201
|
Saharinen P, Eklund L, Alitalo K. Therapeutic targeting of the angiopoietin-TIE pathway. Nat Rev Drug Discov 2017; 16:635-661. [PMID: 28529319 DOI: 10.1038/nrd.2016.278] [Citation(s) in RCA: 375] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The endothelial angiopoietin (ANG)-TIE growth factor receptor pathway regulates vascular permeability and pathological vascular remodelling during inflammation, tumour angiogenesis and metastasis. Drugs that target the ANG-TIE pathway are in clinical development for oncological and ophthalmological applications. The aim is to complement current vascular endothelial growth factor (VEGF)-based anti-angiogenic therapies in cancer, wet age-related macular degeneration and macular oedema. The unique function of the ANG-TIE pathway in vascular stabilization also renders this pathway an attractive target in sepsis, organ transplantation, atherosclerosis and vascular complications of diabetes. This Review covers key aspects of the function of the ANG-TIE pathway in vascular disease and describes the recent development of novel therapeutics that target this pathway.
Collapse
Affiliation(s)
- Pipsa Saharinen
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, P.O. Box 63, FI-00014 Helsinki, Finland
| | - Lauri Eklund
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, Aapistie 5A, University of Oulu, 90220 Oulu, Finland
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, P.O. Box 63, FI-00014 Helsinki, Finland
| |
Collapse
|
202
|
Soni D, Regmi SC, Wang DM, DebRoy A, Zhao YY, Vogel SM, Malik AB, Tiruppathi C. Pyk2 phosphorylation of VE-PTP downstream of STIM1-induced Ca 2+ entry regulates disassembly of adherens junctions. Am J Physiol Lung Cell Mol Physiol 2017; 312:L1003-L1017. [PMID: 28385807 DOI: 10.1152/ajplung.00008.2017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/28/2017] [Accepted: 03/29/2017] [Indexed: 11/22/2022] Open
Abstract
Vascular endothelial protein tyrosine phosphatase (VE-PTP) stabilizes endothelial adherens junctions (AJs) through constitutive dephosphorylation of VE-cadherin. Here we investigated the role of stromal interaction molecule 1 (STIM1) activation of store-operated Ca2+ entry (SOCE) in regulating AJ assembly. We observed that SOCE induced by STIM1 activated Pyk2 in human lung microvascular endothelial cells (ECs) and induced tyrosine phosphorylation of VE-PTP at Y1981. Pyk2-induced tyrosine phosphorylation of VE-PTP promoted Src binding to VE-PTP, Src activation, and subsequent VE-cadherin phosphorylation and thereby increased the endothelial permeability response. The increase in permeability was secondary to disassembly of AJs. Pyk2-mediated responses were blocked in EC-restricted Stim1 knockout mice, indicating the requirement for STIM1 in initiating the signaling cascade. A peptide derived from the Pyk2 phosphorylation site on VE-PTP abolished the STIM1/SOCE-activated permeability response. Thus Pyk2 activation secondary to STIM1-induced SOCE causes tyrosine phosphorylation of VE-PTP, and VE-PTP, in turn, binds to and activates Src, thereby phosphorylating VE-cadherin to increase endothelial permeability through disassembly of AJs. Our results thus identify a novel signaling mechanism by which STIM1-induced Ca2+ signaling activates Pyk2 to inhibit the interaction of VE-PTP and VE-cadherin and hence increase endothelial permeability. Therefore, targeting the Pyk2 activation pathway may be a potentially important anti-inflammatory strategy.
Collapse
Affiliation(s)
- Dheeraj Soni
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois
| | - Sushil C Regmi
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois
| | - Dong-Mei Wang
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois
| | - Auditi DebRoy
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois
| | - You-Yang Zhao
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois
| | - Stephen M Vogel
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois
| | - Asrar B Malik
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois
| | - Chinnaswamy Tiruppathi
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois
| |
Collapse
|
203
|
Dragoni S, Hudson N, Kenny BA, Burgoyne T, McKenzie JA, Gill Y, Blaber R, Futter CE, Adamson P, Greenwood J, Turowski P. Endothelial MAPKs Direct ICAM-1 Signaling to Divergent Inflammatory Functions. THE JOURNAL OF IMMUNOLOGY 2017; 198:4074-4085. [PMID: 28373581 PMCID: PMC5421301 DOI: 10.4049/jimmunol.1600823] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 03/07/2017] [Indexed: 12/30/2022]
Abstract
Lymphocyte transendothelial migration (TEM) is critically dependent on intraendothelial signaling triggered by adhesion to ICAM-1. Here we show that endothelial MAPKs ERK, p38, and JNK mediate diapedesis-related and diapedesis-unrelated functions of ICAM-1 in cerebral and dermal microvascular endothelial cells (MVECs). All three MAPKs were activated by ICAM-1 engagement, either through lymphocyte adhesion or Ab-mediated clustering. MAPKs were involved in ICAM-1-dependent expression of TNF-α in cerebral and dermal MVECs, and CXCL8, CCL3, CCL4, VCAM-1, and cyclooxygenase 2 (COX-2) in cerebral MVECs. Endothelial JNK and to a much lesser degree p38 were the principal MAPKs involved in facilitating diapedesis of CD4+ lymphocytes across both types of MVECs, whereas ERK was additionally required for TEM across dermal MVECs. JNK activity was critical for ICAM-1-induced F-actin rearrangements. Furthermore, activation of endothelial ICAM-1/JNK led to phosphorylation of paxillin, its association with VE-cadherin, and internalization of the latter. Importantly ICAM-1-induced phosphorylation of paxillin was required for lymphocyte TEM and converged functionally with VE-cadherin phosphorylation. Taken together we conclude that during lymphocyte TEM, ICAM-1 signaling diverges into pathways regulating lymphocyte diapedesis, and other pathways modulating gene expression thereby contributing to the long-term inflammatory response of the endothelium.
Collapse
Affiliation(s)
- Silvia Dragoni
- Department of Cell Biology, Institute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom
| | - Natalie Hudson
- Department of Cell Biology, Institute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom
| | - Bridget-Ann Kenny
- Department of Cell Biology, Institute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom
| | - Thomas Burgoyne
- Department of Cell Biology, Institute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom
| | - Jenny A McKenzie
- Department of Cell Biology, Institute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom
| | - Yadvinder Gill
- Department of Cell Biology, Institute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom
| | - Robert Blaber
- Department of Cell Biology, Institute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom
| | - Clare E Futter
- Department of Cell Biology, Institute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom
| | - Peter Adamson
- Department of Cell Biology, Institute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom
| | - John Greenwood
- Department of Cell Biology, Institute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom
| | - Patric Turowski
- Department of Cell Biology, Institute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom
| |
Collapse
|
204
|
Delgado-Bellido D, Serrano-Saenz S, Fernández-Cortés M, Oliver FJ. Vasculogenic mimicry signaling revisited: focus on non-vascular VE-cadherin. Mol Cancer 2017; 16:65. [PMID: 28320399 PMCID: PMC5359927 DOI: 10.1186/s12943-017-0631-x] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Accepted: 03/06/2017] [Indexed: 12/11/2022] Open
Abstract
Vasculogenic mimicry (VM) is a blood supply system independent of endothelial vessels in tumor cells from different origins. It reflects the plasticity of aggressive tumor cells that express vascular cell markers and line tumor vasculature. The presence of VM is associated with a high tumor grade, short survival, invasion and metastasis. Endothelial cells (ECs) express various members of the cadherin superfamily, in particular vascular endothelial (VE-) cadherin, which is the main adhesion receptor of endothelial adherent junctions. Aberrant extra-vascular expression of VE-cadherin has been observed in certain cancer types associated with VM. In this review we focus on non-endothelial VE-cadherin as a prominent factor involved in the acquisition of tubules-like structures by aggressive tumor cells and we summarize the specific signaling pathways, the association with trans-differentiation and stem-like phenotype and the therapeutic opportunities derived from the in-depth knowledge of the peculiarities of the biology of VE-cadherin and other key components of VM.
Collapse
Affiliation(s)
| | | | | | - F Javier Oliver
- IPBLN, CSIC, CIBERONC, Granada, Spain. .,IPBLN, CSIC, Av. Conocimiento s/n, 18016, Granada, Spain.
| |
Collapse
|
205
|
Ena/VASP proteins regulate activated T-cell trafficking by promoting diapedesis during transendothelial migration. Proc Natl Acad Sci U S A 2017; 114:E2901-E2910. [PMID: 28320969 DOI: 10.1073/pnas.1701886114] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Vasodilator-stimulated phosphoprotein (VASP) and Ena-VASP-like (EVL) are cytoskeletal effector proteins implicated in regulating cell morphology, adhesion, and migration in various cell types. However, the role of these proteins in T-cell motility, adhesion, and in vivo trafficking remains poorly understood. This study identifies a specific role for EVL and VASP in T-cell diapedesis and trafficking. We demonstrate that EVL and VASP are selectively required for activated T-cell trafficking but are not required for normal T-cell development or for naïve T-cell trafficking to lymph nodes and spleen. Using a model of multiple sclerosis, we show an impairment in trafficking of EVL/VASP-deficient activated T cells to the inflamed central nervous system of mice with experimental autoimmune encephalomyelitis. Additionally, we found a defect in trafficking of EVL/VASP double-knockout (dKO) T cells to the inflamed skin and secondary lymphoid organs. Deletion of EVL and VASP resulted in the impairment in α4 integrin (CD49d) expression and function. Unexpectedly, EVL/VASP dKO T cells did not exhibit alterations in shear-resistant adhesion to, or in crawling on, primary endothelial cells under physiologic shear forces. Instead, deletion of EVL and VASP impaired T-cell diapedesis. Furthermore, T-cell diapedesis became equivalent between control and EVL/VASP dKO T cells upon α4 integrin blockade. Overall, EVL and VASP selectively mediate activated T-cell trafficking by promoting the diapedesis step of transendothelial migration in a α4 integrin-dependent manner.
Collapse
|
206
|
Kourtzelis I, Mitroulis I, von Renesse J, Hajishengallis G, Chavakis T. From leukocyte recruitment to resolution of inflammation: the cardinal role of integrins. J Leukoc Biol 2017; 102:677-683. [PMID: 28292945 DOI: 10.1189/jlb.3mr0117-024r] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 02/21/2017] [Accepted: 02/23/2017] [Indexed: 12/23/2022] Open
Abstract
Integrins constitute a large group of adhesion receptors that are formed as heterodimers of α and β subunits. Their presence and activation status on the surface of leukocytes modulate a broad spectrum of processes in inflammation and immunity. This mini review critically outlines research advances with regard to the function of leukocyte integrins in regulating and integrating the onset and resolution of acute inflammation. Specifically, we summarize and discuss relevant, current literature that supports the multifunctional role of integrins and their partners. The latter include molecules that physically associate with integrins or regulate their activity in the context of the following: 1) leukocyte recruitment to an inflamed tissue, 2) recognition and phagocytosis of apoptotic neutrophils (efferocytosis), and 3) egress of efferocytic macrophages from the inflamed site to lymphoid tissues. The understanding of the fine-tuning mechanisms of the aforementioned processes by integrins and their functional partners may enable the design of therapeutic tools to counteract destructive inflammation and promote more efficient resolution of inflammation.
