1
|
Aldirawi M, Ghanbari P, Mietkowska M, März S, Odenthal-Schnittler M, Franz J, Wegner J, Currie S, Kipcke JP, Taha M, Giglmaier M, Blanque A, Schillers H, Raz E, Vestweber D, Rottner K, Schnittler H. A specific role for endothelial EPLIN-isoform-regulated actin dynamics in neutrophil transmigration. Sci Rep 2025; 15:15698. [PMID: 40325158 PMCID: PMC12053001 DOI: 10.1038/s41598-025-98192-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 04/09/2025] [Indexed: 05/07/2025] Open
Abstract
Proinflammatory cytokines such as TNF-α or IL-1β activate the endothelium promoting leukocyte transendothelial migration (TEM) via expression of cell adhesion molecules (CAM) and cause actin remodelling. However, the function of endothelial actin remodelling in TEM remains elusive, despite its involvement in the formation of docking structures, diapedesis pores and pore resealing. Here, we establish EPLIN-isoforms, EPLIN-β and EPLIN-α, as differential regulators of TNF-α-inducedactin-remodelling significantly affecting TEM. We find EPLIN-β-induced stress fiber formation upon TNF-α-treatment weakens endothelial junctions, upregulates junctional dynamics and facilitates intercellular gaps for TEM. Increased junctional dynamics involves branched actin filaments under the control of EPLIN-α, including docking structure formation and transmigratory pore closure. We further establish by EPLIN deletion and re-expression studies that EPLIN-α-mediated termination of branched actin filaments maintains TNF-α-induced junctional dynamics and intercellular gaps facilitating TEM. These findings highlight the critical role of TNF-α-induced differential actin dynamics, controlled by EPLIN isoforms, in TEM. These results also offer a wider understanding of inflammation-induced TEM by incorporating altered junctional dynamics alongside upregulation of cell adhesion molecules.
Collapse
Affiliation(s)
- Mohammed Aldirawi
- Institute of Anatomy and Vascular Biology, University Münster, Vesaliusweg 2-4, Münster, Germany
| | - Parisa Ghanbari
- Institute of Anatomy and Vascular Biology, University Münster, Vesaliusweg 2-4, Münster, Germany
- Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, Münster, Germany
| | - Magdalena Mietkowska
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106, Braunschweig, Germany
- Molecular Cell Biology Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Sigrid März
- Institute of Anatomy and Vascular Biology, University Münster, Vesaliusweg 2-4, Münster, Germany
| | - Maria Odenthal-Schnittler
- Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, Münster, Germany
- Institute of Neuropathology, University of Münster, Pottkamp 2, 48149, Münster, Germany
| | - Jonas Franz
- Institute of Anatomy and Vascular Biology, University Münster, Vesaliusweg 2-4, Münster, Germany
- Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, Münster, Germany
- Institute of Neuropathology, University of Münster, Pottkamp 2, 48149, Münster, Germany
| | - Julian Wegner
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, 48149, Münster, Germany
| | - Silke Currie
- Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, Münster, Germany
| | - Jan Philip Kipcke
- Institute of Anatomy and Vascular Biology, University Münster, Vesaliusweg 2-4, Münster, Germany
- Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, Münster, Germany
- Institute of Neuropathology, University of Münster, Pottkamp 2, 48149, Münster, Germany
| | - Muna Taha
- Institute of Anatomy and Vascular Biology, University Münster, Vesaliusweg 2-4, Münster, Germany
| | - Marcus Giglmaier
- Institute of Aerodynamics and Fluid Mechanics, Technical University of Munich, Boltzmannstr. 15, 85748, Garching, Germany
| | - Anja Blanque
- Institute of Physiology, University Münster, Robert-Koch Strasse 27a, 48149, Münster, Germany
| | - Hermann Schillers
- Institute of Physiology, University Münster, Robert-Koch Strasse 27a, 48149, Münster, Germany
| | - Erez Raz
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, 48149, Münster, Germany
| | - Dietmar Vestweber
- Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, Münster, Germany
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106, Braunschweig, Germany
- Molecular Cell Biology Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Hans Schnittler
- Institute of Anatomy and Vascular Biology, University Münster, Vesaliusweg 2-4, Münster, Germany.
- Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, Münster, Germany.
- Institute of Neuropathology, University of Münster, Pottkamp 2, 48149, Münster, Germany.
| |
Collapse
|
2
|
Trevino TN, Almousawi AA, Martins-Goncalves R, Ochoa-Raya A, Robinson KF, Abad GL, Tai LM, Oliveira SD, Minshall RD, Lutz SE. A Brain Endothelial Cell Caveolin-1/CXCL10 Axis Promotes T Cell Transcellular Migration Across the Blood-Brain Barrier. ASN Neuro 2025; 17:2472070. [PMID: 40063988 PMCID: PMC12047051 DOI: 10.1080/17590914.2025.2472070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/05/2024] [Accepted: 01/06/2025] [Indexed: 05/04/2025] Open
Abstract
The mechanisms that govern whether T cells cross blood-brain barrier (BBB) endothelium by transcellular versus paracellular routes are unclear. Caveolin-1 is a membrane scaffolding and signaling protein associated with transcellular transmigration through the endothelial cytoplasm. Here, we report that the neuroinflammatory chemokine CXCL10 induced transcellular, caveolar transmigration of CXCR3+ CD4+ T cells. Specifically, data revealed that CXCL10-induced transcellular transmigration requires expression of Caveolin-1 and ICAM-1 in brain endothelial cells and of the CXCL10 receptor, CXCR3, and LFA-1 in T cells. Moreover, Caveolin-1 promoted CXCL10 aggregation into brain endothelial cytoplasmic stores, providing a mechanism for activation and recruitment of CXCR3+ T cells to migrate at cytoplasmic locations, distal to cell-cell junctions. Consistent with our in vitro data, genetic ablation of Caveolin-1 reduces infiltration of CXCR3+ CD4+ T cells into the CNS in experimental autoimmune encephalomyelitis. Our findings establish a novel mechanism by which brain endothelial cells utilize Caveolin-1 dependent CXCL10 intracellular stores to license T cells for transcellular migration across the blood-brain barrier.
Collapse
Affiliation(s)
- Troy N. Trevino
- Departments of Anatomy and Cell Biology, University of Illinois at Chicago, College of Medicine, Chicago, Illinois, USA
| | - Ali A. Almousawi
- Departments of Anatomy and Cell Biology, University of Illinois at Chicago, College of Medicine, Chicago, Illinois, USA
| | - Remy Martins-Goncalves
- Departments of Anatomy and Cell Biology, University of Illinois at Chicago, College of Medicine, Chicago, Illinois, USA
| | - Andrea Ochoa-Raya
- Departments of Anatomy and Cell Biology, University of Illinois at Chicago, College of Medicine, Chicago, Illinois, USA
| | - KaReisha F. Robinson
- Departments of Anatomy and Cell Biology, University of Illinois at Chicago, College of Medicine, Chicago, Illinois, USA
| | - Genesis L. Abad
- Departments of Anatomy and Cell Biology, University of Illinois at Chicago, College of Medicine, Chicago, Illinois, USA
| | - Leon M. Tai
- Departments of Anatomy and Cell Biology, University of Illinois at Chicago, College of Medicine, Chicago, Illinois, USA
| | - Suellen D. Oliveira
- Anesthesiology, University of Illinois at Chicago, College of Medicine, Chicago, Illinois, USA
- Physiology and Biophysics, University of Illinois at Chicago, College of Medicine, Chicago, Illinois, USA
| | - Richard D. Minshall
- Anesthesiology, University of Illinois at Chicago, College of Medicine, Chicago, Illinois, USA
- Pharmacology and Regenerative Medicine, University of Illinois at Chicago, College of Medicine, Chicago, Illinois, USA
| | - Sarah E. Lutz
- Departments of Anatomy and Cell Biology, University of Illinois at Chicago, College of Medicine, Chicago, Illinois, USA
| |
Collapse
|
3
|
Hansen CE, Hollaus D, Kamermans A, de Vries HE. Tension at the gate: sensing mechanical forces at the blood-brain barrier in health and disease. J Neuroinflammation 2024; 21:325. [PMID: 39696463 PMCID: PMC11657007 DOI: 10.1186/s12974-024-03321-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024] Open
Abstract
Microvascular brain endothelial cells tightly limit the entry of blood components and peripheral cells into the brain by forming the blood-brain barrier (BBB). The BBB is regulated by a cascade of mechanical and chemical signals including shear stress and elasticity of the adjacent endothelial basement membrane (BM). During physiological aging, but especially in neurological diseases including multiple sclerosis (MS), stroke, small vessel disease, and Alzheimer's disease (AD), the BBB is exposed to inflammation, rigidity changes of the BM, and disturbed cerebral blood flow (CBF). These altered forces lead to increased vascular permeability, reduced endothelial reactivity to vasoactive mediators, and promote leukocyte transmigration. Whereas the molecular players involved in leukocyte infiltration have been described in detail, the importance of mechanical signalling throughout this process has only recently been recognized. Here, we review relevant features of mechanical forces acting on the BBB under healthy and pathological conditions, as well as the endothelial mechanosensory elements detecting and responding to altered forces. We demonstrate the underlying complexity by focussing on the family of transient receptor potential (TRP) ion channels. A better understanding of these processes will provide insights into the pathogenesis of several neurological disorders and new potential leads for treatment.
