMC9385638 DOI: 10.1038/s41467-022-32102-9] [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: 07/27/2021] [Accepted: 07/15/2022] [Indexed: 12/02/2022] Open
Abstract
A fundamental property of higher eukaryotes that underpins their evolutionary success is stable cell-cell cohesion. Yet, how intrinsic cell rheology and stiffness contributes to junction stabilization and maturation is poorly understood. We demonstrate that localized modulation of cell rheology governs the transition of a slack, undulated cell-cell contact (weak adhesion) to a mature, straight junction (optimal adhesion). Cell pairs confined on different geometries have heterogeneous elasticity maps and control their own intrinsic rheology co-ordinately. More compliant cell pairs grown on circles have slack contacts, while stiffer triangular cell pairs favour straight junctions with flanking contractile thin bundles. Counter-intuitively, straighter cell-cell contacts have reduced receptor density and less dynamic junctional actin, suggesting an unusual adaptive mechano-response to stabilize cell-cell adhesion. Our modelling informs that slack junctions arise from failure of circular cell pairs to increase their own intrinsic stiffness and resist the pressures from the neighbouring cell. The inability to form a straight junction can be reversed by increasing mechanical stress artificially on stiffer substrates. Our data inform on the minimal intrinsic rheology to generate a mature junction and provide a springboard towards understanding elements governing tissue-level mechanics.
Collapse
Affiliation(s)
- K Sri-Ranjan
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - J L Sanchez-Alonso
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - P Swiatlowska
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - S Rothery
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - P Novak
- School of Engineering and Materials Science, Queen Mary University, London, UK
| | - S Gerlach
- Institute of Biological Information Processing, IBI-2: Mechanobiology, Julich, Germany
| | - D Koeninger
- Institute of Biological Information Processing, IBI-2: Mechanobiology, Julich, Germany
| | - B Hoffmann
- Institute of Biological Information Processing, IBI-2: Mechanobiology, Julich, Germany
| | - R Merkel
- Institute of Biological Information Processing, IBI-2: Mechanobiology, Julich, Germany
| | - M M Stevens
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering Imperial College London, London, UK
| | - S X Sun
- Department of Mechanical Engineering and Institute of NanoBioTechnology, Johns Hopkins University, Baltimore Maryland, USA
| | - J Gorelik
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK.
| | - Vania M M Braga
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
24
|
Canales Coutiño B, Mayor R. Neural crest mechanosensors: Seeing old proteins in a new light. Dev Cell 2022; 57:1792-1801. [PMID: 35901790 DOI: 10.1016/j.devcel.2022.07.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/26/2022] [Accepted: 07/05/2022] [Indexed: 11/25/2022]
Abstract
Mechanical forces exerted on neural crest cells control their collective migration and differentiation. This perspective discusses our current understanding of neural crest mechanotransduction during cell migration and differentiation. Additionally, we describe proteins that have mechanosensitive functions in other systems, such as mechanosensitive G-protein-coupled receptors, mechanosensitive ion channels, cell-cell adhesion, and cell-matrix-interacting proteins, and highlight that these same proteins have in the past been studied in neural crest development from a purely signaling point of view. We propose that future studies elucidate the mechanosensitive functions these receptors may play in neural crest development and integrate this with their known molecular role.
Collapse
Affiliation(s)
- Brenda Canales Coutiño
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
25
|
Adherens junctions stimulate and spatially guide integrin activation and extracellular matrix deposition. Cell Rep 2022; 40:111091. [PMID: 35858563 DOI: 10.1016/j.celrep.2022.111091] [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: 11/30/2021] [Revised: 05/04/2022] [Accepted: 06/22/2022] [Indexed: 11/03/2022] Open
Abstract
Cadherins and integrins are intrinsically linked through the actin cytoskeleton and are largely responsible for the mechanical integrity and organization of tissues. We show that cadherin clustering stimulates and spatially guides integrin activation. Adherens junction (AJ)-associated integrin activation depends on locally generated tension and does not require extracellular matrix ligands. It leads to the creation of primed integrin clusters, which spatially determine where focal adhesions will form if ligands are present and where ligands will be deposited. AJs that display integrin activation are targeted by microtubules facilitating their disassembly via caveolin-based endocytosis, showing that integrin activation impacts the stability of the core cadherin complex. Thus, the interplay between cadherins and integrins is more intimate than what was once believed and is rooted in the capacity of active integrins to be stabilized via AJ-generated tension. Altogether, our data establish a mechanism of cross-regulation between cadherins and integrins.
Collapse
|
26
|
Silvani G, Bradbury P, Basirun C, Mehner C, Zalli D, Poole K, Chou J. Testing 3D printed biological platform for advancing simulated microgravity and space mechanobiology research. NPJ Microgravity 2022; 8:19. [PMID: 35662260 PMCID: PMC9166742 DOI: 10.1038/s41526-022-00207-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 05/13/2022] [Indexed: 12/02/2022] Open
Abstract
The advancement of microgravity simulators is helping many researchers better understanding the impact of the mechanically unloaded space environment on cellular function and disfunction. However, performing microgravity experiments on Earth, using simulators such as the Random Positioning Machine, introduces some unique practical challenges, including air bubble formation and leakage of growth medium from tissue culture flask and plates, all of which limit research progress. Here, we developed an easy-to-use hybrid biological platform designed with the precision of 3D printing technologies combined with PDMS microfluidic fabrication processes to facilitate reliable and reproducible microgravity cellular experiments. The system has been characterized for applications in the contest of brain cancer research by exposing glioblastoma and endothelial cells to 24 h of simulated microgravity condition to investigate the triggered mechanosensing pathways involved in cellular adaptation to the new environment. The platform demonstrated compatibility with different biological assays, i.e., proliferation, viability, morphology, protein expression and imaging of molecular structures, showing advantages over the conventional usage of culture flask. Our results indicated that both cell types are susceptible when the gravitational vector is disrupted, confirming the impact that microgravity has on both cancer and healthy cells functionality. In particular, we observed deactivation of Yap-1 molecule in glioblastoma cells and the remodeling of VE-Cadherin junctional protein in endothelial cells. The study provides support for the application of the proposed biological platform for advancing space mechanobiology research, also highlighting perspectives and strategies for developing next generation of brain cancer molecular therapies, including targeted drug delivery strategies.
