1
|
Hein M, Qambari H, An D, Balaratnasingam C. Current understanding of subclinical diabetic retinopathy informed by histology and high-resolution in vivo imaging. Clin Exp Ophthalmol 2024; 52:464-484. [PMID: 38363022 DOI: 10.1111/ceo.14363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/21/2024] [Accepted: 01/26/2024] [Indexed: 02/17/2024]
Abstract
The escalating incidence of diabetes mellitus has amplified the global impact of diabetic retinopathy. There are known structural and functional changes in the diabetic retina that precede the fundus photography abnormalities which currently are used to diagnose clinical diabetic retinopathy. Understanding these subclinical alterations is important for effective disease management. Histology and high-resolution clinical imaging reveal that the entire neurovascular unit, comprised of retinal vasculature, neurons and glial cells, is affected in subclinical disease. Early functional manifestations are seen in the form of blood flow and electroretinography disturbances. Structurally, there are alterations in the cellular components of vasculature, glia and the neuronal network. On clinical imaging, changes to vessel density and thickness of neuronal layers are observed. How these subclinical disturbances interact and ultimately manifest as clinical disease remains elusive. However, this knowledge reveals potential early therapeutic targets and the need for imaging modalities that can detect subclinical changes in a clinical setting.
Collapse
Affiliation(s)
- Martin Hein
- Physiology and Pharmacology Group, Lions Eye Institute, Perth, Western Australia, Australia
- Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Western Australia, Australia
| | - Hassanain Qambari
- Physiology and Pharmacology Group, Lions Eye Institute, Perth, Western Australia, Australia
- Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Western Australia, Australia
| | - Dong An
- Physiology and Pharmacology Group, Lions Eye Institute, Perth, Western Australia, Australia
- Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Western Australia, Australia
| | - Chandrakumar Balaratnasingam
- Physiology and Pharmacology Group, Lions Eye Institute, Perth, Western Australia, Australia
- Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Western Australia, Australia
- Department of Ophthalmology, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| |
Collapse
|
2
|
Li Y, Ni N, Lee M, Wei W, Andrikopoulos N, Kakinen A, Davis TP, Song Y, Ding F, Leong DT, Ke PC. Endothelial leakiness elicited by amyloid protein aggregation. Nat Commun 2024; 15:613. [PMID: 38242873 PMCID: PMC10798980 DOI: 10.1038/s41467-024-44814-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 01/05/2024] [Indexed: 01/21/2024] Open
Abstract
Alzheimer's disease (AD) is a major cause of dementia debilitating the global ageing population. Current understanding of the AD pathophysiology implicates the aggregation of amyloid beta (Aβ) as causative to neurodegeneration, with tauopathies, apolipoprotein E and neuroinflammation considered as other major culprits. Curiously, vascular endothelial barrier dysfunction is strongly associated with Aβ deposition and 80-90% AD subjects also experience cerebral amyloid angiopathy. Here we show amyloid protein-induced endothelial leakiness (APEL) in human microvascular endothelial monolayers as well as in mouse cerebral vasculature. Using signaling pathway assays and discrete molecular dynamics, we revealed that the angiopathy first arose from a disruption to vascular endothelial (VE)-cadherin junctions exposed to the nanoparticulates of Aβ oligomers and seeds, preceding the earlier implicated proinflammatory and pro-oxidative stressors to endothelial leakiness. These findings were analogous to nanomaterials-induced endothelial leakiness (NanoEL), a major phenomenon in nanomedicine depicting the paracellular transport of anionic inorganic nanoparticles in the vasculature. As APEL also occurred in vitro with the oligomers and seeds of alpha synuclein, this study proposes a paradigm for elucidating the vascular permeation, systemic spread, and cross-seeding of amyloid proteins that underlie the pathogeneses of AD and Parkinson's disease.
Collapse
Affiliation(s)
- Yuhuan Li
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Fudan University, Shanghai, 200032, China
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Nengyi Ni
- National University of Singapore, Department of Chemical and Biomolecular Engineering, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Myeongsang Lee
- Department of Physics and Astronomy, Clemson University, Clemson, SC, 29634, USA
| | - Wei Wei
- College of Veterinary Medicine, Southwest University, Chongqing, 402460, China
| | - Nicholas Andrikopoulos
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- The Nanomedicine Center, The Great Bay Area National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
| | - Aleksandr Kakinen
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Thomas P Davis
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Yang Song
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China.
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC, 29634, USA.
| | - David Tai Leong
- National University of Singapore, Department of Chemical and Biomolecular Engineering, 4 Engineering Drive 4, Singapore, 117585, Singapore.
| | - Pu Chun Ke
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
- The Nanomedicine Center, The Great Bay Area National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China.
| |
Collapse
|
3
|
Li Y, Liu Z, Han X, Liang F, Zhang Q, Huang X, Shi X, Huo H, Han M, Liu X, Zhu H, He L, Shen L, Hu X, Wang J, Wang QD, Smart N, Zhou B, He B. Dynamics of Endothelial Cell Generation and Turnover in Arteries During Homeostasis and Diseases. Circulation 2024; 149:135-154. [PMID: 38084582 DOI: 10.1161/circulationaha.123.064301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 10/06/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND Endothelial cell (EC) generation and turnover by self-proliferation contributes to vascular repair and regeneration. The ability to accurately measure the dynamics of EC generation would advance our understanding of cellular mechanisms of vascular homeostasis and diseases. However, it is currently challenging to evaluate the dynamics of EC generation in large vessels such as arteries because of their infrequent proliferation. METHODS By using dual recombination systems based on Cre-loxP and Dre-rox, we developed a genetic system for temporally seamless recording of EC proliferation in vivo. We combined genetic recording of EC proliferation with single-cell RNA sequencing and gene knockout to uncover cellular and molecular mechanisms underlying EC generation in arteries during homeostasis and disease. RESULTS Genetic proliferation tracing reveals that ≈3% of aortic ECs undergo proliferation per month in adult mice during homeostasis. The orientation of aortic EC division is generally parallel to blood flow in the aorta, which is regulated by the mechanosensing protein Piezo1. Single-cell RNA sequencing analysis reveals 4 heterogeneous aortic EC subpopulations with distinct proliferative activity. EC cluster 1 exhibits transit-amplifying cell features with preferential proliferative capacity and enriched expression of stem cell markers such as Sca1 and Sox18. EC proliferation increases in hypertension but decreases in type 2 diabetes, coinciding with changes in the extent of EC cluster 1 proliferation. Combined gene knockout and proliferation tracing reveals that Hippo/vascular endothelial growth factor receptor 2 signaling pathways regulate EC proliferation in large vessels. CONCLUSIONS Genetic proliferation tracing quantitatively delineates the dynamics of EC generation and turnover, as well as EC division orientation, in large vessels during homeostasis and disease. An EC subpopulation in the aorta exhibits more robust cell proliferation during homeostasis and type 2 diabetes, identifying it as a potential therapeutic target for vascular repair and regeneration.
Collapse
Affiliation(s)
- Yi Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Zixin Liu
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Ximeng Han
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Feng Liang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| | - Qianyu Zhang
- School of Life Science and Technology, ShanghaiTech University, China (Q.Z., M.H., B.Z.)
| | - Xiuzhen Huang
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Xin Shi
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| | - Huanhuan Huo
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| | - Maoying Han
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
- School of Life Science and Technology, ShanghaiTech University, China (Q.Z., M.H., B.Z.)
| | - Xiuxiu Liu
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Huan Zhu
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Lingjuan He
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China (L.H.)
| | - Linghong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| | - Xinyang Hu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China (X.H., J.W.)
| | - Jian'an Wang
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China (X.H., J.W.)
| | - Qing-Dong Wang
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (Q.D.W.)
| | - Nicola Smart
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, UK (N.S.)
| | - Bin Zhou
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
- School of Life Science and Technology, ShanghaiTech University, China (Q.Z., M.H., B.Z.)
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, China (B.Z.)
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| |
Collapse
|
4
|
Arce FT, Younger S, Gaber AA, Mascarenhas JB, Rodriguez M, Dudek SM, Garcia JGN. Lamellipodia dynamics and microrheology in endothelial cell paracellular gap closure. Biophys J 2023; 122:4730-4747. [PMID: 37978804 PMCID: PMC10754712 DOI: 10.1016/j.bpj.2023.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 05/06/2023] [Accepted: 11/16/2023] [Indexed: 11/19/2023] Open
Abstract
Vascular endothelial cells (ECs) form a semipermeable barrier separating vascular contents from the interstitium, thereby regulating the movement of water and molecular solutes across small intercellular gaps, which are continuously forming and closing. Under inflammatory conditions, however, larger EC gaps form resulting in increased vascular leakiness to circulating fluid, proteins, and cells, which results in organ edema and dysfunction responsible for key pathophysiologic findings in numerous inflammatory disorders. In this study, we extend our earlier work examining the biophysical properties of EC gap formation and now address the role of lamellipodia, thin sheet-like membrane projections from the leading edge, in modulating EC spatial-specific contractile properties and gap closure. Micropillars, fabricated by soft lithography, were utilized to form reproducible paracellular gaps in human lung ECs. Using time-lapse imaging via optical microscopy, rates of EC gap closure and motility were measured with and without EC stimulation with the barrier-enhancing sphingolipid, sphingosine-1-phosphate. Peripheral ruffle formation was ubiquitous during gap closure. Kymographs were generated to quantitatively compare the lamellipodia dynamics of sphingosine-1-phosphate-stimulated and -unstimulated ECs. Utilizing atomic force microscopy, we characterized the viscoelastic behavior of EC lamellipodia. Our results indicate decreased stiffness and increased liquid-like behavior of expanding lamellipodia compared with regions away from the cellular edge (lamella and cell body) during EC gap closure, results in sync with the rapid kinetics of protrusion/retraction motion. We hypothesize this dissipative EC behavior during gap closure is linked to actomyosin cytoskeletal rearrangement and decreased cross-linking during lamellipodia expansion. In summary, these studies of the kinetic and mechanical properties of EC lamellipodia and ruffles at gap boundaries yield insights into the mechanisms of vascular barrier restoration and potentially a model system for examining the druggability of lamellipodial protein targets to enhance vascular barrier integrity.
Collapse
Affiliation(s)
- Fernando Teran Arce
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, Florida.
| | - Scott Younger
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona
| | - Amir A Gaber
- Department of Medicine, University of Arizona, Tucson, Arizona
| | | | - Marisela Rodriguez
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, Florida; Department of Medicine, University of Arizona, Tucson, Arizona
| | - Steven M Dudek
- Department of Medicine, The University of Illinois at Chicago, Chicago, Illinois
| | - Joe G N Garcia
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, Florida.
| |
Collapse
|
5
|
Breslin JW. Edema and lymphatic clearance: molecular mechanisms and ongoing challenges. Clin Sci (Lond) 2023; 137:1451-1476. [PMID: 37732545 PMCID: PMC11025659 DOI: 10.1042/cs20220314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/18/2023] [Accepted: 08/31/2023] [Indexed: 09/22/2023]
Abstract
Resolution of edema remains a significant clinical challenge. Conditions such as traumatic shock, sepsis, or diabetes often involve microvascular hyperpermeability, which leads to tissue and organ dysfunction. Lymphatic insufficiency due to genetic causes, surgical removal of lymph nodes, or infections, leads to varying degrees of tissue swelling that impair mobility and immune defenses. Treatment options are limited to management of edema as there are no specific therapeutics that have demonstrated significant success for ameliorating microvascular leakage or impaired lymphatic function. This review examines current knowledge about the physiological, cellular, and molecular mechanisms that control microvascular permeability and lymphatic clearance, the respective processes for interstitial fluid formation and removal. Clinical conditions featuring edema, along with potential future directions are discussed.