Collapse
Affiliation(s)
- Ioannis Kourtzelis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; and
| | - Ioannis Mitroulis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; and
| | - Janusz von Renesse
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; and
| | - George Hajishengallis
- Department of Microbiology, University of Pennsylvania, School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; and
| |
Collapse
|
207
|
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: 1.9] [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
|
208
|
Géraud C, Koch PS, Zierow J, Klapproth K, Busch K, Olsavszky V, Leibing T, Demory A, Ulbrich F, Diett M, Singh S, Sticht C, Breitkopf-Heinlein K, Richter K, Karppinen SM, Pihlajaniemi T, Arnold B, Rodewald HR, Augustin HG, Schledzewski K, Goerdt S. GATA4-dependent organ-specific endothelial differentiation controls liver development and embryonic hematopoiesis. J Clin Invest 2017; 127:1099-1114. [PMID: 28218627 DOI: 10.1172/jci90086] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/20/2016] [Indexed: 12/23/2022] Open
Abstract
Microvascular endothelial cells (ECs) are increasingly recognized as organ-specific gatekeepers of their microenvironment. Microvascular ECs instruct neighboring cells in their organ-specific vascular niches through angiocrine factors, which include secreted growth factors (angiokines), extracellular matrix molecules, and transmembrane proteins. However, the molecular regulators that drive organ-specific microvascular transcriptional programs and thereby regulate angiodiversity are largely elusive. In contrast to other ECs, which form a continuous cell layer, liver sinusoidal ECs (LSECs) constitute discontinuous, permeable microvessels. Here, we have shown that the transcription factor GATA4 controls murine LSEC specification and function. LSEC-restricted deletion of Gata4 caused transformation of discontinuous liver sinusoids into continuous capillaries. Capillarization was characterized by ectopic basement membrane deposition, formation of a continuous EC layer, and increased expression of VE-cadherin. Correspondingly, ectopic expression of GATA4 in cultured continuous ECs mediated the downregulation of continuous EC-associated transcripts and upregulation of LSEC-associated genes. The switch from discontinuous LSECs to continuous ECs during embryogenesis caused liver hypoplasia, fibrosis, and impaired colonization by hematopoietic progenitor cells, resulting in anemia and embryonic lethality. Thus, GATA4 acts as master regulator of hepatic microvascular specification and acquisition of organ-specific vascular competence, which are indispensable for liver development. The data also establish an essential role of the hepatic microvasculature in embryonic hematopoiesis.
Collapse
|
209
|
Modality of tumor endothelial VEGFR2 silencing-mediated improvement in intratumoral distribution of lipid nanoparticles. J Control Release 2017; 251:1-10. [PMID: 28192155 DOI: 10.1016/j.jconrel.2017.02.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 01/16/2017] [Accepted: 02/08/2017] [Indexed: 01/04/2023]
Abstract
The vascular endothelial growth factor (VEGF)-mediated enhancement in vascular permeability is considered to be a major factor in tumor-targeting delivery via the enhanced permeability and retention (EPR) effect. We previously reported that the silencing of the endothelial VEGF receptor (VEGFR2) by a liposomal siRNA system (RGD-MEND) resulted in an enhanced intratumoral distribution of polyethylene glycol (PEG)-modified liposomes (LPs) in a renal cell carcinoma, a type of hypervascularized cancer, although the inhibition of VEGF signaling would be expected to decrease the permeability of the tumor vasculature. We herein report that the enhancement in the intratumoral distribution of LPs by VEGFR2 inhibition was dependent on the vascular type of the tumor (stroma vessel type; SV and tumor vessel type; TV). In the case of TV-type tumors (renal cell carcinoma and hepatocellular carcinoma), inhibiting VEGFR2 improved intratumoral distribution, while no effect was found in the case of SV-type tumors (colorectal cancer). Moreover, through a comparison of the intratumoral distribution of LPs with a variety of physical properties (100nm vs 400nm, neutral vs negative vs positive), VEGFR2 inhibition was found to alter the tumor microenvironment, including heparan sulfate proteoglycans (HSPGs). In addition, the results regarding the effect of the size of nanoparticles indicated that VEGFR2 inhibition improved the penetration of nanoparticles through the vessel wall, but not via permeability, suggesting the involvement of an unknown mechanism. Our findings suggest that a combination of anti-angiogenic therapy and delivery via the EPR effect would be useful in certain cases, and that altering the tumor microenvironment by VEGFR2 blockade has a drastic effect on the intratumoral distribution of nanoparticles.
Collapse
|
210
|
Ma X, Xiaokaiti Y, Lei H, Liu W, Xu J, Sun Y, Zhao X, Pu X, Zhai S. Epinephrine inhibits vascular hyperpermeability during platelet-activating factor- or ovalbumin-induced anaphylaxis. RSC Adv 2017. [DOI: 10.1039/c7ra09268g] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Platelet-activating factor (PAF) has been shown to play a critical role in mediating vascular hyperpermeability during anaphylaxis.
Collapse
Affiliation(s)
- Xiang Ma
- Department of Pharmacy
- Peking University Third Hospital
- Beijing
- China
| | - Yilixiati Xiaokaiti
- State Key Laboratory of Natural and Biomimetic Drugs
- Peking University
- Beijing
- China
- School of Pharmacy and Pharmaceutical Science
| | - Hui Lei
- State Key Laboratory of Natural and Biomimetic Drugs
- Peking University
- Beijing
- China
- Department of Molecular and Cellular Pharmacology
| | - Wei Liu
- Department of Pharmacy
- Peking University Third Hospital
- Beijing
- China
| | - Jiamin Xu
- State Key Laboratory of Natural and Biomimetic Drugs
- Peking University
- Beijing
- China
- Department of Molecular and Cellular Pharmacology
| | - Yi Sun
- State Key Laboratory of Natural and Biomimetic Drugs
- Peking University
- Beijing
- China
- Department of Molecular and Cellular Pharmacology
| | - Xin Zhao
- State Key Laboratory of Natural and Biomimetic Drugs
- Peking University
- Beijing
- China
- Department of Molecular and Cellular Pharmacology
| | - Xiaoping Pu
- State Key Laboratory of Natural and Biomimetic Drugs
- Peking University
- Beijing
- China
- Department of Molecular and Cellular Pharmacology
| | - Suodi Zhai
- Department of Pharmacy
- Peking University Third Hospital
- Beijing
- China
| |
Collapse
|
211
|
Leukocyte Kinetics and Migration in the Lungs. Respir Med 2017. [DOI: 10.1007/978-3-319-41912-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
212
|
Abstract
Endothelial cells are a constitutive part of the heart and vasculature and form a crucial link between the cardiovascular system and the immune system. Besides their commonly accepted roles in angiogenesis, hemostasis, and the regulation of vascular tone, they are an essential and active component of immune responses. Expression of a range of innate pattern recognition receptors allows them to respond to inflammatory stimulation, and they control immune cell recruitment and extravasation into target tissues throughout the body.In this chapter, I will therefore summarize classical endothelial cell properties and functions and their cross talk with the immune system as well as the operational immunological role of endothelial cells in facilitating immune responses.
Collapse
Affiliation(s)
- Caterina Sturtzel
- Innovative Cancer Models, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung e.V, Vienna, Austria.
| |
Collapse
|
213
|
Dephosphorylation of Y685-VE-Cadherin Involved in Pulmonary Microvascular Endothelial Barrier Injury Induced by Angiotensin II. Mediators Inflamm 2016; 2016:8696481. [PMID: 28119542 PMCID: PMC5227173 DOI: 10.1155/2016/8696481] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 11/04/2016] [Accepted: 11/22/2016] [Indexed: 11/23/2022] Open
Abstract
Angiotensin II (AngII) caused pulmonary microvascular endothelial barrier injury, which induced acute aortic dissection (AAD) combined with acute lung injury (ALI). However, the exact mechanism is unclear. We investigated the role of dephosphorylation of Y685-VE-cadherin in the AngII induced pulmonary microvascular endothelial barrier injury. Mice or pulmonary microvascular endothelial cells (PMVECs) were divided into control group, AngII group, AngII+PP2 (Src kinase inhibitor) group, and PP2 group. PP2 was used to inhibit the phosphorylation of Y685-VE-cadherin. Pathological changes, infiltration of macrophages and neutrophils, and pulmonary microvascular permeability were used to determine the pulmonary microvascular endothelial barrier function. Flow cytometry was used to determine the apoptosis of PMVECs, and immunofluorescence was used to determine the skeletal arrangement. Transendothelial resistance was used to detect the permeability of endothelial barrier. Phosphorylation of Y685-VE-cadherin was significantly reduced after AngII stimulation (P < 0.05), together with skeletal rearrangement, and elevation of endothelial permeability which finally induced endothelial barrier injury. After PP2 interference, the phosphorylation of Y685-VE-cadherin was further reduced and the endothelial permeability was further elevated. These data indicated that AngII could induce pulmonary injury by triggering endothelial barrier injury, and such process may be related to the dephosphorylation of Y685-VE-cadherin and the endothelial skeletal rearrangement.
Collapse
|
214
|
Ebrahim NA, Leach L. Transendothelial migration of human umbilical mesenchymal stem cells across uterine endothelial monolayers: Junctional dynamics and putative mechanisms. Placenta 2016; 48:87-98. [DOI: 10.1016/j.placenta.2016.10.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 10/19/2016] [Accepted: 10/20/2016] [Indexed: 11/28/2022]
|
215
|
Laakkonen JP, Lappalainen JP, Theelen TL, Toivanen PI, Nieminen T, Jauhiainen S, Kaikkonen MU, Sluimer JC, Ylä-Herttuala S. Differential regulation of angiogenic cellular processes and claudin-5 by histamine and VEGF via PI3K-signaling, transcription factor SNAI2 and interleukin-8. Angiogenesis 2016; 20:109-124. [DOI: 10.1007/s10456-016-9532-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 11/07/2016] [Indexed: 01/19/2023]
|
216
|
Garrett JP, Lowery AM, Adam AP, Kowalczyk AP, Vincent PA. Regulation of endothelial barrier function by p120-catenin∙VE-cadherin interaction. Mol Biol Cell 2016; 28:85-97. [PMID: 27852896 PMCID: PMC5221632 DOI: 10.1091/mbc.e16-08-0616] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 10/06/2016] [Accepted: 11/08/2016] [Indexed: 01/06/2023] Open
Abstract
Maintaining VE-cadherin levels by inhibiting its endocytosis through p120-catenin binding is not sufficient for forming a restrictive barrier. Instead, p120-catenin binding to VE-cadherin is required to allow tyrosine-phosphorylated VE-cadherin to contribute to barrier formation. Endothelial p120-catenin (p120) maintains the level of vascular endothelial cadherin (VE-Cad) by inhibiting VE-Cad endocytosis. Loss of p120 results in a decrease in VE-Cad levels, leading to the formation of monolayers with decreased barrier function (as assessed by transendothelial electrical resistance [TEER]), whereas overexpression of p120 increases VE-Cad levels and promotes a more restrictive monolayer. To test whether reduced endocytosis mediated by p120 is required for VE-Cad formation of a restrictive barrier, we restored VE-Cad levels using an endocytic-defective VE-Cad mutant. This endocytic-defective mutant was unable to rescue the loss of TEER associated with p120 or VE-Cad depletion. In contrast, the endocytic-defective mutant was able to prevent sprout formation in a fibrin bead assay, suggesting that p120•VE-Cad interaction regulates barrier function and angiogenic sprouting through different mechanisms. Further investigation found that depletion of p120 increases Src activity and that loss of p120 binding results in increased VE-Cad phosphorylation. In addition, expression of a Y658F–VE-Cad mutant or an endocytic-defective Y658F–VE-Cad double mutant were both able to rescue TEER independently of p120 binding. Our results show that in addition to regulating endocytosis, p120 also allows the phosphorylated form of VE-Cad to participate in the formation of a restrictive monolayer.
