1
|
Atkin-Smith GK, Santavanond JP, Light A, Rimes JS, Samson AL, Er J, Liu J, Johnson DN, Le Page M, Rajasekhar P, Yip RKH, Geoghegan ND, Rogers KL, Chang C, Bryant VL, Margetts M, Keightley MC, Kilpatrick TJ, Binder MD, Tran S, Lee EF, Fairlie WD, Ozkocak DC, Wei AH, Hawkins ED, Poon IKH. In situ visualization of endothelial cell-derived extracellular vesicle formation in steady state and malignant conditions. Nat Commun 2024; 15:8802. [PMID: 39438460 PMCID: PMC11496675 DOI: 10.1038/s41467-024-52867-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
Endothelial cells are integral components of all vasculature within complex organisms. As they line the blood vessel wall, endothelial cells are constantly exposed to a variety of molecular factors and shear force that can induce cellular damage and stress. However, how endothelial cells are removed or eliminate unwanted cellular contents, remains unclear. The generation of large extracellular vesicles (EVs) has emerged as a key mechanism for the removal of cellular waste from cells that are dying or stressed. Here, we used intravital microscopy of the bone marrow to directly measure the kinetics of EV formation from endothelial cells in vivo under homoeostatic and malignant conditions. These large EVs are mitochondria-rich, expose the 'eat me' signal phosphatidylserine, and can interact with immune cell populations as a potential clearance mechanism. Elevated levels of circulating EVs correlates with degradation of the bone marrow vasculature caused by acute myeloid leukaemia. Together, our study provides in vivo spatio-temporal characterization of EV formation in the murine vasculature and suggests that circulating, large endothelial cell-derived EVs can provide a snapshot of vascular damage at distal sites.
Collapse
Affiliation(s)
- Georgia K Atkin-Smith
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia.
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia.
- Research Centre for Extracellular Vesicles, La Trobe University, Melbourne, VIC, Australia.
| | - Jascinta P Santavanond
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
- Research Centre for Extracellular Vesicles, La Trobe University, Melbourne, VIC, Australia
| | - Amanda Light
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Joel S Rimes
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Andre L Samson
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Jeremy Er
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- Clinical Haematology Department, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Joy Liu
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Darryl N Johnson
- Materials Characterisation and Fabrication Platform, Department of Chemical Engineering, University of Melbourne, Parkville, VIC, Australia
| | - Mélanie Le Page
- ARAFlowCore, Alfred Research Alliance, Monash University, Melbourne, VIC, Australia
| | - Pradeep Rajasekhar
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Raymond K H Yip
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Niall D Geoghegan
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Kelly L Rogers
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Catherine Chang
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Vanessa L Bryant
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Clinical Immunology and Allergy, Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Mai Margetts
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - M Cristina Keightley
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
- Department of Rural Clinical Sciences, La Trobe Rural Health School, Bendigo, VIC, Australia
| | - Trevor J Kilpatrick
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Michele D Binder
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - Sharon Tran
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Erinna F Lee
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Walter D Fairlie
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Dilara C Ozkocak
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
- Research Centre for Extracellular Vesicles, La Trobe University, Melbourne, VIC, Australia
| | - Andrew H Wei
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- Clinical Haematology Department, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Edwin D Hawkins
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia.
| | - Ivan K H Poon
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia.
- Research Centre for Extracellular Vesicles, La Trobe University, Melbourne, VIC, Australia.
| |
Collapse
|
2
|
D’Aversa E, Salvatori F, Vaccarezza M, Antonica B, Grisafi M, Singh AV, Secchiero P, Zauli G, Tisato V, Gemmati D. circRNAs as Epigenetic Regulators of Integrity in Blood-Brain Barrier Architecture: Mechanisms and Therapeutic Strategies in Multiple Sclerosis. Cells 2024; 13:1316. [PMID: 39195206 PMCID: PMC11352526 DOI: 10.3390/cells13161316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/30/2024] [Accepted: 08/03/2024] [Indexed: 08/29/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory neurodegenerative disease leading to progressive demyelination and neuronal loss, with extensive neurological symptoms. As one of the most widespread neurodegenerative disorders, with an age onset of about 30 years, it turns out to be a socio-health and economic issue, thus necessitating therapeutic interventions currently unavailable. Loss of integrity in the blood-brain barrier (BBB) is one of the distinct MS hallmarks. Brain homeostasis is ensured by an endothelial cell-based monolayer at the interface between the central nervous system (CNS) and systemic bloodstream, acting as a selective barrier. MS results in enhanced barrier permeability, mainly due to the breakdown of tight (TJs) and adherens junctions (AJs) between endothelial cells. Specifically, proinflammatory mediator release causes failure in cytoplasmic exposure of junctions, resulting in compromised BBB integrity that enables blood cells to cross the barrier, establishing iron deposition and neuronal impairment. Cells with a compromised cytoskeletal protein network, fiber reorganization, and discontinuous junction structure can occur, resulting in BBB dysfunction. Recent investigations on spatial transcriptomics have proven circularRNAs (circRNAs) to be powerful multi-functional molecules able to epigenetically regulate transcription and structurally support proteins. In the present review, we provide an overview of the recent role ascribed to circRNAs in maintaining BBB integrity/permeability via cytoskeletal stability. Increased knowledge of the mechanisms responsible for impairment and circRNA's role in driving BBB damage and dysfunction might be helpful for the recognition of novel therapeutic targets to overcome BBB damage and unrestrained neurodegeneration.
Collapse
Affiliation(s)
- Elisabetta D’Aversa
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Francesca Salvatori
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Mauro Vaccarezza
- Curtin Medical School & Curtin Health Innovation Research Institute (CHIRI), Faculty of Health Sciences, Curtin University, Bentley, WA 6102, Australia
| | - Bianca Antonica
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Miriana Grisafi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Ajay Vikram Singh
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), 10589 Berlin, Germany
| | - Paola Secchiero
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Giorgio Zauli
- Research Department, King Khaled Eye Specialistic Hospital, Riyadh 11462, Saudi Arabia
| | - Veronica Tisato
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- University Strategic Centre for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
- LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
| | - Donato Gemmati
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- University Strategic Centre for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
- Centre Haemostasis & Thrombosis, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
3
|
Chikh A, Raimondi C. Endothelial Neuropilin-1: a multifaced signal transducer with an emerging role in inflammation and atherosclerosis beyond angiogenesis. Biochem Soc Trans 2024; 52:137-150. [PMID: 38323651 PMCID: PMC10903451 DOI: 10.1042/bst20230329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 02/08/2024]
Abstract
Neuropilin-1 (NRP1) is a transmembrane glycoprotein expressed by several cell types including, neurons, endothelial cells (ECs), smooth muscle cells, cardiomyocytes and immune cells comprising macrophages, dendritic cells and T cell subsets. Since NRP1 discovery in 1987 as an adhesion molecule in the frog nervous system, more than 2300 publications on PubMed investigated the function of NRP1 in physiological and pathological contexts. NRP1 has been characterised as a coreceptor for class 3 semaphorins and several members of the vascular endothelial growth factor (VEGF) family. Because the VEGF family is the main regulator of blood and lymphatic vessel growth in addition to promoting neurogenesis, neuronal patterning, neuroprotection and glial growth, the role of NRP1 in these biological processes has been extensively investigated. It is now established that NRP1 promotes the physiological growth of new vessels from pre-existing ones in the process of angiogenesis. Furthermore, several studies have shown that NRP1 mediates signalling pathways regulating pathological vascular growth in ocular neovascular diseases and tumour development. Less defined are the roles of NRP1 in maintaining the function of the quiescent established vasculature in an adult organism. This review will focus on the opposite roles of NRP1 in regulating transforming growth factor β signalling pathways in different cell types, and on the emerging role of endothelial NRP1 as an atheroprotective, anti-inflammatory factor involved in the response of ECs to shear stress.
Collapse
Affiliation(s)
- Anissa Chikh
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, London SW17 0RE, U.K
| | - Claudio Raimondi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, U.K
| |
Collapse
|
4
|
Panagiotides NG, Poledniczek M, Andreas M, Hülsmann M, Kocher AA, Kopp CW, Piechota-Polanczyk A, Weidenhammer A, Pavo N, Wadowski PP. Myocardial Oedema as a Consequence of Viral Infection and Persistence-A Narrative Review with Focus on COVID-19 and Post COVID Sequelae. Viruses 2024; 16:121. [PMID: 38257821 PMCID: PMC10818479 DOI: 10.3390/v16010121] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Microvascular integrity is a critical factor in myocardial fluid homeostasis. The subtle equilibrium between capillary filtration and lymphatic fluid removal is disturbed during pathological processes leading to inflammation, but also in hypoxia or due to alterations in vascular perfusion and coagulability. The degradation of the glycocalyx as the main component of the endothelial filtration barrier as well as pericyte disintegration results in the accumulation of interstitial and intracellular water. Moreover, lymphatic dysfunction evokes an increase in metabolic waste products, cytokines and inflammatory cells in the interstitial space contributing to myocardial oedema formation. This leads to myocardial stiffness and impaired contractility, eventually resulting in cardiomyocyte apoptosis, myocardial remodelling and fibrosis. The following article reviews pathophysiological inflammatory processes leading to myocardial oedema including myocarditis, ischaemia-reperfusion injury and viral infections with a special focus on the pathomechanisms evoked by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. In addition, clinical implications including potential long-term effects due to viral persistence (long COVID), as well as treatment options, are discussed.
Collapse
Affiliation(s)
- Noel G. Panagiotides
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Michael Poledniczek
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| | - Martin Andreas
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria; (M.A.); (A.A.K.)
| | - Martin Hülsmann
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Alfred A. Kocher
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria; (M.A.); (A.A.K.)
| | - Christoph W. Kopp
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| | | | - Annika Weidenhammer
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Noemi Pavo
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Patricia P. Wadowski
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| |
Collapse
|
5
|
Tang J, Kang Y, Zhou Y, Shang N, Li X, Wang H, Lan J, Wang S, Wu L, Peng Y. TIMP2 ameliorates blood-brain barrier disruption in traumatic brain injury by inhibiting Src-dependent VE-cadherin internalization. J Clin Invest 2023; 134:e164199. [PMID: 38015626 PMCID: PMC10849766 DOI: 10.1172/jci164199] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/21/2023] [Indexed: 11/30/2023] Open
Abstract
Blood-brain barrier (BBB) disruption is a serious pathological consequence of traumatic brain injury (TBI), for which there are limited therapeutic strategies. Tissue inhibitor of metalloproteinase-2 (TIMP2), a molecule with dual functions of inhibiting MMP activity and displaying cytokine-like activity through receptor binding, has been reported to inhibit VEGF-induced vascular hyperpermeability. Here, we investigate the ability of TIMP2 to ameliorate BBB disruption in TBI and the underlying molecular mechanisms. Both TIMP2 and AlaTIMP2, a TIMP2 mutant without MMP-inhibiting activity, attenuated neurological deficits and BBB leakage in TBI mice; they also inhibited junctional protein degradation and translocation to reduce paracellular permeability in human brain microvascular endothelial cells (ECs) exposed to hypoxic plus inflammatory insult. Mechanistic studies revealed that TIMP2 interacted with α3β1 integrin on ECs, inhibiting Src activation-dependent VE-cadherin phosphorylation, VE-cadherin/catenin complex destabilization, and subsequent VE-cadherin internalization. Notably, localization of VE-cadherin on the membrane was critical for TIMP2-mediated EC barrier integrity. Furthermore, TIMP2-mediated increased membrane localization of VE-cadherin enhanced the level of active Rac1, thereby inhibiting stress fiber formation. All together, our studies have identified an MMP-independent mechanism by which TIMP2 regulates EC barrier integrity after TBI. TIMP2 may be a therapeutic agent for TBI and other neurological disorders involving BBB breakdown.