Collapse
Affiliation(s)
- Jerome W Breslin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, FL, U.S.A
| |
Collapse
|
6
|
Wen L, Yan W, Zhu L, Tang C, Wang G. The role of blood flow in vessel remodeling and its regulatory mechanism during developmental angiogenesis. Cell Mol Life Sci 2023; 80:162. [PMID: 37221410 PMCID: PMC11072276 DOI: 10.1007/s00018-023-04801-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 04/06/2023] [Accepted: 05/06/2023] [Indexed: 05/25/2023]
Abstract
Vessel remodeling is essential for a functional and mature vascular network. According to the difference in endothelial cell (EC) behavior, we classified vessel remodeling into vessel pruning, vessel regression and vessel fusion. Vessel remodeling has been proven in various organs and species, such as the brain vasculature, subintestinal veins (SIVs), and caudal vein (CV) in zebrafish and yolk sac vessels, retina, and hyaloid vessels in mice. ECs and periendothelial cells (such as pericytes and astrocytes) contribute to vessel remodeling. EC junction remodeling and actin cytoskeleton dynamic rearrangement are indispensable for vessel pruning. More importantly, blood flow has a vital role in vessel remodeling. In recent studies, several mechanosensors, such as integrins, platelet endothelial cell adhesion molecule-1 (PECAM-1)/vascular endothelial cell (VE-cadherin)/vascular endothelial growth factor receptor 2 (VEGFR2) complex, and notch1, have been shown to contribute to mechanotransduction and vessel remodeling. In this review, we highlight the current knowledge of vessel remodeling in mouse and zebrafish models. We further underline the contribution of cellular behavior and periendothelial cells to vessel remodeling. Finally, we discuss the mechanosensory complex in ECs and the molecular mechanisms responsible for vessel remodeling.
Collapse
Affiliation(s)
- Lin Wen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Wenhua Yan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Li Zhu
- Cyrus Tang Hematology Center, Cyrus Tang Medical Institute, Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Suzhou, 215123, China
| | - Chaojun Tang
- Cyrus Tang Hematology Center, Cyrus Tang Medical Institute, Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Suzhou, 215123, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China.
- JinFeng Laboratory, Chongqing, 401329, China.
| |
Collapse
|
7
|
Tokarz VL, Pereira RVS, Jaldin-Fincati JR, Mylvaganam S, Klip A. Junctional integrity and directional mobility of lymphatic endothelial cell monolayers are disrupted by saturated fatty acids. Mol Biol Cell 2023; 34:ar28. [PMID: 36735487 PMCID: PMC10092641 DOI: 10.1091/mbc.e22-08-0367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The lymphatic circulation regulates transfer of tissue fluid and immune cells toward the venous circulation. While obesity impairs lymphatic vessel function, the contribution of lymphatic endothelial cells (LEC) to metabolic disease phenotypes is poorly understood. LEC of lymphatic microvessels are in direct contact with the interstitial fluid, whose composition changes during the development of obesity, markedly by increases in saturated fatty acids. Palmitate, the most prevalent saturated fatty acid in lymph and blood, is detrimental to metabolism and function of diverse tissues, but its impact on LEC function is relatively unknown. Here, palmitate (but not its unsaturated counterpart palmitoleate) destabilized adherens junctions in human microvascular LEC in culture, visualized as changes in VE-cadherin, α-catenin, and β-catenin localization. Detachment of these proteins from cortical actin filaments was associated with abundant actomyosin stress fibers. The effects were Rho-associated protein kinase (ROCK)- and myosin-dependent, as inhibition with Y27632 or blebbistatin, respectively, prevented stress fiber accumulation and preserved junctions. Without functional junctions, palmitate-treated LEC failed to directionally migrate to close wounds in two dimensions and failed to form endothelial tubes in three dimensions. A reorganization of the lymphatic endothelial actin cytoskeleton may contribute to lymphatic dysfunction in obesity and could be considered as a therapeutic target.
Collapse
Affiliation(s)
- Victoria L Tokarz
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Physiology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Rafaela V S Pereira
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | | | - Sivakami Mylvaganam
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Physiology, University of Toronto, Toronto, ON M5S 1A1, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A1, Canada.,Department of Paediatrics, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
8
|
Bhaskar H, Kleinjan D, Oi C, Gidden Z, Rosser SJ, Horrocks MH, Regan L. Live-cell super-resolution imaging of actin using LifeAct-14 with a PAINT-based approach. Protein Sci 2023; 32:e4558. [PMID: 36585831 PMCID: PMC9878614 DOI: 10.1002/pro.4558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/01/2023]
Abstract
We present direct-LIVE-PAINT, an easy-to-implement approach for the nanoscopic imaging of protein structures in live cells using labeled binding peptides. We demonstrate the feasibility of direct-LIVE-PAINT with an actin-binding peptide fused to EGFP, the location of which can be accurately determined as it transiently binds to actin filaments. We show that direct-LIVE-PAINT can be used to image actin structures below the diffraction-limit of light and have used it to observe the dynamic nature of actin in live cells. We envisage a similar approach could be applied to imaging other proteins within live mammalian cells.
Collapse
Affiliation(s)
- Haresh Bhaskar
- The School of Biological SciencesUniversity of EdinburghEdinburghUK,EaStCHEM School of ChemistryUniversity of EdinburghEdinburghUK
| | - Dirk‐Jan Kleinjan
- Centre for Synthetic and Systems Biology and UK Centre for Mammalian Synthetic Biology, School of Biological SciencesUniversity of EdinburghEdinburghUK
| | - Curran Oi
- Department of Genome SciencesUniversity of WashingtonSeattleWashingtonUSA
| | - Zoe Gidden
- The School of Biological SciencesUniversity of EdinburghEdinburghUK,EaStCHEM School of ChemistryUniversity of EdinburghEdinburghUK
| | - Susan J. Rosser
- Centre for Synthetic and Systems Biology and UK Centre for Mammalian Synthetic Biology, School of Biological SciencesUniversity of EdinburghEdinburghUK
| | | | - Lynne Regan
- The School of Biological SciencesUniversity of EdinburghEdinburghUK
| |
Collapse
|
9
|
Pathak S, Gupta R, Parkar H, Joshi N, Nagotu S, Kale A. The role of Colchicine on actin polymerization dynamics: as a potent anti-angiogenic factor. J Biomol Struct Dyn 2022; 40:11729-11743. [PMID: 34424806 DOI: 10.1080/07391102.2021.1965911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Over the years, cancer research has focused on different strategies to discover drugs and therapies to treat the metastatic stage of cancer. This stage depends upon the type, and the cause of cancer. One of the central facts about any cancer invasion is the formation of new blood vessels that provide nutrients to these uncontrollably dividing cells. This phenomenon is called angiogenesis and is responsible for tumor progression and metastasis. Tumor angiogenesis is a sequential process wherein various angiogenic factors produced by tumor cells bind to receptors of endothelial cells. This stimulates the cytoskeletal protein, especially actin to reorganize themselves and undergo the process of canalization. The driving force for such membrane transformation is spatially and temporally-regulated by polymerization of submembrane actin filaments. So far, Colchicine has been studied for its effectiveness in controlling microtubule reorganization during cell division, but its role is far from understood on actin polymerization. In our current study, we report the effect of Colchicine on actin polymerization dynamics using biophysical analysis like Right light scattering (RLS), Dynamic light scattering (DLS), Circular dichroism (CD) analysis, Scanning electron microscopy (SEM) study. Isothermal titration calorimetry (ITC) and kinetic measurements. Isothermal titration calorimetry (ITC) indicates multiple site binding for colchicine with actin aggregates. We have checked the in vivo effect of colchicine using end3 cells of Saccharomyces cerevisiae. We also report the anti-angiogenesis activity of colchicine via ex-ovo chicken chorioallantoic membrane (CAM) assay. We predict the target site of binding for the drug by docking studies. Based on our findings, we suggest the 'drug-repurposed' function for colchicine as a potential anti-angiogenic candidate.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Samridhi Pathak
- School of Chemical Sciences, UM-DAE Center for excellence in basic sciences, University of Mumbai, Mumbai, Maharashtra, India
| | - Rahul Gupta
- School of Chemical Sciences, UM-DAE Center for excellence in basic sciences, University of Mumbai, Mumbai, Maharashtra, India
| | - Haifa Parkar
- School of Chemical Sciences, UM-DAE Center for excellence in basic sciences, University of Mumbai, Mumbai, Maharashtra, India
| | - Neha Joshi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Shirisha Nagotu
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Avinash Kale
- School of Chemical Sciences, UM-DAE Center for excellence in basic sciences, University of Mumbai, Mumbai, Maharashtra, India
| |
Collapse
|
10
|
Hirano M, Hirano K. Critical role of Rho proteins in myosin light chain di-phosphorylation during early phase of endothelial barrier disruption. J Physiol Sci 2022; 72:32. [PMID: 36476233 DOI: 10.1186/s12576-022-00857-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022]
Abstract
We previously reported the Rho-associated coiled-coil containing protein kinase (ROCK)-mediated di-phosphorylation of myosin light chain (MLC) and actin bundle formation at the cell periphery as early events of the endothelial barrier disruption. We herein examined the role of RhoA during early events of barrier disruption. Treatment of cultured porcine aortic endothelial cells with simvastatin prevented the decrease in trans-endothelial electrical resistance, MLC di-phosphorylation and peripheral actin bundle formation seen 3 min after thrombin stimulation. Co-treatment with geranylgeranyl pyrophosphate rescued the thrombin-induced events. Thrombin increased a GTP-bound form of RhoA and phosphorylation of myosin phosphatase target subunit 1 (MYPT1) at the ROCK site. The intracellular introduction of the inhibitory protein of RhoA inhibited the thrombin-induced di-phosphorylation of MLC. However, knockdown of either one of RhoA, RhoB or RhoC failed to inhibit thrombin-induced MLC di-phosphorylation. The findings suggest that Rho proteins play a critical role during early events of thrombin-induced barrier disruption.
Collapse
Affiliation(s)
- Mayumi Hirano
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-Gun, Kagawa, Japan
| | - Katsuya Hirano
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-Gun, Kagawa, Japan.
| |
Collapse
|
11
|
Paudel SS, deWeever A, Sayner S, Stevens T, Tambe DT. Substrate stiffness modulates migration and local intercellular membrane motion in pulmonary endothelial cell monolayers. Am J Physiol Cell Physiol 2022; 323:C936-C949. [PMID: 35912996 PMCID: PMC9467474 DOI: 10.1152/ajpcell.00339.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 07/22/2022] [Accepted: 07/22/2022] [Indexed: 11/22/2022]
Abstract
The pulmonary artery endothelium forms a semipermeable barrier that limits macromolecular flux through intercellular junctions. This barrier is maintained by an intrinsic forward protrusion of the interacting membranes between adjacent cells. However, the dynamic interactions of these membranes have been incompletely quantified. Here, we present a novel technique to quantify the motion of the peripheral membrane of the cells, called paracellular morphological fluctuations (PMFs), and to assess the impact of substrate stiffness on PMFs. Substrate stiffness impacted large-length scale morphological changes such as cell size and motion. Cell size was larger on stiffer substrates, whereas the speed of cell movement was decreased on hydrogels with stiffness either larger or smaller than 1.25 kPa, consistent with cells approaching a jammed state. Pulmonary artery endothelial cells moved fastest on 1.25 kPa hydrogel, a stiffness consistent with a healthy pulmonary artery. Unlike these large-length scale morphological changes, the baseline of PMFs was largely insensitive to the substrate stiffness on which the cells were cultured. Activation of store-operated calcium channels using thapsigargin treatment triggered a transient increase in PMFs beyond the control treatment. However, in hypocalcemic conditions, such an increase in PMFs was absent on 1.25 kPa hydrogel but was present on 30 kPa hydrogel-a stiffness consistent with that of a hypertensive pulmonary artery. These findings indicate that 1) PMFs occur in cultured endothelial cell clusters, irrespective of the substrate stiffness; 2) PMFs increase in response to calcium influx through store-operated calcium entry channels; and 3) stiffer substrate promotes PMFs through a mechanism that does not require calcium influx.