Collapse
Affiliation(s)
- Giulia Silvani
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia
| | - Peta Bradbury
- Institut Curie, Paris Sciences et Lettres Research University, Mechanics and Genetics of Embryonic and Tumoral Development Group, Paris, France
| | - Carin Basirun
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia
| | - Christine Mehner
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, USA
| | - Detina Zalli
- Institute of Continuing Education, University of Cambridge, Camridge, UK
| | - Kate Poole
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, Faculty of Medicine & Health, Sydney, NSW, Australia
| | - Joshua Chou
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia.
| |
Collapse
|
27
|
Small molecule compound M12 reduces vascular permeability in obese mice via blocking endothelial TRPV4-Nox2 interaction. Acta Pharmacol Sin 2022; 43:1430-1440. [PMID: 34654876 PMCID: PMC9160247 DOI: 10.1038/s41401-021-00780-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 09/17/2021] [Indexed: 02/07/2023]
Abstract
Transient receptor potential channel TRPV4 and nicotinamide adenine dinucleotide phosphate oxidase (Nox2) are involved in oxidative stress that increases endothelial permeability. It has been shown that obesity enhances the physical association of TRPV4 and Nox2, but the role of TRPV4-Nox2 association in obesity has not been clarified. In this study we investigated the function of TRPV4-Nox2 complex in reducing oxidative stress and regulating abnormal vascular permeability in obesity. Obesity was induced in mice by feeding a high-fat diet (HFD) for 14 weeks. The physical interaction between TRPV4 and Nox2 was measured using FRET, co-immunoprecipitation and GST pull-down assays. The functional interaction was measured by rhodamine phalloidin, CM-H2DCFDA in vitro, the fluorescent dye dihydroethidium (DHE) staining assay, and the Evans blue permeability assay in vivo. We demonstrated that TRPV4 physically and functionally associated with Nox2, and this physical association was enhanced in aorta of obese mice. Furthermore, we showed that interrupting TRPV4-Nox2 coupling by TRPV4 knockout, or by treatment with a specific Nox2 inhibitor Nox2 dstat or a specific TRPV4 inhibitor HC067046 significantly attenuated obesity-induced ROS overproduction in aortic endothelial cells, and reversed the abnormal endothelial cytoskeletal structure. In order to discover small molecules disrupting the over-coupling of TPRV4 and Nox2 in obesity, we performed molecular docking analysis and found that compound M12 modulated TRPV4-Nox2 association, reduced ROS production, and finally reversed disruption of the vascular barrier in obesity. Together, this study, for the first time, provides evidence for the TRPV4 physically interacting with Nox2. TRPV4-Nox2 complex is a potential drug target in improving oxidative stress and disruption of the vascular barrier in obesity. Compound M12 targeting TRPV4-Nox2 complex can improve vascular barrier function in obesity.
Collapse
|
28
|
Kotini MP, van der Stoel MM, Yin J, Han MK, Kirchmaier B, de Rooij J, Affolter M, Huveneers S, Belting HG. Vinculin controls endothelial cell junction dynamics during vascular lumen formation. Cell Rep 2022; 39:110658. [PMID: 35417696 DOI: 10.1016/j.celrep.2022.110658] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 02/01/2022] [Accepted: 03/20/2022] [Indexed: 12/20/2022] Open
Abstract
Blood vessel morphogenesis is driven by coordinated endothelial cell behaviors. Active remodeling of cell-cell junctions promotes cellular plasticity while preserving vascular integrity. Here, we analyze the dynamics of endothelial adherens junctions during lumen formation in angiogenic sprouts in vivo. Live imaging in zebrafish reveals that lumen expansion is accompanied by the formation of transient finger-shaped junctions. Junctional fingers are positively regulated by blood pressure, whereas flow inhibition prevents their formation. Using fluorescent reporters, we show that junctional fingers contain the mechanotransduction protein vinculin. Furthermore, genetic deletion of vinculin prevents finger formation, a junctional defect that could be rescued by transient endothelial expression of vinculin. Our findings suggest a mechanism whereby lumen expansion leads to an increase in junctional tension, triggering recruitment of vinculin and formation of junctional fingers. We propose that endothelial cells employ force-dependent junctional remodeling to counteract external forces in order to maintain vascular integrity during sprouting angiogenesis.
Collapse
Affiliation(s)
- Maria P Kotini
- Biozentrum der Universität Basel, Spitalstrasse 41, 4056 Basel, Switzerland
| | - Miesje M van der Stoel
- Amsterdam UMC, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Jianmin Yin
- Biozentrum der Universität Basel, Spitalstrasse 41, 4056 Basel, Switzerland
| | - Mitchell K Han
- Department of Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Stratenum 3.231, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Bettina Kirchmaier
- Institute for Cell Biology and Neuroscience, Biologicum/Campus Riedberg, Goethe-Universität Frankfurt Am Main, 60438 Frankfurt Am Main, Germany
| | - Johan de Rooij
- Department of Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Stratenum 3.231, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Markus Affolter
- Biozentrum der Universität Basel, Spitalstrasse 41, 4056 Basel, Switzerland
| | - Stephan Huveneers
- Amsterdam UMC, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands.
| | - Heinz-Georg Belting
- Biozentrum der Universität Basel, Spitalstrasse 41, 4056 Basel, Switzerland.