Collapse
Affiliation(s)
| | | | - Alejandro P Adam
- Department of Molecular and Cellular Physiology and.,Department of Ophthalmology, Albany Medical College, Albany, NY 12208
| | - Andrew P Kowalczyk
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30307.,Department of Dermatology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30307
| | | |
Collapse
|
217
|
Schnittler H. Contraction of endothelial cells: 40 years of research, but the debate still lives. Histochem Cell Biol 2016; 146:651-656. [PMID: 27680546 DOI: 10.1007/s00418-016-1501-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2016] [Indexed: 12/23/2022]
Abstract
Force generation in non-muscle cells is vital for many cellular and tissue functions. Force-generating mechanisms include actomyosin-mediated contraction, actin polymerization that drives plasma membrane protrusions and filopodia as well as kinesin- and dynein-controlled transport of vesicles and organelles along the microtubule cytoskeleton. The actomyosin-mediated contractility and actin remodeling in both epithelium and endothelium were shown to have significant impact on cell migration, shape change and formation and control of intercellular junctions. In endothelium, contraction is supposed to control permeability for fluid and solutes. However, recent studies demonstrated the constitutive appearance of junction-associated intermittent lamellipodia (JAIL) that drive vascular endothelial cadherin (VE-cadherin) dynamics and control endothelial permeability. Since thrombin blocks JAIL formation and thus increases endothelial permeability, the concept of a simple Rho GTPase-controlled contraction, which is supposed to open endothelial junctions, becomes challenged. Furthermore, specific tyrosine phosphorylation sites of VE-cadherin and catenins have been shown to be involved in control of VE-cadherin-mediated cell adhesion. How the causal-mechanistic interdependency between contractility, VE-cadherin and catenin phosphorylation and JAIL-mediated dynamic remodeling of VE-cadherin is regulated is still an open question and needs to be further addressed.
Collapse
Affiliation(s)
- Hans Schnittler
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Vesaliusweg 2-4, 48149, Münster, Germany.
| |
Collapse
|
218
|
Abstract
The entry of leukocytes into tissues requires well-coordinated interactions between the immune cells and endothelial cells which form the inner lining of blood vessels. The molecular basis for recognition, capture, and adhesion of leukocytes to the endothelial apical surface is well studied. This review will focus on recent advances in our understanding of events following the firm interaction of leukocytes with the inner surface of the blood vessel wall. We will discuss how leukocytes initiate the transmigration (diapedesis) process, trigger the opening of gaps in the endothelial barrier, and eventually move through this boundary.
Collapse
|
219
|
Bolinger MT, Antonetti DA. Moving Past Anti-VEGF: Novel Therapies for Treating Diabetic Retinopathy. Int J Mol Sci 2016; 17:E1498. [PMID: 27618014 PMCID: PMC5037775 DOI: 10.3390/ijms17091498] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/22/2016] [Accepted: 08/30/2016] [Indexed: 12/25/2022] Open
Abstract
Diabetic retinopathy is the leading cause of blindness in working age adults, and is projected to be a significant future health concern due to the rising incidence of diabetes. The recent advent of anti-vascular endothelial growth factor (VEGF) antibodies has revolutionized the treatment of diabetic retinopathy but a significant subset of patients fail to respond to treatment. Accumulating evidence indicates that inflammatory cytokines and chemokines other than VEGF may contribute to the disease process. The current review examines the presence of non-VEGF cytokines in the eyes of patients with diabetic retinopathy and highlights mechanistic pathways in relevant animal models. Finally, novel drug targets including components of the kinin-kallikrein system and emerging treatments such as anti-HPTP (human protein tyrosine phosphatase) β antibodies are discussed. Recognition of non-VEGF contributions to disease pathogenesis may lead to novel therapeutics to enhance existing treatments for patients who do not respond to anti-VEGF therapies.
Collapse
Affiliation(s)
- Mark T Bolinger
- Departments of Ophthalmology and Visual Sciences, Kellogg Eye Center, and Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48105, USA.
| | - David A Antonetti
- Departments of Ophthalmology and Visual Sciences, Kellogg Eye Center, and Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48105, USA.
| |
Collapse
|
220
|
Adam AP, Lowery AM, Martino N, Alsaffar H, Vincent PA. Src Family Kinases Modulate the Loss of Endothelial Barrier Function in Response to TNF-α: Crosstalk with p38 Signaling. PLoS One 2016; 11:e0161975. [PMID: 27603666 PMCID: PMC5014308 DOI: 10.1371/journal.pone.0161975] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/15/2016] [Indexed: 01/23/2023] Open
Abstract
Activation of Src Family Kinase (SFK) signaling is required for the increase in endothelial permeability induced by a variety of cytokines and growth factors. However, we previously demonstrated that activation of endogenous SFKs by expression of dominant negative C-terminal Src Kinase (DN-Csk) is not sufficient to decrease endothelial adherens junction integrity. Basal SFK activity has been observed in normal venular endothelia and was not associated with increased basal permeability. The basal SFK activity however was found to contribute to increased sensitivity of the venular endothelium to inflammatory mediator-induced leakage. How SFK activation achieves this is still not well understood. Here, we show that SFK activation renders human dermal microvascular endothelial cells susceptible to low doses of TNF-α. Treatment of DN-Csk-expressing cells with 50 pg/ml TNF-α induced a loss of TEER as well as drastic changes in the actin cytoskeleton and focal adhesion proteins. This synergistic effect was independent of ROCK or NF-κB activity. TNF-α-induced p38 signaling was required for the synergistic effect on barrier function, and activation of the p38 MAPK alone was also able to induce changes in permeability only in monolayers with active SFKs. These results suggest that the activation of endogenous levels of SFK renders the endothelial barrier more susceptible to low, physiologic doses of TNF-α through activation of p38 which leads to a loss of endothelial tight junctions.
Collapse
Affiliation(s)
- Alejandro P. Adam
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, United States of America
- Department of Ophthalmology, Albany Medical College, Albany, New York, United States of America
- * E-mail: (PAV); (APA)
| | - Anthony M. Lowery
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, United States of America
| | - Nina Martino
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, United States of America
| | - Hiba Alsaffar
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, United States of America
| | - Peter A. Vincent
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, United States of America
- * E-mail: (PAV); (APA)
| |
Collapse
|
221
|
Wylezinski LS, Hawiger J. Interleukin 2 Activates Brain Microvascular Endothelial Cells Resulting in Destabilization of Adherens Junctions. J Biol Chem 2016; 291:22913-22923. [PMID: 27601468 DOI: 10.1074/jbc.m116.729038] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Indexed: 11/06/2022] Open
Abstract
The pleiotropic cytokine interleukin 2 (IL2) disrupts the blood-brain barrier and alters brain microcirculation, underlying vascular leak syndrome that complicates cancer immunotherapy with IL2. The microvascular effects of IL2 also play a role in the development of multiple sclerosis and other chronic neurological disorders. The mechanism of IL2-induced disruption of brain microcirculation has not been determined previously. We found that both human and murine brain microvascular endothelial cells express constituents of the IL2 receptor complex. Then we established that signaling through this receptor complex leads to activation of the transcription factor, nuclear factor κB, resulting in expression of proinflammatory interleukin 6 and monocyte chemoattractant protein 1. We also discovered that IL2 induces disruption of adherens junctions, concomitant with cytoskeletal reorganization, ultimately leading to increased endothelial cell permeability. IL2-induced phosphorylation of vascular endothelial cadherin (VE-cadherin), a constituent of adherens junctions, leads to dissociation of its stabilizing adaptor partners, p120-catenin and β-catenin. Increased phosphorylation of VE-cadherin was also accompanied by a reduction of Src homology 2 domain-containing protein-tyrosine phosphatase 2, known to maintain vascular barrier function. These results unravel the mechanism of deleterious effects induced by IL2 on brain microvascular endothelial cells and may inform the development of new measures to improve IL2 cancer immunotherapy, as well as treatments for autoimmune diseases affecting the central nervous system.
Collapse
Affiliation(s)
| | - Jacek Hawiger
- From the Departments of Molecular Physiology and Biophysics and .,Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Vanderbilt University Medical Center and.,Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee 37232-2363
| |
Collapse
|
222
|
Abstract
Inflammation is part of the complex biological response of body tissues to harmful stimuli, such as pathogens. It serves as a protective response that involves leukocytes, blood vessels and molecular mediators with the purpose to eliminate the initial cause of cell injury and to initiate tissue repair. Inflammation is tightly regulated by the body and is associated with transient crossing of leukocytes through the blood vessel wall, a process called transendothelial migration (TEM) or diapedesis. TEM is a close collaboration between leukocytes on one hand and the endothelium on the other. Limiting vascular leakage during TEM but also when the leukocyte has crossed the endothelium is essential for maintaining vascular homeostasis. Although many details have been uncovered during the recent years, the molecular mechanisms from the vascular part that drive TEM still shows significant gaps in our understanding. This review will focus on the local signals that are induced in the endothelium that regulate leukocyte TEM and simultaneous preservation of endothelial barrier function.
Collapse
Affiliation(s)
- Lilian Schimmel
- a Department of Molecular Cell Biology , Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands
| | - Niels Heemskerk
- a Department of Molecular Cell Biology , Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands
| | - Jaap D van Buul
- a Department of Molecular Cell Biology , Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands
| |
Collapse
|
223
|
Reglero-Real N, Colom B, Bodkin JV, Nourshargh S. Endothelial Cell Junctional Adhesion Molecules: Role and Regulation of Expression in Inflammation. Arterioscler Thromb Vasc Biol 2016; 36:2048-2057. [PMID: 27515379 DOI: 10.1161/atvbaha.116.307610] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 07/27/2016] [Indexed: 12/30/2022]
Abstract
Endothelial cells line the lumen of all blood vessels and play a critical role in maintaining the barrier function of the vasculature. Sealing of the vessel wall between adjacent endothelial cells is facilitated by interactions involving junctionally expressed transmembrane proteins, including tight junctional molecules, such as members of the junctional adhesion molecule family, components of adherence junctions, such as VE-Cadherin, and other molecules, such as platelet endothelial cell adhesion molecule. Of importance, a growing body of evidence indicates that the expression of these molecules is regulated in a spatiotemporal manner during inflammation: responses that have significant implications for the barrier function of blood vessels against blood-borne macromolecules and transmigrating leukocytes. This review summarizes key aspects of our current understanding of the dynamics and mechanisms that regulate the expression of endothelial cells junctional molecules during inflammation and discusses the associated functional implications of such events in acute and chronic scenarios.