Collapse
|
6
|
Bosseboeuf E, Chikh A, Chaker AB, Mitchell TP, Vignaraja D, Rajendrakumar R, Khambata RS, Nightingale TD, Mason JC, Randi AM, Ahluwalia A, Raimondi C. Neuropilin-1 interacts with VE-cadherin and TGFBR2 to stabilize adherens junctions and prevent activation of endothelium under flow. Sci Signal 2023; 16:eabo4863. [PMID: 37220183 PMCID: PMC7614756 DOI: 10.1126/scisignal.abo4863] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 05/03/2023] [Indexed: 05/25/2023]
Abstract
Linear and disturbed flow differentially regulate gene expression, with disturbed flow priming endothelial cells (ECs) for a proinflammatory, atheroprone expression profile and phenotype. Here, we investigated the role of the transmembrane protein neuropilin-1 (NRP1) in ECs exposed to flow using cultured ECs, mice with an endothelium-specific knockout of NRP1, and a mouse model of atherosclerosis. We demonstrated that NRP1 was a constituent of adherens junctions that interacted with VE-cadherin and promoted its association with p120 catenin, stabilizing adherens junctions and inducing cytoskeletal remodeling in alignment with the direction of flow. We also showed that NRP1 interacted with transforming growth factor-β (TGF-β) receptor II (TGFBR2) and reduced the plasma membrane localization of TGFBR2 and TGF-β signaling. NRP1 knockdown increased the abundance of proinflammatory cytokines and adhesion molecules, resulting in increased leukocyte rolling and atherosclerotic plaque size. These findings describe a role for NRP1 in promoting endothelial function and reveal a mechanism by which NRP1 reduction in ECs may contribute to vascular disease by modulating adherens junction signaling and promoting TGF-β signaling and inflammation.
Collapse
Affiliation(s)
- Emy Bosseboeuf
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Anissa Chikh
- Molecular and Clinical Sciences Research Institute, St. George’s, University of London, London SW17 0RE, UK
| | - Ahmed Bey Chaker
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Tom P. Mitchell
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre for Microvascular Research, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Dhilakshani Vignaraja
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Ridhi Rajendrakumar
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Rayomand S. Khambata
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Thomas D. Nightingale
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre for Microvascular Research, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Justin C. Mason
- Vascular Sciences, National Heart & Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| | - Anna M. Randi
- Vascular Sciences, National Heart & Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| | - Amrita Ahluwalia
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Claudio Raimondi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
7
|
Simulated Microgravity Increases the Permeability of HUVEC Monolayer through Up-Regulation of Rap1GAP and Decreased Rap2 Activation. Int J Mol Sci 2022; 23:ijms23020630. [PMID: 35054818 PMCID: PMC8776081 DOI: 10.3390/ijms23020630] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/30/2021] [Accepted: 12/30/2021] [Indexed: 02/01/2023] Open
Abstract
Space microgravity condition has great physiological influence on astronauts’ health. The interaction of endothelial cells, which control vascular permeability and immune responses, is sensitive to mechanical stress. However, whether microgravity has significant effects on the physiological function of the endothelium has not been investigated. In order to address such a question, a clinostat-based culture model with a HUVEC monolayer being inside the culture vessel under the simulated microgravity (SMG) was established. The transmittance of FITC-tagged dextran was used to estimate the change of integrity of the adherens junction of the HUVEC monolayer. Firstly, we found that the permeability of the HUVEC monolayer was largely increased after SMG treatment. To elucidate the mechanism of the increased permeability of the HUVEC monolayer under SMG, the levels of total expression and activated protein levels of Rap1 and Rap2 in HUVEC cells, which regulate the adherens junction of endothelial cells, were detected by WB and GST pull-down after SMG. As the activation of both Rap1 and Rap2 was significantly decreased under SMG, the expression of Rap1GEF1 (C3G) and Rap1GAP in HUVECs, which regulate the activation of them, was further determined. The results indicate that both C3G and Rap1GAP showed a time-dependent increase with the expression of Rap1GAP being dominant at 48 h after SMG. The down-regulation of the expression of junctional proteins, VE-cadherin and β-catenin, in HUVEC cells was also confirmed by WB and immunofluorescence after SMG. To clarify whether up-regulation of Rap1GAP is necessary for the increased permeability of the HUVEC monolayer after SMG, the expression of Rap1GAP was knocked down by Rap1GAP-shRNA, and the change of permeability of the HUVEC monolayer was detected. The results indicate that knock-down of Rap1GAP reduced SMG-induced leaking of the HUVEC monolayer in a time-dependent manner. In total, our results indicate that the Rap1GAP-Rap signal axis was necessary for the increased permeability of the HUVEC monolayer along with the down-regulation of junctional molecules including VE-cadherin and β-catenin.
Collapse
|
8
|
Zhu X, Yang M, Zhao P, Li S, Zhang L, Huang L, Huang Y, Fei P, Yang Y, Zhang S, Xu H, Yuan Y, Zhang X, Zhu X, Ma S, Hao F, Sundaresan P, Zhu W, Yang Z. Catenin α 1 mutations cause familial exudative vitreoretinopathy by overactivating Norrin/β-catenin signaling. J Clin Invest 2021; 131:139869. [PMID: 33497368 DOI: 10.1172/jci139869] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 01/22/2021] [Indexed: 12/19/2022] Open
Abstract
Familial exudative vitreoretinopathy (FEVR) is a severe retinal vascular disease that causes blindness. FEVR has been linked to mutations in several genes associated with inactivation of the Norrin/β-catenin signaling pathway, but these account for only approximately 50% of cases. We report that mutations in α-catenin (CTNNA1) cause FEVR by overactivating the β-catenin pathway and disrupting cell adherens junctions. We identified 3 heterozygous mutations in CTNNA1 (p.F72S, p.R376Cfs*27, and p.P893L) by exome sequencing and further demonstrated that FEVR-associated mutations led to overactivation of Norrin/β-catenin signaling as a result of impaired protein interactions within the cadherin-catenin complex. The clinical features of FEVR were reproduced in mice lacking Ctnna1 in vascular endothelial cells (ECs) or with overactivated β-catenin signaling by an EC-specific gain-of-function allele of Ctnnb1. In isolated mouse lung ECs, both CTNNA1-P893L and F72S mutants failed to rescue either the disrupted F-actin arrangement or the VE-cadherin and CTNNB1 distribution. Moreover, we discovered that compound heterozygous Ctnna1 F72S and a deletion allele could cause a similar phenotype. Furthermore, in a FEVR family, we identified a mutation of LRP5, which activates Norrin/β-catenin signaling, and the corresponding knockin mice exhibited a partial FEVR-like phenotype. Our study demonstrates that the precise regulation of β-catenin activation is critical for retinal vascular development and provides new insights into the pathogenesis of FEVR.
Collapse
Affiliation(s)
- Xianjun Zhu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Mu Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Peiquan Zhao
- Department of Ophthalmology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shujin Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Lin Zhang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Lulin Huang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yi Huang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ping Fei
- Department of Ophthalmology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yeming Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Shanshan Zhang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Huijuan Xu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ye Yuan
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xiang Zhang
- Department of Ophthalmology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xiong Zhu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Shi Ma
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Fang Hao
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Periasamy Sundaresan
- Department of Genetics, Aravind Medical Research Foundation, Aravind Eye Hospital, Madurai, Tamil Nadu, India
| | - Weiquan Zhu
- Department of Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Zhenglin Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| |
Collapse
|
9
|
Arts JJG, Mahlandt EK, Grönloh MLB, Schimmel L, Noordstra I, Gordon E, van Steen ACI, Tol S, Walzog B, van Rijssel J, Nolte MA, Postma M, Khuon S, Heddleston JM, Wait E, Chew TL, Winter M, Montanez E, Goedhart J, van Buul JD. Endothelial junctional membrane protrusions serve as hotspots for neutrophil transmigration. eLife 2021; 10:66074. [PMID: 34431475 PMCID: PMC8437435 DOI: 10.7554/elife.66074] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 08/22/2021] [Indexed: 12/27/2022] Open
Abstract
Upon inflammation, leukocytes rapidly transmigrate across the endothelium to enter the inflamed tissue. Evidence accumulates that leukocytes use preferred exit sites, alhough it is not yet clear how these hotspots in the endothelium are defined and how they are recognized by the leukocyte. Using lattice light sheet microscopy, we discovered that leukocytes prefer endothelial membrane protrusions at cell junctions for transmigration. Phenotypically, these junctional membrane protrusions are present in an asymmetric manner, meaning that one endothelial cell shows the protrusion and the adjacent one does not. Consequently, leukocytes cross the junction by migrating underneath the protruding endothelial cell. These protrusions depend on Rac1 activity and by using a photo-activatable Rac1 probe, we could artificially generate local exit-sites for leukocytes. Overall, we have discovered a new mechanism that uses local induced junctional membrane protrusions to facilitate/steer the leukocyte escape/exit from inflamed vessel walls.
Collapse
Affiliation(s)
- Janine JG Arts
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of AmsterdamAmsterdamNetherlands
| | - Eike K Mahlandt
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of AmsterdamAmsterdamNetherlands
| | - Max LB Grönloh
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of AmsterdamAmsterdamNetherlands
| | - Lilian Schimmel
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of AmsterdamAmsterdamNetherlands
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of QueenslandBrisbaneQLDAustralia
| | - Ivar Noordstra
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of QueenslandBrisbaneQLDAustralia
| | - Emma Gordon
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of QueenslandBrisbaneQLDAustralia
| | - Abraham CI van Steen
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
| | - Simon Tol
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
| | - Barbara Walzog
- Department of Cardiovascular Physiology and Pathophysiology, Walter Brendel Center of Experimental Medicine, Biomedical Center, Ludwig-Maximilians-Universität MünchenPlanegg-MartinsriedGermany
| | - Jos van Rijssel
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
| | - Martijn A Nolte
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
| | - Marten Postma
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of AmsterdamAmsterdamNetherlands
| | - Satya Khuon
- Advanced Imaging Center at Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - John M Heddleston
- Advanced Imaging Center at Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
- Microscopy Facility at the Cleveland Clinic Florida Research and Innovation CenterPort St. LucieUnited States
| | - Eric Wait
- Advanced Imaging Center at Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Teng Leong Chew
- Advanced Imaging Center at Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Mark Winter
- Zuckerman Postdoctoral Fellow, Department of Marine Sciences, University of HaifaHaifaIsrael
| | - Eloi Montanez
- Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of BarcelonaBarcelonaSpain
| | - Joachim Goedhart
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of AmsterdamAmsterdamNetherlands
| | - Jaap D van Buul
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of AmsterdamAmsterdamNetherlands
| |
Collapse
|
10
|
A ligand-insensitive UNC5B splicing isoform regulates angiogenesis by promoting apoptosis. Nat Commun 2021; 12:4872. [PMID: 34381052 PMCID: PMC8358048 DOI: 10.1038/s41467-021-24998-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
The Netrin-1 receptor UNC5B is an axon guidance regulator that is also expressed in endothelial cells (ECs), where it finely controls developmental and tumor angiogenesis. In the absence of Netrin-1, UNC5B induces apoptosis that is blocked upon Netrin-1 binding. Here, we identify an UNC5B splicing isoform (called UNC5B-Δ8) expressed exclusively by ECs and generated through exon skipping by NOVA2, an alternative splicing factor regulating vascular development. We show that UNC5B-Δ8 is a constitutively pro-apoptotic splicing isoform insensitive to Netrin-1 and required for specific blood vessel development in an apoptosis-dependent manner. Like NOVA2, UNC5B-Δ8 is aberrantly expressed in colon cancer vasculature where its expression correlates with tumor angiogenesis and poor patient outcome. Collectively, our data identify a mechanism controlling UNC5B’s necessary apoptotic function in ECs and suggest that the NOVA2/UNC5B circuit represents a post-transcriptional pathway regulating angiogenesis. UNC5B is a Netrin-1 receptor expressed in endothelial cells that in the absence of ligand induces apoptosis. Here the authors identify an UNC5B splicing isoform that is insensitive to the pro-survival ligand Netrin-1 and is required for apoptosis-dependent blood vessel development.
Collapse
|
11
|
Okamoto T, Park EJ, Kawamoto E, Usuda H, Wada K, Taguchi A, Shimaoka M. Endothelial connexin-integrin crosstalk in vascular inflammation. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166168. [PMID: 33991620 DOI: 10.1016/j.bbadis.2021.166168] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/18/2021] [Accepted: 05/02/2021] [Indexed: 02/06/2023]
Abstract
Cardiovascular diseases including blood vessel disorders represent a major cause of death globally. The essential roles played by local and systemic vascular inflammation in the pathogenesis of cardiovascular diseases have been increasingly recognized. Vascular inflammation triggers the aberrant activation of endothelial cells, which leads to the functional and structural abnormalities in vascular vessels. In addition to humoral mediators such as pro-inflammatory cytokines and prostaglandins, the alteration of physical and mechanical microenvironment - including vascular stiffness and shear stress - modify the gene expression profiles and metabolic profiles of endothelial cells via mechano-transduction pathways, thereby contributing to the pathogenesis of vessel disorders. Notably, connexins and integrins crosstalk each other in response to the mechanical stress, and, thereby, play an important role in regulating the mechano-transduction of endothelial cells. Here, we provide an overview on how the inter-play between connexins and integrins in endothelial cells unfold during the mechano-transduction in vascular inflammation.