Collapse
Affiliation(s)
- Cathrin E Hansen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - David Hollaus
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Alwin Kamermans
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands.
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
4
|
Schwartz AB, Kandasamy A, Del Álamo JC, Yeh YT. Neutrophils exhibit distinct migration phenotypes that are regulated by transendothelial migration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.17.618860. [PMID: 39677773 PMCID: PMC11642774 DOI: 10.1101/2024.10.17.618860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
The extravasation of polymorphonuclear neutrophils (PMNs) is a critical component of the innate immune response that involves transendothelial migration (TEM) and interstitial migration. TEM-mediated interactions between PMNs and vascular endothelial cells (VECs) trigger a cascade of biochemical and mechanobiological signals whose effects on interstitial migration are currently unclear. To address this question, we cultured human VECs on a fibronectin-treated transwell insert to model the endothelium and basement membrane, loaded PMN-like differentiated HL60 (dHL-60) cells in the upper chamber of the insert, and collected the PMNs that crossed the membrane-supported monolayer from the lower chamber. The 3D chemotactic migration of the TEM-conditioned PMNs through collagen matrices was then quantified. Data collected from over 50,000 trajectories showed two distinct migratory phenotypes, i.e., a high-persistence phenotype and a low-persistence phenotype. These phenotypes were conserved across treatment conditions, and their existence was confirmed in human primary PMNs. The high-persistence phenotype was characterized by more straight trajectories and faster migration speeds, whereas the low-persistence one exhibited more frequent sharp turns and loitering periods. A key finding of our study is that TEM induced a phenotypic shift in PMNs from high-persistence migration to low-persistence migration. Changes in the relative proportion of high-persistence and low-persistence populations correlated with GRK2 expression levels. Inhibiting GRK2 hindered the TEM-induced shift in migratory phenotype and impaired the phagocytic function of PMNs. Overall, our study suggests that TEM-mediated GRK2 signaling primes PMNs for a migration phenotype better suited for spatial exploration and inflammation resolution. These observations provide novel insight into the biophysical impacts of TEM that priming PMNs is essential to conduct sentinel functions.
Collapse
|
5
|
Dupas A, Goetz JG, Osmani N. Extravasation of immune and tumor cells from an endothelial perspective. J Cell Sci 2024; 137:jcs262066. [PMID: 39530179 DOI: 10.1242/jcs.262066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Crossing the vascular endothelium is a necessary stage for circulating cells aiming to reach distant organs. Leukocyte passage through the endothelium, known as transmigration, is a multistep process during which immune cells adhere to the vascular wall, migrate and crawl along the endothelium until they reach their exit site. Similarly, circulating tumor cells (CTCs), which originate from the primary tumor or reseed from early metastatic sites, disseminate using the blood circulation and also must cross the endothelial barrier to set new colonies in distant organs. CTCs are thought to mimic arrest and extravasation utilized by leukocytes; however, their extravasation also requires processes that, from an endothelial perspective, are specific to cancer cells. Although leukocyte extravasation relies on maintaining endothelial impermeability, it appears that cancer cells can indoctrinate endothelial cells into promoting their extravasation independently of their normal functions. In this Review, we summarize the common and divergent mechanisms of endothelial responses during extravasation of leukocytes (in inflammation) and CTCs (in metastasis), and highlight how these might be leveraged in the development of anti-metastatic treatments.
Collapse
Affiliation(s)
- Amandine Dupas
- Tumor Biomechanics lab, INSERM UMR_S 1109, CRBS, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg Cedex, France
- Université de Strasbourg, Strasbourg, F-67000, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, F-67000, France
- Equipe Labellisée Ligue Contre le Cancer, France
| | - Jacky G Goetz
- Tumor Biomechanics lab, INSERM UMR_S 1109, CRBS, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg Cedex, France
- Université de Strasbourg, Strasbourg, F-67000, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, F-67000, France
- Equipe Labellisée Ligue Contre le Cancer, France
| | - Naël Osmani
- Tumor Biomechanics lab, INSERM UMR_S 1109, CRBS, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg Cedex, France
- Université de Strasbourg, Strasbourg, F-67000, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, F-67000, France
- Equipe Labellisée Ligue Contre le Cancer, France
| |
Collapse
|
6
|
Dooling LJ, Anlaş AA, Tobin MP, Ontko NM, Marchena T, Wang M, Andrechak JC, Discher DE. Clustered macrophages cooperate to eliminate tumors via coordinated intrudopodia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613918. [PMID: 39345601 PMCID: PMC11430028 DOI: 10.1101/2024.09.19.613918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Macrophages often pervade solid tumors, but their nearest neighbor organization is understudied and potentially enables key functions such as phagocytosis. Here, we observe dynamic macrophage clusters in tumors under conditions that maximize cancer cell phagocytosis and use reductionist approaches to uncover pathways to cluster formation and roles for tumor-intrusive pseudopodia, which we term 'intrudopodia'. Macrophage clusters form over hours on low- adhesion substrates after M1 polarization with interferons, including T cell-derived cytokines, and yet clusters prove fluid on timescales of minutes. Clusters also sort from M2 macrophages that disperse on the same substrates. M1 macrophages upregulate specific cell-cell adhesion receptors but suppress actomyosin contractility, and while both pathways contribute to cluster formation, decreased cortical tension was predicted to unleash pseudopodia. Macrophage neighbors in tumor spheroids indeed extend intrudopodia between adjacent cancer cell junctions - at least when phagocytosis conditions are maximized, and coordinated intrudopodia help detach and individualize cancer cells for rapid engulfment. Macrophage clusters thereby provide a cooperative advantage for phagocytosis to overcome solid tumor cohesion.
Collapse
|
7
|
Liaqat I, Hilska I, Saario M, Jakobsson E, Crivaro M, Peränen J, Vaahtomeri K. Spatially targeted chemokine exocytosis guides transmigration at lymphatic endothelial multicellular junctions. EMBO J 2024; 43:3141-3174. [PMID: 38877304 PMCID: PMC11294460 DOI: 10.1038/s44318-024-00129-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 06/16/2024] Open
Abstract
Migrating cells preferentially breach and integrate epithelial and endothelial monolayers at multicellular vertices. These sites are amenable to forces produced by the migrating cell and subsequent opening of the junctions. However, the cues that guide migrating cells to these entry portals, and eventually drive the transmigration process, are poorly understood. Here, we show that lymphatic endothelium multicellular junctions are the preferred sites of dendritic cell transmigration in both primary cell co-cultures and in mouse dermal explants. Dendritic cell guidance to multicellular junctions was dependent on the dendritic cell receptor CCR7, whose ligand, lymphatic endothelial chemokine CCL21, was exocytosed at multicellular junctions. Characterization of lymphatic endothelial secretory routes indicated Golgi-derived RAB6+ vesicles and RAB3+/27+ dense core secretory granules as intracellular CCL21 storage vesicles. Of these, RAB6+ vesicles trafficked CCL21 to the multicellular junctions, which were enriched with RAB6 docking factor ELKS (ERC1). Importantly, inhibition of RAB6 vesicle exocytosis attenuated dendritic cell transmigration. These data exemplify how spatially-restricted exocytosis of guidance cues helps to determine where dendritic cells transmigrate.
Collapse
Affiliation(s)
- Inam Liaqat
- Translational Cancer Medicine Research Program, University of Helsinki, Biomedicum Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
| | - Ida Hilska
- Translational Cancer Medicine Research Program, University of Helsinki, Biomedicum Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
| | - Maria Saario
- Translational Cancer Medicine Research Program, University of Helsinki, Biomedicum Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
| | - Emma Jakobsson
- Translational Cancer Medicine Research Program, University of Helsinki, Biomedicum Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
| | - Marko Crivaro
- Light Microscopy Unit, Institute of Biotechnology, HiLIFE, University of Helsinki, FI-00014, Helsinki, Finland
| | - Johan Peränen
- Institute of Biotechnology, HiLIFE, University of Helsinki, FI-00014, Helsinki, Finland
| | - Kari Vaahtomeri
- Translational Cancer Medicine Research Program, University of Helsinki, Biomedicum Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland.
- Wihuri Research Institute, Biomedicum Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland.
| |
Collapse
|
8
|
Bywaters BC, Trache A, Rivera GM. Modulation of arterial intima stiffness by disturbed blood flow. Exp Biol Med (Maywood) 2024; 249:10090. [PMID: 39143955 PMCID: PMC11323813 DOI: 10.3389/ebm.2024.10090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/09/2024] [Indexed: 08/16/2024] Open
Abstract
The intima, comprising the endothelium and the subendothelial matrix, plays a crucial role in atherosclerosis pathogenesis. The mechanical stress arising from disturbed blood flow (d-flow) and the stiffening of the arterial wall contributes to endothelial dysfunction. However, the specific impacts of these physical forces on the mechanical environment of the intima remain undetermined. Here, we investigated whether inhibiting collagen crosslinking could ameliorate the detrimental effects of persistent d-flow on the mechanical properties of the intima. Partial ligation of the left carotid artery (LCA) was performed in C57BL/6J mice, inducing d-flow. The right carotid artery (RCA) served as an internal control. Carotids were collected 2 days and 2 weeks after surgery to study acute and chronic effects of d-flow on the mechanical phenotype of the intima. The chronic effects of d-flow were decoupled from the ensuing arterial wall stiffening by administration of β-aminopropionitrile (BAPN), an inhibitor of collagen crosslinking by lysyl oxidase (LOX) enzymes. Atomic force microscopy (AFM) was used to determine stiffness of the endothelium and the denuded subendothelial matrix in en face carotid preparations. The stiffness of human aortic endothelial cells (HAEC) cultured on soft and stiff hydrogels was also determined. Acute exposure to d-flow caused a slight decrease in endothelial stiffness in male mice but had no effect on the stiffness of the subendothelial matrix in either sex. Regardless of sex, the intact endothelium was softer than the subendothelial matrix. In contrast, exposure to chronic d-flow led to a substantial increase in the endothelial and subendothelial stiffness in both sexes. The effects of chronic d-flow were largely prevented by concurrent BAPN administration. In addition, HAEC displayed reduced stiffness when cultured on soft vs. stiff hydrogels. We conclude that chronic d-flow results in marked stiffening of the arterial intima, which can be effectively prevented by inhibition of collagen crosslinking.