| |
Collapse
|
29
|
Yao Q, Ke HJ, Yang Q, Liao GY, Liu P. Study on the Mechanism of MicroRNA551b-5p in Severe Acute Pancreatitis Capillary Leakage Syndrome. DISEASE MARKERS 2022; 2022:6373757. [PMID: 35256892 PMCID: PMC8898106 DOI: 10.1155/2022/6373757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/26/2022] [Accepted: 01/31/2022] [Indexed: 11/30/2022]
Abstract
Objective This study focused on investigating the effects of microRNA551b-5p (miR-551b-5p) on severe acute pancreatitis. Methods Initially, quantitative real-time polymerase chain reaction (qPCR) is employed to determine the expression of miR-551b-5p in differentiated human umbilical vein endothelial cells (HUVECs). Further, the effects of aberrantly expressed miR-551b-5p in HUVECs Transwell assay. The expressions of proteins associated with severe acute pancreatitis capillary leakage syndrome are determined by Western blot, FITC-phalloidin, and immunofluorescence stainings. Finally, the correlative factor and the target genes of miR-551b-5p, as well as their contributions, are assessed. Results We observed that overexpression of miR-551b-5p distinctly promoted the expression of EGFR, AKT3, and AQP5, while it suppressed the expression of JAM3, AQP1, and occludin. Functionally, the cytoskeleton of the miR-551b-5p overexpression was relatively loose with apparent vacuoles, and overexpression of miR-551b-5p increased the permeability of HUVECs. Conclusion miR-551b-5p overexpression promoted changes in vascular endothelial permeability via upregulation of the EGFR/AKT3 pathway and downregulation of occludin and JAM3.
Collapse
Affiliation(s)
- Qian Yao
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Hua-Jing Ke
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qin Yang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Gen-You Liao
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Pi Liu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
30
|
Keen AN, Payne LA, Mehta V, Rice A, Simpson LJ, Pang KL, del Rio Hernandez A, Reader JS, Tzima E. Eukaryotic initiation factor 6 regulates mechanical responses in endothelial cells. J Cell Biol 2022; 221:e202005213. [PMID: 35024764 PMCID: PMC8763864 DOI: 10.1083/jcb.202005213] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 10/11/2021] [Accepted: 12/08/2021] [Indexed: 12/22/2022] Open
Abstract
The repertoire of extratranslational functions of components of the protein synthesis apparatus is expanding to include control of key cell signaling networks. However, very little is known about noncanonical functions of members of the protein synthesis machinery in regulating cellular mechanics. We demonstrate that the eukaryotic initiation factor 6 (eIF6) modulates cellular mechanobiology. eIF6-depleted endothelial cells, under basal conditions, exhibit unchanged nascent protein synthesis, polysome profiles, and cytoskeleton protein expression, with minimal effects on ribosomal biogenesis. In contrast, using traction force and atomic force microscopy, we show that loss of eIF6 leads to reduced stiffness and force generation accompanied by cytoskeletal and focal adhesion defects. Mechanistically, we show that eIF6 is required for the correct spatial mechanoactivation of ERK1/2 via stabilization of an eIF6-RACK1-ERK1/2-FAK mechanocomplex, which is necessary for force-induced remodeling. These results reveal an extratranslational function for eIF6 and a novel paradigm for how mechanotransduction, the cellular cytoskeleton, and protein translation constituents are linked.
Collapse
Affiliation(s)
- Adam N. Keen
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Luke A. Payne
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Vedanta Mehta
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Alistair Rice
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, UK
| | - Lisa J. Simpson
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Kar Lai Pang
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Armando del Rio Hernandez
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, UK
| | - John S. Reader
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Ellie Tzima
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
31
|
Tefft JB, Bays JL, Lammers A, Kim S, Eyckmans J, Chen CS. Notch1 and Notch3 coordinate for pericyte-induced stabilization of vasculature. Am J Physiol Cell Physiol 2022; 322:C185-C196. [PMID: 34878922 PMCID: PMC8791789 DOI: 10.1152/ajpcell.00320.2021] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The Notch pathway regulates complex patterning events in many species and is critical for the proper formation and function of the vasculature. Despite this importance, how the various components of the Notch pathway work in concert is still not well understood. For example, NOTCH1 stabilizes homotypic endothelial junctions, but the role of NOTCH1 in heterotypic interactions is not entirely clear. NOTCH3, on the other hand, is essential for heterotypic interactions of pericytes with the endothelium, but how NOTCH3 signaling in pericytes impacts the endothelium remains elusive. Here, we use in vitro vascular models to investigate whether pericyte-induced stabilization of the vasculature requires the cooperation of NOTCH1 and NOTCH3. We observe that both pericyte NOTCH3 and endothelial NOTCH1 are required for the stabilization of the endothelium. Loss of either NOTCH3 or NOTCH1 decreases the accumulation of VE-cadherin at endothelial adherens junctions and increases the frequency of wider, more motile junctions. We found that DLL4 was the key ligand for simulating NOTCH1 activation in endothelial cells and observed that DLL4 expression in pericytes is dependent on NOTCH3. Altogether, these data suggest that an interplay between pericyte NOTCH3 and endothelial NOTCH1 is critical for pericyte-induced vascular stabilization.