Collapse
Affiliation(s)
- Takayuki Okamoto
- Department of Pharmacology, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo-city, Shimane 693-8501, Japan.
| | - Eun Jeong Park
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie 514-8507, Japan
| | - Eiji Kawamoto
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie 514-8507, Japan; Department of Emergency and Disaster Medicine, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie 514-8507, Japan
| | - Haruki Usuda
- Department of Pharmacology, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo-city, Shimane 693-8501, Japan
| | - Koichiro Wada
- Department of Pharmacology, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo-city, Shimane 693-8501, Japan
| | - Akihiko Taguchi
- Department of Regenerative Medicine Research, Foundation for Biomedical Research and Innovation at Kobe, 2-2 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-city, Mie 514-8507, Japan.
| |
Collapse
|
12
|
Epac1 Is Crucial for Maintenance of Endothelial Barrier Function through A Mechanism Partly Independent of Rac1. Cells 2020; 9:cells9102170. [PMID: 32992982 PMCID: PMC7601253 DOI: 10.3390/cells9102170] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/17/2020] [Accepted: 09/23/2020] [Indexed: 12/20/2022] Open
Abstract
Epac1 (exchange protein activated by cAMP) stabilizes the endothelial barrier, but detailed studies are limited by the side effects of pharmacological Epac1 modulators and transient transfections. Here, we compare the key properties of barriers between endothelial cells derived from wild-type (WT) and Epac1-knockout (KO) mice myocardium. We found that KO cell layers, unlike WT layers, had low and cAMP-insensitive trans-endothelial resistance (TER). They also had fragmented VE-cadherin staining despite having augmented cAMP levels and increased protein expression of Rap1, Rac1, RhoA, and VE-cadherin. The simultaneous direct activation of Rac1 and RhoA by CN04 compensated Epac1 loss, since TER was increased. In KO-cells, inhibition of Rac1 activity had no additional effect on TER, suggesting that other mechanisms compensate the inhibition of the Rac1 function to preserve barrier properties. In summary, Epac1 is crucial for baseline and cAMP-mediated barrier stabilization through mechanisms that are at least partially independent of Rac1.
Collapse
|
13
|
Li J, Zhao Y, Choi J, Ting KK, Coleman P, Chen J, Cogger VC, Wan L, Shi Z, Moller T, Zheng X, Vadas MA, Gamble JR. Targeting miR-27a/VE-cadherin interactions rescues cerebral cavernous malformations in mice. PLoS Biol 2020; 18:e3000734. [PMID: 32502201 PMCID: PMC7299406 DOI: 10.1371/journal.pbio.3000734] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/17/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022] Open
Abstract
Cerebral cavernous malformations (CCMs) are vascular lesions predominantly developing in the central nervous system (CNS), with no effective treatments other than surgery. Loss-of-function mutation in CCM1/krev interaction trapped 1 (KRIT1), CCM2, or CCM3/programmed cell death 10 (PDCD10) causes lesions that are characterized by abnormal vascular integrity. Vascular endothelial cadherin (VE-cadherin), a major regulator of endothelial cell (EC) junctional integrity is strongly disorganized in ECs lining the CCM lesions. We report here that microRNA-27a (miR-27a), a negative regulator of VE-cadherin, is elevated in ECs isolated from mouse brains developing early CCM lesions and in cultured ECs with CCM1 or CCM2 depletion. Furthermore, we show miR-27a acts downstream of kruppel-like factor (KLF)2 and KLF4, two known key transcription factors involved in CCM lesion development. Using CD5-2 (a target site blocker [TSB]) to prevent the miR-27a/VE-cadherin mRNA interaction, we present a potential therapy to increase VE-cadherin expression and thus rescue the abnormal vascular integrity. In CCM1- or CCM2-depleted ECs, CD5-2 reduces monolayer permeability, and in Ccm1 heterozygous mice, it restores dermal vessel barrier function. In a neonatal mouse model of CCM disease, CD5-2 normalizes vasculature and reduces vascular leakage in the lesions, inhibits the development of large lesions, and significantly reduces the size of established lesions in the hindbrain. Furthermore, CD5-2 limits the accumulation of inflammatory cells in the lesion area. Our work has established that VE-cadherin is a potential therapeutic target for normalization of the vasculature and highlights that targeting miR-27a/VE-cadherin interaction by CD5-2 is a potential novel therapy for the devastating disease, CCM. Cerebral cavernous malformation (CCM) is a disease for which, hitherto, surgery has been the only option. This study shows that a potential therapeutic, CD5-2, inhibits lesion development and vascular leak in the brains of CCM neonatal mice by targeting the endothelial cell–specific adhesion molecule VE-cadherin and restoring the vascular integrity of CCM lesions.
Collapse
Affiliation(s)
- Jia Li
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, Australia
- Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Yang Zhao
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, Australia
| | - Jaesung Choi
- Laboratory of Cardiovascular Signaling, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, Australia
| | - Ka Ka Ting
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, Australia
| | - Paul Coleman
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, Australia
| | - Jinbiao Chen
- Liver Injury and Cancer Program, Centenary Institute, The University of Sydney, Sydney, Australia
| | - Victoria C. Cogger
- Aging and Alzheimers Institute and ANZAC Research Institute and Concord Hospital, Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Li Wan
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, Australia
| | - Zhongsong Shi
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | | | - Xiangjian Zheng
- Laboratory of Cardiovascular Signaling, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, Australia
| | - Mathew A. Vadas
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, Australia
| | - Jennifer R. Gamble
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, Australia
- * E-mail:
| |
Collapse
|
14
|
Abstract
Epithelial cells form highly organized polarized sheets with characteristic cell morphologies and tissue architecture. Cell–cell adhesion and intercellular communication are prerequisites of such cohesive sheets of cells, and cell connectivity is mediated through several junctional assemblies, namely desmosomes, adherens, tight and gap junctions. These cell–cell junctions form signalling hubs that not only mediate cell–cell adhesion but impact on multiple aspects of cell behaviour, helping to coordinate epithelial cell shape, polarity and function. This review will focus on the tight and adherens junctions, constituents of the apical junctional complex, and aims to provide a comprehensive overview of the complex signalling that underlies junction assembly, integrity and plasticity.
Collapse
Affiliation(s)
- Alexandra D Rusu
- School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| | - Marios Georgiou
- School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| |
Collapse
|
15
|
Belloni E, Di Matteo A, Pradella D, Vacca M, Wyatt CDR, Alfieri R, Maffia A, Sabbioneda S, Ghigna C. Gene Expression Profiles Controlled by the Alternative Splicing Factor Nova2 in Endothelial Cells. Cells 2019; 8:cells8121498. [PMID: 31771184 PMCID: PMC6953062 DOI: 10.3390/cells8121498] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/11/2019] [Accepted: 11/20/2019] [Indexed: 02/07/2023] Open
Abstract
Alternative splicing (AS) plays an important role in expanding the complexity of the human genome through the production of specialized proteins regulating organ development and physiological functions, as well as contributing to several pathological conditions. How AS programs impact on the signaling pathways controlling endothelial cell (EC) functions and vascular development is largely unknown. Here we identified, through RNA-seq, changes in mRNA steady-state levels in ECs caused by the neuro-oncological ventral antigen 2 (Nova2), a key AS regulator of the vascular morphogenesis. Bioinformatics analyses identified significant enrichment for genes regulated by peroxisome proliferator-activated receptor-gamma (Ppar-γ) and E2F1 transcription factors. We also showed that Nova2 in ECs controlled the AS profiles of Ppar-γ and E2F dimerization partner 2 (Tfdp2), thus generating different protein isoforms with distinct function (Ppar-γ) or subcellular localization (Tfdp2). Collectively, our results supported a mechanism whereby Nova2 integrated splicing decisions in order to regulate Ppar-γ and E2F1 activities. Our data added a layer to the sequential series of events controlled by Nova2 in ECs to orchestrate vascular biology.
Collapse
Affiliation(s)
- Elisa Belloni
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
| | - Anna Di Matteo
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
| | - Davide Pradella
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
| | - Margherita Vacca
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
| | - Christopher D. R. Wyatt
- Centre for Biodiversity and Environment Research, University College London, Gower Street, London WC1E 6BT, UK
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr Aiguader 88, 08003 Barcelona, Spain
- Universitat Pompeu Fabra, Plaça de la Mercè, 10-12, 08002 Barcelona, Spain
| | - Roberta Alfieri
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
| | - Antonio Maffia
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
| | - Simone Sabbioneda
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
| | - Claudia Ghigna
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
- Correspondence:
| |
Collapse
|
16
|
Mundi S, Massaro M, Scoditti E, Carluccio MA, van Hinsbergh VWM, Iruela-Arispe ML, De Caterina R. Endothelial permeability, LDL deposition, and cardiovascular risk factors-a review. Cardiovasc Res 2019; 114:35-52. [PMID: 29228169 DOI: 10.1093/cvr/cvx226] [Citation(s) in RCA: 216] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 12/05/2017] [Indexed: 12/21/2022] Open
Abstract
Early atherosclerosis features functional and structural changes in the endothelial barrier function that affect the traffic of molecules and solutes between the vessel lumen and the vascular wall. Such changes are mechanistically related to the development of atherosclerosis. Proatherogenic stimuli and cardiovascular risk factors, such as dyslipidaemias, diabetes, obesity, and smoking, all increase endothelial permeability sharing a common signalling denominator: an imbalance in the production/disposal of reactive oxygen species (ROS), broadly termed oxidative stress. Mostly as a consequence of the activation of enzymatic systems leading to ROS overproduction, proatherogenic factors lead to a pro-inflammatory status that translates in changes in gene expression and functional rearrangements, including changes in the transendothelial transport of molecules, leading to the deposition of low-density lipoproteins (LDL) and the subsequent infiltration of circulating leucocytes in the intima. In this review, we focus on such early changes in atherogenesis and on the concept that proatherogenic stimuli and risk factors for cardiovascular disease, by altering the endothelial barrier properties, co-ordinately trigger the accumulation of LDL in the intima and ultimately plaque formation.
Collapse
Affiliation(s)
- Santa Mundi
- Department of Biological and Environmental Science and Technology (DISTEBA), University of Salento, via Monteroni, 73100, Lecce, Italy
| | - Marika Massaro
- National Research Council (CNR), Department of Biomedical sciences, Institute of Clinical Physiology, Via Monteroni, 73100, Lecce, Italy
| | - Egeria Scoditti
- National Research Council (CNR), Department of Biomedical sciences, Institute of Clinical Physiology, Via Monteroni, 73100, Lecce, Italy
| | - Maria Annunziata Carluccio
- National Research Council (CNR), Department of Biomedical sciences, Institute of Clinical Physiology, Via Monteroni, 73100, Lecce, Italy
| | - Victor W M van Hinsbergh
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat, NL-1081 BT, Amsterdam, The Netherlands
| | - Marial Luisa Iruela-Arispe
- Department of Molecular, Cell and Developmental Biology and Molecular Biology Institute, University of California, 610 Charles E Young Dr S, 90095, Los Angeles, USA; and
| | - Raffaele De Caterina
- Department of Neuroscience, Imaging and Clinical Science and Institute of Advanced Biomedical Technologies, University G. D'Annunzio, via dei Vestini, 66100 Chieti, Italy
| |
Collapse
|
17
|
Angiolini F, Belloni E, Giordano M, Campioni M, Forneris F, Paronetto MP, Lupia M, Brandas C, Pradella D, Di Matteo A, Giampietro C, Jodice G, Luise C, Bertalot G, Freddi S, Malinverno M, Irimia M, Moulton JD, Summerton J, Chiapparino A, Ghilardi C, Giavazzi R, Nyqvist D, Gabellini D, Dejana E, Cavallaro U, Ghigna C. A novel L1CAM isoform with angiogenic activity generated by NOVA2-mediated alternative splicing. eLife 2019; 8:44305. [PMID: 30829570 PMCID: PMC6398979 DOI: 10.7554/elife.44305] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 02/08/2019] [Indexed: 12/13/2022] Open
Abstract
The biological players involved in angiogenesis are only partially defined. Here, we report that endothelial cells (ECs) express a novel isoform of the cell-surface adhesion molecule L1CAM, termed L1-ΔTM. The splicing factor NOVA2, which binds directly to L1CAM pre-mRNA, is necessary and sufficient for the skipping of L1CAM transmembrane domain in ECs, leading to the release of soluble L1-ΔTM. The latter exerts high angiogenic function through both autocrine and paracrine activities. Mechanistically, L1-ΔTM-induced angiogenesis requires fibroblast growth factor receptor-1 signaling, implying a crosstalk between the two molecules. NOVA2 and L1-ΔTM are overexpressed in the vasculature of ovarian cancer, where L1-ΔTM levels correlate with tumor vascularization, supporting the involvement of NOVA2-mediated L1-ΔTM production in tumor angiogenesis. Finally, high NOVA2 expression is associated with poor outcome in ovarian cancer patients. Our results point to L1-ΔTM as a novel, EC-derived angiogenic factor which may represent a target for innovative antiangiogenic therapies. Growing tumors stimulate the formation of new blood vessels to supply the oxygen and nutrients the cancerous cells need to stay alive. Stopping tumors from forming the blood vessels could therefore help us to treat cancer. To do so, we need to understand how different proteins control when and how blood vessels develop. Cells make proteins by first ‘transcribing’ genes to form RNA molecules. In many cases, the RNA then goes through a process called alternative splicing. Proteins known as splicing factors cut out different segments of the RNA molecule and stick together the remaining segments to form templates for protein production. This enables a single gene to produce many different variants of a protein. Angiolini, Belloni, Giordano et al. have now studied mouse and human versions of the cells that line the blood vessels grown by tumors. This revealed that a splicing factor called NOVA2 targets a protein called L1CAM, which is normally responsible for gluing adjacent cells together. Angiolini et al. found that NOVA2 splices L1CAM into a form not seen before. Instead of remaining anchored to cell surfaces, the newly identified form of L1CAM is released into the blood circulation, where it stimulates new blood vessels to grow. Samples taken from the blood vessels of human ovarian tumors showed high levels of both NOVA2 and the modified form of L1CAM, while blood vessels in healthy tissue contain no, or very low levels of both proteins. Therefore, if the new form of L1CAM can be detected in the blood, it could be used to help cancer diagnosis, and to indicate which patients would benefit from treatments that restrict the growth of blood vessels in tumors. Further work is now needed to explore these possibilities.