Collapse
Affiliation(s)
- Briana C. Bywaters
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
| | - Andreea Trache
- Department of Medical Physiology, Texas A&M Health Science Center, Bryan, TX, United States
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| | - Gonzalo M. Rivera
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
| |
Collapse
|
9
|
Calo CJ, Patil T, Palizzi M, Wheeler N, Hind LE. Collagen concentration regulates neutrophil extravasation and migration in response to infection in an endothelium dependent manner. Front Immunol 2024; 15:1405364. [PMID: 39021568 PMCID: PMC11251947 DOI: 10.3389/fimmu.2024.1405364] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Introduction As the body's first line of defense against disease and infection, neutrophils must efficiently navigate to sites of inflammation; however, neutrophil dysregulation contributes to the pathogenesis of numerous diseases that leave people susceptible to infections. Many of these diseases are also associated with changes to the protein composition of the extracellular matrix. While it is known that neutrophils and endothelial cells, which play a key role in neutrophil activation, are sensitive to the mechanical and structural properties of the extracellular matrix, our understanding of how protein composition in the matrix affects the neutrophil response to infection is incomplete. Methods To investigate the effects of extracellular matrix composition on the neutrophil response to infection, we used an infection-on-a-chip microfluidic device that replicates a portion of a blood vessel endothelium surrounded by a model extracellular matrix. Model blood vessels were fabricated by seeding human umbilical vein endothelial cells on 2, 4, or 6 mg/mL type I collagen hydrogels. Primary human neutrophils were loaded into the endothelial lumens and stimulated by adding the bacterial pathogen Pseudomonas aeruginosa to the surrounding matrix. Results Collagen concentration did not affect the cell density or barrier function of the endothelial lumens. Upon infectious challenge, we found greater neutrophil extravasation into the 4 mg/mL collagen gels compared to the 6 mg/mL collagen gels. We further found that extravasated neutrophils had the highest migration speed and distance in 2mg/mL gels and that these values decreased with increasing collagen concentration. However, these phenomena were not observed in the absence of an endothelial lumen. Lastly, no differences in the percent of extravasated neutrophils producing reactive oxygen species were observed across the various collagen concentrations. Discussion Our study suggests that neutrophil extravasation and migration in response to an infectious challenge are regulated by collagen concentration in an endothelial cell-dependent manner. The results demonstrate how the mechanical and structural aspects of the tissue microenvironment affect the neutrophil response to infection. Additionally, these findings underscore the importance of developing and using microphysiological systems for studying the regulatory factors that govern the neutrophil response.
Collapse
Affiliation(s)
| | | | | | | | - Laurel E. Hind
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, United States
| |
Collapse
|
10
|
Xia M, Stegmeyer RI, Shirakura K, Butz S, Thiriot A, von Andrian UH, Vestweber D. Conditions that promote transcellular neutrophil migration in vivo. Sci Rep 2024; 14:14471. [PMID: 38914623 PMCID: PMC11196655 DOI: 10.1038/s41598-024-65173-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 06/18/2024] [Indexed: 06/26/2024] Open
Abstract
Circulating leukocytes enter tissue either through endothelial junctions (paracellular) or via a pore through the body of endothelial cells (transcellular). We have previously shown that genetically replacing VE-cadherin with a VE-cadherin-α-catenin (VEC-αC) fusion construct-which binds constitutively to actin-obstructs junctions, and blocks leukocyte extravasation in lung, skin and postcapillary venules of cremaster muscle. However, neutrophil recruitment into the inflamed peritoneal cavity was unimpaired. Investigating reasons for this, here, we visualized neutrophil diapedesis by 3D intravital video microscopy in the cremaster muscle and omentum, the major site of neutrophil recruitment into the peritoneal cavity. We found that 80% of neutrophil-extravasation occurred through HEVs in the omentum, which was unimpaired by VEC-αC. In addition, in larger venules (60-85 µm) of both tissues, less than 15% of neutrophils extravasated transcellularly in WT mice. However, in VEC-α-C mice, transcellular diapedesis increased severalfold in the omentum, but not in the cremaster. In line with this, omental venules expressed higher levels of ICAM-1 and atypical chemokine receptor 1. Furthermore, only in the omentum, VEC-αC expression caused reduced elongation of venular endothelium in flow-direction, suggesting different biomechanical properties. Collectively, VEC-αC does not inhibit paracellular transmigration in all types of venules and can modulate the diapedesis route.
Collapse
Affiliation(s)
- Min Xia
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149, Münster, Germany
| | - Rebekka I Stegmeyer
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149, Münster, Germany
| | - Keisuke Shirakura
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149, Münster, Germany
| | - Stefan Butz
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149, Münster, Germany
| | - Aude Thiriot
- Department of Immunology and Center for Immune Imaging, Harvard Medical School, Boston, MA, USA
| | - Ulrich H von Andrian
- Department of Immunology and Center for Immune Imaging, Harvard Medical School, Boston, MA, USA
| | - Dietmar Vestweber
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149, Münster, Germany.
| |
Collapse
|
11
|
Cassisa A, Cima L. Cutaneous vasculitis: insights into pathogenesis and histopathological features. Pathologica 2024; 116:119-133. [PMID: 38767544 PMCID: PMC11138767 DOI: 10.32074/1591-951x-985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 03/01/2024] [Indexed: 05/22/2024] Open
Abstract
The mechanisms underlying the onset and progression of vasculitis remain poorly understood. This condition is characterized by damage to the vascular wall, recruitment of inflammatory cells, and subsequent structural remodeling, which are hallmarks of vasculitis. The histopathological classification of vasculitis relies on the size of the affected vessel and the predominant type of inflammatory cell involved - neutrophils in acute cases, lymphocytes in chronic conditions, and histiocytes in granulomatous forms. Pathological changes progress in every context, and a single vasculitic pattern can be associated with various systemic conditions. Conversely, a single causative agent may lead to multiple distinct clinical and pathological manifestations of vasculitis. Moreover, many cases of vasculitis have no identifiable cause. A foundational understanding of the normal structure of the cutaneous vascular network is crucial. Similarly, identifying the cellular and molecular participants and their roles in forming the "dermal microvascular unit" is propedeutical. This review aims to elucidate the complex mechanisms involved in the initiation and progression of vasculitis, offering a comprehensive overview of its histopathological classification, underlying causes, and the significant role of the cutaneous vascular network and cellular dynamics. By integrating the latest insights from studies on NETosis and the implications of lymphocytic infiltration in autoimmune diseases, we seek to bridge gaps in current knowledge and highlight areas for future research. Our discussion extends to the clinical implications of vasculitis, emphasizing the importance of identifying etiological agents and understanding the diverse histopathological manifestations to improve diagnostic accuracy and treatment outcomes.
Collapse
Affiliation(s)
- Angelo Cassisa
- Department of Oncology, Section of Pathology, San Giovanni di Dio Hospital, USL Centro Toscana, Florence, Italy
| | - Luca Cima
- Department of Laboratory Medicine, Pathology Unit, Santa Chiara Hospital, Trento, Italy
| |
Collapse
|
12
|
Kim J, Mooren OL, Onken MD, Cooper JA. Septin and actin contributions to endothelial cell-cell junctions and monolayer integrity. Cytoskeleton (Hoboken) 2023; 80:228-241. [PMID: 36205643 PMCID: PMC10079785 DOI: 10.1002/cm.21732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/02/2022] [Accepted: 10/04/2022] [Indexed: 11/10/2022]
Abstract
Septins in endothelial cells (ECs) have important roles supporting the integrity of the endothelial monolayer. Cell-cell junctions in EC monolayers are highly dynamic, with continuous retractions and protrusions. Depletion of septins in ECs leads to disruption of cell-cell junctions, which are composed of VE-cadherin and other junctional proteins. In EC monolayers, septins are concentrated at the plasma membrane at sites of cell-cell contact, in curved- and scallop-shaped patterns. These membrane-associated septin accumulations are located in regions of positive membrane curvature, and those regions are often associated with and immediately adjacent to actin-rich protrusions with negative membrane curvature. EC septins associate directly with plasma membrane lipids, based on findings with site-specific mutations of septins in ECs, which is consistent with biochemical and cell biological studies in other systems. Loss of septins leads to disruption of the EC monolayer, and gaps form between cells. The number and breadth of cell-cell contacts and junctions decreases, and the number and frequency of retractions, ruffles, and protrusions at cell edges also decreases. In addition, loss of septins leads to decreased amounts of F-actin at the cortical membrane, along with increased amounts of F-actin in stress fibers of the cytoplasm. Endothelial monolayer disruption from loss of septins is also associated with decreased transendothelial electric resistance (TEER) and increased levels of transendothelial migration (TEM) by immune and cancer cells, owing to the gaps in the monolayer. A current working model is that assembly of septin filaments at regions of positive membrane curvature contributes to a mechanical footing or base for actin-based protrusive forces generated at adjoining regions of the membrane. Specific molecular interactions between the septin and actin components of the cytoskeleton may also be important contributors. Regulators of actin assembly may promote and support the assembly of septin filaments at the membrane, as part of a molecular feedback loop between the assembly of septin and actin filaments.