Collapse
Affiliation(s)
- Juliann B. Tefft
- 1The Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Jennifer L. Bays
- 1The Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, Massachusetts,2The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts
| | - Alex Lammers
- 1The Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, Massachusetts,2The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts
| | - Sudong Kim
- 1The Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, Massachusetts,2The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts
| | - Jeroen Eyckmans
- 1The Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, Massachusetts,2The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts
| | - Christopher S. Chen
- 1The Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, Massachusetts,2The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts
| |
Collapse
|
32
|
Sullivan B, Light T, Vu V, Kapustka A, Hristova K, Leckband D. Mechanical disruption of E-cadherin complexes with epidermal growth factor receptor actuates growth factor-dependent signaling. Proc Natl Acad Sci U S A 2022; 119:e2100679119. [PMID: 35074920 PMCID: PMC8794882 DOI: 10.1073/pnas.2100679119] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 12/10/2021] [Indexed: 12/21/2022] Open
Abstract
Increased intercellular tension is associated with enhanced cell proliferation and tissue growth. Here, we present evidence for a force-transduction mechanism that links mechanical perturbations of epithelial (E)-cadherin (CDH1) receptors to the force-dependent activation of epidermal growth factor receptor (EGFR, ERBB1)-a key regulator of cell proliferation. Here, coimmunoprecipitation studies first show that E-cadherin and EGFR form complexes at the plasma membrane that are disrupted by either epidermal growth factor (EGF) or increased tension on homophilic E-cadherin bonds. Although force on E-cadherin bonds disrupts the complex in the absence of EGF, soluble EGF is required to mechanically activate EGFR at cadherin adhesions. Fully quantified spectral imaging fluorescence resonance energy transfer further revealed that E-cadherin and EGFR directly associate to form a heterotrimeric complex of two cadherins and one EGFR protein. Together, these results support a model in which the tugging forces on homophilic E-cadherin bonds trigger force-activated signaling by releasing EGFR monomers to dimerize, bind EGF ligand, and signal. These findings reveal the initial steps in E-cadherin-mediated force transduction that directly link intercellular force fluctuations to the activation of growth regulatory signaling cascades.
Collapse
Affiliation(s)
- Brendan Sullivan
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Taylor Light
- Department of Materials Science and Engineering, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218
| | - Vinh Vu
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Adrian Kapustka
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Kalina Hristova
- Department of Materials Science and Engineering, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218;
| | - Deborah Leckband
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801;
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Center for Quantitative Biology and Biophysics, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| |
Collapse
|
33
|
Brandt MM, Cheng C, Merkus D, Duncker DJ, Sorop O. Mechanobiology of Microvascular Function and Structure in Health and Disease: Focus on the Coronary Circulation. Front Physiol 2022; 12:771960. [PMID: 35002759 PMCID: PMC8733629 DOI: 10.3389/fphys.2021.771960] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/11/2021] [Indexed: 12/19/2022] Open
Abstract
The coronary microvasculature plays a key role in regulating the tight coupling between myocardial perfusion and myocardial oxygen demand across a wide range of cardiac activity. Short-term regulation of coronary blood flow in response to metabolic stimuli is achieved via adjustment of vascular diameter in different segments of the microvasculature in conjunction with mechanical forces eliciting myogenic and flow-mediated vasodilation. In contrast, chronic adjustments in flow regulation also involve microvascular structural modifications, termed remodeling. Vascular remodeling encompasses changes in microvascular diameter and/or density being largely modulated by mechanical forces acting on the endothelium and vascular smooth muscle cells. Whereas in recent years, substantial knowledge has been gathered regarding the molecular mechanisms controlling microvascular tone and how these are altered in various diseases, the structural adaptations in response to pathologic situations are less well understood. In this article, we review the factors involved in coronary microvascular functional and structural alterations in obstructive and non-obstructive coronary artery disease and the molecular mechanisms involved therein with a focus on mechanobiology. Cardiovascular risk factors including metabolic dysregulation, hypercholesterolemia, hypertension and aging have been shown to induce microvascular (endothelial) dysfunction and vascular remodeling. Additionally, alterations in biomechanical forces produced by a coronary artery stenosis are associated with microvascular functional and structural alterations. Future studies should be directed at further unraveling the mechanisms underlying the coronary microvascular functional and structural alterations in disease; a deeper understanding of these mechanisms is critical for the identification of potential new targets for the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Maarten M Brandt
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Caroline Cheng
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Walter Brendel Center of Experimental Medicine (WBex), LMU Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Oana Sorop
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
34
|
Sunderland K, Jiang J, Zhao F. Disturbed flow's impact on cellular changes indicative of vascular aneurysm initiation, expansion, and rupture: A pathological and methodological review. J Cell Physiol 2022; 237:278-300. [PMID: 34486114 PMCID: PMC8810685 DOI: 10.1002/jcp.30569] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/06/2021] [Accepted: 08/16/2021] [Indexed: 01/03/2023]
Abstract
Aneurysms are malformations within the arterial vasculature brought on by the structural breakdown of the microarchitecture of the vessel wall, with aneurysms posing serious health risks in the event of their rupture. Blood flow within vessels is generally laminar with high, unidirectional wall shear stressors that modulate vascular endothelial cell functionality and regulate vascular smooth muscle cells. However, altered vascular geometry induced by bifurcations, significant curvature, stenosis, or clinical interventions can alter the flow, generating low stressor disturbed flow patterns. Disturbed flow is associated with altered cellular morphology, upregulated expression of proteins modulating inflammation, decreased regulation of vascular permeability, degraded extracellular matrix, and heightened cellular apoptosis. The understanding of the effects disturbed flow has on the cellular cascades which initiate aneurysms and promote their subsequent growth can further elucidate the nature of this complex pathology. This review summarizes the current knowledge about the disturbed flow and its relation to aneurysm pathology, the methods used to investigate these relations, as well as how such knowledge has impacted clinical treatment methodologies. This information can contribute to the understanding of the development, growth, and rupture of aneurysms and help develop novel research and aneurysmal treatment techniques.