Collapse
Affiliation(s)
- Sunita Subedi Paudel
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama
- Department of Mechanical Aerospace and Biomedical Engineering, University of South Alabama, Mobile, Alabama
- Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Althea deWeever
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama
- Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Sarah Sayner
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama
- Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Troy Stevens
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama
- Department of Internal Medicine, University of South Alabama, Mobile, Alabama
- Department of Mechanical Aerospace and Biomedical Engineering, University of South Alabama, Mobile, Alabama
| | - Dhananjay T Tambe
- Department of Mechanical Aerospace and Biomedical Engineering, University of South Alabama, Mobile, Alabama
- Department of Pharmacology, University of South Alabama, Mobile, Alabama
- Center for Lung Biology, University of South Alabama, Mobile, Alabama
| |
Collapse
|
12
|
Hall JD, Farzaneh S, Babakhani Galangashi R, Pujari A, Sweet DT, Kahn ML, Jiménez JM. Lymphoedema conditions disrupt endothelial barrier function in vitro. J R Soc Interface 2022; 19:20220223. [PMID: 36000230 PMCID: PMC9399713 DOI: 10.1098/rsif.2022.0223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 07/27/2022] [Indexed: 11/12/2022] Open
Abstract
Lymphatic vessel contractions generate net antegrade pulsatile lymph flow. By contrast, impaired lymphatic vessels are often associated with lymphoedema and altered lymph flow. The effect of lymphoedema on the lymph flow field and endothelium is not completely known. Here, we characterized the lymphatic flow field of a platelet-specific receptor C-type lectin-like receptor 2 (CLEC2) deficient lymphoedema mouse model. In regions of lymphoedema, collecting vessels were significantly distended, vessel contractility was greatly diminished and pulsatile lymph flow was replaced by quasi-steady flow. In vitro exposure of human dermal lymphatic endothelial cells (LECs) to lymphoedema-like quasi-steady flow conditions increased intercellular gap formation and permeability in comparison to normal pulsatile lymph flow. In the absence of flow, LECs exposed to steady pressure (SP) increased intercellular gap formation in contrast with pulsatile pressure (PP). The absence of pulsatility in steady fluid flow and SP conditions without flow-induced upregulation of myosin light chain (MLCs) regulatory subunits 9 and 12B mRNA expression and phosphorylation of MLCs, in contrast with pulsatile flow and PP without flow. These studies reveal that the loss of pulsatility, which can occur with lymphoedema, causes LEC contraction and an increase in intercellular gap formation mediated by MLC phosphorylation.
Collapse
Affiliation(s)
- Joshua D. Hall
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, MA, USA
| | - Sina Farzaneh
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, MA, USA
| | - Reza Babakhani Galangashi
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, MA, USA
| | - Akshay Pujari
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, MA, USA
| | - Daniel T. Sweet
- Department of Medicine and Division of Cardiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark L. Kahn
- Department of Medicine and Division of Cardiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Juan M. Jiménez
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, MA, USA
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, USA
| |
Collapse
|
13
|
Silvani G, Bradbury P, Basirun C, Mehner C, Zalli D, Poole K, Chou J. Testing 3D printed biological platform for advancing simulated microgravity and space mechanobiology research. NPJ Microgravity 2022; 8:19. [PMID: 35662260 PMCID: PMC9166742 DOI: 10.1038/s41526-022-00207-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 05/13/2022] [Indexed: 12/02/2022] Open
Abstract
The advancement of microgravity simulators is helping many researchers better understanding the impact of the mechanically unloaded space environment on cellular function and disfunction. However, performing microgravity experiments on Earth, using simulators such as the Random Positioning Machine, introduces some unique practical challenges, including air bubble formation and leakage of growth medium from tissue culture flask and plates, all of which limit research progress. Here, we developed an easy-to-use hybrid biological platform designed with the precision of 3D printing technologies combined with PDMS microfluidic fabrication processes to facilitate reliable and reproducible microgravity cellular experiments. The system has been characterized for applications in the contest of brain cancer research by exposing glioblastoma and endothelial cells to 24 h of simulated microgravity condition to investigate the triggered mechanosensing pathways involved in cellular adaptation to the new environment. The platform demonstrated compatibility with different biological assays, i.e., proliferation, viability, morphology, protein expression and imaging of molecular structures, showing advantages over the conventional usage of culture flask. Our results indicated that both cell types are susceptible when the gravitational vector is disrupted, confirming the impact that microgravity has on both cancer and healthy cells functionality. In particular, we observed deactivation of Yap-1 molecule in glioblastoma cells and the remodeling of VE-Cadherin junctional protein in endothelial cells. The study provides support for the application of the proposed biological platform for advancing space mechanobiology research, also highlighting perspectives and strategies for developing next generation of brain cancer molecular therapies, including targeted drug delivery strategies.
Collapse
Affiliation(s)
- Giulia Silvani
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia
| | - Peta Bradbury
- Institut Curie, Paris Sciences et Lettres Research University, Mechanics and Genetics of Embryonic and Tumoral Development Group, Paris, France
| | - Carin Basirun
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia
| | - Christine Mehner
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, USA
| | - Detina Zalli
- Institute of Continuing Education, University of Cambridge, Camridge, UK
| | - Kate Poole
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, Faculty of Medicine & Health, Sydney, NSW, Australia
| | - Joshua Chou
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia.
| |
Collapse
|
14
|
Jia C, Shi J, Han T, Yu ACH, Qin P. Spatiotemporal Dynamics and Mechanisms of Actin Cytoskeletal Re-modeling in Cells Perforated by Ultrasound-Driven Microbubbles. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:760-777. [PMID: 35190224 DOI: 10.1016/j.ultrasmedbio.2021.12.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 12/18/2021] [Accepted: 12/22/2021] [Indexed: 06/14/2023]
Abstract
To develop new strategies for improving the efficacy and biosafety of sonoporation-based macromolecule delivery, it is essential to understand the mechanisms underlying plasma membrane re-sealing and function recovery of the cells perforated by ultrasound-driven microbubbles. However, we lack a clear understanding of the spatiotemporal dynamics of the disrupted actin cytoskeleton and its role in the re-sealing of sonoporated cells. Here we used a customized experimental setup for single-pulse ultrasound (133.33-µs duration and 0.70-MPa peak negative pressure) exposure to microbubbles and for real-time recording of single-cell (human umbilical vein endothelial cell) responses by laser confocal microscopic imaging. We found that in reversibly sonoporated cells, the locally disrupted actin cytoskeleton, which was spatially correlated with the perforated plasma membrane, underwent three successive phases (expansion; contraction and re-sealing; and recovery) to re-model and that each phase of the disrupted actin cytoskeleton was approximately synchronized with that of the perforated plasma membrane. Moreover, compared with the closing time of the perforated plasma membrane, the same time was used for the re-sealing of the actin cytoskeleton in mildly sonoporated cells and a longer time was required in moderately sonoporated cells. Further, the generation, directional migration, accumulation and re-polymerization of globular actin polymers during the three phases drove the re-modeling of the actin cytoskeleton. However, in irreversibly sonoporated cells, the actin cytoskeleton, which underwent expansion and no contraction, was progressively de-polymerized and could not be re-sealed. Finally, we found that intracellular calcium transients were essential for the recruitment of globular actin and the re-modeling of the actin cytoskeleton. These results provide new insight into the role of actin cytoskeleton dynamics in the re-sealing of sonoporated cells and serve to guide the design of new strategies for sonoporation-based delivery.
Collapse
Affiliation(s)
- Caixia Jia
- School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jianmin Shi
- School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Han
- School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Alfred C H Yu
- Schlegel Research Institute for Aging, University of Waterloo, Waterloo, Ontario, Canada
| | - Peng Qin
- School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
15
|
CCL4 induces inflammatory signalling and barrier disruption in the neurovascular endothelium. Brain Behav Immun Health 2021; 18:100370. [PMID: 34755124 PMCID: PMC8560974 DOI: 10.1016/j.bbih.2021.100370] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 12/27/2022] Open
Abstract
Background During neuroinflammation many chemokines alter the function of the blood-brain barrier (BBB) that regulates the entry of macromolecules and immune cells into the brain. As the milieu of the brain is altered, biochemical and structural changes contribute to the pathogenesis of neuroinflammation and may impact on neurogenesis. The chemokine CCL4, previously known as MIP-1β, is upregulated in a wide variety of central nervous system disorders, including multiple sclerosis, where it is thought to play a key role in the neuroinflammatory process. However, the effect of CCL4 on BBB endothelial cells (ECs) is unknown. Materials and methods Expression and distribution of CCR5, phosphorylated p38, F-actin, zonula occludens-1 (ZO-1) and vascular endothelial cadherin (VE-cadherin) were analysed in the human BBB EC line hCMEC/D3 by Western blot and/or immunofluorescence in the presence and absence of CCL4. Barrier modulation in response to CCL4 using hCMEC/D3 monolayers was assessed by measuring molecular flux of 70 kDa RITC-dextran and transendothelial lymphocyte migration. Permeability changes in response to CCL4 in vivo were measured by an occlusion technique in pial microvessels of Wistar rats and by fluorescein angiography in mouse retinae. Results CCR5, the receptor for CCL4, was expressed in hCMEC/D3 cells. CCL4 stimulation led to phosphorylation of p38 and the formation of actin stress fibres, both indicative of intracellular chemokine signalling. The distribution of junctional proteins was also altered in response to CCL4: junctional ZO-1 was reduced by circa 60% within 60 min. In addition, surface VE-cadherin was redistributed through internalisation. Consistent with these changes, CCL4 induced hyperpermeability in vitro and in vivo and increased transmigration of lymphocytes across monolayers of hCMEC/D3 cells. Conclusion These results show that CCL4 can modify BBB function and may contribute to disease pathogenesis. The chemokine CCL4 induced phosphorylation of P38 in an in vitro model of the blood-brain barrier (BBB). CCL4 treatment resulted in reduction of plasma membrane VE-cadherin and junctional ZO-1. CCL4 induced neurovascular barrier breakdown in vitro and in vivo.
Collapse
|
16
|
Gamma irradiation exposure for collapsed cell junctions and reduced angiogenesis of 3-D in vitro blood vessels. Sci Rep 2021; 11:18230. [PMID: 34521931 PMCID: PMC8440565 DOI: 10.1038/s41598-021-97692-8] [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: 12/08/2020] [Accepted: 08/17/2021] [Indexed: 02/08/2023] Open
Abstract
During radiotherapy, microenvironments neighboring the tumor are also exposed to gamma irradiation; this results in unexpected side effects. Blood vessels can serve as microenvironments for tumors and they play an important role in providing nutrients to tumors. This is mostly related to tumor progression, metastasis, and relapse after therapy. Many studies have been performed to obtain a better understanding of tumor vasculature after radiotherapy with in vitro models. However, compared to 3-D models, 2-D in vitro endothelial monolayers cannot physiologically reflect in vivo blood vessels. We previously remodeled the extracellular matrix (ECM) hydrogel that enhanced the tight barrier formation of 3-D blood vessels and the vascular endothelial growth factor (VEGF) gradient induced angiogenesis in a microfluidic device. In this study, the blood vessel model is further introduced to understand how gamma irradiation affects the endothelial monolayer. After the gamma irradiation exposure, we observed a collapsed endothelial barrier and a reduced angiogenic potential. Changes in the cell behaviors of the tip and stalk cells were also detected in the angiogenesis model after irradiation, which is difficult to observe in 2-D monolayer models. Therefore, the 3-D in vitro blood vessel model can be used to understand radiation-induced endothelial injuries.
Collapse
|
17
|
Silvani G, Basirun C, Wu H, Mehner C, Poole K, Bradbury P, Chou J. A 3D‐Bioprinted Vascularized Glioblastoma‐on‐a‐Chip for Studying the Impact of Simulated Microgravity as a Novel Pre‐Clinical Approach in Brain Tumor Therapy. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100106] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Giulia Silvani
- School of Biomedical Engineering, Faculty of Engineering and Information Technology University of Technology Sydney Sydney Australia
| | - Carin Basirun
- School of Biomedical Engineering, Faculty of Engineering and Information Technology University of Technology Sydney Sydney Australia
| | - Hanjie Wu
- School of Biomedical Engineering, Faculty of Engineering and Information Technology University of Technology Sydney Sydney Australia
| | - Christine Mehner
- Department of Physiology and Biomedical Engineering Mayo Clinic Jacksonville FL USA
| | - Kate Poole
- EMBL Australia node in Single Molecule Science, School of Medical Sciences, Faculty of Medicine University of New South Wales Sydney 2052 Australia
| | - Peta Bradbury
- Institut Curie, Paris Sciences et Lettres Research University Mechanics and Genetics of Embryonic and Tumoral Development Group Paris France
| | - Joshua Chou
- School of Biomedical Engineering, Faculty of Engineering and Information Technology University of Technology Sydney Sydney Australia
| |
Collapse
|
18
|
Huang D, Ding Q, Chen S, Lü S, Zhang Y, Long M. E-selectin negatively regulates polymorphonuclear neutrophil transmigration through altered endothelial junction integrity. FASEB J 2021; 35:e21521. [PMID: 33811691 DOI: 10.1096/fj.202000662rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 01/28/2021] [Accepted: 02/26/2021] [Indexed: 11/11/2022]
Abstract
Transendothelial migration (TEM) of neutrophils under blood flow is critical in the inflammatory cascade. However, the role of endothelial plasticity in this process is not fully understood. Therefore, we used an in vitro model to test the dynamics of human polymorphonuclear neutrophil (PMN) TEM across lipopolysaccharide-treated human umbilical vein endothelial cell (HUVEC) monolayers. Interestingly, shRNA-E-selectin knockdown in HUVECs destabilized endothelial junctional integrity by reducing actin branching and increasing stress fiber at cell-cell junctions. This process is accomplished by downregulating the activation of cortactin and Arp2/3, which in turn alters the adhesive function of VE-cadherin, enhancing PMN transmigration. Meanwhile, redundant P-selectins possess overlapping functions in E-selectin-mediated neutrophil adhesion, and transmigration. These results demonstrate, to our knowledge, for the first time, that E-selectins negatively regulate neutrophil transmigration through alterations in endothelial plasticity. Furthermore, it improves our understanding of the mechanisms underlying actin remodeling, and junctional integrity, in endothelial cells mediating leukocyte TEM.