Collapse
Affiliation(s)
- Natalia Reglero-Real
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Bartomeu Colom
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.,Wellcome Trust Sanger Institute, Cambridge CB10 1SA, UK
| | - Jennifer Victoria Bodkin
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Sussan Nourshargh
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
224
|
Cell-cell junctional mechanotransduction in endothelial remodeling. Cell Mol Life Sci 2016; 74:279-292. [PMID: 27506620 PMCID: PMC5219012 DOI: 10.1007/s00018-016-2325-8] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 07/15/2016] [Accepted: 08/03/2016] [Indexed: 02/06/2023]
Abstract
The vasculature is one of the most dynamic tissues that encounter numerous mechanical cues derived from pulsatile blood flow, blood pressure, activity of smooth muscle cells in the vessel wall, and transmigration of immune cells. The inner layer of blood and lymphatic vessels is covered by the endothelium, a monolayer of cells which separates blood from tissue, an important function that it fulfills even under the dynamic circumstances of the vascular microenvironment. In addition, remodeling of the endothelial barrier during angiogenesis and trafficking of immune cells is achieved by specific modulation of cell-cell adhesion structures between the endothelial cells. In recent years, there have been many new discoveries in the field of cellular mechanotransduction which controls the formation and destabilization of the vascular barrier. Force-induced adaptation at endothelial cell-cell adhesion structures is a crucial node in these processes that challenge the vascular barrier. One of the key examples of a force-induced molecular event is the recruitment of vinculin to the VE-cadherin complex upon pulling forces at cell-cell junctions. Here, we highlight recent advances in the current understanding of mechanotransduction responses at, and derived from, endothelial cell-cell junctions. We further discuss their importance for vascular barrier function and remodeling in development, inflammation, and vascular disease.
Collapse
|
225
|
Häuselmann I, Roblek M, Protsyuk D, Huck V, Knopfova L, Grässle S, Bauer AT, Schneider SW, Borsig L. Monocyte Induction of E-Selectin-Mediated Endothelial Activation Releases VE-Cadherin Junctions to Promote Tumor Cell Extravasation in the Metastasis Cascade. Cancer Res 2016; 76:5302-12. [PMID: 27488527 DOI: 10.1158/0008-5472.can-16-0784] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 06/27/2016] [Indexed: 12/24/2022]
Abstract
Tumor cells interact with blood constituents and these interactions promote metastasis. Selectins are vascular receptors facilitating interactions of tumor cells with platelets, leukocytes, and endothelium, but the role of endothelial E-selectin remains unclear. Here we show that E-selectin is a major receptor for monocyte recruitment to tumor cell-activated endothelium. Experimental and spontaneous lung metastasis using murine tumor cells, without E-selectin ligands, were attenuated in E-selectin-deficient mice. Tumor cell-derived CCL2 promoted endothelial activation, resulting in enhanced endothelial E-selectin expression. The recruitment of inflammatory monocytes to metastasizing tumor cells was dependent on the local endothelial activation and the presence of E-selectin. Monocytes promoted transendothelial migration of tumor cells through the induction of E-selectin-dependent endothelial retractions and a subsequent modulation of tight junctions through dephosphorylation of VE-cadherin. Thus, endothelial E-selectin shapes the tumor microenvironment through the recruitment, adhesion, and activation of monocytes that facilitate tumor cell extravasation and thereby metastasis. These findings provide evidence that endothelial E-selectin is a novel factor contributing to endothelial retraction required for efficient lung metastasis. Cancer Res; 76(18); 5302-12. ©2016 AACR.
Collapse
Affiliation(s)
- Irina Häuselmann
- Institute of Physiology, University of Zürich and Zürich Center for Integrative Human Physiology, Zurich, Switzerland
| | - Marko Roblek
- Institute of Physiology, University of Zürich and Zürich Center for Integrative Human Physiology, Zurich, Switzerland
| | - Darya Protsyuk
- Institute of Physiology, University of Zürich and Zürich Center for Integrative Human Physiology, Zurich, Switzerland
| | - Volker Huck
- Department of Dermatology, Experimental Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lucia Knopfova
- International Clinical Research Center, Center for Biological and Cellular Engineering, St. Anne's University Hospital and Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Sandra Grässle
- Department of Dermatology, Experimental Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Alexander T Bauer
- Department of Dermatology, Experimental Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefan W Schneider
- Department of Dermatology, Experimental Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lubor Borsig
- Institute of Physiology, University of Zürich and Zürich Center for Integrative Human Physiology, Zurich, Switzerland.
| |
Collapse
|
226
|
Rathnakumar K, Savant S, Giri H, Ghosh A, Fisslthaler B, Fleming I, Ram U, Bera AK, Augustin HG, Dixit M. Angiopoietin-2 mediates thrombin-induced monocyte adhesion and endothelial permeability. J Thromb Haemost 2016; 14:1655-67. [PMID: 27241812 DOI: 10.1111/jth.13376] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 05/04/2016] [Indexed: 02/03/2023]
Abstract
UNLABELLED Essentials Mechanism of thrombin-induced inflammation is not fully understood. Thrombin induced monocyte adhesion and barrier loss require Angiopoietin-2 (Ang-2). Ang-2 mediates vessel leakage and monocyte adhesion through SHP-2/p38MAPK pathway. Calcium dependent SHP2/p38MAPK activation regulates Ang-2 expression through a feedback loop. SUMMARY Background Thrombin imparts an inflammatory phenotype to the endothelium by promoting increased monocyte adhesion and vascular permeability. However, the molecular players that govern these events are incompletely understood. Objective The aim of this study was to determine whether Angiopoietin-2 (Ang-2) has a role, if any, in regulating inflammatory signals initiated by thrombin. Methods Assessment of vascular leakage by Miles assay was performed by intra-dermal injection on the foot paw. Surface levels of intercellular adhesion molecule-1 (ICAM-1) were determined by flow cytometry. Overexpression, knockdown and phosphorylation of proteins were determined by Western blotting. Results In time-course experiments, thrombin-stimulated Ang-2 up-regulation, peaked prior to the expression of adhesion molecule ICAM-1 in human umbilical vein-derived endothelial cells (HUVECs). Knockdown of Ang-2 blocked both thrombin-induced monocyte adhesion and ICAM-1 expression. In addition, Ang-2(-/-) mice displayed defective vascular leakage when treated with thrombin. Introducing Ang-2 protein in Ang-2(-/-) mice failed to recover a wild-type phenotype. Mechanistically, Ang-2 appears to regulate the thrombin-activated calcium spike that is required for tyrosine phosphatase SHP2 and p38 MAPK activation. Further, down-regulation of SHP2 attenuated both thrombin-induced Ang-2 expression and monocyte adhesion. Down-regulation of the adaptor protein Gab1, a co-activator of SHP2, as well as overexpression of the Gab1 mutant incapable of interacting with SHP2 (YFGab1), inhibited thrombin-mediated effects, including downstream activation of p38 MAPK, which in turn was required for Ang-2 expression. Conclusions The data establish an essential role of the Gab1/SHP2/p38MAPK signaling pathway and Ang-2 in regulating thrombin-induced monocyte adhesion and vascular leakage.
Collapse
Affiliation(s)
- K Rathnakumar
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences and Bioengineering, Indian Institute of Technology Madras, Chennai, India
| | - S Savant
- Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - H Giri
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences and Bioengineering, Indian Institute of Technology Madras, Chennai, India
| | - A Ghosh
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences and Bioengineering, Indian Institute of Technology Madras, Chennai, India
| | - B Fisslthaler
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt, Germany
| | - I Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt, Germany
| | - U Ram
- Seethapathy Clinic and Hospital, Chennai, India
| | - A K Bera
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences and Bioengineering, Indian Institute of Technology Madras, Chennai, India
| | - H G Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany
- Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim (CBTM), Heidelberg University, Heidelberg, Germany
| | - M Dixit
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences and Bioengineering, Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
227
|
Bradfield PF, Menon A, Miljkovic-Licina M, Lee BP, Fischer N, Fish RJ, Kwak B, Fisher EA, Imhof BA. Divergent JAM-C Expression Accelerates Monocyte-Derived Cell Exit from Atherosclerotic Plaques. PLoS One 2016; 11:e0159679. [PMID: 27442505 PMCID: PMC4956249 DOI: 10.1371/journal.pone.0159679] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 07/06/2016] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis, caused in part by monocytes in plaques, continues to be a disease that afflicts the modern world. Whilst significant steps have been made in treating this chronic inflammatory disease, questions remain on how to prevent monocyte and macrophage accumulation in atherosclerotic plaques. Junctional Adhesion Molecule C (JAM-C) expressed by vascular endothelium directs monocyte transendothelial migration in a unidirectional manner leading to increased inflammation. Here we show that interfering with JAM-C allows reverse-transendothelial migration of monocyte-derived cells, opening the way back out of the inflamed environment. To study the role of JAM-C in plaque regression we used a mouse model of atherosclerosis, and tested the impact of vascular JAM-C expression levels on monocyte reverse transendothelial migration using human cells. Studies in-vitro under inflammatory conditions revealed that overexpression or gene silencing of JAM-C in human endothelium exposed to flow resulted in higher rates of monocyte reverse-transendothelial migration, similar to antibody blockade. We then transplanted atherosclerotic, plaque-containing aortic arches from hyperlipidemic ApoE-/- mice into wild-type normolipidemic recipient mice. JAM-C blockade in the recipients induced greater emigration of monocyte-derived cells and further diminished the size of atherosclerotic plaques. Our findings have shown that JAM-C forms a one-way vascular barrier for leukocyte transendothelial migration only when present at homeostatic copy numbers. We have also shown that blocking JAM-C can reduce the number of atherogenic monocytes/macrophages in plaques by emigration, providing a novel therapeutic strategy for chronic inflammatory pathologies.