Collapse
Affiliation(s)
- Francesca Angiolini
- Unit of Gynecological Oncology Research, Program of Gynecological Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Elisa Belloni
- Istituto di Genetica Molecolare, Consiglio Nazionale delle Ricerche, Pavia, Italy
| | - Marco Giordano
- Unit of Gynecological Oncology Research, Program of Gynecological Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Matteo Campioni
- The Armenise-Harvard Laboratory of Structural Biology, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Federico Forneris
- The Armenise-Harvard Laboratory of Structural Biology, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Maria Paola Paronetto
- Department of Movement, Human and Health Sciences, Università degli Studi di Roma "Foro Italico", Rome, Italy
| | - Michela Lupia
- Unit of Gynecological Oncology Research, Program of Gynecological Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Chiara Brandas
- Istituto di Genetica Molecolare, Consiglio Nazionale delle Ricerche, Pavia, Italy
| | - Davide Pradella
- Istituto di Genetica Molecolare, Consiglio Nazionale delle Ricerche, Pavia, Italy.,Università degli Studi di Pavia, Pavia, Italy
| | - Anna Di Matteo
- Istituto di Genetica Molecolare, Consiglio Nazionale delle Ricerche, Pavia, Italy
| | | | - Giovanna Jodice
- Molecular Medicine Program, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Chiara Luise
- Molecular Medicine Program, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Giovanni Bertalot
- Molecular Medicine Program, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Stefano Freddi
- Molecular Medicine Program, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Manuel Irimia
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | | | | | - Antonella Chiapparino
- The Armenise-Harvard Laboratory of Structural Biology, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Carmen Ghilardi
- Laboratory of Biology and Treatment of Metastasis, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Raffaella Giavazzi
- Laboratory of Biology and Treatment of Metastasis, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Daniel Nyqvist
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Davide Gabellini
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisabetta Dejana
- FIRC Institute of Molecular Oncology, Milan, Italy.,Rudbeck Laboratory and Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ugo Cavallaro
- Unit of Gynecological Oncology Research, Program of Gynecological Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Claudia Ghigna
- Istituto di Genetica Molecolare, Consiglio Nazionale delle Ricerche, Pavia, Italy
| |
Collapse
|
18
|
Szymborska A, Gerhardt H. Hold Me, but Not Too Tight-Endothelial Cell-Cell Junctions in Angiogenesis. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a029223. [PMID: 28851748 DOI: 10.1101/cshperspect.a029223] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Endothelial cell-cell junctions must perform seemingly incompatible tasks during vascular development-providing stable connections that prevent leakage, while allowing dynamic cellular rearrangements during sprouting, anastomosis, lumen formation, and functional remodeling of the vascular network. This review aims to highlight recent insights into the molecular mechanisms governing endothelial cell-cell adhesion in the context of vascular development.
Collapse
Affiliation(s)
- Anna Szymborska
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin
| | - Holger Gerhardt
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany.,Vascular Patterning Laboratory, Center for Cancer Biology, VIB, Department of Oncology, KU Leuven, 3000 Leuven, Belgium.,DZHK (German Centre for Cardiovascular Research), partner site Berlin.,Berlin Institute of Health (BIH), 10178 Berlin, Germany
| |
Collapse
|
19
|
Helbing T, Arnold L, Wiltgen G, Hirschbihl E, Gabelmann V, Hornstein A, Esser JS, Diehl P, Grundmann S, Busch HJ, Fink K, Bode C, Moser M. Endothelial BMP4 Regulates Leukocyte Diapedesis and Promotes Inflammation. Inflammation 2018; 40:1862-1874. [PMID: 28755278 DOI: 10.1007/s10753-017-0627-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Leukocyte recruitment is a fundamental event in the response of the innate immune system to injury. This process is promoted in part by the opening of endothelial cell adherens junctions that allows leukocyte extravasation through gaps between adjacent endothelial cells. VE-cadherin is a key component of endothelial cell adherens junctions and a negative regulator of leukocyte emigration. Accumulating evidence implicates bone morphogenetic protein (BMP) 4 as a critical regulator in vascular biology, but its role in leukocyte extravasation in vitro and in vivo has not been investigated so far. To assess the impact of BMP4 on leukocyte emigration in vivo, we used the thioglycollate-induced peritonitis model. C57BL/6 mice were intraperitoneally (i.p.) injected with recombinant BMP4 in addition to thioglycollate. Compared to solvent-treated controls, we observed higher accumulation of leukocytes in the peritoneal lavage of BMP4-treated mice indicating that BMP4 promotes leukocyte diapedesis into the inflamed peritoneal cavity. Endothelial cell-specific deletion of BMP4 in mice markedly diminished leukocyte diapedesis following thioglycollate administration suggesting that endothelial BMP4 is required for leukocyte recruitment. Consistent with these in vivo results, transwell migration assays with human umbilical vein endothelial cells (HUVECs) in vitro revealed that recombinant BMP4 enhanced leukocyte transmigration through the endothelial monolayer. Conversely, silencing of endothelial BMP4 by siRNA dampened leukocyte diapedesis in vitro. Mechanistic studies showed that loss of BMP4 improved endothelial junction stability by upregulation of VE-cadherin expression in vitro and in vivo. Vice versa, treatment of HUVECs with recombinant BMP4 decreased expression of VE-cadherin and impaired endothelial junction stability shown by Western blotting and immunocytochemistry. Finally, severe endothelial damage in HUVECs in response to serum of patients collected 24 h after survived cardiac arrest was accompanied by increase in leukocyte migration in transwell assays and activation of the BMP pathway most probably by upregulation of endothelial BMP4 RNA and protein expression. Collectively, the present study provides novel evidence that endothelial BMP4 controls leukocyte recruitment through a VE-cadherin-dependent mechanism and that BMP4-induced inflammation might be involved in the pathogenesis of endothelial cell damage following successful resuscitation after cardiac arrest.
Collapse
Affiliation(s)
- Thomas Helbing
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany.
| | - Linus Arnold
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
| | - Gwendoline Wiltgen
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
| | - Eva Hirschbihl
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
| | - Valentin Gabelmann
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
| | - Alexandra Hornstein
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
| | - Jennifer S Esser
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
| | - Philipp Diehl
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
| | - Sebastian Grundmann
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
| | - Hans-Jörg Busch
- Department of Emergency Medicine, University Hospital of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Katrin Fink
- Department of Emergency Medicine, University Hospital of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph Bode
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
| | - Martin Moser
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
| |
Collapse
|
20
|
Chepizhko O, Lionetti MC, Malinverno C, Giampietro C, Scita G, Zapperi S, La Porta CAM. From jamming to collective cell migration through a boundary induced transition. SOFT MATTER 2018; 14:3774-3782. [PMID: 29713711 DOI: 10.1039/c8sm00128f] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Cell monolayers provide an interesting example of active matter, exhibiting a phase transition from flowing to jammed states as they age. Here we report experiments and numerical simulations illustrating how a jammed cellular layer rapidly reverts to a flowing state after a wound. Quantitative comparison between experiments and simulations shows that cells change their self-propulsion and alignment strength so that the system crosses a phase transition line, which we characterize by finite-size scaling in an active particle model. This wound-induced unjamming transition is found to occur generically in epithelial, endothelial and cancer cells.
Collapse
Affiliation(s)
- Oleksandr Chepizhko
- Institut für Theoretische Physik, Leopold-Franzens-Universität Innsbruck, Technikerstrasse 21a, A-6020 Innsbruck, Austria
| | | | | | | | | | | | | |
Collapse
|
21
|
Kovacs-Kasa A, Kim KM, Cherian-Shaw M, Black SM, Fulton DJ, Verin AD. Extracellular adenosine-induced Rac1 activation in pulmonary endothelium: Molecular mechanisms and barrier-protective role. J Cell Physiol 2018; 233:5736-5746. [PMID: 29168172 DOI: 10.1002/jcp.26281] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 11/14/2017] [Indexed: 12/12/2022]
Abstract
We have previously shown that Gs-coupled adenosine receptors (A2a) are primarily involved in adenosine-induced human pulmonary artery endothelial cell (HPAEC) barrier enhancement. However, the downstream events that mediate the strengthening of the endothelial cell (EC) barrier via adenosine signaling are largely unknown. In the current study, we tested the overall hypothesis that adenosine-induced Rac1 activation and EC barrier enhancement is mediated by Gs-dependent stimulation of cAMP-dependent Epac1-mediated signaling cascades. Adenoviral transduction of HPAEC with constitutively-active (C/A) Rac1 (V12Rac1) significantly increases transendothelial electrical resistance (TER) reflecting an enhancement of the EC barrier. Conversely, expression of an inactive Rac1 mutant (N17Rac1) decreases TER reflecting a compromised EC barrier. The adenosine-induced increase in TER was accompanied by activation of Rac1, decrease in contractility (MLC dephosphorylation), but not Rho inhibition. Conversely, inhibition of Rac1 activity attenuates adenosine-induced increase in TER. We next examined the role of cAMP-activated Epac1 and its putative downstream targets Rac1, Vav2, Rap1, and Tiam1. Depletion of Epac1 attenuated the adenosine-induced Rac1 activation and the increase in TER. Furthermore, silencing of Rac1 specific guanine nucleotide exchange factors (GEFs), Vav2 and Rap1a expression significantly attenuated adenosine-induced increases in TER and activation of Rac1. Depletion of Rap1b only modestly impacted adenosine-induced increases in TER and Tiam1 depletion had no effect on adenosine-induced Rac1 activation and TER. Together these data strongly suggest that Rac1 activity is required for adenosine-induced EC barrier enhancement and that the activation of Rac1 and ability to strengthen the EC barrier depends, at least in part, on cAMP-dependent Epac1/Vav2/Rap1-mediated signaling.