Collapse
Affiliation(s)
- Dandan Huang
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Qihan Ding
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Shenbao Chen
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Shouqin Lü
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yan Zhang
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Mian Long
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China.,Lead Contact, Beijing, China
| |
Collapse
|
19
|
Bayir E, Sendemir A. Role of Intermediate Filaments in Blood-Brain Barrier in Health and Disease. Cells 2021; 10:cells10061400. [PMID: 34198868 PMCID: PMC8226756 DOI: 10.3390/cells10061400] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/11/2022] Open
Abstract
The blood–brain barrier (BBB) is a highly selective cellular monolayer unique to the microvasculature of the central nervous system (CNS), and it mediates the communication of the CNS with the rest of the body by regulating the passage of molecules into the CNS microenvironment. Limitation of passage of substances through the BBB is mainly due to tight junctions (TJ) and adherens junctions (AJ) between brain microvascular endothelial cells. The importance of actin filaments and microtubules in establishing and maintaining TJs and AJs has been indicated; however, recent studies have shown that intermediate filaments are also important in the formation and function of cell–cell junctions. The most common intermediate filament protein in endothelial cells is vimentin. Vimentin plays a role in blood–brain barrier permeability in both cell–cell and cell–matrix interactions by affecting the actin and microtubule reorganization and by binding directly to VE-cadherin or integrin proteins. The BBB permeability increases due to the formation of stress fibers and the disruption of VE–cadherin interactions between two neighboring cells in various diseases, disrupting the fiber network of intermediate filament vimentin in different ways. Intermediate filaments may be long ignored key targets in regulation of BBB permeability in health and disease.
Collapse
Affiliation(s)
- Ece Bayir
- Ege University Central Research Test and Analysis Laboratory Application and Research Center (EGE-MATAL), Ege University, 35100 Izmir, Turkey;
| | - Aylin Sendemir
- Department of Bioengineering, Faculty of Engineering, Ege University, 35100 Izmir, Turkey
- Department of Biomedical Technologies, Graduate School of Natural and Applied Science, Ege University, 35100 Izmir, Turkey
- Correspondence: ; Tel.: +90-232-3114817
| |
Collapse
|
20
|
Seebach J, Klusmeier N, Schnittler H. Autoregulatory "Multitasking" at Endothelial Cell Junctions by Junction-Associated Intermittent Lamellipodia Controls Barrier Properties. Front Physiol 2021; 11:586921. [PMID: 33488392 PMCID: PMC7815704 DOI: 10.3389/fphys.2020.586921] [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: 07/24/2020] [Accepted: 11/30/2020] [Indexed: 01/12/2023] Open
Abstract
Vascular endothelial cell (EC) junctions are key structures controlling tissue homeostasis in physiology. In the last three decades, excellent studies have addressed many aspects of this complex and highly dynamic regulation, including cell signaling, remodeling processes of the proteins of tight junctions, adherens junctions, and gap junctions, the cytoskeleton, and post-transcriptional modifications, transcriptional activation, and gene silencing. In this dynamic process, vascular endothelial cadherin (VE-cadherin) provides the core structure of EC junctions mediating the physical adhesion of cells as well as the control of barrier function and monolayer integrity via remodeling processes, regulation of protein expression and post-translational modifications. In recent years, research teams have documented locally restricted dynamics of EC junctions in which actin-driven protrusions in plasma membranes play a central role. In this regard, our research group showed that the dynamics of VE-cadherin is driven by small (1-5 μm) actin-mediated protrusions in plasma membranes that, due to this specific function, were named "junction-associated intermittent lamellipodia" (JAIL). JAIL form at overlapping, adjacent cells, and exactly at this site new VE-cadherin interactions occur, leading to new VE-cadherin adhesion sites, a process that restores weak or lost VE-cadherin adhesion. Mechanistically, JAIL formation occurs locally restricted (1-5 μm) and underlies autoregulation in which the local VE-cadherin concentration is an important parameter. A decrease in the local concentration of VE-cadherin stimulates JAIL formation, whereas an increase in the concentration of VE-cadherin blocks it. JAIL mediated VE-cadherin remodeling at the subjunctional level have been shown to be of crucial importance in angiogenesis, wound healing, and changes in permeability during inflammation. The concept of subjunctional regulation of EC junctions is strongly supported by permeability assays, which can be employed to quantify actin-driven subjunctional changes. In this brief review, we summarize and discuss the current knowledge and concepts of subjunctional regulation in the endothelium.
Collapse
Affiliation(s)
- Jochen Seebach
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Nadine Klusmeier
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Hans Schnittler
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Münster, Germany
| |
Collapse
|
21
|
Ben-Saadon S, Gavriel M, Zaretsky U, Jaffa AJ, Grisaru D, Elad D. Tissue-engineered arterial intima model exposed to steady wall shear stresses. J Biomech 2021; 117:110236. [PMID: 33508722 DOI: 10.1016/j.jbiomech.2021.110236] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 12/16/2022]
Abstract
The arterial intima is continuously under pulsatile wall shear stresses (WSS) imposed by the circulating blood. The knowledge of the contribution of smooth muscle cells (SMC) to the response of endothelial cell (EC) to WSS is still incomplete. We developed a co-culture model of EC on top of SMC that mimics the inner in vivo structure of the arterial intima of large arteries. The co-cultured model, as well as a monolayer model of EC, were developed in custom-designed wells that allowed for mechanobiology experiments. Both the monolayer and co-culture models were exposed to steady flow induced WSS of up to 24 dyne/cm2 and for lengths of 60 min. Quantification of WSS induced alterations in the cytoskeletal actin filaments (F-actin) and vascular endothelial cadherin (VE-cadherin) junctions were utilized from confocal images and flow cytometry. High confluency of both models was observed even after exposure to the high WSS. The quantitive analysis revealed larger post WSS amounts of EC F-actin polymerization in the monolayer, which may be explained by the relative help of the SMC to resist the external load of WSS. The VE-cadherin demonstrated morphological alterations in the monolayer model, but without significant changes in their content. The SMC in the co-culture maintained their contractile phenotype post high WSS which is more physiological, but not post low WSS. Generally, the results of this work demonstrate the active role of SMC in the intima performance to resist flow induced WSS.
Collapse
Affiliation(s)
- Sara Ben-Saadon
- Department of Biomedical Engineering, Faculty of Engineering, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Mark Gavriel
- Department of Biomedical Engineering, Faculty of Engineering, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Uri Zaretsky
- Department of Biomedical Engineering, Faculty of Engineering, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Ariel J Jaffa
- Department of Obstetrics and Gynecology, Lis Maternity Hospital, Tel-Aviv Medical Center, Tel Aviv 64239, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Dan Grisaru
- Department of Gynecological Oncology, Lis Maternity Hospital, Tel-Aviv Medical Center, Tel Aviv 64239, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - David Elad
- Department of Biomedical Engineering, Faculty of Engineering, Tel-Aviv University, Tel-Aviv 69978, Israel.
| |
Collapse
|
22
|
Kim J, Cooper JA. Junctional Localization of Septin 2 Is Required for Organization of Junctional Proteins in Static Endothelial Monolayers. Arterioscler Thromb Vasc Biol 2021; 41:346-359. [PMID: 33147991 PMCID: PMC7769918 DOI: 10.1161/atvbaha.120.315472] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 10/20/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Septin 2 is localized at junctions in human microvascular endothelial monolayers. The junctional localization of septin 2 is necessary for organization of cell-cell adhesion proteins of endothelial cells. Approach and Results: Septin 2 was depleted at junctions by suppression of expression using shRNA, treatment with inflammatory cytokine, TNF (tumor necrosis factor)-α, and ectopic overexpression of septin 2 phosphatidylinositol 4,5-bisphosphate binding mutant defect in interaction with plasma membrane. Under those conditions, organizations and expression levels of various junctional proteins were analyzed. Confocal images of immunofluorescence staining showed substantial disorganization of adherens junctional proteins, nectin-2 and afadin, TJP (tight junction protein), ZO (zonula occludens)-1, and intercellular adhesion protein, PECAM-1 (platelet-endothelial cell adhesion molecule-1). Immunoblots for those proteins did not show significant changes in expression except for nectin-2 that highly increased in expression. Significant differential gene expression profiles and biological pathway analysis by septin 2 suppression and by TNF-α treatment using RNA-seq showed common overlapping pathways. The commonalities in expression may be consistent with the similar effects on the overall organization of cell-cell adhesion proteins. CONCLUSIONS Localization of septin 2 at cell junctions are required for the arrangement of junctional proteins and the integrity of the barrier formed by endothelial monolayers.
Collapse
Affiliation(s)
- Joanna Kim
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO, USA
| | - John A. Cooper
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
23
|
Daum R, Mrsic I, Hutterer J, Junginger A, Hinderer S, Meixner AJ, Gauglitz G, Chassé T, Schenke-Layland K. Fibronectin adsorption on oxygen plasma-treated polyurethane surfaces modulates endothelial cell response. J Mater Chem B 2021; 9:1647-1660. [DOI: 10.1039/d0tb02757j] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Fibronectin coating increases implant biocompatibility by enhancing surface endothelialization via integrin-mediated binding.
Collapse
Affiliation(s)
- Ruben Daum
- NMI Natural and Medical Sciences
- Institute at the University of Tübingen
- 72770 Reutlingen
- Germany
- Department of Women's Health
| | - Ivana Mrsic
- Institute of Physical and Theoretical Chemistry
- Eberhard Karls University Tübingen
- 72076 Tübingen
- Germany
| | - Johanna Hutterer
- Institute of Physical and Theoretical Chemistry
- Eberhard Karls University Tübingen
- 72076 Tübingen
- Germany
| | - Achim Junginger
- Institute of Physical and Theoretical Chemistry
- Eberhard Karls University Tübingen
- 72076 Tübingen
- Germany
| | - Svenja Hinderer
- NMI Natural and Medical Sciences
- Institute at the University of Tübingen
- 72770 Reutlingen
- Germany
- Department of Women's Health
| | - Alfred J. Meixner
- Institute of Physical and Theoretical Chemistry
- Eberhard Karls University Tübingen
- 72076 Tübingen
- Germany
- Center for Light–Matter Interaction
| | - Günter Gauglitz
- Institute of Physical and Theoretical Chemistry
- Eberhard Karls University Tübingen
- 72076 Tübingen
- Germany
| | - Thomas Chassé
- Institute of Physical and Theoretical Chemistry
- Eberhard Karls University Tübingen
- 72076 Tübingen
- Germany
- Center for Light–Matter Interaction
| | - Katja Schenke-Layland
- NMI Natural and Medical Sciences
- Institute at the University of Tübingen
- 72770 Reutlingen
- Germany
- Department of Women's Health
| |
Collapse
|
24
|
Taha M, Aldirawi M, März S, Seebach J, Odenthal-Schnittler M, Bondareva O, Bojovic V, Schmandra T, Wirth B, Mietkowska M, Rottner K, Schnittler H. EPLIN-α and -β Isoforms Modulate Endothelial Cell Dynamics through a Spatiotemporally Differentiated Interaction with Actin. Cell Rep 2020; 29:1010-1026.e6. [PMID: 31644899 DOI: 10.1016/j.celrep.2019.09.043] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 08/08/2019] [Accepted: 09/13/2019] [Indexed: 12/14/2022] Open
Abstract
Actin-binding proteins are essential for linear and branched actin filament dynamics that control shape change, cell migration, and cell junction remodeling in vascular endothelium (endothelial cells [ECs]). The epithelial protein lost in neoplasm (EPLIN) is an actin-binding protein, expressed as EPLIN-α and EPLIN-β by alternative promoters; however, the isoform-specific functions are not yet understood. Aortic compared to cava vein ECs and shear stress-exposed cultured ECs express increased EPLIN-β levels that stabilize stress fibers. In contrast, EPLIN-α expression is increased in growing and migrating ECs, is targeted to membrane protrusions, and terminates their growth via interaction with the Arp2/3 complex. The data indicate that EPLIN-α controls protrusion dynamics while EPLIN-β has an actin filament stabilizing role, which is consistent with FRAP analyses demonstrating a lower EPLIN-β turnover rate compared to EPLIN-α. Together, EPLIN isoforms differentially control actin dynamics in ECs, essential in shear stress responses, cell migration, and barrier function.