Collapse
Affiliation(s)
- Joanna Kim
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, Missouri, USA
| | - Olivia L Mooren
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, Missouri, USA
| | - Michael D Onken
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, Missouri, USA
| | - John A Cooper
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
13
|
Watson SA, Javanmardi Y, Zanieri L, Shahreza S, Ragazzini R, Bonfanti P, Moeendarbary E. Integrated role of human thymic stromal cells in hematopoietic stem cell extravasation. Bioeng Transl Med 2023; 8:e10454. [PMID: 36925684 PMCID: PMC10013751 DOI: 10.1002/btm2.10454] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 11/19/2022] Open
Abstract
The human thymus is the site of T-cell maturation and induction of central tolerance. Hematopoietic stem cell (HSC)-derived progenitors are recruited to the thymus from the fetal liver during early prenatal development and from bone marrow at later stages and postnatal life. The mechanism by which HSCs are recruited to the thymus is poorly understood in humans, though mouse models have indicated the critical role of thymic stromal cells (TSC). Here, we developed a 3D microfluidic assay based on human cells to model HSC extravasation across the endothelium into the extracellular matrix. We found that the presence of human TSC consisting of cultured thymic epithelial cells (TEC) and interstitial cells (TIC) increases the HSC extravasation rates by 3-fold. Strikingly, incorporating TEC or TIC alone is insufficient to perturb HSC extravasation rates. Furthermore, we identified complex gene expressions from interactions between endothelial cells, TEC and TIC modulates the HSCs extravasation. Our results suggest that comprehensive signaling from the complex thymic microenvironment is crucial for thymus seeding and that our system will allow manipulation of these signals with the potential to increase thymocyte migration in a therapeutic setting.
Collapse
Affiliation(s)
- Sara A. Watson
- Department of Mechanical EngineeringUCLLondonUK
- Epithelial Stem Cell Biology and Regenerative Medicine LabThe Francis Crick InstituteLondonUK
| | | | - Luca Zanieri
- Epithelial Stem Cell Biology and Regenerative Medicine LabThe Francis Crick InstituteLondonUK
- Institute of Immunity and TransplantationDivision of Infection & Immunity, UCLLondonUK
| | | | - Roberta Ragazzini
- Epithelial Stem Cell Biology and Regenerative Medicine LabThe Francis Crick InstituteLondonUK
- Institute of Immunity and TransplantationDivision of Infection & Immunity, UCLLondonUK
| | - Paola Bonfanti
- Epithelial Stem Cell Biology and Regenerative Medicine LabThe Francis Crick InstituteLondonUK
- Institute of Immunity and TransplantationDivision of Infection & Immunity, UCLLondonUK
| | | |
Collapse
|
14
|
Caveolin-1 is a primary determinant of endothelial stiffening associated with dyslipidemia, disturbed flow, and ageing. Sci Rep 2022; 12:17822. [PMID: 36280774 PMCID: PMC9592578 DOI: 10.1038/s41598-022-20713-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 09/16/2022] [Indexed: 01/20/2023] Open
Abstract
Endothelial stiffness is emerging as a major determinant in endothelial function. Here, we analyzed the role of caveolin-1 (Cav-1) in determining the stiffness of endothelial cells (EC) exposed to oxidized low density lipoprotein (oxLDL) under static and hemodynamic conditions in vitro and of aortic endothelium in vivo in mouse models of dyslipidemia and ageing. Elastic moduli of cultured ECs and of the endothelial monolayer of freshly isolated mouse aortas were measured using atomic force microscopy (AFM). We found that a loss of Cav-1 abrogates the uptake of oxLDL and oxLDL-induced endothelial stiffening, as well as endothelial stiffening induced by disturbed flow (DF), which was also oxLDL dependent. Mechanistically, Cav-1 is required for the expression of CD36 (cluster of differentiation 36) scavenger receptor. Genetic deletion of Cav-1 abrogated endothelial stiffening observed in the DF region of the aortic arch, and induced by a high fat diet (4-6 weeks) and significantly blunted endothelial stiffening that develops with advanced age. This effect was independent of stiffening of the sub-endothelium layer. Additionally, Cav-1 expression significantly increased with age. No differences in elastic modulus were observed between the sexes in advanced aged wild type and Cav-1 knockout mice. Taken together, this study demonstrates that Cav-1 plays a critical role in endothelial stiffening induced by oxLDL in vitro and by dyslipidemia, disturbed flow and ageing in vivo.
Collapse
|
15
|
Banerjee S, Nara R, Chakraborty S, Chowdhury D, Haldar S. Integrin Regulated Autoimmune Disorders: Understanding the Role of Mechanical Force in Autoimmunity. Front Cell Dev Biol 2022; 10:852878. [PMID: 35372360 PMCID: PMC8971850 DOI: 10.3389/fcell.2022.852878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
The pathophysiology of autoimmune disorders is multifactorial, where immune cell migration, adhesion, and lymphocyte activation play crucial roles in its progression. These immune processes are majorly regulated by adhesion molecules at cell–extracellular matrix (ECM) and cell–cell junctions. Integrin, a transmembrane focal adhesion protein, plays an indispensable role in these immune cell mechanisms. Notably, integrin is regulated by mechanical force and exhibit bidirectional force transmission from both the ECM and cytosol, regulating the immune processes. Recently, integrin mechanosensitivity has been reported in different immune cell processes; however, the underlying mechanics of these integrin-mediated mechanical processes in autoimmunity still remains elusive. In this review, we have discussed how integrin-mediated mechanotransduction could be a linchpin factor in the causation and progression of autoimmune disorders. We have provided an insight into how tissue stiffness exhibits a positive correlation with the autoimmune diseases’ prevalence. This provides a plausible connection between mechanical load and autoimmunity. Overall, gaining insight into the role of mechanical force in diverse immune cell processes and their dysregulation during autoimmune disorders will open a new horizon to understand this physiological anomaly.
Collapse
|
16
|
Kamnev A, Lacouture C, Fusaro M, Dupré L. Molecular Tuning of Actin Dynamics in Leukocyte Migration as Revealed by Immune-Related Actinopathies. Front Immunol 2021; 12:750537. [PMID: 34867982 PMCID: PMC8634686 DOI: 10.3389/fimmu.2021.750537] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/12/2021] [Indexed: 01/13/2023] Open
Abstract
Motility is a crucial activity of immune cells allowing them to patrol tissues as they differentiate, sample or exchange information, and execute their effector functions. Although all immune cells are highly migratory, each subset is endowed with very distinct motility patterns in accordance with functional specification. Furthermore individual immune cell subsets adapt their motility behaviour to the surrounding tissue environment. This review focuses on how the generation and adaptation of diversified motility patterns in immune cells is sustained by actin cytoskeleton dynamics. In particular, we review the knowledge gained through the study of inborn errors of immunity (IEI) related to actin defects. Such pathologies are unique models that help us to uncover the contribution of individual actin regulators to the migration of immune cells in the context of their development and function.
Collapse
Affiliation(s)
- Anton Kamnev
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.,Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Claire Lacouture
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM, CNRS, Toulouse III Paul Sabatier University, Toulouse, France.,Laboratoire De Physique Théorique, IRSAMC, Université De Toulouse (UPS), CNRS, Toulouse, France
| | - Mathieu Fusaro
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM, CNRS, Toulouse III Paul Sabatier University, Toulouse, France
| | - Loïc Dupré
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.,Department of Dermatology, Medical University of Vienna, Vienna, Austria.,Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM, CNRS, Toulouse III Paul Sabatier University, Toulouse, France
| |
Collapse
|
17
|
CCL4 induces inflammatory signalling and barrier disruption in the neurovascular endothelium. Brain Behav Immun Health 2021; 18:100370. [PMID: 34755124 PMCID: PMC8560974 DOI: 10.1016/j.bbih.2021.100370] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 12/27/2022] Open
Abstract
Background During neuroinflammation many chemokines alter the function of the blood-brain barrier (BBB) that regulates the entry of macromolecules and immune cells into the brain. As the milieu of the brain is altered, biochemical and structural changes contribute to the pathogenesis of neuroinflammation and may impact on neurogenesis. The chemokine CCL4, previously known as MIP-1β, is upregulated in a wide variety of central nervous system disorders, including multiple sclerosis, where it is thought to play a key role in the neuroinflammatory process. However, the effect of CCL4 on BBB endothelial cells (ECs) is unknown. Materials and methods Expression and distribution of CCR5, phosphorylated p38, F-actin, zonula occludens-1 (ZO-1) and vascular endothelial cadherin (VE-cadherin) were analysed in the human BBB EC line hCMEC/D3 by Western blot and/or immunofluorescence in the presence and absence of CCL4. Barrier modulation in response to CCL4 using hCMEC/D3 monolayers was assessed by measuring molecular flux of 70 kDa RITC-dextran and transendothelial lymphocyte migration. Permeability changes in response to CCL4 in vivo were measured by an occlusion technique in pial microvessels of Wistar rats and by fluorescein angiography in mouse retinae. Results CCR5, the receptor for CCL4, was expressed in hCMEC/D3 cells. CCL4 stimulation led to phosphorylation of p38 and the formation of actin stress fibres, both indicative of intracellular chemokine signalling. The distribution of junctional proteins was also altered in response to CCL4: junctional ZO-1 was reduced by circa 60% within 60 min. In addition, surface VE-cadherin was redistributed through internalisation. Consistent with these changes, CCL4 induced hyperpermeability in vitro and in vivo and increased transmigration of lymphocytes across monolayers of hCMEC/D3 cells. Conclusion These results show that CCL4 can modify BBB function and may contribute to disease pathogenesis. The chemokine CCL4 induced phosphorylation of P38 in an in vitro model of the blood-brain barrier (BBB). CCL4 treatment resulted in reduction of plasma membrane VE-cadherin and junctional ZO-1. CCL4 induced neurovascular barrier breakdown in vitro and in vivo.