Collapse
Affiliation(s)
- Anita Kovacs-Kasa
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Kyung Mi Kim
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Mary Cherian-Shaw
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Stephen M Black
- Center for Lung Vascular Pathobiology, University of Arizona, Phoenix, Arizona
| | - David J Fulton
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Alexander D Verin
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| |
Collapse
|
22
|
Lagendijk AK, Yap AS, Hogan BM. Notching a New Pathway in Vascular Flow Sensing. Trends Cell Biol 2018; 28:173-175. [DOI: 10.1016/j.tcb.2017.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 12/18/2017] [Indexed: 11/16/2022]
|
23
|
Morini MF, Giampietro C, Corada M, Pisati F, Lavarone E, Cunha SI, Conze LL, O'Reilly N, Joshi D, Kjaer S, George R, Nye E, Ma A, Jin J, Mitter R, Lupia M, Cavallaro U, Pasini D, Calado DP, Dejana E, Taddei A. VE-Cadherin-Mediated Epigenetic Regulation of Endothelial Gene Expression. Circ Res 2018; 122:231-245. [PMID: 29233846 PMCID: PMC5771688 DOI: 10.1161/circresaha.117.312392] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 11/30/2017] [Accepted: 12/11/2016] [Indexed: 01/15/2023]
Abstract
RATIONALE The mechanistic foundation of vascular maturation is still largely unknown. Several human pathologies are characterized by deregulated angiogenesis and unstable blood vessels. Solid tumors, for instance, get their nourishment from newly formed structurally abnormal vessels which present wide and irregular interendothelial junctions. Expression and clustering of the main endothelial-specific adherens junction protein, VEC (vascular endothelial cadherin), upregulate genes with key roles in endothelial differentiation and stability. OBJECTIVE We aim at understanding the molecular mechanisms through which VEC triggers the expression of a set of genes involved in endothelial differentiation and vascular stabilization. METHODS AND RESULTS We compared a VEC-null cell line with the same line reconstituted with VEC wild-type cDNA. VEC expression and clustering upregulated endothelial-specific genes with key roles in vascular stabilization including claudin-5, vascular endothelial-protein tyrosine phosphatase (VE-PTP), and von Willebrand factor (vWf). Mechanistically, VEC exerts this effect by inhibiting polycomb protein activity on the specific gene promoters. This is achieved by preventing nuclear translocation of FoxO1 (Forkhead box protein O1) and β-catenin, which contribute to PRC2 (polycomb repressive complex-2) binding to promoter regions of claudin-5, VE-PTP, and vWf. VEC/β-catenin complex also sequesters a core subunit of PRC2 (Ezh2 [enhancer of zeste homolog 2]) at the cell membrane, preventing its nuclear translocation. Inhibition of Ezh2/VEC association increases Ezh2 recruitment to claudin-5, VE-PTP, and vWf promoters, causing gene downregulation. RNA sequencing comparison of VEC-null and VEC-positive cells suggested a more general role of VEC in activating endothelial genes and triggering a vascular stability-related gene expression program. In pathological angiogenesis of human ovarian carcinomas, reduced VEC expression paralleled decreased levels of claudin-5 and VE-PTP. CONCLUSIONS These data extend the knowledge of polycomb-mediated regulation of gene expression to endothelial cell differentiation and vessel maturation. The identified mechanism opens novel therapeutic opportunities to modulate endothelial gene expression and induce vascular normalization through pharmacological inhibition of the polycomb-mediated repression system.
Collapse
Affiliation(s)
- Marco F Morini
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Costanza Giampietro
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Monica Corada
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Federica Pisati
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Elisa Lavarone
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Sara I Cunha
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Lei L Conze
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Nicola O'Reilly
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Dhira Joshi
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Svend Kjaer
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Roger George
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Emma Nye
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Anqi Ma
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Jian Jin
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Richard Mitter
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Michela Lupia
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Ugo Cavallaro
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Diego Pasini
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Dinis P Calado
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Elisabetta Dejana
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.).
| | - Andrea Taddei
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.).
| |
Collapse
|
24
|
Abstract
Under physiological conditions, the arterial endothelium exerts a powerful protective influence to maintain vascular homeostasis. However, during the development of vascular disease, these protective activities are lost, and dysfunctional endothelial cells actually promote disease pathogenesis. Numerous investigations have analyzed the characteristics of dysfunctional endothelium with a view to understanding the processes responsible for the dysfunction and to determining their role in vascular pathology. This review adopts an alternate approach: reviewing the mechanisms that contribute to the initial formation of a healthy protective endothelium and on how those mechanisms may be disrupted, precipitating the appearance of dysfunctional endothelial cells and the progression of vascular disease. This approach, which highlights the role of endothelial adherens junctions and vascular endothelial-cadherin in endothelial maturation and endothelial dysfunction, provides new insight into the remarkable biology of this important cell layer and its role in vascular protection and vascular disease.
Collapse
|
25
|
Radeva MY, Waschke J. Mind the gap: mechanisms regulating the endothelial barrier. Acta Physiol (Oxf) 2018; 222. [PMID: 28231640 DOI: 10.1111/apha.12860] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 11/21/2016] [Accepted: 02/16/2017] [Indexed: 12/11/2022]
Abstract
The endothelial barrier consists of intercellular contacts localized in the cleft between endothelial cells, which is covered by the glycocalyx in a sievelike manner. Both types of barrier-forming junctions, i.e. the adherens junction (AJ) serving mechanical anchorage and mechanotransduction and the tight junction (TJ) sealing the intercellular space to limit paracellular permeability, are tethered to the actin cytoskeleton. Under resting conditions, the endothelium thereby builds a selective layer controlling the exchange of fluid and solutes with the surrounding tissue. However, in the situation of an inflammatory response such as in anaphylaxis or sepsis intercellular contacts disintegrate in post-capillary venules leading to intercellular gap formation. The resulting oedema can cause shock and multi-organ failure. Therefore, maintenance as well as coordinated opening and closure of interendothelial junctions is tightly regulated. The two principle underlying mechanisms comprise spatiotemporal activity control of the small GTPases Rac1 and RhoA and the balance of the phosphorylation state of AJ proteins. In the resting state, junctional Rac1 and RhoA activity is enhanced by junctional components, actin-binding proteins, cAMP signalling and extracellular cues such as sphingosine-1-phosphate (S1P) and angiopoietin-1 (Ang-1). In addition, phosphorylation of AJ components is prevented by junction-associated phosphatases including vascular endothelial protein tyrosine phosphatase (VE-PTP). In contrast, inflammatory mediators inhibiting cAMP/Rac1 signalling cause strong activation of RhoA and induce AJ phosphorylation finally leading to endocytosis and cleavage of VE-cadherin. This results in dissolution of TJs the outcome of which is endothelial barrier breakdown.
Collapse
Affiliation(s)
- M. Y. Radeva
- Institute of Anatomy and Cell Biology; Ludwig-Maximilians-Universität München; Munich Germany
| | - J. Waschke
- Institute of Anatomy and Cell Biology; Ludwig-Maximilians-Universität München; Munich Germany
| |
Collapse
|
26
|
A CDC42-centered signaling unit is a dominant positive regulator of endothelial integrity. Sci Rep 2017; 7:10132. [PMID: 28860633 PMCID: PMC5579287 DOI: 10.1038/s41598-017-10392-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 08/07/2017] [Indexed: 12/27/2022] Open
Abstract
Endothelial barrier function is carefully controlled to protect tissues from edema and damage inflicted by extravasated leukocytes. RhoGTPases, in conjunction with myriad regulatory proteins, exert both positive and negative effects on the endothelial barrier integrity. Precise knowledge about the relevant mechanisms is currently fragmented and we therefore performed a comprehensive analysis of endothelial barrier regulation by RhoGTPases and their regulators. Combining RNAi with electrical impedance measurements we quantified the relevance of 270 Rho-associated genes for endothelial barrier function. Statistical analysis identified 10 targets of which six promoted- and four reduced endothelial barrier function upon downregulation. We analyzed in more detail two of these which were not previously identified as regulators of endothelial integrity. We found that the Rac1-GEF (Guanine nucleotide Exchange Factor) TIAM2 is a positive regulator and the Cdc42(Rac1)-GAP (GTPase-Activating Protein) SYDE1 is a negative regulator of the endothelial barrier function. Finally, we found that the GAP SYDE1 is part of a Cdc42-centered signaling unit, also comprising the Cdc42-GEF FARP1 and the Cdc42 effector PAK7 which controls the integrity of the endothelial barrier. In conclusion, using a siRNA-based screen, we identified new regulators of barrier function and found that Cdc42 is a dominant positive regulator of endothelial integrity.
Collapse
|
27
|
Yi L, Huang X, Guo F, Zhou Z, Chang M, Huan J. GSK-3Beta-Dependent Activation of GEF-H1/ROCK Signaling Promotes LPS-Induced Lung Vascular Endothelial Barrier Dysfunction and Acute Lung Injury. Front Cell Infect Microbiol 2017; 7:357. [PMID: 28824887 PMCID: PMC5543036 DOI: 10.3389/fcimb.2017.00357] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 07/25/2017] [Indexed: 12/15/2022] Open
Abstract
The bacterial endotoxin or lipopolysaccharide (LPS) leads to the extensive vascular endothelial cells (EC) injury under septic conditions. Guanine nucleotide exchange factor-H1 (GEF-H1)/ROCK signaling not only involved in LPS-induced overexpression of pro-inflammatory mediator in ECs but also implicated in LPS-induced endothelial hyper-permeability. However, the mechanisms behind LPS-induced GEF-H1/ROCK signaling activation in the progress of EC injury remain incompletely understood. GEF-H1 localized on microtubules (MT) and is suppressed in its MT-bound state. MT disassembly promotes GEF-H1 release from MT and stimulates downstream ROCK-specific GEF activity. Since glycogen synthase kinase (GSK-3beta) participates in regulating MT dynamics under pathologic conditions, we examined the pivotal roles for GSK-3beta in modulating LPS-induced activation of GEF-H1/ROCK, increase of vascular endothelial permeability and severity of acute lung injury (ALI). In this study, we found that LPS induced human pulmonary endothelial cell (HPMEC) monolayers disruption accompanied by increase in GSK-3beta activity, activation of GEF-H1/ROCK signaling and decrease in beta-catenin and ZO-1 expression. Inhibition of GSK-3beta reduced HPMEC monolayers hyper-permeability and GEF-H1/ROCK activity in response to LPS. GSK-3beta/GEF-H1/ROCK signaling is implicated in regulating the expression of beta-catenin and ZO-1. In vivo, GSK-3beta inhibition attenuated LPS-induced activation of GEF-H1/ROCK pathway, lung edema and subsequent ALI. These findings present a new mechanism of GSK-3beta-dependent exacerbation of lung micro-vascular hyper-permeability and escalation of ALI via activation of GEF-H1/ROCK signaling and disruption of intracellular junctional proteins under septic condition.
Collapse
Affiliation(s)
- Lei Yi
- Department of Orthopedics, Shanghai Fengxian Central Hospital, Shanghai University of Medicine and Health Sciences Affiliated Sixth People's Hospital South CampusShanghai, China.,Department of Burn and Plastic Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| | - Xiaoqin Huang
- Department of Burn and Plastic Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| | - Feng Guo
- Department of Burn and Plastic Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| | - Zengding Zhou
- Department of Burn and Plastic Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| | - Mengling Chang
- Department of Burn and Plastic Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| | - Jingning Huan
- Department of Burn and Plastic Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai, China
| |
Collapse
|
28
|
Ebnet K, Kummer D, Steinbacher T, Singh A, Nakayama M, Matis M. Regulation of cell polarity by cell adhesion receptors. Semin Cell Dev Biol 2017; 81:2-12. [PMID: 28739340 DOI: 10.1016/j.semcdb.2017.07.032] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/12/2017] [Accepted: 07/20/2017] [Indexed: 01/01/2023]
Abstract
The ability of cells to polarize is an intrinsic property of almost all cells and is required for the devlopment of most multicellular organisms. To develop cell polarity, cells integrate various signals derived from intrinsic as well as extrinsic sources. In the recent years, cell-cell adhesion receptors have turned out as important regulators of cellular polarization. By interacting with conserved cell polarity proteins, they regulate the recruitment of polarity complexes to specific sites of cell-cell adhesion. By initiating intracellular signaling cascades at those sites, they trigger their specific subcellular activation. Not surprisingly, cell-cell adhesion receptors regulate diverse aspects of cell polarity, including apico-basal polarity in epithelial and endothelial cells, front-to-rear polarity in collectively migrating cells, and planar cell polarity during organ development. Here, we review the recent developments highlighting the central roles of cell-cell adhesion molecules in the development of cell polarity.