Collapse
Affiliation(s)
- Kevin Sunderland
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931
| | - Jingfeng Jiang
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931,Corresponding Authors: Feng Zhao, 101 Bizzell Street, College Station, TX 77843-312, Tel : 979-458-1239, , Jingfeng Jiang, 1400 Townsend Dr., Houghton, MI 49931, Tel: 906-487-1943
| | - Feng Zhao
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843,Corresponding Authors: Feng Zhao, 101 Bizzell Street, College Station, TX 77843-312, Tel : 979-458-1239, , Jingfeng Jiang, 1400 Townsend Dr., Houghton, MI 49931, Tel: 906-487-1943
| |
Collapse
|
35
|
Dynamics of the Actin Cytoskeleton at Adhesion Complexes. BIOLOGY 2021; 11:biology11010052. [PMID: 35053050 PMCID: PMC8773209 DOI: 10.3390/biology11010052] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 01/06/2023]
Abstract
The shape of cells is altered to allow cells to adapt to their changing environments, including responding to internally generated and externally applied force. Force is sensed by cell surface adhesion proteins that are enriched in sites where cells bind to the extracellular matrix (focal adhesions) and neighboring cells (cell-cell or adherens junctions). Receptors at these adhesion sites stimulate intracellular signal transduction cascades that culminate in dramatic changes in the actin cytoskeleton. New actin filaments form, and/or new and existing filaments can be cleaved, branched, or bundled. Here, we discuss the actin cytoskeleton and its functions. We will examine the current understanding for how the actin cytoskeleton is tethered to adhesion sites. Finally, we will highlight recent studies describing how the actin cytoskeleton at these adhesion sites is remodeled in response to force.
Collapse
|
36
|
Multifaceted Pathomolecular Mechanism of a VWF Large Deletion Involved in the Pathogenesis of Severe VWD. Blood Adv 2021; 6:1038-1053. [PMID: 34861678 PMCID: PMC8945295 DOI: 10.1182/bloodadvances.2021005895] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/21/2021] [Indexed: 11/20/2022] Open
Abstract
The present study demonstrates the dominant-negative impact of an in-frame large deletion on VWF biosynthesis and biogenesis of the WPBs. The malformed WPBs/altered trafficking of its inflammatory cargos cause distresses in endothelial cell signaling pathways and phenotype.
An in-frame heterozygous large deletion of exons 4 through 34 of the von Willebrand factor (VWF) gene was identified in a type 3 von Willebrand disease (VWD) index patient (IP), as the only VWF variant. The IP exhibited severe bleeding episodes despite prophylaxis treatment, with a short VWF half-life after infusion of VWF/factor VIII concentrates. Transcript analysis confirmed transcription of normal VWF messenger RNA besides an aberrant deleted transcript. The IP endothelial colony-forming cells (ECFCs) exhibited a defect in the VWF multimers and Weibel-Palade bodies (WPBs) biogenesis, although demonstrating normal VWF secretion compared with healthy cells. Immunostaining of IP-ECFCs revealed subcellular mislocalization of WPBs pro-inflammatory cargos angiopoietin-2 (Ang2, nuclear accumulation) and P-selectin. Besides, the RNA-sequencing (RNA-seq) analysis showed upregulation of pro-inflammatory and proangiogenic genes, P-selectin, interleukin 8 (IL-8), IL-6, and GROα, copackaged with VWF into WPBs. Further, whole-transcriptome RNA-seq and subsequent gene ontology (GO) enrichment analysis indicated the most enriched GO-biological process terms among the differentially expressed genes in IP-ECFCs were regulation of cell differentiation, cell adhesion, leukocyte adhesion to vascular endothelial, blood vessel morphogenesis, and angiogenesis, which resemble downstream signaling pathways associated with inflammatory stimuli and Ang2 priming. Accordingly, our functional experiments exhibited an increased endothelial cell adhesiveness and interruption in endothelial cell–cell junctions of the IP-ECFCs. In conclusion, the deleted VWF has a dominant-negative impact on multimer assembly and the biogenesis of WPBs, leading to altered trafficking of their pro-inflammatory cargos uniquely, which, in turn, causes changes in cellular signaling pathways, phenotype, and function of the endothelial cells.
Collapse
|
37
|
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
|
38
|
Bornhorst D, Abdelilah-Seyfried S. Strong as a Hippo's Heart: Biomechanical Hippo Signaling During Zebrafish Cardiac Development. Front Cell Dev Biol 2021; 9:731101. [PMID: 34422841 PMCID: PMC8375320 DOI: 10.3389/fcell.2021.731101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 07/20/2021] [Indexed: 11/13/2022] Open
Abstract
The heart is comprised of multiple tissues that contribute to its physiological functions. During development, the growth of myocardium and endocardium is coupled and morphogenetic processes within these separate tissue layers are integrated. Here, we discuss the roles of mechanosensitive Hippo signaling in growth and morphogenesis of the zebrafish heart. Hippo signaling is involved in defining numbers of cardiac progenitor cells derived from the secondary heart field, in restricting the growth of the epicardium, and in guiding trabeculation and outflow tract formation. Recent work also shows that myocardial chamber dimensions serve as a blueprint for Hippo signaling-dependent growth of the endocardium. Evidently, Hippo pathway components act at the crossroads of various signaling pathways involved in embryonic zebrafish heart development. Elucidating how biomechanical Hippo signaling guides heart morphogenesis has direct implications for our understanding of cardiac physiology and pathophysiology.
Collapse
Affiliation(s)
- Dorothee Bornhorst
- Stem Cell Program, Division of Hematology and Oncology, Boston Children's Hospital, Boston, MA, United States.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, United States
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany.,Institute of Molecular Biology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
39
|
Inman A, Smutny M. Feeling the force: Multiscale force sensing and transduction at the cell-cell interface. Semin Cell Dev Biol 2021; 120:53-65. [PMID: 34238674 DOI: 10.1016/j.semcdb.2021.06.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/11/2021] [Accepted: 06/13/2021] [Indexed: 12/13/2022]
Abstract
A universal principle of all living cells is the ability to sense and respond to mechanical stimuli which is essential for many biological processes. Recent efforts have identified critical mechanosensitive molecules and response pathways involved in mechanotransduction during development and tissue homeostasis. Tissue-wide force transmission and local force sensing need to be spatiotemporally coordinated to precisely regulate essential processes during development such as tissue morphogenesis, patterning, cell migration and organogenesis. Understanding how cells identify and interpret extrinsic forces and integrate a specific response on cell and tissue level remains a major challenge. In this review we consider important cellular and physical factors in control of cell-cell mechanotransduction and discuss their significance for cell and developmental processes. We further highlight mechanosensitive macromolecules that are known to respond to external forces and present examples of how force responses can be integrated into cell and developmental programs.