Collapse
Affiliation(s)
- Muna Taha
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms University of Münster, 48149 Münster, Germany; Cells-in-Motion Cluster of Excellence (EXC 1003 CiM), Westfälische Wilhelms University of Münster, 48149 Münster, Germany
| | - Mohammed Aldirawi
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms University of Münster, 48149 Münster, Germany; Cells-in-Motion Cluster of Excellence (EXC 1003 CiM), Westfälische Wilhelms University of Münster, 48149 Münster, Germany
| | - Sigrid März
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms University of Münster, 48149 Münster, Germany; Cells-in-Motion Cluster of Excellence (EXC 1003 CiM), Westfälische Wilhelms University of Münster, 48149 Münster, Germany
| | - Jochen Seebach
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms University of Münster, 48149 Münster, Germany; Cells-in-Motion Cluster of Excellence (EXC 1003 CiM), Westfälische Wilhelms University of Münster, 48149 Münster, Germany
| | - Maria Odenthal-Schnittler
- Cells-in-Motion Cluster of Excellence (EXC 1003 CiM), Westfälische Wilhelms University of Münster, 48149 Münster, Germany; Department of Ophthalmology, Westfälische Wilhelms University of Münster, Medical Center, 48149 Münster, Germany
| | - Olga Bondareva
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms University of Münster, 48149 Münster, Germany; Cells-in-Motion Cluster of Excellence (EXC 1003 CiM), Westfälische Wilhelms University of Münster, 48149 Münster, Germany
| | - Vesna Bojovic
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms University of Münster, 48149 Münster, Germany
| | - Thomas Schmandra
- Heart and Vascular Clinic Bad Neustadt, Bad Neustadt a.d. Saale, Germany
| | - Benedikt Wirth
- Cells-in-Motion Cluster of Excellence (EXC 1003 CiM), Westfälische Wilhelms University of Münster, 48149 Münster, Germany; Institute for Analysis and Numerics, Westfälische Wilhelms University of Münster, 48149 Münster Germany
| | - Magdalena Mietkowska
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany; Molecular Cell Biology Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany; Molecular Cell Biology Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Hans Schnittler
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms University of Münster, 48149 Münster, Germany; Cells-in-Motion Cluster of Excellence (EXC 1003 CiM), Westfälische Wilhelms University of Münster, 48149 Münster, Germany.
| |
Collapse
|
25
|
Motallebnejad P, Azarin SM. Chemically defined human vascular laminins for biologically relevant culture of hiPSC-derived brain microvascular endothelial cells. Fluids Barriers CNS 2020; 17:54. [PMID: 32912242 PMCID: PMC7488267 DOI: 10.1186/s12987-020-00215-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/25/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND In recent years, differentiation of human induced pluripotent stem cells (hiPSCs) into brain-specific microvascular endothelial cells (iBMECs) has frequently been used to model the blood-brain barrier (BBB). However, there are limitations in the use of iBMECs for in vitro studies, such as transendothelial electrical resistance (TEER) instability, weak junctional expression of VE-cadherin, and lack of proper fluid shear stress response. In vivo, the basement membrane (BM) composition of the BBB evolves throughout development, and laminins become the dominant component of the mature vascular BM. However, laminin isoforms of the endothelial BM have not been used for culture of differentiated iBMECs. The main goal of this study is to investigate the effect of different laminin isoforms of the endothelial BM on iBMEC functionality and to determine whether better recapitulation of the physiological BM in vitro can address the aforementioned limitations of iBMECs. METHODS Using a previously reported method, hiPSCs were differentiated into iBMECs. The influence of main laminins of the endothelial BM, LN 411 and LN 511, on iBMEC functionality was studied and compared to a collagen IV and fibronectin mixture (CN IV-FN). Quantitative RT-PCR, immunocytochemistry, and TEER measurement were utilized to assess gene and protein expression and barrier properties of iBMECs on different extracellular matrices. Single-channel microfluidic devices were used to study the effect of shear stress on iBMECs. RESULTS LN 511, but not LN 411, improved iBMEC barrier properties and resulted in more sustained TEER stability. Immunocytochemistry showed improved junctional protein expression compared to iBMECs cultured on CN IV-FN. iBMECs cultured on LN 511 showed a reduction of stress fibers, indicating resting endothelial phenotype, whereas gene expression analysis revealed upregulation of multiple genes involved in endothelial activation in iBMECs on CN IV-FN. Finally, culturing iBMECs on LN 511 enhanced physiological responses to shear stress, including morphological changes and enhanced junctional protein association. CONCLUSION LN 511 improves the functionality and long-term barrier stability of iBMECs. Our findings suggest that incorporation of physiologically relevant LN 511 in iBMEC culture would be beneficial for disease modeling applications and BBB-on-a-chip platforms that accommodate fluid flow.
Collapse
Affiliation(s)
- Pedram Motallebnejad
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Samira M Azarin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
26
|
Lynn KS, Peterson RJ, Koval M. Ruffles and spikes: Control of tight junction morphology and permeability by claudins. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183339. [PMID: 32389670 DOI: 10.1016/j.bbamem.2020.183339] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/24/2020] [Accepted: 05/01/2020] [Indexed: 02/06/2023]
Abstract
Epithelial barrier function is regulated by a family of transmembrane proteins known as claudins. Functional tight junctions are formed when claudins interact with other transmembrane proteins, cytosolic scaffold proteins and the actin cytoskeleton. The predominant scaffold protein, zonula occludens-1 (ZO-1), directly binds to most claudin C-terminal domains, crosslinking them to the actin cytoskeleton. When imaged by immunofluorescence microscopy, tight junctions most frequently are linear structures that form between tricellular junctions. However, tight junctions also adapt non-linear architectures exhibiting either a ruffled or spiked morphology, which both are responses to changes in claudin engagement of actin filaments. Other terms for ruffled tight junctions include wavy, tortuous, undulating, serpentine or zig-zag junctions. Ruffling is under the control of hypoxia induced factor (HIF) and integrin-mediated signaling, as well as direct mechanical stimulation. Tight junction ruffling is specifically enhanced by claudin-2, antagonized by claudin-1 and requires claudin binding to ZO-1. Tight junction spikes are sites of active vesicle budding and fusion that appear as perpendicular projections oriented towards the nucleus. Spikes share molecular features with focal adherens junctions and tubulobulbar complexes found in Sertoli cells. Lung epithelial cells under stress form spikes due to an increase in claudin-5 expression that directly disrupts claudin-18/ZO-1 interactions. Together this suggests that claudins are not simply passive cargoes controlled by scaffold proteins. We propose a model where claudins specifically influence tight junction scaffold proteins to control interactions with the cytoskeleton as a mechanism that regulates tight junction assembly and function.
Collapse
Affiliation(s)
- K Sabrina Lynn
- Division of Pulmonary, Allergy Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Raven J Peterson
- Division of Pulmonary, Allergy Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Michael Koval
- Division of Pulmonary, Allergy Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
27
|
Pellicani R, Poletto E, Andreuzzi E, Paulitti A, Doliana R, Bizzotto D, Braghetta P, Colladel R, Tarticchio G, Sabatelli P, Bucciotti F, Bressan G, Iozzo RV, Colombatti A, Bonaldo P, Mongiat M. Multimerin-2 maintains vascular stability and permeability. Matrix Biol 2020; 87:11-25. [DOI: 10.1016/j.matbio.2019.08.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 12/12/2022]
|
28
|
Wu J, Rowart P, Jouret F, Gassaway BM, Rajendran V, Rinehart J, Caplan MJ. Mechanisms involved in AMPK-mediated deposition of tight junction components to the plasma membrane. Am J Physiol Cell Physiol 2020; 318:C486-C501. [PMID: 31913699 DOI: 10.1152/ajpcell.00422.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
AMP-activated protein kinase (AMPK) activation promotes early stages of epithelial junction assembly. AMPK activation in MDCK renal epithelial cells facilitates localization of the junction-associated proteins aPKCζ and Par3 to the plasma membrane and promotes conversion of Cdc42, a key regulator of epithelial polarization and junction assembly, to its active GTP bound state. Furthermore, Par3 is an important regulator of AMPK-mediated aPKCζ localization. Both aPKCζ and Par3 serve as intermediates in AMPK-mediated junction assembly, with inhibition of aPKCζ activity or Par3 knockdown disrupting AMPK's ability to facilitate zonula occludens (ZO-1) localization. AMPK phosphorylates the adherens junction protein afadin and regulates its interaction with the tight-junction protein zonula occludens-1. Afadin is phosphorylated at two critical sites, S228 (residing within an aPKCζ consensus site) and S1102 (residing within an AMPK consensus site), that are differentially regulated during junction assembly and that exert different effects on the process. Expression of phospho-defective mutants (S228A and S1102A) perturbed ZO-1 localization to the plasma membrane during AMPK-induced junction assembly. Expression of S228A increased the ZO-1/afadin interaction, while S1102A reduced this interaction during extracellular calcium-induced junction assembly. Inhibition of aPKCζ activity also increased the ZO-1/afadin interaction. Taken together, these data suggest that aPKCζ phosphorylation of afadin terminates the ZO-1/afadin interaction and thus permits the later stages of junction assembly.
Collapse
Affiliation(s)
- Jingshing Wu
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Pascal Rowart
- Groupe Interdisciplinaire de Génoprotéomique Appliquée, Cardiovascular Sciences, University of Liège, Liège, Belgium
| | - Francois Jouret
- Groupe Interdisciplinaire de Génoprotéomique Appliquée, Cardiovascular Sciences, University of Liège, Liège, Belgium
| | - Brandon M Gassaway
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut.,Systems Biology Institute, Yale University, West Haven, Connecticut
| | - Vanathy Rajendran
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Jesse Rinehart
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut.,Systems Biology Institute, Yale University, West Haven, Connecticut
| | - Michael J Caplan
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
29
|
Hiepen C, Jatzlau J, Hildebrandt S, Kampfrath B, Goktas M, Murgai A, Cuellar Camacho JL, Haag R, Ruppert C, Sengle G, Cavalcanti-Adam EA, Blank KG, Knaus P. BMPR2 acts as a gatekeeper to protect endothelial cells from increased TGFβ responses and altered cell mechanics. PLoS Biol 2019; 17:e3000557. [PMID: 31826007 PMCID: PMC6927666 DOI: 10.1371/journal.pbio.3000557] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 12/23/2019] [Accepted: 11/14/2019] [Indexed: 12/12/2022] Open
Abstract
Balanced transforming growth factor-beta (TGFβ)/bone morphogenetic protein (BMP)-signaling is essential for tissue formation and homeostasis. While gain in TGFβ signaling is often found in diseases, the underlying cellular mechanisms remain poorly defined. Here we show that the receptor BMP type 2 (BMPR2) serves as a central gatekeeper of this balance, highlighted by its deregulation in diseases such as pulmonary arterial hypertension (PAH). We show that BMPR2 deficiency in endothelial cells (ECs) does not abolish pan-BMP-SMAD1/5 responses but instead favors the formation of mixed-heteromeric receptor complexes comprising BMPR1/TGFβR1/TGFβR2 that enable enhanced cellular responses toward TGFβ. These include canonical TGFβ-SMAD2/3 and lateral TGFβ-SMAD1/5 signaling as well as formation of mixed SMAD complexes. Moreover, BMPR2-deficient cells express genes indicative of altered biophysical properties, including up-regulation of extracellular matrix (ECM) proteins such as fibrillin-1 (FBN1) and of integrins. As such, we identified accumulation of ectopic FBN1 fibers remodeled with fibronectin (FN) in junctions of BMPR2-deficient ECs. Ectopic FBN1 deposits were also found in proximity to contractile intimal cells in pulmonary artery lesions of BMPR2-deficient heritable PAH (HPAH) patients. In BMPR2-deficient cells, we show that ectopic FBN1 is accompanied by active β1-integrin highly abundant in integrin-linked kinase (ILK) mechano-complexes at cell junctions. Increased integrin-dependent adhesion, spreading, and actomyosin-dependent contractility facilitates the retrieval of active TGFβ from its latent fibrillin-bound depots. We propose that loss of BMPR2 favors endothelial-to-mesenchymal transition (EndMT) allowing cells of myo-fibroblastic character to create a vicious feed-forward process leading to hyperactivated TGFβ signaling. In summary, our findings highlight a crucial role for BMPR2 as a gatekeeper of endothelial homeostasis protecting cells from increased TGFβ responses and integrin-mediated mechano-transduction.