Collapse
Affiliation(s)
- Klaus Ebnet
- Institute-associated Research Group: Cell adhesion and cell polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Germany; Interdisciplinary Clinical Research Center (IZKF), University of Münster, Germany; Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Germany.
| | - Daniel Kummer
- Institute-associated Research Group: Cell adhesion and cell polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Germany; Interdisciplinary Clinical Research Center (IZKF), University of Münster, Germany
| | - Tim Steinbacher
- Institute-associated Research Group: Cell adhesion and cell polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Germany; Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Germany
| | - Amrita Singh
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Germany; Institute of Cell Biology, ZMBE, University of Münster, Germany
| | - Masanori Nakayama
- Laboratory for Cell Polarity and Organogenesis, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Maja Matis
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Germany; Institute of Cell Biology, ZMBE, University of Münster, Germany.
| |
Collapse
|
29
|
β-Catenin Is Required for Endothelial Cyp1b1 Regulation Influencing Metabolic Barrier Function. J Neurosci 2017; 36:8921-35. [PMID: 27559173 DOI: 10.1523/jneurosci.0148-16.2016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 06/27/2016] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED The canonical Wnt/β-catenin signaling pathway is crucial for blood-brain barrier (BBB) formation in brain endothelial cells. Although glucose transporter 1, claudin-3, and plasmalemma vesicular-associated protein have been identified as Wnt/β-catenin targets in brain endothelial cells, further downstream targets relevant to BBB formation and function are incompletely explored. By Affymetrix expression analysis, we show that the cytochrome P450 enzyme Cyp1b1 was significantly decreased in β-catenin-deficient mouse endothelial cells, whereas its close homolog Cyp1a1 was upregulated in an aryl hydrocarbon receptor-dependent manner, hence indicating that β-catenin is indispensable for Cyp1b1 but not for Cyp1a1 expression. Functionally, Cyp1b1 could generate retinoic acid from retinol leading to cell-autonomous induction of the barrier-related ATP-binding cassette transporter P-glycoprotein. Cyp1b1 could also generate 20-hydroxyeicosatetraenoic acid from arachidonic acid, decreasing endothelial barrier function in vitro In mice in vivo pharmacological inhibition of Cyp1b1 increased BBB permeability for small molecular tracers, and Cyp1b1 was downregulated in glioma vessels in which BBB function is lost. Hence, we propose Cyp1b1 as a target of β-catenin indirectly influencing BBB properties via its metabolic activity, and as a potential target for modulating barrier function in endothelial cells. SIGNIFICANCE STATEMENT Wnt/β-catenin signaling is crucial for blood-brain barrier (BBB) development and maintenance; however, its role in regulating metabolic characteristics of endothelial cells is unclear. We provide evidence that β-catenin influences endothelial metabolism by transcriptionally regulating the cytochrome P450 enzyme Cyp1b1 Furthermore, expression of its close homolog Cyp1a1 was inhibited by β-catenin. Functionally, Cyp1b1 generated retinoic acid as well as 20-hydroxyeicosatetraenoic acid that regulated P-glycoprotein and junction proteins, respectively, thereby modulating BBB properties. Inhibition of Cyp1b1 in vivo increased BBB permeability being in line with its downregulation in glioma endothelia, potentially implicating Cyp1b1 in other brain pathologies. In conclusion, Wnt/β-catenin signaling regulates endothelial metabolic barrier function through Cyp1b1 transcription.
Collapse
|
30
|
Chang F, Flavahan S, Flavahan NA. Impaired activity of adherens junctions contributes to endothelial dilator dysfunction in ageing rat arteries. J Physiol 2017; 595:5143-5158. [PMID: 28561330 DOI: 10.1113/jp274189] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/17/2017] [Indexed: 12/18/2022] Open
Abstract
KEY POINTS Ageing-induced endothelial dysfunction contributes to organ dysfunction and progression of cardiovascular disease. VE-cadherin clustering at adherens junctions promotes protective endothelial functions, including endothelium-dependent dilatation. Ageing increased internalization and degradation of VE-cadherin, resulting in impaired activity of adherens junctions. Inhibition of VE-cadherin clustering at adherens junctions (function-blocking antibody; FBA) reduced endothelial dilatation in young arteries but did not affect the already impaired dilatation in old arteries. After junctional disruption with the FBA, dilatation was similar in young and old arteries. Src tyrosine kinase activity and tyrosine phosphorylation of VE-cadherin were increased in old arteries. Src inhibition increased VE-cadherin at adherens junctions and increased endothelial dilatation in old, but not young, arteries. Src inhibition did not increase dilatation in old arteries treated with the VE-cadherin FBA. Ageing impairs the activity of adherens junctions, which contributes to endothelial dilator dysfunction. Restoring the activity of adherens junctions could be of therapeutic benefit in vascular ageing. ABSTRACT Endothelial dilator dysfunction contributes to pathological vascular ageing. Experiments assessed whether altered activity of endothelial adherens junctions (AJs) might contribute to this dysfunction. Aortas and tail arteries were isolated from young (3-4 months) and old (22-24 months) F344 rats. VE-cadherin immunofluorescent staining at endothelial AJs and AJ width were reduced in old compared to young arteries. A 140 kDa VE-cadherin species was present on the cell surface and in TTX-insoluble fractions, consistent with junctional localization. Levels of the 140 kDa VE-cadherin were decreased, whereas levels of a TTX-soluble 115 kDa VE-cadherin species were increased in old compared to young arteries. Acetylcholine caused endothelium-dependent dilatation that was decreased in old compared to young arteries. Disruption of VE-cadherin clustering at AJs (function-blocking antibody, FBA) inhibited dilatation to acetylcholine in young, but not old, arteries. After the FBA, there was no longer any difference in dilatation between old and young arteries. Src activity and tyrosine phosphorylation of VE-cadherin were increased in old compared to young arteries. In old arteries, Src inhibition (saracatinib) increased: (i) 140 kDa VE-cadherin in the TTX-insoluble fraction, (ii) VE-cadherin intensity at AJs, (iii) AJ width, and (iv) acetylcholine dilatation. In old arteries treated with the FBA, saracatinib no longer increased acetylcholine dilatation. Saracatinib did not affect dilatation in young arteries. Therefore, ageing impairs AJ activity, which appears to reflect Src-induced phosphorylation, internalization and degradation of VE-cadherin. Moreover, impaired AJ activity can account for the endothelial dilator dysfunction in old arteries. Restoring endothelial AJ activity may be a novel therapeutic approach to vascular ageing.
Collapse
Affiliation(s)
- Fumin Chang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Sheila Flavahan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Nicholas A Flavahan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
31
|
Ramos CJ, Antonetti DA. The role of small GTPases and EPAC-Rap signaling in the regulation of the blood-brain and blood-retinal barriers. Tissue Barriers 2017. [PMID: 28632993 DOI: 10.1080/21688370.2017.1339768] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Maintenance and regulation of the vascular endothelial cell junctional complex is critical for proper barrier function of the blood-brain barrier (BBB) and the highly related blood-retinal barrier (BRB) that help maintain proper neuronal environment. Recent research has demonstrated that the junctional complex is actively maintained and can be dynamically regulated. Studies focusing on the mechanisms of barrier formation, maintenance, and barrier disruption have been of interest to understanding development of the BBB and BRB and identifying a means for therapeutic intervention for diseases ranging from brain tumors and dementia to blinding eye diseases. Research has increasingly revealed that small GTPases play a critical role in both barrier formation and disruption mechanisms. This review will summarize the current data on small GTPases in barrier regulation with an emphasis on the EPAC-Rap1 signaling pathway to Rho in endothelial barriers, as well as explore its potential involvement in paracellular flux and transcytosis regulation.
Collapse
Affiliation(s)
- Carla J Ramos
- a Department of Ophthalmology and Visual Sciences , University of Michigan , Ann Arbor , MI USA
| | - David A Antonetti
- a Department of Ophthalmology and Visual Sciences , University of Michigan , Ann Arbor , MI USA
| |
Collapse
|
32
|
Komarova YA, Kruse K, Mehta D, Malik AB. Protein Interactions at Endothelial Junctions and Signaling Mechanisms Regulating Endothelial Permeability. Circ Res 2017; 120:179-206. [PMID: 28057793 DOI: 10.1161/circresaha.116.306534] [Citation(s) in RCA: 336] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 10/04/2016] [Accepted: 10/06/2016] [Indexed: 12/31/2022]
Abstract
The monolayer of endothelial cells lining the vessel wall forms a semipermeable barrier (in all tissue except the relatively impermeable blood-brain and inner retinal barriers) that regulates tissue-fluid homeostasis, transport of nutrients, and migration of blood cells across the barrier. Permeability of the endothelial barrier is primarily regulated by a protein complex called adherens junctions. Adherens junctions are not static structures; they are continuously remodeled in response to mechanical and chemical cues in both physiological and pathological settings. Here, we discuss recent insights into the post-translational modifications of junctional proteins and signaling pathways regulating plasticity of adherens junctions and endothelial permeability. We also discuss in the context of what is already known and newly defined signaling pathways that mediate endothelial barrier leakiness (hyperpermeability) that are important in the pathogenesis of cardiovascular and lung diseases and vascular inflammation.
Collapse
Affiliation(s)
- Yulia A Komarova
- From the Department of Pharmacology and the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago
| | - Kevin Kruse
- From the Department of Pharmacology and the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago
| | - Dolly Mehta
- From the Department of Pharmacology and the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago
| | - Asrar B Malik
- From the Department of Pharmacology and the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago.
| |
Collapse
|
33
|
Aragon-Sanabria V, Pohler SE, Eswar VJ, Bierowski M, Gomez EW, Dong C. VE-Cadherin Disassembly and Cell Contractility in the Endothelium are Necessary for Barrier Disruption Induced by Tumor Cells. Sci Rep 2017; 7:45835. [PMID: 28393886 PMCID: PMC5385522 DOI: 10.1038/srep45835] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 03/03/2017] [Indexed: 12/20/2022] Open
Abstract
During metastasis, breakdown of the endothelial barrier is critical for tumor cell extravasation through blood vessel walls and is mediated by a combination of tumor secreted soluble factors and receptor-ligand interactions. However, a complete mechanism governing tumor cell transendothelial migration remains unclear. Here, we investigate the roles of tumor-associated signals in regulating endothelial cell contractility and adherens junction disassembly leading to endothelial barrier breakdown. We show that Src mediates VE-cadherin disassembly in response to metastatic melanoma cells. Through the use of pharmacological inhibitors of cytoskeletal contractility we find that endothelial cell contractility is responsive to interactions with metastatic cancer cells and that reducing endothelial cell contractility abrogates migration of melanoma cells across endothelial monolayers. Furthermore, we find that a combination of tumor secreted soluble factors and receptor-ligand interactions mediate activation of Src within endothelial cells that is necessary for phosphorylation of VE-cadherin and for breakdown of the endothelial barrier. Together, these results provide insight into how tumor cell signals act in concert to modulate cytoskeletal contractility and adherens junctions disassembly during extravasation and may aid in identification of therapeutic targets to block metastasis.
Collapse
Affiliation(s)
- Virginia Aragon-Sanabria
- Department of Biomedical Engineering, Pennsylvania State university, University Park, PA, 16802, USA
| | - Steven E. Pohler
- Department of Chemical Engineering, Pennsylvania State University, University Park, PA, 16802, USA
| | - Vikram J. Eswar
- Department of Biomedical Engineering, Pennsylvania State university, University Park, PA, 16802, USA
| | - Matthew Bierowski
- Department of Chemical Engineering, Pennsylvania State University, University Park, PA, 16802, USA
| | - Esther W. Gomez
- Department of Biomedical Engineering, Pennsylvania State university, University Park, PA, 16802, USA
- Department of Chemical Engineering, Pennsylvania State University, University Park, PA, 16802, USA
| | - Cheng Dong
- Department of Biomedical Engineering, Pennsylvania State university, University Park, PA, 16802, USA
| |
Collapse
|
34
|
Rudimov EG, Buravkova LB. Gravisensitivity of endothelial cells: the role of cytoskeleton and adhesion molecules. ACTA ACUST UNITED AC 2017. [DOI: 10.1134/s0362119716060177] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
35
|
Wang YHW, Meyer RD, Bondzie PA, Jiang Y, Rahimi I, Rezazadeh K, Mehta M, Laver NMV, Costello CE, Rahimi N. IGPR-1 Is Required for Endothelial Cell-Cell Adhesion and Barrier Function. J Mol Biol 2016; 428:5019-5033. [PMID: 27838321 DOI: 10.1016/j.jmb.2016.11.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 11/02/2016] [Accepted: 11/03/2016] [Indexed: 12/22/2022]
Abstract
Endothelial cell (EC) barrier function plays a prevalent regulatory mechanism for the integrity and homeostasis of blood vessels and modulates angiogenesis and immune responses. Cell adhesion molecules (CAMs) play a central role in the barrier function of ECs. Although Ig-containing and proline-rich receptor-1(IGPR-1) was recently identified as a novel CAM expressed in ECs, the molecular mechanisms underlying the function of IGPR-1 in ECs remain uncharacterized. In this report, we investigated the role of IGPR-1 in EC barrier function and the molecular mechanism of its activation in ECs. We demonstrate that IGPR-1 is localized to endothelial adherens junctions and, through trans-homophilic dimerization, regulates endothelial cell-cell adhesion and barrier function. Trans-homophilic dimerization of IGPR-1 stimulates the phosphorylation of serine 220 (Ser220), which is required for IGPR-1 to regulate endothelial barrier function and angiogenesis. Moreover, IGPR-1 chimera, which mimics the trans-homophilic dimerization of IGPR-1, induced a sustained phosphorylation of Ser220 upon stimulation with a ligand. Coordinated dimerization of IGPR-1 and its homophilic interaction modulates its adhesive function and Ser220 phosphorylation. This adhesive function of IGPR-1 contributes to the barrier function of ECs.