Collapse
Affiliation(s)
- Angus Inman
- Centre for Mechanochemical Cell Biology and Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV47AL, UK
| | - Michael Smutny
- Centre for Mechanochemical Cell Biology and Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV47AL, UK.
| |
Collapse
|
40
|
A junctional PACSIN2/EHD4/MICAL-L1 complex coordinates VE-cadherin trafficking for endothelial migration and angiogenesis. Nat Commun 2021; 12:2610. [PMID: 33972531 PMCID: PMC8110786 DOI: 10.1038/s41467-021-22873-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 03/31/2021] [Indexed: 11/24/2022] Open
Abstract
Angiogenic sprouting relies on collective migration and coordinated rearrangements of endothelial leader and follower cells. VE-cadherin-based adherens junctions have emerged as key cell-cell contacts that transmit forces between cells and trigger signals during collective cell migration in angiogenesis. However, the underlying molecular mechanisms that govern these processes and their functional importance for vascular development still remain unknown. We previously showed that the F-BAR protein PACSIN2 is recruited to tensile asymmetric adherens junctions between leader and follower cells. Here we report that PACSIN2 mediates the formation of endothelial sprouts during angiogenesis by coordinating collective migration. We show that PACSIN2 recruits the trafficking regulators EHD4 and MICAL-L1 to the rear end of asymmetric adherens junctions to form a recycling endosome-like tubular structure. The junctional PACSIN2/EHD4/MICAL-L1 complex controls local VE-cadherin trafficking and thereby coordinates polarized endothelial migration and angiogenesis. Our findings reveal a molecular event at force-dependent asymmetric adherens junctions that occurs during the tug-of-war between endothelial leader and follower cells, and allows for junction-based guidance during collective migration in angiogenesis. Communication between endothelial leader and follower cells during collective cell migration is crucial for vascular development. Here, the authors show that PACSIN2 guides collective cell migration and angiogenesis by recruiting a protein trafficking complex to asymmetric cell-cell junctions, controlling local junction plasticity.
Collapse
|
41
|
Molecular Dambusters: What Is Behind Hyperpermeability in Bradykinin-Mediated Angioedema? Clin Rev Allergy Immunol 2021; 60:318-347. [PMID: 33725263 PMCID: PMC7962090 DOI: 10.1007/s12016-021-08851-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2021] [Indexed: 02/08/2023]
Abstract
In the last few decades, a substantial body of evidence underlined the pivotal role of bradykinin in certain types of angioedema. The formation and breakdown of bradykinin has been studied thoroughly; however, numerous questions remained open regarding the triggering, course, and termination of angioedema attacks. Recently, it became clear that vascular endothelial cells have an integrative role in the regulation of vessel permeability. Apart from bradykinin, a great number of factors of different origin, structure, and mechanism of action are capable of modifying the integrity of vascular endothelium, and thus, may participate in the regulation of angioedema formation. Our aim in this review is to describe the most important permeability factors and the molecular mechanisms how they act on endothelial cells. Based on endothelial cell function, we also attempt to explain some of the challenging findings regarding bradykinin-mediated angioedema, where the function of bradykinin itself cannot account for the pathophysiology. By deciphering the complex scenario of vascular permeability regulation and edema formation, we may gain better scientific tools to be able to predict and treat not only bradykinin-mediated but other types of angioedema as well.
Collapse
|
42
|
Heme induces rapid endothelial barrier dysfunction via the MKK3/p38MAPK axis. Blood 2021; 136:749-754. [PMID: 32548640 DOI: 10.1182/blood.2019003986] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 05/11/2020] [Indexed: 12/23/2022] Open
Abstract
Several studies demonstrate that hemolysis and free heme in circulation cause endothelial barrier dysfunction and are associated with severe pathological conditions such as acute respiratory distress syndrome, acute chest syndrome, and sepsis. However, the precise molecular mechanisms involved in the pathology of heme-induced barrier disruption remain to be elucidated. In this study, we investigated the role of free heme in the endothelial barrier integrity and mechanisms of heme-mediated intracellular signaling of human lung microvascular endothelial cells (HLMVECs). Heme, in a dose-dependent manner, induced a rapid drop in the endothelial barrier integrity of HLMVECs. An investigation into barrier proteins revealed that heme primarily affected the tight junction proteins zona occludens-1, claudin-1, and claudin-5, which were significantly reduced after heme exposure. The p38MAPK/HSP27 pathway, involved in the regulation of endothelial cytoskeleton remodeling, was also significantly altered after heme treatment, both in HLMVECs and mice. By using a knockout (KO) mouse for MKK3, a key regulator of the p38MAPK pathway, we showed that this KO effectively decreased heme-induced endothelial barrier dysfunction. Taken together, our results indicate that targeting the p38MAPK pathway may represent a crucial treatment strategy in alleviating hemolytic diseases.