Collapse
Affiliation(s)
- Christian Hiepen
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| | - Jerome Jatzlau
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Germany
| | - Susanne Hildebrandt
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Germany
| | - Branka Kampfrath
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| | - Melis Goktas
- Max Planck Institute of Colloids and Interfaces, Mechano(bio)chemistry, Potsdam, Germany
| | - Arunima Murgai
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Germany
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | | | - Rainer Haag
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| | - Clemens Ruppert
- Universities of Giessen and Marburg Lung Center (UGMLC), Medical Clinic II, Justus Liebig University, Giessen, Germany
| | - Gerhard Sengle
- University of Cologne, Center for Biochemistry, Medical Faculty, Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
| | | | - Kerstin G. Blank
- Max Planck Institute of Colloids and Interfaces, Mechano(bio)chemistry, Potsdam, Germany
| | - Petra Knaus
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| |
Collapse
|
30
|
Colás-Algora N, Millán J. How many cadherins do human endothelial cells express? Cell Mol Life Sci 2019; 76:1299-1317. [PMID: 30552441 PMCID: PMC11105309 DOI: 10.1007/s00018-018-2991-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/16/2018] [Accepted: 12/06/2018] [Indexed: 12/13/2022]
Abstract
The vasculature is the paradigm of a compartment generated by parallel cellular barriers that aims to transport oxygen, nutrients and immune cells in complex organisms. Vascular barrier dysfunction leads to fatal acute and chronic inflammatory diseases. The endothelial barrier lines the inner side of vessels and is the main regulator of vascular permeability. Cadherins comprise a superfamily of 114 calcium-dependent adhesion proteins that contain conserved cadherin motifs and form cell-cell junctions in metazoans. In mature human endothelial cells, only VE (vascular endothelial)-cadherin and N (neural)-cadherin have been investigated in detail. Although both cadherins are essential for regulating endothelial permeability, no comprehensive expression studies to identify which other family members could play a relevant role in endothelial cells has so far been performed. Here, we have reviewed gene and protein expression databases to analyze cadherin expression in mature human endothelium and found that at least 24 cadherin superfamily members are significantly expressed. Based on data obtained from other cell types, organisms and experimental models, we discuss their potential functions, many of them unrelated to the formation of endothelial cell-cell junctions. The expression of this new set of endothelial cadherins highlights the important but still poorly defined roles of planar cell polarity, the Hippo pathway and mitochondria metabolism in human vascular homeostasis.
Collapse
Affiliation(s)
- Natalia Colás-Algora
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, Cantoblanco, 28049, Madrid, Spain
| | - Jaime Millán
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, Cantoblanco, 28049, Madrid, Spain.
| |
Collapse
|
31
|
Rieger J, Hopperdietzel C, Kaessmeyer S, Slosarek I, Diecke S, Richardson K, Plendl J. Human and equine endothelial cells in a live cell imaging scratch assay in vitro. Clin Hemorheol Microcirc 2019; 70:495-509. [DOI: 10.3233/ch-189316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Juliane Rieger
- Freie Universität Berlin, Department of Veterinary Medicine, Institute for Veterinary Anatomy, Germany
| | - Carsten Hopperdietzel
- Freie Universität Berlin, Department of Veterinary Medicine, Institute for Veterinary Anatomy, Germany
| | - Sabine Kaessmeyer
- Freie Universität Berlin, Department of Veterinary Medicine, Institute for Veterinary Anatomy, Germany
| | - Ilka Slosarek
- Freie Universität Berlin, Department of Veterinary Medicine, Institute for Veterinary Anatomy, Germany
| | - Sebastian Diecke
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), DZHK (German Centre for Cardiovascular Research), Partner Site, Germany; Berlin Institute of Health (BIH), Germany
| | - Ken Richardson
- Murdoch University, School of Veterinary and Life Sciences, Murdoch, WA, Australia
| | - Johanna Plendl
- Freie Universität Berlin, Department of Veterinary Medicine, Institute for Veterinary Anatomy, Germany
| |
Collapse
|
32
|
Weng J, Yu L, Chen Z, Su H, Yu S, Zhang Y, Lei X, Chen L, Cui Y, Huang Q, Jiang Y, Guo X. β-Catenin phosphorylation at Y654 and Y142 is crucial for high mobility group box-1 protein-induced pulmonary vascular hyperpermeability. J Mol Cell Cardiol 2019; 127:174-184. [DOI: 10.1016/j.yjmcc.2018.12.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 12/21/2018] [Accepted: 12/25/2018] [Indexed: 12/19/2022]
|
33
|
Cao J, Schnittler H. Putting VE-cadherin into JAIL for junction remodeling. J Cell Sci 2019; 132:132/1/jcs222893. [DOI: 10.1242/jcs.222893] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
ABSTRACT
Junction dynamics of endothelial cells are based on the integration of signal transduction, cytoskeletal remodeling and contraction, which are necessary for the formation and maintenance of monolayer integrity, but also enable repair and regeneration. The VE-cadherin–catenin complex forms the molecular basis of the adherence junctions and cooperates closely with actin filaments. Several groups have recently described small actin-driven protrusions at the cell junctions that are controlled by the Arp2/3 complex, contributing to cell junction regulation. We identified these protrusions as the driving force for VE-cadherin dynamics, as they directly induce new VE-cadherin-mediated adhesion sites, and have accordingly referred to these structures as junction-associated intermittent lamellipodia (JAIL). JAIL extend over only a few microns and thus provide the basis for a subcellular regulation of adhesion. The local (subcellular) VE-cadherin concentration and JAIL formation are directly interdependent, which enables autoregulation. Therefore, this mechanism can contribute a subcellularly regulated adaptation of cell contact dynamics, and is therefore of great importance for monolayer integrity and relative cell migration during wound healing and angiogenesis, as well as for inflammatory responses. In this Review, we discuss the mechanisms and functions underlying these actin-driven protrusions and consider their contribution to the dynamic regulation of endothelial cell junctions.
Collapse
Affiliation(s)
- Jiahui Cao
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Münster Germany
| | - Hans Schnittler
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Münster Germany
| |
Collapse
|
34
|
Histamine causes endothelial barrier disruption via Ca 2+-mediated RhoA activation and tension at adherens junctions. Sci Rep 2018; 8:13229. [PMID: 30185878 PMCID: PMC6125323 DOI: 10.1038/s41598-018-31408-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 08/03/2018] [Indexed: 12/21/2022] Open
Abstract
During inflammation, the disruption of the endothelial barrier leads to increased microvascular permeability. Whether tension along cell junctions contributes to histamine-induced endothelial barrier disruption remains unknown. Rapid Ca2+ influx induced by both histamine and thrombin was accompanied by endothelial barrier breakdown revealed as drop of transendothelial electric resistance in primary human microvascular endothelial cells. Interestingly, GLISA measurements revealed activation of RhoA but not inactivation of Rac1 at the time-point of barrier breakdown. FRET measurements showed activation of RhoA at intercellular junctions after both thrombin and histamine exposure. Breakdown coincided with increased stress fiber formation but not with translocation of vinculin, which was located along junctions in the resting state similar to postcapillary venules ex vivo. Moreover, increased tension at AJs was indicated by immunostaining with a conformation-sensitive antibody targeting the α18-subunit of α-catenin. Ca2+ chelation by BAPTA-AM and ROCK1 inhibition by Y27632 abolished both increase of tension along AJs as well as barrier dysfunction. Moreover, BAPTA-AM decreased RhoA activation following histamine stimulation, indicating a key role of Ca2+ signaling in barrier breakdown. Taken together, in response to histamine, Ca2+ via RhoA/ROCK activation along endothelial adherens junctions (AJs) appears to be critical for barrier disruption and presumably correlated with enhanced tension. However, vinculin appears not to be critical in this process.
Collapse
|
35
|
Fong LY, Ng CT, Yong YK, Hakim MN, Ahmad Z. Asiatic acid stabilizes cytoskeletal proteins and prevents TNF-α-induced disorganization of cell-cell junctions in human aortic endothelial cells. Vascul Pharmacol 2018; 117:15-26. [PMID: 30114509 DOI: 10.1016/j.vph.2018.08.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/12/2018] [Accepted: 08/11/2018] [Indexed: 12/22/2022]
Abstract
Endothelial hyperpermeability represents an initiating step in early atherosclerosis and it often occurs as a result of endothelial barrier dysfunction. Asiatic acid, a major triterpene isolated from Centella asiatica (L.) Urban, has previously been demonstrated to protect against tumor necrosis factor (TNF)-α-induced endothelial barrier dysfunction. The present study aimed to investigate the mechanisms underlying the barrier protective effect of asiatic acid in human aortic endothelial cells (HAECs). The localization of F-actin, diphosphorylated myosin light chain (diphospho-MLC), adherens junctions (AJs) and tight junctions (TJs) was studied using immunocytochemistry techniques and confocal microscopy. Their total protein expressions were examined using western blot analysis. The endothelial permeability was assessed using In Vitro Vascular Permeability Assay kits. In addition, intracellular redistribution of the junctional proteins was evaluated using subcellular fractionation kits. We show that asiatic acid stabilized F-actin and diphospho-MLC at the cell periphery and prevented their rearrangement stimulated by TNF-α. However, asiatic acid failed to attenuate cytochalasin D-induced increased permeability. Besides, asiatic acid abrogated TNF-α-induced structural reorganization of vascular endothelial (VE)-cadherin and β-catenin by preserving their reticulum structures at cell-cell contact areas. In addition, asiatic acid also inhibited TNF-α-induced redistribution of occludin and zona occludens (ZO)-1 in different subcellular fractions. In conclusion, the barrier-stabilizing effect of asiatic acid might be associated with preservation of AJs and prevention of TJ redistribution caused by TNF-α. This study provides evidence to support the potential use of asiatic acid in the prevention of early atherosclerosis, which is initiated by endothelial barrier dysfunction.
Collapse
Affiliation(s)
- Lai Yen Fong
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia; Department of Pre-clinical Sciences, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, 43000 Kajang, Selangor, Malaysia.
| | - Chin Theng Ng
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia; Physiology Unit, Faculty of Medicine, AIMST University, 08100 Bedong, Kedah, Malaysia
| | - Yoke Keong Yong
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Muhammad Nazrul Hakim
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Zuraini Ahmad
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| |
Collapse
|
36
|
Kim J, Cooper JA. Septins regulate junctional integrity of endothelial monolayers. Mol Biol Cell 2018; 29:1693-1703. [PMID: 29771630 PMCID: PMC6080707 DOI: 10.1091/mbc.e18-02-0136] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 04/25/2018] [Accepted: 05/09/2018] [Indexed: 01/03/2023] Open
Abstract
Junctional integrity of endothelial monolayers is crucial to control movement of molecules and cells across the endothelium. Examining the structure and dynamics of cell junctions in endothelial monolayers, we discovered a role for septins. Contacts between adjacent endothelial cells were dynamic, with protrusions extending above or below neighboring cells. Vascular endothelial cadherin (VE-cadherin) was present at cell junctions, with a membrane-associated layer of F-actin. Septins localized at cell-junction membranes, in patterns distinct from VE-cadherin and F-actin. Septins assumed curved and scallop-shaped patterns at junctions, especially in regions of positive membrane curvature associated with actin-rich membrane protrusions. Depletion of septins led to disrupted morphology of VE-cadherin junctions and increased expression of VE-cadherin. In videos, septin-depleted cells displayed remodeling at cell junctions; regions with VE-cadherin were broader, and areas with membrane ruffling were wider. Septin depletion and junction disruption led to functional loss of junctional integrity, revealed by decreased transendothelial electric resistance and increased transmigration of immune cells. We conclude that septins, as cytoskeletal elements associated with the plasma membrane, are important for cell junctions and junctional integrity of endothelial monolayers, functioning at regions of positive curvature in support of actin-rich protrusions to promote cadherin-based cell junctions.