Collapse
|
18
|
Rodenburg WS, van Buul JD. Rho GTPase signalling networks in cancer cell transendothelial migration. VASCULAR BIOLOGY 2021; 3:R77-R95. [PMID: 34738075 PMCID: PMC8558887 DOI: 10.1530/vb-21-0008] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 01/21/2023]
Abstract
Rho GTPases are small signalling G-proteins that are central regulators of cytoskeleton dynamics, and thereby regulate many cellular processes, including the shape, adhesion and migration of cells. As such, Rho GTPases are also essential for the invasive behaviour of cancer cells, and thus involved in several steps of the metastatic cascade, including the extravasation of cancer cells. Extravasation, the process by which cancer cells leave the circulation by transmigrating through the endothelium that lines capillary walls, is an essential step for metastasis towards distant organs. During extravasation, Rho GTPase signalling networks not only regulate the transmigration of cancer cells but also regulate the interactions between cancer and endothelial cells and are involved in the disruption of the endothelial barrier function, ultimately allowing cancer cells to extravasate into the underlying tissue and potentially form metastases. Thus, targeting Rho GTPase signalling networks in cancer may be an effective approach to inhibit extravasation and metastasis. In this review, the complex process of cancer cell extravasation will be discussed in detail. Additionally, the roles and regulation of Rho GTPase signalling networks during cancer cell extravasation will be discussed, both from a cancer cell and endothelial cell point of view.
Collapse
Affiliation(s)
- Wessel S Rodenburg
- Molecular Cell Biology Lab at Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Jaap D van Buul
- Molecular Cell Biology Lab at Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands.,Leeuwenhoek Centre for Advanced Microscopy, Section Molecular Cytology at Swammerdam Institute for Life Sciences at University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
19
|
Arts JJG, Mahlandt EK, Grönloh MLB, Schimmel L, Noordstra I, Gordon E, van Steen ACI, Tol S, Walzog B, van Rijssel J, Nolte MA, Postma M, Khuon S, Heddleston JM, Wait E, Chew TL, Winter M, Montanez E, Goedhart J, van Buul JD. Endothelial junctional membrane protrusions serve as hotspots for neutrophil transmigration. eLife 2021; 10:66074. [PMID: 34431475 PMCID: PMC8437435 DOI: 10.7554/elife.66074] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 08/22/2021] [Indexed: 12/27/2022] Open
Abstract
Upon inflammation, leukocytes rapidly transmigrate across the endothelium to enter the inflamed tissue. Evidence accumulates that leukocytes use preferred exit sites, alhough it is not yet clear how these hotspots in the endothelium are defined and how they are recognized by the leukocyte. Using lattice light sheet microscopy, we discovered that leukocytes prefer endothelial membrane protrusions at cell junctions for transmigration. Phenotypically, these junctional membrane protrusions are present in an asymmetric manner, meaning that one endothelial cell shows the protrusion and the adjacent one does not. Consequently, leukocytes cross the junction by migrating underneath the protruding endothelial cell. These protrusions depend on Rac1 activity and by using a photo-activatable Rac1 probe, we could artificially generate local exit-sites for leukocytes. Overall, we have discovered a new mechanism that uses local induced junctional membrane protrusions to facilitate/steer the leukocyte escape/exit from inflamed vessel walls.
Collapse
Affiliation(s)
- Janine JG Arts
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of AmsterdamAmsterdamNetherlands
| | - Eike K Mahlandt
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of AmsterdamAmsterdamNetherlands
| | - Max LB Grönloh
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of AmsterdamAmsterdamNetherlands
| | - Lilian Schimmel
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of AmsterdamAmsterdamNetherlands
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of QueenslandBrisbaneQLDAustralia
| | - Ivar Noordstra
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of QueenslandBrisbaneQLDAustralia
| | - Emma Gordon
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of QueenslandBrisbaneQLDAustralia
| | - Abraham CI van Steen
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
| | - Simon Tol
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
| | - Barbara Walzog
- Department of Cardiovascular Physiology and Pathophysiology, Walter Brendel Center of Experimental Medicine, Biomedical Center, Ludwig-Maximilians-Universität MünchenPlanegg-MartinsriedGermany
| | - Jos van Rijssel
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
| | - Martijn A Nolte
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
| | - Marten Postma
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of AmsterdamAmsterdamNetherlands
| | - Satya Khuon
- Advanced Imaging Center at Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - John M Heddleston
- Advanced Imaging Center at Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
- Microscopy Facility at the Cleveland Clinic Florida Research and Innovation CenterPort St. LucieUnited States
| | - Eric Wait
- Advanced Imaging Center at Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Teng Leong Chew
- Advanced Imaging Center at Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Mark Winter
- Zuckerman Postdoctoral Fellow, Department of Marine Sciences, University of HaifaHaifaIsrael
| | - Eloi Montanez
- Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of BarcelonaBarcelonaSpain
| | - Joachim Goedhart
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of AmsterdamAmsterdamNetherlands
| | - Jaap D van Buul
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of AmsterdamAmsterdamNetherlands
| |
Collapse
|
20
|
Khan IM, Ulrich BJ, Nelson AS, Sehra S, Kansas GS, Kaplan MH. Selectin Dependence of Allergic Skin Inflammation Is Diminished by Maternal Atopy. Immunohorizons 2021; 5:703-710. [PMID: 34433625 PMCID: PMC8638165 DOI: 10.4049/immunohorizons.2100052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/04/2021] [Indexed: 11/19/2022] Open
Abstract
Allergic skin inflammation requires the influx of inflammatory cells into the skin. Extravasation of leukocytes into the skin requires interactions between endothelial selectins and their glycan ligands on the surface of leukocytes. Selectin-ligand formation requires the activity of several glycosyltransferases, including Fut7 In this report, we tested the importance of Fut7 for the development of allergic skin inflammation in the Stat6VT transgenic mouse model. We observed that Fut7 deficiency was protective but did not eliminate disease. Segregation of the data by gender of the parent that transmitted the Stat6VT transgene, but not by gender of the pups, which were analyzed for disease, revealed that the protective effects of Fut7 deficiency were significantly greater when dams were Stat6VT negative. In contrast, in mice from litters of Stat6VT+ dams, Fut7 deficiency resulted in only modest protection. These findings indicate that pups from atopic dams exhibit a greater propensity for allergic disease, similar to observations in humans, and that the effect of maternal atopy is due to enhanced selectin-independent mechanisms of leukocyte recruitment in their offspring. Together, these results demonstrate that Fut7 deficiency can be protective in a model of atopic dermatitis but that maternal atopy diminishes these protective effects, suggesting alternative pathways for leukocyte recruitment in the absence of Fut7 enzyme activity. These observations have implications for understanding how the environment in utero predisposes for the development of allergic disease.
Collapse
Affiliation(s)
- Ibrahim M Khan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| | - Benjamin J Ulrich
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| | - Andrew S Nelson
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN; and
| | - Sarita Sehra
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN; and
| | - Geoffrey S Kansas
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Mark H Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN;
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN; and
| |
Collapse
|
21
|
Huang D, Ding Q, Chen S, Lü S, Zhang Y, Long M. E-selectin negatively regulates polymorphonuclear neutrophil transmigration through altered endothelial junction integrity. FASEB J 2021; 35:e21521. [PMID: 33811691 DOI: 10.1096/fj.202000662rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 01/28/2021] [Accepted: 02/26/2021] [Indexed: 11/11/2022]
Abstract
Transendothelial migration (TEM) of neutrophils under blood flow is critical in the inflammatory cascade. However, the role of endothelial plasticity in this process is not fully understood. Therefore, we used an in vitro model to test the dynamics of human polymorphonuclear neutrophil (PMN) TEM across lipopolysaccharide-treated human umbilical vein endothelial cell (HUVEC) monolayers. Interestingly, shRNA-E-selectin knockdown in HUVECs destabilized endothelial junctional integrity by reducing actin branching and increasing stress fiber at cell-cell junctions. This process is accomplished by downregulating the activation of cortactin and Arp2/3, which in turn alters the adhesive function of VE-cadherin, enhancing PMN transmigration. Meanwhile, redundant P-selectins possess overlapping functions in E-selectin-mediated neutrophil adhesion, and transmigration. These results demonstrate, to our knowledge, for the first time, that E-selectins negatively regulate neutrophil transmigration through alterations in endothelial plasticity. Furthermore, it improves our understanding of the mechanisms underlying actin remodeling, and junctional integrity, in endothelial cells mediating leukocyte TEM.