Collapse
Affiliation(s)
- Paul F. Bradfield
- Department of Pathology and Immunology, CMU, University of Geneva, 1211, rue Michel Servet 1, Geneva 4, Switzerland
- * E-mail:
| | - Arjun Menon
- Division of Cardiology, New York University Langone Medical Center, New York, New York 10016, United States of America
| | - Marijana Miljkovic-Licina
- Department of Pathology and Immunology, CMU, University of Geneva, 1211, rue Michel Servet 1, Geneva 4, Switzerland
| | - Boris P. Lee
- Department of Pathology and Immunology, CMU, University of Geneva, 1211, rue Michel Servet 1, Geneva 4, Switzerland
| | - Nicolas Fischer
- NovImmune S.A., 14 chemin des Aulx, 1228 Plan-les-Ouates, Geneva, Switzerland
| | - Richard J. Fish
- Department of Genetic Medicine and Development, CMU, University of Geneva, 1211, rue Michel Servet 1, Geneva, Switzerland
| | - Brenda Kwak
- Department of Pathology and Immunology, CMU, University of Geneva, 1211, rue Michel Servet 1, Geneva 4, Switzerland
| | - Edward A. Fisher
- Division of Cardiology, New York University Langone Medical Center, New York, New York 10016, United States of America
| | - Beat A. Imhof
- Department of Pathology and Immunology, CMU, University of Geneva, 1211, rue Michel Servet 1, Geneva 4, Switzerland
| |
Collapse
|
228
|
Gordon EJ, Fukuhara D, Weström S, Padhan N, Sjöström EO, van Meeteren L, He L, Orsenigo F, Dejana E, Bentley K, Spurkland A, Claesson-Welsh L. The endothelial adaptor molecule TSAd is required for VEGF-induced angiogenic sprouting through junctional c-Src activation. Sci Signal 2016; 9:ra72. [PMID: 27436360 DOI: 10.1126/scisignal.aad9256] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Activation of vascular endothelial growth factor (VEGF) receptor 2 (VEGFR2) by VEGF binding is critical for vascular morphogenesis. In addition, VEGF disrupts the endothelial barrier by triggering the phosphorylation and turnover of the junctional molecule VE-cadherin, a process mediated by the VEGFR2 downstream effectors T cell-specific adaptor (TSAd) and the tyrosine kinase c-Src. We investigated whether the VEGFR2-TSAd-c-Src pathway was required for angiogenic sprouting. Indeed, Tsad-deficient embryoid bodies failed to sprout in response to VEGF. Tsad-deficient mice displayed impaired angiogenesis specifically during tracheal vessel development, but not during retinal vasculogenesis, and in VEGF-loaded Matrigel plugs, but not in those loaded with FGF. The SH2 and proline-rich domains of TSAd bridged VEGFR2 and c-Src, and this bridging was critical for the localization of activated c-Src to endothelial junctions and elongation of the growing sprout, but not for selection of the tip cell. These results revealed that vascular sprouting and permeability are both controlled through the VEGFR2-TSAd-c-Src signaling pathway in a subset of tissues, which may be useful in developing strategies to control tissue-specific pathological angiogenesis.
Collapse
Affiliation(s)
- Emma J Gordon
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv 20, Uppsala 75185, Sweden.
| | - Daisuke Fukuhara
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv 20, Uppsala 75185, Sweden
| | - Simone Weström
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv 20, Uppsala 75185, Sweden
| | - Narendra Padhan
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv 20, Uppsala 75185, Sweden
| | - Elisabet O Sjöström
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv 20, Uppsala 75185, Sweden
| | - Laurens van Meeteren
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv 20, Uppsala 75185, Sweden
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv 20, Uppsala 75185, Sweden
| | - Fabrizio Orsenigo
- FIRC Institute of Molecular Oncology Foundation, IFOM, Milan 20139, Italy
| | - Elisabetta Dejana
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv 20, Uppsala 75185, Sweden. FIRC Institute of Molecular Oncology Foundation, IFOM, Milan 20139, Italy
| | - Katie Bentley
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv 20, Uppsala 75185, Sweden. Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Anne Spurkland
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo 0317, Norway
| | - Lena Claesson-Welsh
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv 20, Uppsala 75185, Sweden.
| |
Collapse
|
229
|
Dorland YL, Malinova TS, van Stalborch AMD, Grieve AG, van Geemen D, Jansen NS, de Kreuk BJ, Nawaz K, Kole J, Geerts D, Musters RJP, de Rooij J, Hordijk PL, Huveneers S. The F-BAR protein pacsin2 inhibits asymmetric VE-cadherin internalization from tensile adherens junctions. Nat Commun 2016; 7:12210. [PMID: 27417273 PMCID: PMC4947187 DOI: 10.1038/ncomms12210] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 06/10/2016] [Indexed: 12/14/2022] Open
Abstract
Vascular homoeostasis, development and disease critically depend on the regulation of endothelial cell-cell junctions. Here we uncover a new role for the F-BAR protein pacsin2 in the control of VE-cadherin-based endothelial adhesion. Pacsin2 concentrates at focal adherens junctions (FAJs) that are experiencing unbalanced actomyosin-based pulling. FAJs move in response to differences in local cytoskeletal geometry and pacsin2 is recruited consistently to the trailing end of fast-moving FAJs via a mechanism that requires an intact F-BAR domain. Photoconversion, photobleaching, immunofluorescence and super-resolution microscopy reveal polarized dynamics, and organization of junctional proteins between the front of FAJs and their trailing ends. Interestingly, pacsin2 recruitment inhibits internalization of the VE-cadherin complex from FAJ trailing ends and is important for endothelial monolayer integrity. Together, these findings reveal a novel junction protective mechanism during polarized trafficking of VE-cadherin, which supports barrier maintenance within dynamic endothelial tissue.
Collapse
Affiliation(s)
- Yvonne L Dorland
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam 1066 CX, The Netherlands
| | - Tsveta S Malinova
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam 1105 AZ, The Netherlands
| | - Anne-Marieke D van Stalborch
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam 1066 CX, The Netherlands
| | - Adam G Grieve
- Hubrecht Institute and University Medical Center Utrecht, Utrecht 3584 CT, The Netherlands
| | - Daphne van Geemen
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam 1066 CX, The Netherlands
| | - Nicolette S Jansen
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam 1066 CX, The Netherlands
| | - Bart-Jan de Kreuk
- Department of Medicine, University of California, San Diego, California 92093, USA
| | - Kalim Nawaz
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam 1066 CX, The Netherlands
| | - Jeroen Kole
- Department of Physiology, VU University Medical Center, Amsterdam 1081 HV, The Netherlands
| | - Dirk Geerts
- Department of Pediatric Oncology/Hematology, Erasmus University Medical Center, Rotterdam 3015 GE, The Netherlands
| | - René J P Musters
- Department of Physiology, VU University Medical Center, Amsterdam 1081 HV, The Netherlands
| | - Johan de Rooij
- Department of Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht 3584 CG, The Netherlands
| | - Peter L Hordijk
- Department of Physiology, VU University Medical Center, Amsterdam 1081 HV, The Netherlands
| | - Stephan Huveneers
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam 1066 CX, The Netherlands.,Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam 1105 AZ, The Netherlands
| |
Collapse
|
230
|
Dipeptidyl Peptidase-4 Inhibitor Increases Vascular Leakage in Retina through VE-cadherin Phosphorylation. Sci Rep 2016; 6:29393. [PMID: 27381080 PMCID: PMC4933943 DOI: 10.1038/srep29393] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 06/16/2016] [Indexed: 12/13/2022] Open
Abstract
The inhibitors of CD26 (dipeptidyl peptidase-4; DPP4) have been widely prescribed to control glucose level in diabetic patients. DPP4-inhibitors, however, accumulate stromal cell-derived factor-1α (SDF-1α), a well-known inducer of vascular leakage and angiogenesis both of which are fundamental pathophysiology of diabetic retinopathy. The aim of this study was to investigate the effects of DPP4-inhibitors on vascular permeability and diabetic retinopathy. DPP4-inhibitor (diprotin A or sitagliptin) increased the phosphorylation of Src and vascular endothelial-cadherin (VE-cadherin) in human endothelial cells and disrupted endothelial cell-to-cell junctions, which were attenuated by CXCR4 (receptor of SDF-1α)-blocker or Src-inhibitor. Disruption of endothelial cell-to-cell junctions in the immuno-fluorescence images correlated with the actual leakage of the endothelial monolayer in the transwell endothelial permeability assay. In the Miles assay, vascular leakage was observed in the ears into which SDF-1α was injected, and this effect was aggravated by DPP4-inhibitor. In the model of retinopathy of prematurity, DPP4-inhibitor increased not only retinal vascularity but also leakage. Additionally, in the murine diabetic retinopathy model, DPP4-inhibitor increased the phosphorylation of Src and VE-cadherin and aggravated vascular leakage in the retinas. Collectively, DPP4-inhibitor induced vascular leakage by augmenting the SDF-1α/CXCR4/Src/VE-cadherin signaling pathway. These data highlight safety issues associated with the use of DPP4-inhibitors.
Collapse
|
231
|
Fournier P, Dussault S, Fusco A, Rivard A, Royal I. Tyrosine Phosphatase PTPRJ/DEP-1 Is an Essential Promoter of Vascular Permeability, Angiogenesis, and Tumor Progression. Cancer Res 2016; 76:5080-91. [PMID: 27364551 DOI: 10.1158/0008-5472.can-16-1071] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 06/24/2016] [Indexed: 11/16/2022]
Abstract
The protein tyrosine phosphatase PTPRJ/DEP-1 has been implicated in negative growth regulation in endothelial cells, where its expression varies at transitions between proliferation and contact inhibition. However, in the same cells, DEP-1 has also been implicated in VEGF-dependent Src activation, permeability, and capillary formation, suggesting a positive role in regulating these functions. To resolve this dichotomy in vivo, we investigated postnatal angiogenesis and vascular permeability in a DEP-1-deficient mouse. In this study, we report that DEP-1 is required for Src activation and phosphorylation of its endothelial cell-specific substrate, VE-cadherin, after systemic injection of VEGF. Accordingly, VEGF-induced vascular leakage was abrogated in the DEP-1-deficient mice. Furthermore, capillary formation was impaired in murine aortic tissue rings or Matrigel plugs infused with VEGF. In the absence of DEP-1, angiogenesis triggered by ischemia or during tumor formation was defective, which in the latter case was associated with reduced tumor cell proliferation and increased apoptosis. Macrophage infiltration was also impaired, reflecting reduced vascular permeability in the tumors or a possible cell autonomous effect of DEP-1. Consequently, the formation of spontaneous and experimental lung metastases was strongly decreased in DEP-1-deficient mice. In clinical specimens of cancer, less vascularized tumors exhibited lower microvascular expression of DEP-1. Altogether, our results established DEP-1 as an essential driver of VEGF-dependent permeability, angiogenesis, and metastasis, suggesting a novel therapeutic route to cancer treatment. Cancer Res; 76(17); 5080-91. ©2016 AACR.