Collapse
Affiliation(s)
- Joanna Kim
- Departments of Biochemistry & Molecular Biophysics and Cell Biology & Physiology, Washington University, St. Louis, MO 63110
| | - John A. Cooper
- Departments of Biochemistry & Molecular Biophysics and Cell Biology & Physiology, Washington University, St. Louis, MO 63110
| |
Collapse
|
37
|
Choi HJ, Kim NE, Kim J, An S, Yang SH, Ha J, Cho S, Kwon I, Kim YD, Nam HS, Heo JH. Dabigatran reduces endothelial permeability through inhibition of thrombin-induced cytoskeleton reorganization. Thromb Res 2018; 167:S0049-3848(18)30324-4. [PMID: 29735342 DOI: 10.1016/j.thromres.2018.04.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/28/2018] [Accepted: 04/18/2018] [Indexed: 11/23/2022]
Abstract
Dabigatran etexilate (DE), a new oral anti-coagulant, is a direct thrombin inhibitor. Clinical trials showed the favorable benefit-to-risk profile of DE compared to warfarin for the prevention of ischemic stroke in patients with atrial fibrillation. Remarkably, patients treated with dabigatran showed reduced rates of intracerebral hemorrhage compared to warfarin. As the breakdown of endothelial barrier integrity is associated with hemorrhagic events and as thrombin increases endothelial permeability, we hypothesized that dabigatran preserves the endothelial barrier by inhibiting thrombin-induced permeability. We assessed leakage of fluorescein isothiocyanate (FITC)-dextran through the endothelial monolayer and measured trans-endothelial electrical resistance of the endothelial monolayer after treatment of thrombin or thrombin pre-incubated with dabigatran. Thrombin increased the permeability of endothelial cells. Dabigatran effectively blocked the ability of thrombin to increase permeability. Dabigatran inhibited the formation of actin stress fibers induced by thrombin and inhibited consequent destabilization of junctional protein complexes and intercellular gap formation. The interaction of thrombin with protease activated receptor-1 activates the Rho A guanosine triphosphate (GTP)ase-myosin light chain (MLC) phosphorylation signaling axis, leading to actin cytoskeleton changes. This signaling pathway was effectively inhibited by dabigatran in endothelial cells. Consistently, the number of phosphorylated MLC-positive cells was significantly decreased in ischemic tissue of rat brains. These results indicate dabigatran blocks the ability of thrombin to induce vascular permeability and the resulting underlying signaling cascade in endothelial cells. Our findings provide evidence that dabigatran may confer a lower risk of intracerebral hemorrhage by preserving endothelial barrier integrity.
Collapse
Affiliation(s)
- Hyun-Jung Choi
- Severance Integrative Research Institute for Cerebral & Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Na-Eun Kim
- Severance Integrative Research Institute for Cerebral & Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jayoung Kim
- Severance Integrative Research Institute for Cerebral & Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sunho An
- Severance Integrative Research Institute for Cerebral & Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea; Brain Korea 21 Plus Project for Medical Science, Yonsei University, Seoul 03722, Republic of Korea
| | - Seung-Hee Yang
- Severance Integrative Research Institute for Cerebral & Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jimin Ha
- Severance Integrative Research Institute for Cerebral & Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea; Brain Korea 21 Plus Project for Medical Science, Yonsei University, Seoul 03722, Republic of Korea
| | - Sunghee Cho
- The Burke-Cornell Medical Research Institute, White Plains, NY 10605, United States; Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10021, United States
| | - Il Kwon
- Severance Integrative Research Institute for Cerebral & Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea; Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Dae Kim
- Severance Integrative Research Institute for Cerebral & Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyo Suk Nam
- Severance Integrative Research Institute for Cerebral & Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ji Hoe Heo
- Severance Integrative Research Institute for Cerebral & Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea; Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Brain Korea 21 Plus Project for Medical Science, Yonsei University, Seoul 03722, Republic of Korea.
| |
Collapse
|
38
|
Smit KF, Konkel M, Kerindongo R, Landau MA, Zuurbier CJ, Hollmann MW, Preckel B, Nieuwland R, Albrecht M, Weber NC. Helium alters the cytoskeleton and decreases permeability in endothelial cells cultured in vitro through a pathway involving Caveolin-1. Sci Rep 2018; 8:4768. [PMID: 29555979 PMCID: PMC5859123 DOI: 10.1038/s41598-018-23030-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 03/05/2018] [Indexed: 01/31/2023] Open
Abstract
Caveolins are involved in anaesthetic-induced cardioprotection. Actin filaments are located in close connection to Caveolins in the plasma membrane. We hypothesised that helium might affect the cytoskeleton and induce secretion of Caveolin. HCAEC, HUVEC and Cav-1 siRNA transfected HUVEC were exposed for 20 minutes to either helium (5% CO2, 25% O2, 70% He) or control gas (5% CO2, 25% O2, 70% N2). Cells and supernatants were collected for infrared Western blot analysis, immunofluorescence staining, nanoparticle tracking analysis and permeability measurements. Helium treatment increased the cortical localisation of F-actin fibers in HUVEC. After 6 hours, helium decreased cellular Caveolin-1 (Cav-1) levels and increased Cav-1 levels in the supernatant. Cell permeability was decreased 6 and 12 hours after helium treatment, and increased levels of Vascular Endothelial - Cadherin (VE-Cadherin) and Connexin 43 (Cx43) were observed. Transfection with Cav-1 siRNA abolished the effects of helium treatment on VE-Cadherin, Cx43 levels and permeability. Supernatant obtained after helium treatment reduced cellular permeability in remote HUVEC, indicating that increased levels of Cav-1 are responsible for the observed alterations. These findings suggest that Cav-1 is secreted after helium exposure in vitro, altering the cytoskeleton and increasing VE-Cadherin and Cx43 expression resulting in decreased permeability in HUVEC.
Collapse
Affiliation(s)
- Kirsten F Smit
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A), Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Moritz Konkel
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A), Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
- Department of Anaesthesiology, UKSH, Campus Kiel, Kiel, Germany
| | - Raphaela Kerindongo
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A), Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Maximilian A Landau
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A), Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
- Department of Anaesthesiology, UKSH, Campus Kiel, Kiel, Germany
| | - Coert J Zuurbier
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A), Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Markus W Hollmann
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A), Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Benedikt Preckel
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A), Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, and Vesicle Observation Centre, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Martin Albrecht
- Department of Anaesthesiology, UKSH, Campus Kiel, Kiel, Germany
| | - Nina C Weber
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A), Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands.
| |
Collapse
|
39
|
Efimova N, Svitkina TM. Branched actin networks push against each other at adherens junctions to maintain cell-cell adhesion. J Cell Biol 2018; 217:1827-1845. [PMID: 29507127 PMCID: PMC5940301 DOI: 10.1083/jcb.201708103] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 12/21/2017] [Accepted: 02/12/2018] [Indexed: 12/14/2022] Open
Abstract
Adherens junctions (AJs) are mechanosensitive cadherin-based intercellular adhesions that interact with the actin cytoskeleton and carry most of the mechanical load at cell-cell junctions. Both Arp2/3 complex-dependent actin polymerization generating pushing force and nonmuscle myosin II (NMII)-dependent contraction producing pulling force are necessary for AJ morphogenesis. Which actin system directly interacts with AJs is unknown. Using platinum replica electron microscopy of endothelial cells, we show that vascular endothelial (VE)-cadherin colocalizes with Arp2/3 complex-positive actin networks at different AJ types and is positioned at the interface between two oppositely oriented branched networks from adjacent cells. In contrast, actin-NMII bundles are located more distally from the VE-cadherin-rich zone. After Arp2/3 complex inhibition, linear AJs split, leaving gaps between cells with detergent-insoluble VE-cadherin transiently associated with the gap edges. After NMII inhibition, VE-cadherin is lost from gap edges. We propose that the actin cytoskeleton at AJs acts as a dynamic push-pull system, wherein pushing forces maintain extracellular VE-cadherin transinteraction and pulling forces stabilize intracellular adhesion complexes.
Collapse
Affiliation(s)
- Nadia Efimova
- Department of Biology, University of Pennsylvania, Philadelphia, PA
| | | |
Collapse
|
40
|
The epigenetic factor KDM2B regulates cell adhesion, small rho GTPases, actin cytoskeleton and migration in prostate cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:587-597. [PMID: 29408056 DOI: 10.1016/j.bbamcr.2018.01.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/12/2018] [Accepted: 01/17/2018] [Indexed: 12/19/2022]
Abstract
The histone demethylase KDM2B is an epigenetic factor with oncogenic properties that is regulated by the basic fibroblasts growth factor (FGF-2). It has recently been shown that KDM2B co-operates with Polycomb Group proteins to promote cell migration and angiogenesis in tumors. In the present study we addressed the role of KDM2B in regulating actin cytoskeleton signaling, cell-cell adhesion and migration of prostate tumor cells. We report here that KDM2B is functionally expressed in DU-145 prostate cancer cells, activated by FGF-2 and regulates EZH2. KDM2B knockdown induced potent up-regulation of gene transcription and protein expression of the epithelial markers E-cadherin and ZO-1, while KDM2B overexpression down-regulated the levels of both markers, suggesting control of cell adhesion by KDM2B. RhoA and RhoB protein expression and activity were diminished upon KDM2B-knockdown and upregulated in KDM2B-overexpressing cell clones. In accordance, actin reorganization with formation of stress fibers became evident in KDM2B-overexpressing cells and abolished in the presence of the Rho inhibitor C3 transferase. DU-145 cell migration was significantly enhanced in KDM2B overexpressing cells and abolished in C3-pretreated cells. Conversely, the retardation of cell migration observed in KDM2B knockdown cells was enhanced in C3-pretreated cells. These results establish a clear functional link between the epigenetic factor KDM2B and the regulation of cell adhesion and Rho-GTPases signaling that controls actin reorganization and cell migration.
Collapse
|
41
|
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: 169] [Impact Index Per Article: 28.2] [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
|
42
|
Prystopiuk V, Fels B, Simon CS, Liashkovich I, Pasrednik D, Kronlage C, Wedlich-Söldner R, Oberleithner H, Fels J. A two-phase response of endothelial cells to hydrostatic pressure. J Cell Sci 2018; 131:jcs.206920. [DOI: 10.1242/jcs.206920] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 05/10/2018] [Indexed: 01/15/2023] Open
Abstract
The vascular endothelium is exposed to three types of mechanical forces: blood flow-mediated shear stress, vessel-diameter dependent wall tension and hydrostatic pressure. Despite considerable variations of blood pressure in normal and pathological physiology, little is known about the acute molecular and cellular effects of hydrostatic pressure on endothelial cells. Here, we used a combination of quantitative fluorescence microscopy, atomic force microscopy and molecular perturbations to characterize the specific response of endothelial cells to pressure application. We identified a two-phase response of endothelial cells to acute (1 h) vs. chronic (24 h) pressure application (100 mmHg). While both regimes induce cortical stiffening, the acute response is linked to calcium-mediated myosin activation, whereas the chronic cell response is dominated by increased cortical actin density and a loss in endothelial barrier function. GsMTx-4 and amiloride inhibit the acute pressure response, which suggest the sodium channel ENaC as key player in endothelial pressure sensing. The described two-phase pressure response may participate in the differential effects of transient changes in blood pressure and hypertension.