Collapse
Affiliation(s)
- Dandan Huang
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Qihan Ding
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Shenbao Chen
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Shouqin Lü
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yan Zhang
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Mian Long
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory) and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China.,Lead Contact, Beijing, China
| |
Collapse
|
22
|
Arefi SMA, Yang CWT, Sin DD, Feng JJ. A mechanical test of the tenertaxis hypothesis for leukocyte diapedesis. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2021; 44:93. [PMID: 34236552 PMCID: PMC8264968 DOI: 10.1140/epje/s10189-021-00096-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/21/2021] [Indexed: 06/13/2023]
Abstract
As part of the immune response, leukocytes can directly transmigrate through the body of endothelial cells or through the gap between adjacent endothelial cells. These are known, respectively, as the transcellular and paracellular route of diapedesis. What determines the usage of one route over the other is unclear. A recently proposed tenertaxis hypothesis claims that leukocytes choose the path with less mechanical resistance against leukocyte protrusions. We examined this hypothesis using numerical simulation of the mechanical resistance during paracellular and transcellular protrusions. By using parameters based on human lung endothelium, our results show that the required force to breach the endothelium through the transcellular route is greater than paracellular route, in agreement with experiments. Moreover, experiments have demonstrated that manipulation of the relative strength between the two routes can make the transcellular route preferable. Our simulations have demonstrated this reversal and thus tentatively confirmed the hypothesis of tenertaxis.
Collapse
Affiliation(s)
- S M Amin Arefi
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Cheng Wei Tony Yang
- Centre for Heart Lung Innovation, St Paul's Hospital and University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Don D Sin
- Centre for Heart Lung Innovation, St Paul's Hospital and University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - James J Feng
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
- Department of Mathematics, University of British Columbia, Vancouver, BC, V6T 1Z2, Canada.
| |
Collapse
|
23
|
Certo M, Elkafrawy H, Pucino V, Cucchi D, Cheung KC, Mauro C. Endothelial cell and T-cell crosstalk: Targeting metabolism as a therapeutic approach in chronic inflammation. Br J Pharmacol 2021; 178:2041-2059. [PMID: 31999357 PMCID: PMC8246814 DOI: 10.1111/bph.15002] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/09/2020] [Accepted: 01/15/2020] [Indexed: 12/14/2022] Open
Abstract
The role of metabolic reprogramming in the coordination of the immune response has gained increasing consideration in recent years. Indeed, it has become clear that changes in the metabolic status of immune cells can alter their functional properties. During inflammation, T cells need to generate sufficient energy and biomolecules to support growth, proliferation, and effector functions. Therefore, T cells need to rearrange their metabolism to meet these demands. A similar metabolic reprogramming has been described in endothelial cells, which have the ability to interact with and modulate the function of immune cells. In this overview, we will discuss recent insights in the complex crosstalk between endothelial cells and T cells as well as their metabolic reprogramming following activation. We highlight key components of this metabolic switch that can lead to the development of new therapeutics against chronic inflammatory disorders. LINKED ARTICLES: This article is part of a themed issue on Cellular metabolism and diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.10/issuetoc.
Collapse
Affiliation(s)
- Michelangelo Certo
- Institute of Inflammation and Ageing, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Hagar Elkafrawy
- Medical Biochemistry and Molecular Biology Department, Faculty of MedicineAlexandria UniversityAlexandriaEgypt
| | - Valentina Pucino
- Institute of Inflammation and Ageing, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Danilo Cucchi
- Barts Cancer InstituteQueen Mary University of LondonLondonUK
| | - Kenneth C.P. Cheung
- School of Life SciencesThe Chinese University of Hong KongHong Kong SARChina
| | - Claudio Mauro
- Institute of Inflammation and Ageing, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
- Institute of Cardiovascular Sciences, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
- Institute of Metabolism and Systems Research, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| |
Collapse
|
24
|
Schwartz AB, Campos OA, Criado-Hidalgo E, Chien S, del Álamo JC, Lasheras JC, Yeh YT. Elucidating the Biomechanics of Leukocyte Transendothelial Migration by Quantitative Imaging. Front Cell Dev Biol 2021; 9:635263. [PMID: 33855018 PMCID: PMC8039384 DOI: 10.3389/fcell.2021.635263] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/09/2021] [Indexed: 01/13/2023] Open
Abstract
Leukocyte transendothelial migration is crucial for innate immunity and inflammation. Upon tissue damage or infection, leukocytes exit blood vessels by adhering to and probing vascular endothelial cells (VECs), breaching endothelial cell-cell junctions, and transmigrating across the endothelium. Transendothelial migration is a critical rate-limiting step in this process. Thus, leukocytes must quickly identify the most efficient route through VEC monolayers to facilitate a prompt innate immune response. Biomechanics play a decisive role in transendothelial migration, which involves intimate physical contact and force transmission between the leukocytes and the VECs. While quantifying these forces is still challenging, recent advances in imaging, microfabrication, and computation now make it possible to study how cellular forces regulate VEC monolayer integrity, enable efficient pathfinding, and drive leukocyte transmigration. Here we review these recent advances, paying particular attention to leukocyte adhesion to the VEC monolayer, leukocyte probing of endothelial barrier gaps, and transmigration itself. To offer a practical perspective, we will discuss the current views on how biomechanics govern these processes and the force microscopy technologies that have enabled their quantitative analysis, thus contributing to an improved understanding of leukocyte migration in inflammatory diseases.
Collapse
Affiliation(s)
- Amy B. Schwartz
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
| | - Obed A. Campos
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
| | - Ernesto Criado-Hidalgo
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Juan C. del Álamo
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
- Department of Mechanical Engineering, University of Washington, Seattle, WA, United States
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, United States
| | - Juan C. Lasheras
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Yi-Ting Yeh
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
25
|
Rademakers T, Goedhart M, Hoogenboezem M, Ponce AG, van Rijssel J, Samus M, Schnoor M, Butz S, Huveneers S, Vestweber D, Nolte MA, Voermans C, van Buul JD. Hematopoietic stem and progenitor cells use podosomes to transcellularly cross the bone marrow endothelium. Haematologica 2020; 105:2746-2756. [PMID: 33256374 PMCID: PMC7716366 DOI: 10.3324/haematol.2018.196329] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 01/20/2020] [Indexed: 11/30/2022] Open
Abstract
Bone marrow endothelium plays an important role in the homing of hematopoietic stem and progenitor cells upon transplantation, but surprisingly little is known on how the bone marrow endothelial cells regulate local permeability and hematopoietic stem and progenitor cells transmigration. We show that temporal loss of vascular endothelial-cadherin function promotes vascular permeability in BM, even upon low-dose irradiation. Loss of vascular endothelial-cadherin function also enhances homing of transplanted hematopoietic stem and progenitor cells to the bone marrow of irradiated mice although engraftment is not increased. Intriguingly, stabilizing junctional vascular endothelial-cadherin in vivo reduced bone marrow permeability, but did not prevent hematopoietic stem and progenitor cells migration into the bone marrow, suggesting that hematopoietic stem and progenitor cells use the transcellular migration route to enter the bone marrow. Indeed, using an in vitro migration assay, we show that human hematopoietic stem and progenitor cells predominantly cross bone marrow endothelium in a transcellular manner in homeostasis by inducing podosome-like structures. Taken together, vascular endothelial-cadherin is crucial for BM vascular homeostasis but dispensable for the homing of hematopoietic stem and progenitor cells. These findings are important in the development of potential therapeutic targets to improve hematopoietic stem and progenitor cell homing strategies.
Collapse
Affiliation(s)
- Timo Rademakers
- Department of Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Amsterdam, the Netherlands
| | - Marieke Goedhart
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Amsterdam, the Netherlands
| | - Mark Hoogenboezem
- Department of Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Amsterdam, the Netherlands
| | - Alexander García Ponce
- Department of Molecular Biomedicine, Center of Research and Advanced Studies (CINVESTAV-IPN), Mexico-City, Mexico
| | - Jos van Rijssel
- Department of Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Amsterdam, the Netherlands
| | - Maryna Samus
- Max Planck Institute for Molecular Biomedicine, Munster, Germany
| | - Michael Schnoor
- Department of Molecular Biomedicine, Center of Research and Advanced Studies (CINVESTAV-IPN), Mexico-City, Mexico
| | - Stefan Butz
- Max Planck Institute for Molecular Biomedicine, Munster, Germany
| | - Stephan Huveneers
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, the Netherlands
| | | | - Martijn A. Nolte
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Amsterdam, the Netherlands
| | - Carlijn Voermans
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Amsterdam, the Netherlands
| | - Jaap D. van Buul
- Department of Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
26
|
Kalafati L, Chavakis T. Hematopoietic stem and progenitor cells take the route through the bone marrow endothelium. Haematologica 2020; 105:2700-2701. [PMID: 33256368 PMCID: PMC7716267 DOI: 10.3324/haematol.2020.262113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Lydia Kalafati
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden; National Center for Tumor Diseases, Partner Site Dresden, Dresden and German Cancer Research Center Heidelberg, Heidelberg, Germany
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden.