Collapse
Affiliation(s)
- Patrick Fournier
- CRCHUM - Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada. Institut du cancer de Montréal, Montréal, Quebec, Canada
| | - Sylvie Dussault
- CRCHUM - Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada
| | - Alfredo Fusco
- Dipartimento di Biologia e Patologia Cellulare e Molecolare, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Alain Rivard
- CRCHUM - Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada. Département de Médecine, Université de Montréal, Montréal, Quebec, Canada
| | - Isabelle Royal
- CRCHUM - Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada. Institut du cancer de Montréal, Montréal, Quebec, Canada. Département de Médecine, Université de Montréal, Montréal, Quebec, Canada.
| |
Collapse
|
232
|
Hérodin F, Voir D, Vilgrain I, Courçon M, Drouet M, Boittin FX. Soluble Vascular Endothelial Cadherin as a New Biomarker of Irradiation in Highly Irradiated Baboons with Bone Marrow Protection. HEALTH PHYSICS 2016; 110:598-605. [PMID: 27115227 DOI: 10.1097/hp.0000000000000481] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Vascular endothelial cadherin is the main component of adherens junctions enabling cohesion of the endothelial monolayer in vessels. The extracellular part of vascular endothelial cadherin (VE-cadherin) can be cleaved, releasing soluble fragments in blood (sVE-cadherin). In some diseases with endothelial dysfunction, a correlation between increased blood sVE-cadherin levels and disease state has been proposed. Irradiation is known to induce endothelial damage, but new serum biomarkers are needed to evaluate endothelial damage after irradiation. Here, the authors investigated whether sVE-cadherin may be an interesting biomarker of irradiation in highly irradiated baboons with bone marrow protection. sVE-cadherin was detected in the plasma of young as well as old baboons. Plasma sVE-cadherin levels significantly decrease a few days after irradiation but recover in the late time after irradiation. Kinetic analysis of plasma sVE-cadherin levels suggests a correlation with white blood cell counts in both the acute phase of irradiation and during hematopoietic recovery, suggesting that plasma sVE-cadherin levels may be partly linked to the disappearance and recovery of white blood cells. Interestingly, after hematopoietic recovery was completed, sVE-cadherin levels were found to exceed control values, suggesting that plasma sVE-cadherin may represent a new biomarker of endothelial damage or neovascularization in the late time after irradiation.
Collapse
Affiliation(s)
- Francis Hérodin
- *Institut de Recherche Biomédicale des Armées (IRBA), Brétigny-sur-Orge, France; †Institut National de la Santé et de la Recherche Médicale, U1036, Grenoble, France; Commissariat à l'Energie Atomique et aux Energies Alternatives, Institute of Life Science Research and Technologies, Biology of Cancer and Infection, Grenoble, France; University of Grenoble Alpes, Unité mixte de recherche-S1036, 17, rue des Martyrs, Grenoble, France
| | | | | | | | | | | |
Collapse
|
233
|
Hordijk PL. Recent insights into endothelial control of leukocyte extravasation. Cell Mol Life Sci 2016; 73:1591-608. [PMID: 26794844 PMCID: PMC11108429 DOI: 10.1007/s00018-016-2136-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 01/07/2016] [Accepted: 01/11/2016] [Indexed: 12/30/2022]
Abstract
In the process of leukocyte migration from the circulation across the vascular wall, the crosstalk with endothelial cells that line the blood vessels is essential. It is now firmly established that in endothelial cells important signaling events are initiated upon leukocyte adhesion that impinge on the regulation of cell-cell contact and control the efficiency of transendothelial migration. In addition, several external factors such as shear force and vascular stiffness were recently identified as important regulators of endothelial signaling and, consequently, leukocyte transmigration. Here, I review recent insights into endothelial signaling events that are linked to leukocyte migration across the vessel wall. In this field, protein phosphorylation and Rho-mediated cytoskeletal dynamics are still widely studied using increasingly sophisticated mouse models. In addition, activation of tyrosine phosphatases, changes in endothelial cell stiffness as well as different vascular beds have all been established as important factors in endothelial signaling and leukocyte transmigration. Finally, I address less-well-studied but interesting components in the endothelium that also control transendothelial migration, such as the ephrins and their Eph receptors, that provide novel insights in the complexity associated with this process.
Collapse
Affiliation(s)
- Peter L Hordijk
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Swammerdam Institute for Life Sciences, University of Amsterdam, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands.
- Department of Physiology, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
234
|
Lee RH, Bergmeier W. Platelet immunoreceptor tyrosine-based activation motif (ITAM) and hemITAM signaling and vascular integrity in inflammation and development. J Thromb Haemost 2016; 14:645-54. [PMID: 26749528 DOI: 10.1111/jth.13250] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/24/2015] [Indexed: 01/13/2023]
Abstract
Platelets are essential for maintaining hemostasis following mechanical injury to the vasculature. Besides this established function, novel roles of platelets are becoming increasingly recognized, which are critical in non-injury settings to maintain vascular barrier integrity. For example, during embryogenesis platelets act to support the proper separation of blood and lymphatic vessels. This role continues beyond birth, where platelets prevent leakage of blood into the lymphatic vessel network. During the course of inflammation, platelets are necessary to prevent local hemorrhage due to neutrophil diapedesis and disruption of endothelial cell-cell junctions. Surprisingly, platelets also work to secure tumor-associated blood vessels, inhibiting excessive vessel permeability and intra-tumor hemorrhaging. Interestingly, many of these novel platelet functions depend on immunoreceptor tyrosine-based activation motif (ITAM) signaling but not on signaling via G protein-coupled receptors, which plays a crucial role in platelet plug formation at sites of mechanical injury. Murine platelets express two ITAM-containing receptors: the Fc receptor γ-chain (FcRγ), which functionally associates with the collagen receptor GPVI, and the C-type lectin-like 2 (CLEC-2) receptor, a hemITAM receptor for the mucin-type glycoprotein podoplanin. Human platelets express an additional ITAM receptor, FcγRIIA. These receptors share common downstream effectors, including Syk, SLP-76 and PLCγ2. Here we will review the recent literature that highlights a critical role for platelet GPVI/FcRγ and CLEC-2 in vascular integrity during development and inflammation in mice and discuss the relevance to human disease.
Collapse
Affiliation(s)
- R H Lee
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - W Bergmeier
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
235
|
Schmidt EP, Kuebler WM, Lee WL, Downey GP. Adhesion Molecules: Master Controllers of the Circulatory System. Compr Physiol 2016; 6:945-73. [PMID: 27065171 DOI: 10.1002/cphy.c150020] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This manuscript will review our current understanding of cellular adhesion molecules (CAMs) relevant to the circulatory system, their physiological role in control of vascular homeostasis, innate and adaptive immune responses, and their importance in pathophysiological (disease) processes such as acute lung injury, atherosclerosis, and pulmonary hypertension. This is a complex and rapidly changing area of research that is incompletely understood. By design, we will begin with a brief overview of the structure and classification of the major groups of adhesion molecules and their physiological functions including cellular adhesion and signaling. The role of specific CAMs in the process of platelet aggregation and hemostasis and leukocyte adhesion and transendothelial migration will be reviewed as examples of the complex and cooperative interplay between CAMs during physiological and pathophysiological processes. The role of the endothelial glycocalyx and the glycobiology of this complex system related to inflammatory states such as sepsis will be reviewed. We will then focus on the role of adhesion molecules in the pathogenesis of specific disease processes involving the lungs and cardiovascular system. The potential of targeting adhesion molecules in the treatment of immune and inflammatory diseases will be highlighted in the relevant sections throughout the manuscript.
Collapse
Affiliation(s)
- Eric P Schmidt
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Wolfgang M Kuebler
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
- Departments of Surgery and Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Warren L Lee
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
- Division of Respirology and the Interdepartmental Division of Critical Care Medicine, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Gregory P Downey
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Departments of Medicine, Pediatrics, and Biomedical Research, National Jewish Health, Denver, Colorado, USA
- Departments of Medicine, and Immunology and Microbiology, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
236
|
Millar FR, Summers C, Griffiths MJ, Toshner MR, Proudfoot AG. The pulmonary endothelium in acute respiratory distress syndrome: insights and therapeutic opportunities. Thorax 2016; 71:462-73. [DOI: 10.1136/thoraxjnl-2015-207461] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 02/12/2016] [Indexed: 01/23/2023]
|
237
|
Gonzalez AM, Cyrus BF, Muller WA. Targeted Recycling of the Lateral Border Recycling Compartment Precedes Adherens Junction Dissociation during Transendothelial Migration. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1387-402. [PMID: 26968345 DOI: 10.1016/j.ajpath.2016.01.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 12/24/2015] [Accepted: 01/12/2016] [Indexed: 12/11/2022]
Abstract
Leukocyte transendothelial migration (TEM) requires two major events: local dissociation of adherens junctions manifested as gaps in vascular endothelial (VE)-cadherin staining at the site of TEM and targeted trafficking of the lateral border recycling compartment (LBRC) to the site of TEM. However, the association between LBRC recycling and VE-cadherin gaps remains unknown. We found that when targeting of the LBRC is selectively inhibited using established methods, such as a function blocking anti-platelet endothelial cell adhesion molecule 1 antibody, depolymerizing microtubules, or microinjection of an antibody that inhibits kinesin, VE-cadherin gaps do not form around the blocked leukocyte. This is the first time that the LBRC has been implicated in this process. We obtained similar results for neutrophils and monocytes and in studies using live cell imaging microscopy conducted under fluid shear conditions. Depolymerizing microtubules did not affect the ability of leukocytes to induce tyrosine phosphorylation of VE-cadherin. A VE-cadherin double mutant (Y658F, Y731F) expressed in endothelial cells acted as a dominant negative and inhibited VE-cadherin gap formation and TEM, yet targeting of the LBRC still occurred. These data suggest that targeting of the LBRC to the site of TEM precedes VE-cadherin clearance. Recruitment of the LBRC may play a role in clearing VE-cadherin from the site of TEM.
Collapse
Affiliation(s)
- Annette M Gonzalez
- Department of Pathology, The Feinberg School of Medicine at Northwestern University, Chicago, Illinois
| | - Bita F Cyrus
- Department of Pathology, The Feinberg School of Medicine at Northwestern University, Chicago, Illinois
| | - William A Muller
- Department of Pathology, The Feinberg School of Medicine at Northwestern University, Chicago, Illinois.
| |
Collapse
|
238
|
New insights in the control of vascular permeability: vascular endothelial-cadherin and other players. Curr Opin Hematol 2016; 22:267-72. [PMID: 25767951 DOI: 10.1097/moh.0000000000000137] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW The control of the endothelial barrier function is essential for vascular homeostasis and is mainly mediated by cell-to-cell junctions that tightly regulate permeability to plasma solutes and circulating cells such as leukocytes and tumor cells. While in some circumstances the transient dismantling of endothelial cell junctions might be beneficial, in pathological conditions, such as cancer, severe alterations of endothelial junction composition and function are detrimental, causing massive edema and increased interstitial pressure. Here, we aim to discuss the newly and most recently identified molecular mechanisms that cooperate in the control of vascular permeability. RECENT FINDINGS Although the involvement of vascular endothelial-cadherin in the regulation of vascular leakage is well known, recent findings shed light on additional molecules involved in the control of vascular endothelial-cadherin phosphorylation in physiological and pathological conditions, and identified new unknown regulators of the endothelial barrier function. SUMMARY In the past years, several studies explored the contribution of various signaling pathways in the regulation of vascular leakage. Despite encouraging results, a more comprehensive understanding of the molecular mechanisms involved in this process will define druggable targets for new therapeutic interventions to limit endothelial barrier dysfunctions.