Collapse
Affiliation(s)
- Valeria Prystopiuk
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
- current address: Institute of Life Sciences, Université Catholique de Louvain, Croix du Sud, 4-5, bte L7.07.06, Louvain-la-Neuve B-1348, Belgium
| | - Benedikt Fels
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| | - Caroline Sophie Simon
- Institute of Cell Dynamics and Imaging, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| | - Ivan Liashkovich
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| | - Dzmitry Pasrednik
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| | - Cornelius Kronlage
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| | - Roland Wedlich-Söldner
- Institute of Cell Dynamics and Imaging, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| | - Hans Oberleithner
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| | - Johannes Fels
- Institute of Cell Dynamics and Imaging, University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149, Münster, Germany
| |
Collapse
|
43
|
Parthasarathi K. The Pulmonary Vascular Barrier: Insights into Structure, Function, and Regulatory Mechanisms. MOLECULAR AND FUNCTIONAL INSIGHTS INTO THE PULMONARY VASCULATURE 2018; 228:41-61. [DOI: 10.1007/978-3-319-68483-3_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
44
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
45
|
López-Posadas R, Stürzl M, Atreya I, Neurath MF, Britzen-Laurent N. Interplay of GTPases and Cytoskeleton in Cellular Barrier Defects during Gut Inflammation. Front Immunol 2017; 8:1240. [PMID: 29051760 PMCID: PMC5633683 DOI: 10.3389/fimmu.2017.01240] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 09/19/2017] [Indexed: 12/24/2022] Open
Abstract
An essential role of the intestine is to build and maintain a barrier preventing the luminal gut microbiota from invading the host. This involves two coordinated physical and immunological barriers formed by single layers of intestinal epithelial and endothelial cells, which avoid the activation of local immune responses or the systemic dissemination of microbial agents, and preserve tissue homeostasis. Accordingly, alterations of epithelial and endothelial barrier functions have been associated with gut inflammation, for example during inflammatory bowel disease (IBD). The discriminative control of nutriment uptake and sealing toward potentially pathological microorganisms requires a profound regulation of para- and transcellular permeability. On the subcellular level, the cytoskeleton exerts key regulatory functions in the maintenance of cellular barriers. Increased epithelial/endothelial permeability occurs primarily as a result of a reorganization of cytoskeletal–junctional complexes. Pro-inflammatory mediators such as cytokines can induce cytoskeletal rearrangements, causing inflammation-dependent defects in gut barrier function. In this context, small GTPases of the Rho family and large GTPases from the Dynamin superfamily appear as major cellular switches regulating the interaction between intercellular junctions and actomyosin complexes, and in turn cytoskeleton plasticity. Strikingly, some of these proteins, such as RhoA or guanylate-binding protein-1 (GBP-1) have been associated with gut inflammation and IBD. In this review, we will summarize the role of small and large GTPases for cytoskeleton plasticity and epithelial/endothelial barrier in the context of gut inflammation.
Collapse
Affiliation(s)
| | | | - Imke Atreya
- Universitätsklinikum Erlangen, Erlangen, Germany
| | | | | |
Collapse
|
46
|
Kreutzman A, Colom-Fernández B, Jiménez AM, Ilander M, Cuesta-Mateos C, Pérez-García Y, Arévalo CD, Brück O, Hakanen H, Saarela J, Ortega-Carrión A, de Rosendo A, Juanes-García A, Steegmann JL, Mustjoki S, Vicente-Manzanares M, Muñoz-Calleja C. Dasatinib Reversibly Disrupts Endothelial Vascular Integrity by Increasing Non-Muscle Myosin II Contractility in a ROCK-Dependent Manner. Clin Cancer Res 2017; 23:6697-6707. [DOI: 10.1158/1078-0432.ccr-16-0667] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 08/10/2016] [Accepted: 08/10/2017] [Indexed: 11/16/2022]
|
47
|
Yao Q, Tu C, Lu D, Zou Y, Liu H, Zhang S. Clinicopathological significance of the microRNA-146a/WASP-family verprolin-homologous protein-2 axis in gastric cancer. Cancer Sci 2017; 108:1285-1292. [PMID: 28387985 PMCID: PMC5497796 DOI: 10.1111/cas.13254] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 04/01/2017] [Accepted: 04/05/2017] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is one of the most common malignancies, and cancer invasion and metastasis are the leading causes of cancer‐induced death in GC patients. WASP‐family verprolin‐homologous protein‐2 (WASF2), with a role controlling actin polymerization which is critical in the formation of membrane protrusions involved in cell migration and invasion, has been reported to possess cancer‐promoting effects in several cancers. However, data of WASF2's role in GC are relatively few and even contradictory. In this study, we analyzed WASF2 expression in GC tissues and their corresponding adjacent normal tissues. We found that WASF2 was upregulated in GC tissues and high level of WASF2 was associated with lymph node metastasis of GC. Through gain‐ and loss‐of‐function studies, WASF2 was shown to significantly increase GC cells migration and invasion, but had no effect on proliferation in vitro. Importantly, WASF2 was also found to enhance GC metastasis in vivo. Our previous research suggested that WASF2 was a direct target of microRNA‐146a (miR‐146a). Furthermore, we analyzed miR‐146a's level in GC tissues and their corresponding adjacent normal tissues. We found that miR‐146a was downregulated in GC tissues and low miR‐146a level was associated with advanced TNM stage and lymph node metastasis. The level of WASF2 in GC tissues was negatively correlated with miR‐146a expression and had inverse clinicopathologic features. The newly identified miR‐146a/WASF2 axis may provide a novel therapeutic target for GC.
Collapse
Affiliation(s)
- Qunyan Yao
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chuantao Tu
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Di Lu
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanting Zou
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hongchun Liu
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuncai Zhang
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
48
|
AM966, an Antagonist of Lysophosphatidic Acid Receptor 1, Increases Lung Microvascular Endothelial Permeability through Activation of Rho Signaling Pathway and Phosphorylation of VE-Cadherin. Mediators Inflamm 2017; 2017:6893560. [PMID: 28348461 PMCID: PMC5350330 DOI: 10.1155/2017/6893560] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 01/04/2017] [Accepted: 01/15/2017] [Indexed: 01/15/2023] Open
Abstract
Maintenance of pulmonary endothelial barrier integrity is important for reducing severity of lung injury. Lysophosphatidic acid (LPA) regulates cell motility, cytoskeletal rearrangement, and cell growth. Knockdown of LPA receptor 1 (LPA1) has been shown to mitigate lung injury and pulmonary fibrosis. AM966, an LPA1 antagonist exhibiting an antifibrotic property, has been considered to be a future antifibrotic medicine. Here, we report an unexpected effect of AM966, which increases lung endothelial barrier permeability. An electric cell-substrate sensing (ECIS) system was used to measure permeability in human lung microvascular endothelial cells (HLMVECs). AM966 decreased the transendothelial electrical resistance (TEER) value immediately in a dose-dependent manner. VE-cadherin and f-actin double immunostaining reveals that AM966 increases stress fibers and gap formation between endothelial cells. AM966 induced phosphorylation of myosin light chain (MLC) through activation of RhoA/Rho kinase pathway. Unlike LPA treatment, AM966 had no effect on phosphorylation of extracellular signal-regulated kinases (Erk). Further, in LPA1 silencing cells, we observed that AM966-increased lung endothelial permeability as well as phosphorylation of VE-cadherin and focal adhesion kinase (FAK) were attenuated. This study reveals that AM966 induces lung endothelial barrier dysfunction, which is regulated by LPA1-mediated activation of RhoA/MLC and phosphorylation of VE-cadherin.
Collapse
|
49
|
Belvitch P, Brown ME, Brinley BN, Letsiou E, Rizzo AN, Garcia JGN, Dudek SM. The ARP 2/3 complex mediates endothelial barrier function and recovery. Pulm Circ 2017; 7:200-210. [PMID: 28680579 PMCID: PMC5448540 DOI: 10.1086/690307] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 11/23/2016] [Indexed: 01/03/2023] Open
Abstract
Pulmonary endothelial cell (EC) barrier dysfunction and recovery is critical to the pathophysiology of acute respiratory distress syndrome. Cytoskeletal and subsequent cell membrane dynamics play a key mechanistic role in determination of EC barrier integrity. Here, we characterizAQe the actin related protein 2/3 (Arp 2/3) complex, a regulator of peripheral branched actin polymerization, in human pulmonary EC barrier function through studies of transendothelial electrical resistance (TER), intercellular gap formation, peripheral cytoskeletal structures and lamellipodia. Compared to control, Arp 2/3 inhibition with the small molecule inhibitor CK-666 results in a reduction of baseline barrier function (1,241 ± 53 vs 988 ± 64 ohm; p < 0.01), S1P-induced barrier enhancement and delayed recovery of barrier function after thrombin (143 ± 14 vs 93 ± 6 min; p < 0.01). Functional changes of Arp 2/3 inhibition on barrier integrity are associated temporally with increased intercellular gap area at baseline (0.456 ± 0.02 vs 0.299 ± 0.02; p < 0.05) and thirty minutes after thrombin (0.885 ± 0.03 vs 0.754 ± 0.03; p < 0.05). Immunofluorescent microscopy reveals reduced lamellipodia formation after S1P and during thrombin recovery in Arp 2/3 inhibited cells. Individual lamellipodia demonstrate reduced depth following Arp 2/3 inhibition vs vehicle at baseline (1.83 ± 0.41 vs 2.55 ± 0.46 µm; p < 0.05) and thirty minutes after S1P treatment (1.53 ± 0.37 vs 2.09 ± 0.36 µm; p < 0.05). These results establish a critical role for Arp 2/3 activity in determination of pulmonary endothelial barrier function and recovery through formation of EC lamellipodia and closure of intercellular gaps.
Collapse
Affiliation(s)
- Patrick Belvitch
- Division of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois Hospital and Health Science System, Chicago, IL, USA
| | - Mary E Brown
- Division of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois Hospital and Health Science System, Chicago, IL, USA
| | | | - Eleftheria Letsiou
- Division of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois Hospital and Health Science System, Chicago, IL, USA
| | - Alicia N Rizzo
- Division of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois Hospital and Health Science System, Chicago, IL, USA
| | - Joe G N Garcia
- University of Arizona Health Sciences Center, Tucson, AZ, USA
| | - Steven M Dudek
- Division of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois Hospital and Health Science System, Chicago, IL, USA
| |
Collapse
|
50
|
Domanski D, Zegrocka-Stendel O, Perzanowska A, Dutkiewicz M, Kowalewska M, Grabowska I, Maciejko D, Fogtman A, Dadlez M, Koziak K. Molecular Mechanism for Cellular Response to β-Escin and Its Therapeutic Implications. PLoS One 2016; 11:e0164365. [PMID: 27727329 PMCID: PMC5058498 DOI: 10.1371/journal.pone.0164365] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 09/23/2016] [Indexed: 11/18/2022] Open
Abstract
β-escin is a mixture of triterpene saponins isolated from the horse chestnut seeds (Aesculus hippocastanum L.). The anti-edematous, anti-inflammatory and venotonic properties of β-escin have been the most extensively clinically investigated effects of this plant-based drug and randomized controlled trials have proved the efficacy of β-escin for the treatment of chronic venous insufficiency. However, despite the clinical recognition of the drug its pharmacological mechanism of action still remains largely elusive. To determine the cellular and molecular basis for the therapeutic effectiveness of β-escin we performed discovery and targeted proteomic analyses and in vitro evaluation of cellular and molecular responses in human endothelial cells under inflammatory conditions. Our results demonstrate that in endothelial cells β-escin potently induces cholesterol synthesis which is rapidly followed with marked fall in actin cytoskeleton integrity. The concomitant changes in cell functioning result in a significantly diminished responses to TNF-α stimulation. These include reduced migration, alleviated endothelial monolayer permeability, and inhibition of NFκB signal transduction leading to down-expression of TNF-α-induced effector proteins. Moreover, the study provides evidence for novel therapeutic potential of β-escin beyond the current vascular indications.
Collapse
Affiliation(s)
- Dominik Domanski
- Laboratory of Mass Spectrometry, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Oliwia Zegrocka-Stendel
- Centre for Preclinical Research and Technology, Department of Immunology, Biochemistry and Nutrition, Medical University of Warsaw, Warsaw, Poland
| | - Anna Perzanowska
- Laboratory of Mass Spectrometry, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Malgorzata Dutkiewicz
- Centre for Preclinical Research and Technology, Department of Immunology, Biochemistry and Nutrition, Medical University of Warsaw, Warsaw, Poland
| | - Magdalena Kowalewska
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Iwona Grabowska
- Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Miecznikowa 1, 02–096 Warsaw, Poland
| | - Dorota Maciejko
- Centre for Preclinical Research and Technology, Department of Immunology, Biochemistry and Nutrition, Medical University of Warsaw, Warsaw, Poland
| | - Anna Fogtman
- Laboratory for Microarray Analysis CORELAB, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Michal Dadlez
- Laboratory of Mass Spectrometry, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Koziak
- Centre for Preclinical Research and Technology, Department of Immunology, Biochemistry and Nutrition, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|