Collapse
|
43
|
Barcelona‐Estaje E, Dalby MJ, Cantini M, Salmeron‐Sanchez M. You Talking to Me? Cadherin and Integrin Crosstalk in Biomaterial Design. Adv Healthc Mater 2021; 10:e2002048. [PMID: 33586353 DOI: 10.1002/adhm.202002048] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/20/2021] [Indexed: 12/21/2022]
Abstract
While much work has been done in the design of biomaterials to control integrin-mediated adhesion, less emphasis has been put on functionalization of materials with cadherin ligands. Yet, cell-cell contacts in combination with cell-matrix interactions are key in driving embryonic development, collective cell migration, epithelial to mesenchymal transition, and cancer metastatic processes, among others. This review focuses on the incorporation of both cadherin and integrin ligands in biomaterial design, to promote what is called the "adhesive crosstalk." First, the structure and function of cadherins and their role in eliciting mechanotransductive processes, by themselves or in combination with integrin mechanosensing, are introduced. Then, biomaterials that mimic cell-cell interactions, and recent applications to get insights in fundamental biology and tissue engineering, are critically discussed.
Collapse
Affiliation(s)
- Eva Barcelona‐Estaje
- Centre for the Cellular Microenvironment University of Glasgow Glasgow G12 8QQ UK
| | - Matthew J. Dalby
- Centre for the Cellular Microenvironment University of Glasgow Glasgow G12 8QQ UK
| | - Marco Cantini
- Centre for the Cellular Microenvironment University of Glasgow Glasgow G12 8QQ UK
| | | |
Collapse
|
44
|
Comparison of Endothelial Barrier Functional Recovery After Implantation of a Novel Biodegradable-Polymer Sirolimus-Eluting Stent in Comparison to Durable- and Biodegradable-Polymer Everolimus-Eluting Stents. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2021; 24:1-10. [DOI: 10.1016/j.carrev.2020.08.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/07/2020] [Accepted: 08/10/2020] [Indexed: 11/24/2022]
|
45
|
Paracellular and Transcellular Leukocytes Diapedesis Are Divergent but Interconnected Evolutionary Events. Genes (Basel) 2021; 12:genes12020254. [PMID: 33578809 PMCID: PMC7916592 DOI: 10.3390/genes12020254] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/03/2021] [Accepted: 02/06/2021] [Indexed: 12/15/2022] Open
Abstract
Infiltration of the endothelial layer of the blood-brain barrier by leukocytes plays a critical role in health and disease. When passing through the endothelial layer during the diapedesis process lymphocytes can either follow a paracellular route or a transcellular one. There is a debate whether these two processes constitute one mechanism, or they form two evolutionary distinct migration pathways. We used artificial intelligence, phylogenetic analysis, HH search, ancestor sequence reconstruction to investigate further this intriguing question. We found that the two systems share several ancient components, such as RhoA protein that plays a critical role in controlling actin movement in both mechanisms. However, some of the key components differ between these two transmigration processes. CAV1 genes emerged during Trichoplax adhaerens, and it was only reported in transcellular process. Paracellular process is dependent on PECAM1. PECAM1 emerged from FASL5 during Zebrafish divergence. Lastly, both systems employ late divergent genes such as ICAM1 and VECAM1. Taken together, our results suggest that these two systems constitute two different mechanical sensing mechanisms of immune cell infiltrations of the brain, yet these two systems are connected. We postulate that the mechanical properties of the cellular polarity is the main driving force determining the migration pathway. Our analysis indicates that both systems coevolved with immune cells, evolving to a higher level of complexity in association with the evolution of the immune system.
Collapse
|
46
|
Wolpe AG, Ruddiman CA, Hall PJ, Isakson BE. Polarized Proteins in Endothelium and Their Contribution to Function. J Vasc Res 2021; 58:65-91. [PMID: 33503620 DOI: 10.1159/000512618] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022] Open
Abstract
Protein localization in endothelial cells is tightly regulated to create distinct signaling domains within their tight spatial restrictions including luminal membranes, abluminal membranes, and interendothelial junctions, as well as caveolae and calcium signaling domains. Protein localization in endothelial cells is also determined in part by the vascular bed, with differences between arteries and veins and between large and small arteries. Specific protein polarity and localization is essential for endothelial cells in responding to various extracellular stimuli. In this review, we examine protein localization in the endothelium of resistance arteries, with occasional references to other vessels for contrast, and how that polarization contributes to endothelial function and ultimately whole organism physiology. We highlight the protein localization on the luminal surface, discussing important physiological receptors and the glycocalyx. The protein polarization to the abluminal membrane is especially unique in small resistance arteries with the presence of the myoendothelial junction, a signaling microdomain that regulates vasodilation, feedback to smooth muscle cells, and ultimately total peripheral resistance. We also discuss the interendothelial junction, where tight junctions, adherens junctions, and gap junctions all convene and regulate endothelial function. Finally, we address planar cell polarity, or axial polarity, and how this is regulated by mechanosensory signals like blood flow.
Collapse
Affiliation(s)
- Abigail G Wolpe
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Claire A Ruddiman
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Phillip J Hall
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA, .,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia, USA,
| |
Collapse
|
47
|
Cohesive cancer invasion of the biophysical barrier of smooth muscle. Cancer Metastasis Rev 2021; 40:205-219. [PMID: 33398621 DOI: 10.1007/s10555-020-09950-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 12/15/2020] [Indexed: 01/22/2023]
Abstract
Smooth muscle is found around organs in the digestive, respiratory, and reproductive tracts. Cancers arising in the bladder, prostate, stomach, colon, and other sites progress from low-risk disease to high-risk, lethal metastatic disease characterized by tumor invasion into, within, and through the biophysical barrier of smooth muscle. We consider here the unique biophysical properties of smooth muscle and how cohesive clusters of tumor use mechanosensing cell-cell and cell-ECM (extracellular matrix) adhesion receptors to move through a structured muscle and withstand the biophysical forces to reach distant sites. Understanding integrated mechanosensing features within tumor cluster and smooth muscle and potential triggers within adjacent adipose tissue, such as the unique damage-associated molecular pattern protein (DAMP), eNAMPT (extracellular nicotinamide phosphoribosyltransferase), or visfatin, offers an opportunity to prevent the first steps of invasion and metastasis through the structured muscle.