Collapse
Affiliation(s)
- Yun Hwa Walter Wang
- Department of Pathology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Rosana D Meyer
- Department of Pathology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Philip A Bondzie
- Department of Pathology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Yan Jiang
- Department of Biochemistry and Center for Biomedical Mass Spectrometry, School of Medicine, Boston University, Boston, MA, 02118 USA
| | - Ida Rahimi
- Department of Pathology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Kobra Rezazadeh
- Department of Pathology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Manisha Mehta
- Department of Pathology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Nora M V Laver
- Department of Ophthalmology, Tufts University School of Medicine, Boston, MA 02116, USA
| | - Catherine E Costello
- Department of Biochemistry and Center for Biomedical Mass Spectrometry, School of Medicine, Boston University, Boston, MA, 02118 USA
| | - Nader Rahimi
- Department of Pathology, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
36
|
Abstract
Dense monolayers of living cells display intriguing relaxation dynamics, reminiscent of soft and glassy materials close to the jamming transition, and migrate collectively when space is available, as in wound healing or in cancer invasion. Here we show that collective cell migration occurs in bursts that are similar to those recorded in the propagation of cracks, fluid fronts in porous media, and ferromagnetic domain walls. In analogy with these systems, the distribution of activity bursts displays scaling laws that are universal in different cell types and for cells moving on different substrates. The main features of the invasion dynamics are quantitatively captured by a model of interacting active particles moving in a disordered landscape. Our results illustrate that collective motion of living cells is analogous to the corresponding dynamics in driven, but inanimate, systems.
Collapse
|
37
|
Breast cancer-secreted miR-939 downregulates VE-cadherin and destroys the barrier function of endothelial monolayers. Cancer Lett 2016; 384:94-100. [PMID: 27693459 DOI: 10.1016/j.canlet.2016.09.013] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 08/30/2016] [Accepted: 09/01/2016] [Indexed: 11/21/2022]
Abstract
Exosomes-secreted microRNAs play an important role in metastatic spread. During this process breast cancer cells acquire the ability to transmigrate through blood vessels by inducing changes in the endothelial barrier. We focused on miR-939 that is predicted to target VE-cadherin, a component of adherens junction involved in vessel permeability. By in silico analysis miR-939 was found highly expressed in the basal-like tumor subtypes and in our cohort of 63 triple-negative breast cancers (TNBCs) its expression significantly interacted with lymph node status in predicting disease-free survival probability. We demonstrated, in vitro, that miR-939 directly targets VE-cadherin leading to an increase in HUVECs monolayer permeability. MDA-MB-231 cells transfected with a miR-939 mimic, released miR-939 in exosomes that, once internalized in endothelial cells, favored trans-endothelial migration of MDA-MB-231-GFP cells by the disruption of the endothelial barrier. Notably, when up taken in endothelial cells exosomes caused VE-cadherin down-regulation specifically through miR-939 as we demonstrated by inhibiting miR-939 expression in exosomes-releasing TNBC cells. Together, our data indentify an extracellular pro-tumorigenic role for tumor-derived, exosome-associated miR-939 that can explain its association with worse prognosis in TNBCs.
Collapse
|
38
|
Barabutis N, Verin A, Catravas JD. Regulation of pulmonary endothelial barrier function by kinases. Am J Physiol Lung Cell Mol Physiol 2016; 311:L832-L845. [PMID: 27663990 DOI: 10.1152/ajplung.00233.2016] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 09/15/2016] [Indexed: 12/15/2022] Open
Abstract
The pulmonary endothelium is the target of continuous physiological and pathological stimuli that affect its crucial barrier function. The regulation, defense, and repair of endothelial barrier function require complex biochemical processes. This review examines the role of endothelial phosphorylating enzymes, kinases, a class with profound, interdigitating influences on endothelial permeability and lung function.
Collapse
Affiliation(s)
- Nektarios Barabutis
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia
| | - Alexander Verin
- Vascular Biology Center, Augusta University, Augusta, Georgia; and
| | - John D Catravas
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia, .,School of Medical Diagnostic and Translational Sciences, College of Health Sciences, Old Dominion University, Norfolk, Virginia
| |
Collapse
|
39
|
Giampietro C. VE-cadherin complex plasticity: EPS8 and YAP play relay at adherens junctions. Tissue Barriers 2016; 4:e1232024. [PMID: 28123926 DOI: 10.1080/21688370.2016.1232024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 08/24/2016] [Accepted: 08/29/2016] [Indexed: 01/24/2023] Open
Abstract
The vascular endothelium is a selective barrier that separates the organs from the circulating blood. The endothelium has a wide variety of functions controlled by cell-to-cell junctions and in particular by Vascular Endothelial cadherin (VE-cadherin) complexes. Recent research identified the epidermal growth factor receptor kinase substrate 8 (EPS8) and the co-transcriptional regulator yes-associated protein (YAP) as new components of the adherens junction complexes. The binding of these 2 proteins to VE-cadherin determines the formation of different specialized adhesive structures contributing to the dynamic control of vascular permeability. This commentary will summarize what is currently known about the role of EPS8 and YAP in the modification of molecular organization and intracellular signaling of adherens junction complexes, and their potential multiple effects on vascular homeostasis.
Collapse
Affiliation(s)
- Costanza Giampietro
- Department of Biosciences, University of Milan, Milan, Italy; IFOM-The FIRC Institute of Molecular Oncology, Milan, Italy
| |
Collapse
|
40
|
Brinkmann BF, Steinbacher T, Hartmann C, Kummer D, Pajonczyk D, Mirzapourshafiyi F, Nakayama M, Weide T, Gerke V, Ebnet K. VE-cadherin interacts with cell polarity protein Pals1 to regulate vascular lumen formation. Mol Biol Cell 2016; 27:2811-21. [PMID: 27466317 PMCID: PMC5025268 DOI: 10.1091/mbc.e16-02-0127] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 07/19/2016] [Indexed: 02/06/2023] Open
Abstract
Blood vessel tubulogenesis requires the establishment of apicobasal polarity of endothelial cells. A novel interaction is described of the cell adhesion molecule VE-cadherin with the cell polarity protein Pals1. The activity of VE-cadherin in regulation of endothelial lumen formation depends on its interaction with both Pals1 and Par3. Blood vessel tubulogenesis requires the formation of stable cell-to-cell contacts and the establishment of apicobasal polarity of vascular endothelial cells. Cell polarity is regulated by highly conserved cell polarity protein complexes such as the Par3-aPKC-Par6 complex and the CRB3-Pals1-PATJ complex, which are expressed by many different cell types and regulate various aspects of cell polarity. Here we describe a functional interaction of VE-cadherin with the cell polarity protein Pals1. Pals1 directly interacts with VE-cadherin through a membrane-proximal motif in the cytoplasmic domain of VE-cadherin. VE-cadherin clusters Pals1 at cell–cell junctions. Mutating the Pals1-binding motif in VE-cadherin abrogates the ability of VE-cadherin to regulate apicobasal polarity and vascular lumen formation. In a similar way, deletion of the Par3-binding motif at the C-terminus of VE-cadherin impairs apicobasal polarity and vascular lumen formation. Our findings indicate that the biological activity of VE-cadherin in regulating endothelial polarity and vascular lumen formation is mediated through its interaction with the two cell polarity proteins Pals1 and Par3.
Collapse
Affiliation(s)
- Benjamin F Brinkmann
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity,", University of Münster, 48419 Münster, Germany Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, 48419 Münster, Germany Interdisciplinary Clinical Research Center, University of Münster, 48419 Münster, Germany
| | - Tim Steinbacher
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity,", University of Münster, 48419 Münster, Germany Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, 48419 Münster, Germany
| | - Christian Hartmann
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity,", University of Münster, 48419 Münster, Germany Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, 48419 Münster, Germany Interdisciplinary Clinical Research Center, University of Münster, 48419 Münster, Germany
| | - Daniel Kummer
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity,", University of Münster, 48419 Münster, Germany Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, 48419 Münster, Germany Interdisciplinary Clinical Research Center, University of Münster, 48419 Münster, Germany
| | - Denise Pajonczyk
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity,", University of Münster, 48419 Münster, Germany Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, 48419 Münster, Germany
| | - Fatemeh Mirzapourshafiyi
- Laboratory for Cell Polarity and Organogenesis, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Masanori Nakayama
- Laboratory for Cell Polarity and Organogenesis, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Thomas Weide
- Department of Internal Medicine D, Division of Molecular Nephrology, University Hospital Münster, Albert-Schweitzer-Campus 1, University of Münster, 48419 Münster, Germany
| | - Volker Gerke
- Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, 48419 Münster, Germany Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), University of Münster, 48419 Münster, Germany
| | - Klaus Ebnet
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity,", University of Münster, 48419 Münster, Germany Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, 48419 Münster, Germany Interdisciplinary Clinical Research Center, University of Münster, 48419 Münster, Germany
| |
Collapse
|
41
|
van Buul JD, Timmerman I. Small Rho GTPase-mediated actin dynamics at endothelial adherens junctions. Small GTPases 2016; 7:21-31. [PMID: 26825121 DOI: 10.1080/21541248.2015.1131802] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
VE-cadherin-based cell-cell junctions form the major restrictive barrier of the endothelium to plasma proteins and blood cells. The function of VE-cadherin and the actin cytoskeleton are intimately linked. Vascular permeability factors and adherent leukocytes signal through small Rho GTPases to tightly regulate actin cytoskeletal rearrangements in order to open and re-assemble endothelial cell-cell junctions in a rapid and controlled manner. The Rho GTPases are activated by guanine nucleotide exchange factors (GEFs), conferring specificity and context-dependent control of cell-cell junctions. Although the molecular mechanisms that couple cadherins to actin filaments are beginning to be elucidated, specific stimulus-dependent regulation of the actin cytoskeleton at VE-cadherin-based junctions remains unexplained. Accumulating evidence has suggested that depending on the vascular permeability factor and on the subcellular localization of GEFs, cell-cell junction dynamics and organization are differentially regulated by one specific Rho GTPase. In this Commentary, we focus on new insights how the junctional actin cytoskeleton is specifically and locally regulated by Rho GTPases and GEFs in the endothelium.
Collapse
Affiliation(s)
- Jaap D van Buul
- a Department of Molecular Cell Biology , Sanquin Research and Landsteiner Laboratory, Academic Medical Center Amsterdam, University of Amsterdam , Amsterdam , the Netherlands
| | - Ilse Timmerman
- b Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory , Academic Medical Center Amsterdam, University of Amsterdam , Amsterdam , the Netherlands
| |
Collapse
|
42
|
Tsuji-Tamura K, Ogawa M. Inhibition of the PI3K-Akt and mTORC1 signaling pathways promotes the elongation of vascular endothelial cells. J Cell Sci 2016; 129:1165-78. [PMID: 26826185 DOI: 10.1242/jcs.178434] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 01/21/2016] [Indexed: 12/20/2022] Open
Abstract
Endothelial cell morphology needs to be properly regulated during angiogenesis. Vascular endothelial growth factor (VEGF) induces endothelial cell elongation, which promotes sprouting of pre-existing vessels. However, therapeutic angiogenesis using VEGF has been hampered by side effects such as elevated vascular permeability. Here, we attempted to induce endothelial cell elongation without an overdose of VEGF. By screening a library of chemical inhibitors, we identified phosphatidylinositol 3-kinase (PI3K)-Akt pathway inhibitors and mammalian target of rapamycin complex 1 (mTORC1) inhibitors as potent inducers of endothelial cell elongation. The elongation required VEGF at a low concentration, which was insufficient to elicit the same effect by itself. The elongation also depended on Foxo1, a transcription factor indispensable for angiogenesis. Interestingly, the Foxo1 dependency of the elongation was overridden by inhibition of mTORC1, but not by PI3K-Akt, under stimulation by a high concentration of VEGF. Dual inhibition of mTORC1 and mTORC2 failed to induce cell elongation, revealing mTORC2 as a positive regulator of elongation. Our findings suggest that the PI3K-Akt-Foxo1 and mTORC1-mTORC2 pathways differentially regulate endothelial cell elongation, depending on the microenvironmental levels of VEGF.