| |
Collapse
|
27
|
Sorokin V, Vickneson K, Kofidis T, Woo CC, Lin XY, Foo R, Shanahan CM. Role of Vascular Smooth Muscle Cell Plasticity and Interactions in Vessel Wall Inflammation. Front Immunol 2020; 11:599415. [PMID: 33324416 PMCID: PMC7726011 DOI: 10.3389/fimmu.2020.599415] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/27/2020] [Indexed: 12/25/2022] Open
Abstract
The pathobiology of atherosclerotic disease requires further elucidation to discover new approaches to address its high morbidity and mortality. To date, over 17 million cardiovascular-related deaths have been reported annually, despite a multitude of surgical and nonsurgical interventions and advances in medical therapy. Existing strategies to prevent disease progression mainly focus on management of risk factors, such as hypercholesterolemia. Even with optimum current medical therapy, recurrent cardiovascular events are not uncommon in patients with atherosclerosis, and their incidence can reach 10–15% per year. Although treatments targeting inflammation are under investigation and continue to evolve, clinical breakthroughs are possible only if we deepen our understanding of vessel wall pathobiology. Vascular smooth muscle cells (VSMCs) are one of the most abundant cells in vessel walls and have emerged as key players in disease progression. New technologies, including in situ hybridization proximity ligation assays, in vivo cell fate tracing with the CreERT2-loxP system and single-cell sequencing technology with spatial resolution, broaden our understanding of the complex biology of these intriguing cells. Our knowledge of contractile and synthetic VSMC phenotype switching has expanded to include macrophage-like and even osteoblast-like VSMC phenotypes. An increasing body of data suggests that VSMCs have remarkable plasticity and play a key role in cell-to-cell crosstalk with endothelial cells and immune cells during the complex process of inflammation. These are cells that sense, interact with and influence the behavior of other cellular components of the vessel wall. It is now more obvious that VSMC plasticity and the ability to perform nonprofessional phagocytic functions are key phenomena maintaining the inflammatory state and senescent condition and actively interacting with different immune competent cells.
Collapse
Affiliation(s)
- Vitaly Sorokin
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Cardiac, Thoracic and Vascular Surgery, National University Hospital, National University Health System, Singapore, Singapore
| | - Keeran Vickneson
- School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Theo Kofidis
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Cardiac, Thoracic and Vascular Surgery, National University Hospital, National University Health System, Singapore, Singapore
| | - Chin Cheng Woo
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xiao Yun Lin
- Department of Cardiac, Thoracic and Vascular Surgery, National University Hospital, National University Health System, Singapore, Singapore
| | - Roger Foo
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Genome Institute of Singapore, ASTAR, Singapore, Singapore
| | - Catherine M Shanahan
- School of Cardiovascular Medicine and Sciences, James Black Centre, King's College London, London, United Kingdom
| |
Collapse
|
28
|
Duru G, van Egmond M, Heemskerk N. A Window of Opportunity: Targeting Cancer Endothelium to Enhance Immunotherapy. Front Immunol 2020; 11:584723. [PMID: 33262763 PMCID: PMC7686513 DOI: 10.3389/fimmu.2020.584723] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/19/2020] [Indexed: 12/19/2022] Open
Abstract
Vascular abnormalities in tumors have a major impact on the immune microenvironment in tumors. The consequences of abnormal vasculature include increased hypoxia, acidosis, high intra-tumoral fluid pressure, and angiogenesis. This introduces an immunosuppressive microenvironment that alters immune cell maturation, activation, and trafficking, which supports tumor immune evasion and dissemination of tumor cells. Increasing data suggests that cancer endothelium is a major barrier for traveling leukocytes, ranging from a partial blockade resulting in a selective endothelial barrier, to a complete immune infiltration blockade associated with immune exclusion and immune desert cancer phenotypes. Failed immune cell trafficking as well as immunosuppression within the tumor microenvironment limits the efficacy of immunotherapeutic approaches. As such, targeting proteins with key roles in angiogenesis may potentially reduce immunosuppression and might restore infiltration of anti-tumor immune cells, creating a therapeutic window for successful immunotherapy. In this review, we provide a comprehensive overview of established as well as more controversial endothelial pathways that govern selective immune cell trafficking across cancer endothelium. Additionally, we discuss recent insights and strategies that target tumor vasculature in order to increase infiltration of cytotoxic immune cells during the therapeutic window of vascular normalization hereby improving the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Gizem Duru
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection & Immunity, Amsterdam, Netherlands
| | - Marjolein van Egmond
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection & Immunity, Amsterdam, Netherlands
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Surgery, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Niels Heemskerk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection & Immunity, Amsterdam, Netherlands
| |
Collapse
|
29
|
Principles of Leukocyte Migration Strategies. Trends Cell Biol 2020; 30:818-832. [DOI: 10.1016/j.tcb.2020.06.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 12/14/2022]
|
30
|
Zhao Y, Li J, Ting KK, Chen J, Coleman P, Liu K, Wan L, Moller T, Vadas MA, Gamble JR. The VE-Cadherin/β-catenin signalling axis regulates immune cell infiltration into tumours. Cancer Lett 2020; 496:1-15. [PMID: 32991950 DOI: 10.1016/j.canlet.2020.09.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/14/2020] [Accepted: 09/24/2020] [Indexed: 12/20/2022]
Abstract
Vascular normalisation, the process that reverses the structural and functional abnormalities seen in tumour-associated vessels, is also accompanied by changes in leucocyte trafficking. Our previous studies have shown the normalisation effects of the agent CD5-2 which acts to stabilise VE-Cadherin leading to increased penetration of CD8+ T cells but decreased infiltration of neutrophils (CD11b+Gr1hi) into tumour parenchyma. In the present study, we demonstrate that VE-Cadherin stabilisation through CD5-2 treatment of purified endothelial cells (ECs) results in a similar leucocyte-selective regulation of transmigration, suggesting the existence of an endothelial specific intrinsic mechanism. Further, we show by RNA sequencing (RNA-seq)-based transcriptomic analysis, that treatment of ECs with CD5-2 regulates chemokines known to be involved in leucocyte transmigration, including upregulation of CCL2 and CXCL10 that facilitate CD8+ T cell transmigration. Both in vitro and in vivo mechanistic studies revealed that the increased CCL2 expression was dependent on expression of VE-Cadherin and downstream activation of the AKT/GSK3β/β-catenin/TCF4 signalling pathway. CD5-2 treatment also contributed to the reorganisation of the cytoskeleton, inducing reorganisation of stress fibres to circumferential actin, which previously has been described as associated with the stabilisation of the endothelial barrier, and amplification of the transcellular migration of CD8+ T cells. Thus, we propose that promotion of endothelial junctional integrity during vascular normalisation not only inhibits vascular leak but also resets the endothelial dependent regulation of immune cell infiltration.
Collapse
Affiliation(s)
- Yang Zhao
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, 2050, Australia
| | - Jia Li
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, 2050, Australia
| | - Ka Ka Ting
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, 2050, Australia
| | - Jinbiao Chen
- Liver Injury and Cancer Program, Centenary Institute, The University of Sydney, Sydney, 2050, Australia
| | - Paul Coleman
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, 2050, Australia
| | - Ken Liu
- Liver Injury and Cancer Program, Centenary Institute, The University of Sydney, Sydney, 2050, Australia
| | - Li Wan
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, 2050, Australia
| | | | - Mathew A Vadas
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, 2050, Australia
| | - Jennifer R Gamble
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, 2050, Australia.
| |
Collapse
|
31
|
Le Master E, Ahn SJ, Levitan I. Mechanisms of endothelial stiffening in dyslipidemia and aging: Oxidized lipids and shear stress. CURRENT TOPICS IN MEMBRANES 2020; 86:185-215. [PMID: 33837693 PMCID: PMC8168803 DOI: 10.1016/bs.ctm.2020.08.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Vascular stiffening of the arterial walls is well-known as a key factor in aging and the development of cardiovascular disease; however, the role of endothelial stiffness in vascular dysfunction is still an emerging topic. In this review, the authors discuss the impact of dyslipidemia, oxidized lipids, substrate stiffness, age and pro-atherogenic disturbed flow have on endothelial stiffness. Furthermore, we investigate several mechanistic pathways that are key contributors in endothelial stiffness and discuss their physiological effects in the onset of atherogenesis in the disturbed flow regions of the aortic vasculature. The findings in this chapter describe a novel paradigm of synergistic interaction of plasma dyslipidemia/oxidized lipids and pro-atherogenic disturbed shear stress, as well as aging has on endothelial stiffness and vascular dysfunction.
Collapse
Affiliation(s)
- Elizabeth Le Master
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Sang Joon Ahn
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Irena Levitan
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Illinois at Chicago, Chicago, IL, United States.
| |
Collapse
|
32
|
VanderBurgh JA, Potharazu AV, Schwager SC, Reinhart-King CA. A discrete interface in matrix stiffness creates an oscillatory pattern of endothelial monolayer disruption. J Cell Sci 2020; 133:jcs244533. [PMID: 32878941 PMCID: PMC7520461 DOI: 10.1242/jcs.244533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 08/17/2020] [Indexed: 01/07/2023] Open
Abstract
Intimal stiffening upregulates endothelial cell contractility, disrupting barrier integrity; however, intimal stiffening is non-uniform. The impact of local changes in intimal stiffness on proximal and distal cell-cell interactions is unknown. To investigate the range at which matrix stiffness heterogeneities impact neighboring endothelial cells within a monolayer, we built a micropillar system with adjacent regions of stiff and compliant matrix. The stiffness interface results in an oscillatory pattern of neutrophil transendothelial migration, symmetrical about the interface and well-fit by a sinusoid function. 'Peaks' of the sinusoid were found to have increased cellular contractility and decreased barrier function relative to 'troughs' of the sinusoid. Pharmacological modulation of contractility was observed to break symmetry, altering the amplitude and wavelength of the sinusoid, indicating that contractility may regulate this effect. This work illuminates a novel biophysical phenomenon of the role of stiffness-mediated cell-matrix interactions on cell-cell interactions at a distance. Additionally, it provides insight into the range at which intimal matrix stiffness heterogeneities will impact endothelial barrier function and potentially contribute to atherogenesis.