Collapse
|
239
|
F-actin-rich contractile endothelial pores prevent vascular leakage during leukocyte diapedesis through local RhoA signalling. Nat Commun 2016; 7:10493. [PMID: 26814335 PMCID: PMC4737874 DOI: 10.1038/ncomms10493] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 12/14/2015] [Indexed: 12/17/2022] Open
Abstract
During immune surveillance and inflammation, leukocytes exit the vasculature through transient openings in the endothelium without causing plasma leakage. However, the exact mechanisms behind this intriguing phenomenon are still unknown. Here we report that maintenance of endothelial barrier integrity during leukocyte diapedesis requires local endothelial RhoA cycling. Endothelial RhoA depletion in vitro or Rho inhibition in vivo provokes neutrophil-induced vascular leakage that manifests during the physical movement of neutrophils through the endothelial layer. Local RhoA activation initiates the formation of contractile F-actin structures that surround emigrating neutrophils. These structures that surround neutrophil-induced endothelial pores prevent plasma leakage through actomyosin-based pore confinement. Mechanistically, we found that the initiation of RhoA activity involves ICAM-1 and the Rho GEFs Ect2 and LARG. In addition, regulation of actomyosin-based endothelial pore confinement involves ROCK2b, but not ROCK1. Thus, endothelial cells assemble RhoA-controlled contractile F-actin structures around endothelial pores that prevent vascular leakage during leukocyte extravasation. Endothelial cells can support leukocyte extravasation without causing vascular leakage, but the exact mechanism underlying this process has not been fully elucidated. Here the authors show that it is regulated through actomyosin-based endothelial pore confinement, which requires local endothelial RhoA activation.
Collapse
|
240
|
Timmerman I, Daniel AE, Kroon J, van Buul JD. Leukocytes Crossing the Endothelium: A Matter of Communication. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 322:281-329. [PMID: 26940521 DOI: 10.1016/bs.ircmb.2015.10.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Leukocytes cross the endothelial vessel wall in a process called transendothelial migration (TEM). The purpose of leukocyte TEM is to clear the causing agents of inflammation in underlying tissues, for example, bacteria and viruses. During TEM, endothelial cells initiate signals that attract and guide leukocytes to sites of tissue damage. Leukocytes react by attaching to these sites and signal their readiness to move back to endothelial cells. Endothelial cells in turn respond by facilitating the passage of leukocytes while retaining overall integrity. In this review, we present recent findings in the field and we have endeavored to synthesize a coherent picture of the intricate interplay between endothelial cells and leukocytes during TEM.
Collapse
Affiliation(s)
- Ilse Timmerman
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Anna E Daniel
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Jeffrey Kroon
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Jaap D van Buul
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands.
| |
Collapse
|
241
|
Abstract
Acute respiratory distress syndrome (ARDS) was first described in 1967, and since then there have been a large number of studies addressing its pathogenesis and therapies. Despite intense research efforts, very few therapies for ARDS have been shown to be effective other than the use of lung protection strategies. The scarcity of therapeutic choices is related to the intricate pathogenesis of the syndrome and to insensitive and aspecific criteria to diagnose this profound acute respiratory failure. The aim of this paper is to summarize advances of new ARDS definitions and provide an overview of new relevant signaling pathways that mediate acute lung injury.
Collapse
|
242
|
Chen J, Wang J, Su C, Qian W, Sun L, Sun H, Chen J, Zhang H, Zhang J. Urinary trypsin inhibitor attenuates LPS-induced endothelial barrier dysfunction by upregulation of vascular endothelial-cadherin expression. Inflamm Res 2015; 65:213-24. [PMID: 26681130 DOI: 10.1007/s00011-015-0907-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 11/07/2015] [Accepted: 11/16/2015] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Urinary trypsin inhibitor (UTI) decreases inflammatory cytokine levels and mortality in experimental animal models of inflammation. Here, we observed the effect of UTI on lipopolysaccharide (LPS)-induced hyperpermeability in human umbilical vein endothelial cells (HUVECs) and explored the role of vascular endothelial-cadherin (VE-cadherin) in its effect. METHODS The effect of UTI on endothelial barrier hyperpermeability was detected by an electrical cell-substrate impedance sensing (ECIS) system and a transwell chamber system. The expression of VE-cadherin in HUVECs was examined by real-time PCR and western blot. RESULTS We demonstrated that the alleviation of LPS-induced barrier dysfunction could be achieved by pretreatment with 3000 U/mL of UTI. VE-cadherin monoclonal antibody (mAb) could inhibit the protective effects. UTI maintained VE-cadherin expression by increasing protein stability at both the transcriptional and post-transcriptional levels. Meanwhile, VE-cadherin expression on the cell surface increased when the cells were pretreated with UTI. Furthermore, pretreatment with UTI decreased the phosphorylation of VE-cadherin at Tyr658 but not Tyr731. CONCLUSIONS Our data show that prophylactic UTI maintains the endothelial barrier function, increases VE-cadherin expression, and inhibits the phosphorylation of VE-cadherin at Tyr658 under inflammatory conditions. It suggests a scientific and potential clinical therapeutic importance of UTI in treatment of inflammatory disorders.
Collapse
Affiliation(s)
- Jie Chen
- Department of Emergency, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People's Republic of China
| | - Jun Wang
- The Laboratory of Neurotoxicology, School of Public Health, Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Chenglei Su
- Department of Emergency, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, 221000, People's Republic of China
| | - Wenyi Qian
- The Laboratory of Neurotoxicology, School of Public Health, Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Li Sun
- Department of Basic Medical College, Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Hao Sun
- Department of Emergency, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People's Republic of China
| | - Junjie Chen
- Department of Emergency, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People's Republic of China
| | - Huazhong Zhang
- Department of Emergency, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People's Republic of China
| | - Jinsong Zhang
- Department of Emergency, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People's Republic of China.
| |
Collapse
|
243
|
Frye M, Dierkes M, Küppers V, Vockel M, Tomm J, Zeuschner D, Rossaint J, Zarbock A, Koh GY, Peters K, Nottebaum AF, Vestweber D. Interfering with VE-PTP stabilizes endothelial junctions in vivo via Tie-2 in the absence of VE-cadherin. J Exp Med 2015; 212:2267-87. [PMID: 26642851 PMCID: PMC4689167 DOI: 10.1084/jem.20150718] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 10/16/2015] [Indexed: 12/31/2022] Open
Abstract
Inhibition of VE-PTP counters vascular leakage in inflammation via TIE-2, even in the absence of VE-cadherin. Vascular endothelial (VE)–protein tyrosine phosphatase (PTP) associates with VE-cadherin, thereby supporting its adhesive activity and endothelial junction integrity. VE-PTP also associates with Tie-2, dampening the tyrosine kinase activity of this receptor that can support stabilization of endothelial junctions. Here, we have analyzed how interference with VE-PTP affects the stability of endothelial junctions in vivo. Blocking VE-PTP by antibodies, a specific pharmacological inhibitor (AKB-9778), and gene ablation counteracted vascular leak induction by inflammatory mediators. In addition, leukocyte transmigration through the endothelial barrier was attenuated. Interference with Tie-2 expression in vivo reversed junction-stabilizing effects of AKB-9778 into junction-destabilizing effects. Furthermore, lack of Tie-2 was sufficient to weaken the vessel barrier. Mechanistically, inhibition of VE-PTP stabilized endothelial junctions via Tie-2, which triggered activation of Rap1, which then caused the dissolution of radial stress fibers via Rac1 and suppression of nonmuscle myosin II. Remarkably, VE-cadherin gene ablation did not abolish the junction-stabilizing effect of the VE-PTP inhibitor. Collectively, we conclude that inhibition of VE-PTP stabilizes challenged endothelial junctions in vivo via Tie-2 by a VE-cadherin–independent mechanism. In the absence of Tie-2, however, VE-PTP inhibition destabilizes endothelial barrier integrity in agreement with the VE-cadherin–supportive effect of VE-PTP.
Collapse
Affiliation(s)
- Maike Frye
- Max Planck Institute for Molecular Biomedicine, D-48149 Münster, Germany
| | - Martina Dierkes
- Max Planck Institute for Molecular Biomedicine, D-48149 Münster, Germany
| | - Verena Küppers
- Max Planck Institute for Molecular Biomedicine, D-48149 Münster, Germany
| | - Matthias Vockel
- Max Planck Institute for Molecular Biomedicine, D-48149 Münster, Germany
| | - Janina Tomm
- Max Planck Institute for Molecular Biomedicine, D-48149 Münster, Germany
| | - Dagmar Zeuschner
- Electron Microscopy Unit, Max Planck Institute for Molecular Biomedicine, D-48149 Münster, Germany
| | - Jan Rossaint
- Department of Anesthesiology and Critical Care Medicine, University of Münster, D-48149 Münster, Germany
| | - Alexander Zarbock
- Department of Anesthesiology and Critical Care Medicine, University of Münster, D-48149 Münster, Germany
| | - Gou Young Koh
- Center for Vascular Research, Institute of Basic Science, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Republic of Korea Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Republic of Korea
| | | | | | - Dietmar Vestweber
- Max Planck Institute for Molecular Biomedicine, D-48149 Münster, Germany
| |
Collapse
|
244
|
Benn A, Bredow C, Casanova I, Vukičević S, Knaus P. VE-cadherin facilitates BMP-induced endothelial cell permeability and signaling. J Cell Sci 2015; 129:206-18. [PMID: 26598555 PMCID: PMC4732303 DOI: 10.1242/jcs.179960] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 11/16/2015] [Indexed: 12/22/2022] Open
Abstract
Several vascular disorders, such as aberrant angiogenesis, atherosclerosis and pulmonary hypertension, have been linked to dysfunctional BMP signaling. Vascular hyperpermeability via distortion of endothelial cell adherens junctions is a common feature of these diseases, but the role of BMPs in this process has not been investigated. BMP signaling is initiated by binding of ligand to, and activation of, BMP type I (BMPRI) and type II (BMPRII) receptors. Internalization of VE-cadherin as well as c-Src kinase-dependent phosphorylation have been implicated in the loosening of cell–cell contacts, thereby modulating vascular permeability. Here we demonstrate that BMP6 induces hyperpermeabilization of human endothelial cells by inducing internalization and c-Src-dependent phosphorylation of VE-cadherin. Furthermore, we show BMP-dependent physical interaction of VE-cadherin with the BMP receptor ALK2 (BMPRI) and BMPRII, resulting in stabilization of the BMP receptor complex and, thereby, the support of BMP6-Smad signaling. Our results provide first insights into the molecular mechanism of BMP-induced vascular permeability, a hallmark of various vascular diseases, and provide the basis for further investigations of BMPs as regulators of vascular integrity, both under physiological and pathophysiological conditions. Summary: We reveal the molecular mechanism by which BMP6 induces hyperpermeabilization of the endothelium. This provides first insights into the mechanism of BMP-dependent vascular integrity in normal physiology and disease.