Collapse
|
48
|
VanderBurgh JA, Potharazu AV, Schwager SC, Reinhart-King CA. A discrete interface in matrix stiffness creates an oscillatory pattern of endothelial monolayer disruption. J Cell Sci 2020; 133:jcs244533. [PMID: 32878941 PMCID: PMC7520461 DOI: 10.1242/jcs.244533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 08/17/2020] [Indexed: 01/07/2023] Open
Abstract
Intimal stiffening upregulates endothelial cell contractility, disrupting barrier integrity; however, intimal stiffening is non-uniform. The impact of local changes in intimal stiffness on proximal and distal cell-cell interactions is unknown. To investigate the range at which matrix stiffness heterogeneities impact neighboring endothelial cells within a monolayer, we built a micropillar system with adjacent regions of stiff and compliant matrix. The stiffness interface results in an oscillatory pattern of neutrophil transendothelial migration, symmetrical about the interface and well-fit by a sinusoid function. 'Peaks' of the sinusoid were found to have increased cellular contractility and decreased barrier function relative to 'troughs' of the sinusoid. Pharmacological modulation of contractility was observed to break symmetry, altering the amplitude and wavelength of the sinusoid, indicating that contractility may regulate this effect. This work illuminates a novel biophysical phenomenon of the role of stiffness-mediated cell-matrix interactions on cell-cell interactions at a distance. Additionally, it provides insight into the range at which intimal matrix stiffness heterogeneities will impact endothelial barrier function and potentially contribute to atherogenesis.
Collapse
Affiliation(s)
- Jacob A VanderBurgh
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Archit V Potharazu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Samantha C Schwager
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Cynthia A Reinhart-King
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
49
|
Yin X, Fang T, Zhang L, Lin X, Yang Y, Lou S, Li C, Yu X, Xue Y. Impact of CD144 gene expression on outcomes in stage III gastric cancer patients. Pathology 2020; 52:657-669. [PMID: 32859388 DOI: 10.1016/j.pathol.2020.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/06/2020] [Accepted: 05/13/2020] [Indexed: 10/23/2022]
Abstract
CD144 has been shown to promote tumour angiogenesis, invasion and metastasis in malignant tumours. The purpose of the present study was to investigate the clinical prognostic significance of CD144 in advanced gastric cancer (GC) to complement the American Joint Committee on Cancer (AJCC) 8th Edition convention. We established that CD144 was highly related to angiogenesis using The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) public databases. We randomly selected 173 stage III GC patients who received curative gastrectomy. The expression level of CD144 was assessed by immunohistochemistry and Image-Pro Plus software. After survival analysis, nomograms were created to predict the risk of stage III gastric cancer patients' 5-years survival. In this study, the median value of the CD144 positive area/total area under the microscope was 5.6%, and this was defined as the cut-off value. The expression of CD144 assisted further subgrouping of stage Ⅲa, Ⅲb, and Ⅲc GC patients. To evaluate the disease-free survival (DFS) and overall survival (OS) of patients, univariate and multivariate analysis was performed, which showed that the expression of CD144 was an independent predictor for DFS, and Borrmann type and expression of CD144 were independent predictors for OS (p<0.05). Nomograms were used to evaluate the risk of stage III GC by combining Borrmann type and the expression level of CD144. In advanced GC patients, the expression level of CD144 is a useful prognostic indicator in evaluating the risk of disease prognosis.
Collapse
Affiliation(s)
- Xin Yin
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin, P.R. China
| | - Tianyi Fang
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin, P.R. China
| | - Lei Zhang
- Department of Pathology, Harbin Medical University, Harbin, P.R. China
| | - Xuan Lin
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, P.R. China
| | - Yongheng Yang
- Department of Pathology, Harbin Medical University, Harbin, P.R. China
| | - Shenghan Lou
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin, P.R. China
| | - Chunfeng Li
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin, P.R. China
| | - Xuefeng Yu
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin, P.R. China
| | - Yingwei Xue
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin, P.R. China.
| |
Collapse
|
50
|
Broussard JA, Jaiganesh A, Zarkoob H, Conway DE, Dunn AR, Espinosa HD, Janmey PA, Green KJ. Scaling up single-cell mechanics to multicellular tissues - the role of the intermediate filament-desmosome network. J Cell Sci 2020; 133:jcs228031. [PMID: 32179593 PMCID: PMC7097224 DOI: 10.1242/jcs.228031] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cells and tissues sense, respond to and translate mechanical forces into biochemical signals through mechanotransduction, which governs individual cell responses that drive gene expression, metabolic pathways and cell motility, and determines how cells work together in tissues. Mechanotransduction often depends on cytoskeletal networks and their attachment sites that physically couple cells to each other and to the extracellular matrix. One way that cells associate with each other is through Ca2+-dependent adhesion molecules called cadherins, which mediate cell-cell interactions through adherens junctions, thereby anchoring and organizing the cortical actin cytoskeleton. This actin-based network confers dynamic properties to cell sheets and developing organisms. However, these contractile networks do not work alone but in concert with other cytoarchitectural elements, including a diverse network of intermediate filaments. This Review takes a close look at the intermediate filament network and its associated intercellular junctions, desmosomes. We provide evidence that this system not only ensures tissue integrity, but also cooperates with other networks to create more complex tissues with emerging properties in sensing and responding to increasingly stressful environments. We will also draw attention to how defects in intermediate filament and desmosome networks result in both chronic and acquired diseases.
Collapse
Affiliation(s)
- Joshua A Broussard
- Departments of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Avinash Jaiganesh
- Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Hoda Zarkoob
- Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Daniel E Conway
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Alexander R Dunn
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Horacio D Espinosa
- Department of Mechanical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Paul A Janmey
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kathleen J Green
- Departments of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|