Collapse
Affiliation(s)
- Kiyomi Tsuji-Tamura
- Department of Cell Differentiation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Minetaro Ogawa
- Department of Cell Differentiation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| |
Collapse
|
43
|
|
44
|
p21-Activated Kinase 2 Regulates Endothelial Development and Function through the Bmk1/Erk5 Pathway. Mol Cell Biol 2015; 35:3990-4005. [PMID: 26391956 DOI: 10.1128/mcb.00630-15] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 09/08/2015] [Indexed: 02/03/2023] Open
Abstract
p21-activated kinases (Paks) have been shown to regulate cytoskeleton rearrangements, cell proliferation, attachment, and migration in a variety of cellular contexts, including endothelial cells. However, the role of endothelial Pak in embryo development has not been reported, and currently, there is no consensus on the endothelial function of individual Pak isoforms, in particular p21-activated kinase 2 (Pak2), the main Pak isoform expressed in endothelial cells. In this work, we employ genetic and molecular studies that show that Pak2, but not Pak1, is a critical mediator of development and maintenance of endothelial cell function. Endothelial depletion of Pak2 leads to early embryo lethality due to flawed blood vessel formation in the embryo body and yolk sac. In adult endothelial cells, Pak2 depletion leads to severe apoptosis and acute angiogenesis defects, and in adult mice, endothelial Pak2 deletion leads to increased vascular permeability. Furthermore, ubiquitous Pak2 deletion is lethal in adult mice. We show that many of these defects are mediated through a newly unveiled Pak2/Bmk1 pathway. Our results demonstrate that endothelial Pak2 is essential during embryogenesis and also for adult blood vessel maintenance, and they also pinpoint the Bmk1/Erk5 pathway as a critical mediator of endothelial Pak2 signaling.
Collapse
|
45
|
Timmerman I, Heemskerk N, Kroon J, Schaefer A, van Rijssel J, Hoogenboezem M, van Unen J, Goedhart J, Gadella TWJ, Yin T, Wu Y, Huveneers S, van Buul JD. A local VE-cadherin and Trio-based signaling complex stabilizes endothelial junctions through Rac1. J Cell Sci 2015; 128:3041-54. [PMID: 26116572 DOI: 10.1242/jcs.168674] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 06/22/2015] [Indexed: 01/08/2023] Open
Abstract
Endothelial cell-cell junctions maintain a restrictive barrier that is tightly regulated to allow dynamic responses to permeability-inducing angiogenic factors, as well as to inflammatory agents and adherent leukocytes. The ability of these stimuli to transiently remodel adherens junctions depends on Rho-GTPase-controlled cytoskeletal rearrangements. How the activity of Rho-GTPases is spatio-temporally controlled at endothelial adherens junctions by guanine-nucleotide exchange factors (GEFs) is incompletely understood. Here, we identify a crucial role for the Rho-GEF Trio in stabilizing junctions based around vascular endothelial (VE)-cadherin (also known as CDH5). Trio interacts with VE-cadherin and locally activates Rac1 at adherens junctions during the formation of nascent contacts, as assessed using a novel FRET-based Rac1 biosensor and biochemical assays. The Rac-GEF domain of Trio is responsible for the remodeling of junctional actin from radial into cortical actin bundles, a crucial step for junction stabilization. This promotes the formation of linear adherens junctions and increases endothelial monolayer resistance. Collectively, our data show the importance of spatio-temporal regulation of the actin cytoskeleton through Trio and Rac1 at VE-cadherin-based cell-cell junctions in the maintenance of the endothelial barrier.
Collapse
Affiliation(s)
- Ilse Timmerman
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam 1066 CX, The Netherlands
| | - Niels Heemskerk
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam 1066 CX, The Netherlands
| | - Jeffrey Kroon
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam 1066 CX, The Netherlands
| | - Antje Schaefer
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam 1066 CX, The Netherlands
| | - Jos van Rijssel
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam 1066 CX, The Netherlands
| | - Mark Hoogenboezem
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam 1066 CX, The Netherlands
| | - Jakobus van Unen
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam 1098 XH, The Netherlands
| | - Joachim Goedhart
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam 1098 XH, The Netherlands
| | - Theodorus W J Gadella
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam 1098 XH, The Netherlands
| | - Taofei Yin
- Center for Cell Analysis and Modelling, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Yi Wu
- Center for Cell Analysis and Modelling, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Stephan Huveneers
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam 1066 CX, The Netherlands
| | - Jaap D van Buul
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam 1066 CX, The Netherlands
| |
Collapse
|
46
|
Abstract
The unique characteristics of pulmonary circulation and alveolar-epithelial capillary-endothelial barrier allow for maintenance of the air-filled, fluid-free status of the alveoli essential for facilitating gas exchange, maintaining alveolar stability, and defending the lung against inhaled pathogens. The hallmark of pathophysiology in acute respiratory distress syndrome is the loss of the alveolar capillary permeability barrier and the presence of protein-rich edema fluid in the alveoli. This alteration in permeability and accumulation of fluid in the alveoli accompanies damage to the lung epithelium and vascular endothelium along with dysregulated inflammation and inappropriate activity of leukocytes and platelets. In addition, there is uncontrolled activation of coagulation along with suppression of fibrinolysis and loss of surfactant. These pathophysiological changes result in the clinical manifestations of acute respiratory distress syndrome, which include hypoxemia, radiographic opacities, decreased functional residual capacity, increased physiologic deadspace, and decreased lung compliance. Resolution of acute respiratory distress syndrome involves the migration of cells to the site of injury and re-establishment of the epithelium and endothelium with or without the development of fibrosis. Most of the data related to acute respiratory distress syndrome, however, originate from studies in adults or in mature animals with very few studies performed in children or juvenile animals. The lack of studies in children is particularly problematic because the lungs and immune system are still developing during childhood and consequently the pathophysiology of pediatric acute respiratory distress syndrome may differ in significant ways from that seen in acute respiratory distress syndrome in adults. This article describes what is known of the pathophysiologic processes of pediatric acute respiratory distress syndrome as we know it today while also presenting the much greater body of evidence on these processes as elucidated by adult and animal studies. It is also our expressed intent to generate enthusiasm for larger and more in-depth investigations of the mechanisms of disease and repair specific to children in the years to come.
Collapse
|
47
|
van Buul JD, Geerts D, Huveneers S. Rho GAPs and GEFs: controling switches in endothelial cell adhesion. Cell Adh Migr 2015; 8:108-24. [PMID: 24622613 PMCID: PMC4049857 DOI: 10.4161/cam.27599] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Within blood vessels, endothelial cell–cell and cell–matrix adhesions are crucial to preserve barrier function, and these adhesions are tightly controlled during vascular development, angiogenesis, and transendothelial migration of inflammatory cells. Endothelial cellular signaling that occurs via the family of Rho GTPases coordinates these cell adhesion structures through cytoskeletal remodelling. In turn, Rho GTPases are regulated by GTPase-activating proteins (GAPs) and guanine nucleotide exchange factors (GEFs). To understand how endothelial cells initiate changes in the activity of Rho GTPases, and thereby regulate cell adhesion, we will discuss the role of Rho GAPs and GEFs in vascular biology. Many potentially important Rho regulators have not been studied in detail in endothelial cells. We therefore will first overview which GAPs and GEFs are highly expressed in endothelium, based on comparative gene expression analysis of human endothelial cells compared with other tissue cell types. Subsequently, we discuss the relevance of Rho GAPs and GEFs for endothelial cell adhesion in vascular homeostasis and disease.
Collapse
Affiliation(s)
- Jaap D van Buul
- Department of Molecular Cell Biology; Sanquin Research and Swammerdam Institute for Life Sciences; University of Amsterdam; The Netherlands
| | - Dirk Geerts
- Department of Pediatric Oncology/Hematology; Erasmus University Medical Center; Rotterdam, The Netherlands
| | - Stephan Huveneers
- Department of Molecular Cell Biology; Sanquin Research and Swammerdam Institute for Life Sciences; University of Amsterdam; The Netherlands
| |
Collapse
|
48
|
Abstract
The endothelium forms a selective semi-permeable barrier controlling bidirectional transfer between blood vessel and irrigated tissues. This crucial function relies on the dynamic architecture of endothelial cell–cell junctions, and in particular, VE -cadherin-mediated contacts. VE -cadherin indeed chiefly organizes the opening and closing of the endothelial barrier, and is central in permeability changes. In this review, the way VE -cadherin-based contacts are formed and maintained is first presented, including molecular traits of its expression, partners, and signaling. In a second part, the mechanisms by which VE -cadherin adhesion can be disrupted, leading to cell–cell junction weakening and endothelial permeability increase, are described. Overall, the molecular basis for VE -cadherin control of the endothelial barrier function is of high interest for biomedical research, as vascular leakage is observed in many pathological conditions and human diseases.
Collapse
|
49
|
Erasmus JC, Welsh NJ, Braga VMM. Cooperation of distinct Rac-dependent pathways to stabilise E-cadherin adhesion. Cell Signal 2015; 27:1905-13. [PMID: 25957131 PMCID: PMC4508347 DOI: 10.1016/j.cellsig.2015.04.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 04/18/2015] [Accepted: 04/28/2015] [Indexed: 11/25/2022]
Abstract
The precise mechanisms via which Rac1 is activated by cadherin junctions are not fully known. In keratinocytes Rac1 activation by cadherin junctions requires EGFR signalling, but how EGFR does so is unclear. To address which activator could mediate E-cadherin signalling to Rac1, we investigated EGFR and two Rac1 GEFs, SOS1 and DOCK180. EGFR RNAi prevented junction-induced Rac1 activation and led to fragmented localization of E-cadherin at cadherin contacts. In contrast, depletion of another EGFR family member, ErbB3, did not interfere with either process. DOCK180 RNAi, but not SOS1, prevented E-cadherin-induced Rac1 activation. However, in a strong divergence from EGFR RNAi phenotype, DOCK180 depletion did not perturb actin recruitment or cadherin localisation at junctions. Rather, reduced DOCK180 levels impaired the resistance to mechanical stress of pre-formed cell aggregates. Thus, within the same cell type, EGFR and DOCK180 regulate Rac1 activation by newly-formed contacts, but control separate cellular events that cooperate to stabilise junctions.
Collapse
Affiliation(s)
- Jennifer C Erasmus
- Molecular Medicine, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, SW7 2AZ London, UK
| | - Natalie J Welsh
- Molecular Medicine, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, SW7 2AZ London, UK
| | - Vania M M Braga
- Molecular Medicine, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, SW7 2AZ London, UK.
| |
Collapse
|
50
|
Daneshjou N, Sieracki N, van Nieuw Amerongen GP, Conway DE, Schwartz MA, Komarova YA, Malik AB. Rac1 functions as a reversible tension modulator to stabilize VE-cadherin trans-interaction. ACTA ACUST UNITED AC 2015; 208:23-32. [PMID: 25559184 PMCID: PMC4284224 DOI: 10.1083/jcb.201409108] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The role of the RhoGTPase Rac1 in stabilizing mature endothelial adherens junctions (AJs) is not well understood. In this paper, using a photoactivatable probe to control Rac1 activity at AJs, we addressed the relationship between Rac1 and the dynamics of vascular endothelial cadherin (VE-cadherin). We demonstrated that Rac1 activation reduced the rate of VE-cadherin dissociation, leading to increased density of VE-cadherin at AJs. This response was coupled to a reduction in actomyosin-dependent tension across VE-cadherin adhesion sites. We observed that inhibiting myosin II directly or through photo-release of the caged Rho kinase inhibitor also reduced the rate of VE-cadherin dissociation. Thus, Rac1 functions by stabilizing VE-cadherin trans-dimers in mature AJs by counteracting the actomyosin tension. The results suggest a new model of VE-cadherin adhesive interaction mediated by Rac1-induced reduction of mechanical tension at AJs, resulting in the stabilization of VE-cadherin adhesions.
Collapse
Affiliation(s)
- Nazila Daneshjou
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL 60612
| | - Nathan Sieracki
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL 60612
| | - Geerten P van Nieuw Amerongen
- Department of Physiology, Institute for Cardiovascular Research, Vrije University of Amsterdam, 1081 HV Amsterdam, Netherlands
| | - Daniel E Conway
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510
| | - Martin A Schwartz
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284
| | - Yulia A Komarova
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510
| | - Asrar B Malik
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510
| |
Collapse
|