Collapse
Affiliation(s)
- Andreas Benn
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin 14195, Germany DFG Graduate School 1093 Berlin School of Integrative Oncology, Berlin 13353, Germany DFG Graduate School 203 Berlin-Brandenburg School for Regenerative Therapies, Berlin 13353, Germany
| | - Clara Bredow
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin 14195, Germany
| | - Isabel Casanova
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin 14195, Germany
| | - Slobodan Vukičević
- Center for Translational and Clinical Research, University of Zagreb, Zagreb 10000, Croatia
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin 14195, Germany DFG Graduate School 1093 Berlin School of Integrative Oncology, Berlin 13353, Germany DFG Graduate School 203 Berlin-Brandenburg School for Regenerative Therapies, Berlin 13353, Germany
| |
Collapse
|
245
|
Muller WA. Localized signals that regulate transendothelial migration. Curr Opin Immunol 2015; 38:24-9. [PMID: 26584476 DOI: 10.1016/j.coi.2015.10.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 10/26/2015] [Indexed: 01/13/2023]
Abstract
Transendothelial migration (TEM) of leukocytes is the step in leukocyte emigration in which the leukocyte actually leaves the blood vessel to carry out its role in the inflammatory response. It is therefore, arguably the most critical step in emigration. This review focuses on two of the many aspects of this process that have seen important recent developments. The adhesion molecules, PECAM (CD31) and CD99 that regulate two major steps in TEM, do so by regulating specific signals. PECAM initiates the signaling pathway responsible for the calcium flux that is required for TEM. Calcium enters through the cation channel TRPC6 and recruits the first wave of trafficking of membrane from the lateral border recycling compartment (LBRC). CD99 signals through soluble adenylate cyclase to activate protein kinase A to recruit a second wave of LBRC trafficking. Another process that is critical for TEM is transient removal of VE-cadherin from the site of TEM. However, the local signaling pathways that are responsible for this appear to be different from those that open the junctions to increase vascular permeability.
Collapse
Affiliation(s)
- William A Muller
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60201, USA.
| |
Collapse
|
246
|
Zhang P, Feng S, Liu G, Wang H, Zhu H, Ren Q, Bai H, Fu C, Dong C. Mutant B-Raf(V600E) Promotes Melanoma Paracellular Transmigration by Inducing Thrombin-mediated Endothelial Junction Breakdown. J Biol Chem 2015; 291:2087-106. [PMID: 26504080 DOI: 10.1074/jbc.m115.696419] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Indexed: 01/04/2023] Open
Abstract
Tumor invasiveness depends on the ability of tumor cells to breach endothelial barriers. In this study, we investigated the mechanism by which the adhesion of melanoma cells to endothelium regulates adherens junction integrity and modulates tumor transendothelial migration (TEM) by initiating thrombin generation. We found that the B-Raf(V600E) mutation in metastatic melanoma cells up-regulated tissue factor (TF) expression on cell membranes and promoted thrombin production. Co-culture of endothelial monolayers with metastatic melanoma cells mediated the opening of inter-endothelial spaces near melanoma cell contact sites in the presence of platelet-free plasma (PFP). By using small interfering RNA (siRNA), we demonstrated that B-Raf(V600E) and TF silencing attenuated the focal disassembly of adherens junction induced by tumor contact. Vascular endothelial-cadherin (VE-cadherin) disassembly was dependent on phosphorylation of p120-catenin on Ser-879 and VE-cadherin on Tyr-658, Tyr-685, and Tyr-731, which can be prevented by treatment with the thrombin inhibitor, hirudin, or by silencing the thrombin receptor, protease-activated receptor-1, in endothelial cells. We also provided strong evidence that tumor-derived thrombin enhanced melanoma TEM by inducing ubiquitination-coupled VE-cadherin internalization, focal adhesion formation, and actin assembly in endothelium. Confocal microscopic analysis of tumor TEM revealed that junctions transiently opened and resealed as tumor cells accomplished TEM. In addition, in the presence of PFP, tumor cells preferentially transmigrated via paracellular routes. PFP supported melanoma transmigration under shear conditions via a B-Raf(V600E)-thrombin-dependent mechanism. We concluded that the activation of thrombin generation by cancer cells in plasma is an important process regulating melanoma extravasation by disrupting endothelial junction integrity.
Collapse
Affiliation(s)
- Pu Zhang
- From the Key Laboratory of Luminescence and Real Time Analytical Chemistry, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China, the Department of Bioengineering, Pennsylvania State University, University Park, Pennsylvania 16801, and
| | - Shan Feng
- From the Key Laboratory of Luminescence and Real Time Analytical Chemistry, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Gentao Liu
- the Shanghai Pulmonary Hospital, Tongji University School of Medicine, 507 Zhengmin Road, Shanghai 200433, China
| | - Heyong Wang
- the Shanghai Pulmonary Hospital, Tongji University School of Medicine, 507 Zhengmin Road, Shanghai 200433, China
| | - Huifeng Zhu
- From the Key Laboratory of Luminescence and Real Time Analytical Chemistry, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Qiao Ren
- From the Key Laboratory of Luminescence and Real Time Analytical Chemistry, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Huiyuan Bai
- From the Key Laboratory of Luminescence and Real Time Analytical Chemistry, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Changliang Fu
- the Department of Bioengineering, Pennsylvania State University, University Park, Pennsylvania 16801, and
| | - Cheng Dong
- the Department of Bioengineering, Pennsylvania State University, University Park, Pennsylvania 16801, and
| |
Collapse
|
247
|
Regulation of Endothelial Adherens Junctions by Tyrosine Phosphorylation. Mediators Inflamm 2015; 2015:272858. [PMID: 26556953 PMCID: PMC4628659 DOI: 10.1155/2015/272858] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 08/16/2015] [Indexed: 12/14/2022] Open
Abstract
Endothelial cells form a semipermeable, regulated barrier that limits the passage of fluid, small molecules, and leukocytes between the bloodstream and the surrounding tissues. The adherens junction, a major mechanism of intercellular adhesion, is comprised of transmembrane cadherins forming homotypic interactions between adjacent cells and associated cytoplasmic catenins linking the cadherins to the cytoskeleton. Inflammatory conditions promote the disassembly of the adherens junction and a loss of intercellular adhesion, creating openings or gaps in the endothelium through which small molecules diffuse and leukocytes transmigrate. Tyrosine kinase signaling has emerged as a central regulator of the inflammatory response, partly through direct phosphorylation and dephosphorylation of the adherens junction components. This review discusses the findings that support and those that argue against a direct effect of cadherin and catenin phosphorylation in the disassembly of the adherens junction. Recent findings indicate a complex interaction between kinases, phosphatases, and the adherens junction components that allow a fine regulation of the endothelial permeability to small molecules, leukocyte migration, and barrier resealing.
Collapse
|
248
|
Abstract
Immune responses depend on the ability of leukocytes to move from the circulation into tissue. This is enabled by mechanisms that guide leukocytes to the right exit sites and allow them to cross the barrier of the blood vessel wall. This process is regulated by a concerted action between endothelial cells and leukocytes, whereby endothelial cells activate leukocytes and direct them to extravasation sites, and leukocytes in turn instruct endothelial cells to open a path for transmigration. This Review focuses on recently described mechanisms that control and open exit routes for leukocytes through the endothelial barrier.
Collapse
|
249
|
Gralinski LE, Ferris MT, Aylor DL, Whitmore AC, Green R, Frieman MB, Deming D, Menachery VD, Miller DR, Buus RJ, Bell TA, Churchill GA, Threadgill DW, Katze MG, McMillan L, Valdar W, Heise MT, Pardo-Manuel de Villena F, Baric RS. Genome Wide Identification of SARS-CoV Susceptibility Loci Using the Collaborative Cross. PLoS Genet 2015; 11:e1005504. [PMID: 26452100 PMCID: PMC4599853 DOI: 10.1371/journal.pgen.1005504] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 08/15/2015] [Indexed: 01/21/2023] Open
Abstract
New systems genetics approaches are needed to rapidly identify host genes and genetic networks that regulate complex disease outcomes. Using genetically diverse animals from incipient lines of the Collaborative Cross mouse panel, we demonstrate a greatly expanded range of phenotypes relative to classical mouse models of SARS-CoV infection including lung pathology, weight loss and viral titer. Genetic mapping revealed several loci contributing to differential disease responses, including an 8.5Mb locus associated with vascular cuffing on chromosome 3 that contained 23 genes and 13 noncoding RNAs. Integrating phenotypic and genetic data narrowed this region to a single gene, Trim55, an E3 ubiquitin ligase with a role in muscle fiber maintenance. Lung pathology and transcriptomic data from mice genetically deficient in Trim55 were used to validate its role in SARS-CoV-induced vascular cuffing and inflammation. These data establish the Collaborative Cross platform as a powerful genetic resource for uncovering genetic contributions of complex traits in microbial disease severity, inflammation and virus replication in models of outbred populations.
Collapse
Affiliation(s)
- Lisa E. Gralinski
- Department of Epidemiology, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Martin T. Ferris
- Department of Genetics, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - David L. Aylor
- Department of Genetics, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Alan C. Whitmore
- Department of Genetics, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Richard Green
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Matthew B. Frieman
- Department of Epidemiology, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Damon Deming
- Department of Epidemiology, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Vineet D. Menachery
- Department of Epidemiology, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Darla R. Miller
- Department of Genetics, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Ryan J. Buus
- Department of Genetics, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Timothy A. Bell
- Department of Genetics, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | | | - David W. Threadgill
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - Michael G. Katze
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Leonard McMillan
- Department of Computer Science, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - William Valdar
- Department of Genetics, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Mark T. Heise
- Department of Genetics, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Fernando Pardo-Manuel de Villena
- Department of Genetics, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Ralph S. Baric
- Department of Epidemiology, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
250
|
Endothelial-Leukocyte Interaction in Severe Malaria: Beyond the Brain. Mediators Inflamm 2015; 2015:168937. [PMID: 26491221 PMCID: PMC4605361 DOI: 10.1155/2015/168937] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 08/25/2015] [Accepted: 09/01/2015] [Indexed: 01/23/2023] Open
Abstract
Malaria is the most important parasitic disease worldwide, accounting for 1 million deaths each year. Severe malaria is a systemic illness characterized by dysfunction of brain tissue and of one or more peripheral organs as lungs and kidney. The most severe and most studied form of malaria is associated with cerebral complications due to capillary congestion and the adhesion of infected erythrocytes, platelets, and leukocytes to brain vasculature. Thus, leukocyte rolling and adhesion in the brain vascular bed during severe malaria is singular and distinct from other models of inflammation. The leukocyte/endothelium interaction and neutrophil accumulation are also observed in the lungs. However, lung interactions differ from brain interactions, likely due to differences in the blood-brain barrier and blood-air barrier tight junction composition of the brain and lung endothelium. Here, we review the importance of endothelial dysfunction and the mechanism of leukocyte/endothelium interaction during severe malaria. Furthermore, we hypothesize a possible use of adjunctive therapies to antimalarial drugs that target the interaction between the leukocytes and the endothelium.
Collapse
|