Collapse
Affiliation(s)
- Jacob A VanderBurgh
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Archit V Potharazu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Samantha C Schwager
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Cynthia A Reinhart-King
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
33
|
Chaudhuri PK, Wang MS, Black CT, Huse M, Kam LC. Modulating T Cell Activation Using Depth Sensing Topographic Cues. ACTA ACUST UNITED AC 2020; 4:e2000143. [PMID: 32744809 DOI: 10.1002/adbi.202000143] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/16/2020] [Indexed: 12/11/2022]
Abstract
This report examines how sensing of substrate topography can be used to modulate T cell activation, a key coordinating step in the adaptive immune response. Inspired by the native T cell-antigen presenting cell interface, micrometer scale pits with varying depth are fabricated into planar substrates. Primary CD4+ T cells extend actin-rich protrusions into the micropits. T cell activation, reflected in secretion of cytokines interleukin-2 and interferon gamma, is sensitive to the micropit depth. Surprisingly, arrays of micropits with 4 μm depth enhance activation compared to flat substrates but deeper micropits are less effective at increasing cell response, revealing a biphasic dependence in activation as a function of feature dimensions. Inhibition of cell contractility abrogates the enhanced activation associated with the micropits. In conclusion, this report demonstrates that the 3D, microscale topography can be used to enhance T cell activation, an ability that most directly can be used to improve production of these cells for immunotherapy.
Collapse
Affiliation(s)
| | - Mitchell S Wang
- Pharmacology Graduate Program, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Charles T Black
- Center for Functional Nanomaterials, Brookhaven National Laboratory, Upton, NY, 11973, USA
| | - Morgan Huse
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Lance C Kam
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| |
Collapse
|
34
|
Roy NH, Mammadli M, Burkhardt JK, Karimi M. CrkL is required for donor T cell migration to GvHD target organs. Oncotarget 2020; 11:1505-1514. [PMID: 32391120 PMCID: PMC7197453 DOI: 10.18632/oncotarget.27509] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 02/17/2020] [Indexed: 01/21/2023] Open
Abstract
The success of cancer therapies based on allogeneic hematopoietic stem cell transplant relies on the ability to separate graft-versus-host disease (GvHD) from graft-versus-tumor (GVT) responses. Controlling donor T cell migration into peripheral tissues is a viable option to limit unwanted tissue damage, but a lack of specific targets limits progress on this front. Here, we show that the adaptor protein CrkL, but not the closely related family members CrkI or CrkII, is a crucial regulator of T cell migration. In vitro, CrkL-deficient T cells fail to polymerize actin in response to the integrin ligand ICAM-1, resulting in defective migration. Using a mouse model of GvHD/GVT, we found that while CrkL-deficient T cells can efficiently eliminate hematopoietic tumors they are unable to migrate into inflamed organs, such as the liver and small intestine, and thus do not cause GvHD. These results suggest a specific role for CrkL in trafficking to peripheral organs but not the lymphatic system. In line with this, we found that although CrkL-deficient T cells could clear hematopoietic tumors, they failed to clear the same tumor growing subcutaneously, highlighting the role of CrkL in controlling T cell migration into peripheral tissues. Our results define a unique role for CrkL in controlling T cell migration, and suggest that CrkL function could be therapeutically targeted to enhance the efficacy of immunotherapies involving allogeneic donor cells.
Collapse
Affiliation(s)
- Nathan H Roy
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mahinbanu Mammadli
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Janis K Burkhardt
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mobin Karimi
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
35
|
Abstract
AbstractAtomic force microscopy (AFM) has been widely used to acquire surface topography upon different scanning modes and to quantify mechanical properties of a cell using single-point ramp force mode. However, these traditional measurements need massive force curves originating from multiple points of a cell to exclude the potential errors resulted from limited and factitious selections of testing points, making the measurements time-consuming and highly localized. PeakForce Quantitative NanoMechanics (PF QNM) is a high-speed (faster by 3–4 order of magnitude) and global surface mechanical property mapping method with high spatial resolution, overcoming the drawbacks of traditional ramp mode especially used for a live cell with high heterogeneity. In this protocol, we elaborated how to run PF QNM measurements for live cells and relevant modification may be needed when extending this method to other cell-like soft materials.
Collapse
|
36
|
Wettschureck N, Strilic B, Offermanns S. Passing the Vascular Barrier: Endothelial Signaling Processes Controlling Extravasation. Physiol Rev 2019; 99:1467-1525. [PMID: 31140373 DOI: 10.1152/physrev.00037.2018] [Citation(s) in RCA: 195] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A central function of the vascular endothelium is to serve as a barrier between the blood and the surrounding tissue of the body. At the same time, solutes and cells have to pass the endothelium to leave or to enter the bloodstream to maintain homeostasis. Under pathological conditions, for example, inflammation, permeability for fluid and cells is largely increased in the affected area, thereby facilitating host defense. To appropriately function as a regulated permeability filter, the endothelium uses various mechanisms to allow solutes and cells to pass the endothelial layer. These include transcellular and paracellular pathways of which the latter requires remodeling of intercellular junctions for its regulation. This review provides an overview on endothelial barrier regulation and focuses on the endothelial signaling mechanisms controlling the opening and closing of paracellular pathways for solutes and cells such as leukocytes and metastasizing tumor cells.
Collapse
Affiliation(s)
- Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Boris Strilic
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| |
Collapse
|
37
|
Borjini N, Paouri E, Tognatta R, Akassoglou K, Davalos D. Imaging the dynamic interactions between immune cells and the neurovascular interface in the spinal cord. Exp Neurol 2019; 322:113046. [PMID: 31472115 DOI: 10.1016/j.expneurol.2019.113046] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/16/2019] [Accepted: 08/27/2019] [Indexed: 12/19/2022]
Abstract
Imaging the dynamic interactions between immune cells, glia, neurons and the vasculature in living rodents has revolutionized our understanding of physiological and pathological mechanisms of the CNS. Emerging microscopy and imaging technologies have enabled longitudinal tracking of structural and functional changes in a plethora of different cell types in the brain. The development of novel methods also allowed stable and longitudinal optical access to the spinal cord with minimum tissue perturbation. These important advances facilitated the application of in vivo imaging using two-photon microscopy for studies of the healthy, diseased, or injured spinal cord. Indeed, decoding the interactions between peripheral and resident cells with the spinal cord vasculature has shed new light on neuroimmune and vascular mechanisms regulating the onset and progression of neurological diseases. This review focuses on imaging studies of the interactions between the vasculature and peripheral immune cells or microglia, with emphasis on their contribution to neuroinflammation. We also discuss in vivo imaging studies highlighting the importance of neurovascular changes following spinal cord injury. Real-time imaging of blood-brain barrier (BBB) permeability and other vascular changes, perivascular glial responses, and immune cell entry has revealed unanticipated cellular mechanisms and novel molecular pathways that can be targeted to protect the injured or diseased CNS. Imaging the cell-cell interactions between the vasculature, immune cells, and neurons as they occur in real time, is a powerful tool both for testing the efficacy of existing therapeutic approaches, and for identifying new targets for limiting damage or enhancing the potential for repair of the affected spinal cord tissue.
Collapse
Affiliation(s)
- Nozha Borjini
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Evi Paouri
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Katerina Akassoglou
- Gladstone Institutes, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Dimitrios Davalos
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
38
|
Sleep disorders, nocturnal blood pressure, and cardiovascular risk: A translational perspective. Auton Neurosci 2019; 218:31-42. [DOI: 10.1016/j.autneu.2019.02.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 02/16/2019] [Accepted: 02/21/2019] [Indexed: 12/12/2022]
|
39
|
Escribano J, Chen MB, Moeendarbary E, Cao X, Shenoy V, Garcia-Aznar JM, Kamm RD, Spill F. Balance of mechanical forces drives endothelial gap formation and may facilitate cancer and immune-cell extravasation. PLoS Comput Biol 2019; 15:e1006395. [PMID: 31048903 PMCID: PMC6497229 DOI: 10.1371/journal.pcbi.1006395] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 12/10/2018] [Indexed: 11/29/2022] Open
Abstract
The formation of gaps in the endothelium is a crucial process underlying both cancer and immune cell extravasation, contributing to the functioning of the immune system during infection, the unfavorable development of chronic inflammation and tumor metastasis. Here, we present a stochastic-mechanical multiscale model of an endothelial cell monolayer and show that the dynamic nature of the endothelium leads to spontaneous gap formation, even without intervention from the transmigrating cells. These gaps preferentially appear at the vertices between three endothelial cells, as opposed to the border between two cells. We quantify the frequency and lifetime of these gaps, and validate our predictions experimentally. Interestingly, we find experimentally that cancer cells also preferentially extravasate at vertices, even when they first arrest on borders. This suggests that extravasating cells, rather than initially signaling to the endothelium, might exploit the autonomously forming gaps in the endothelium to initiate transmigration.
Collapse
Affiliation(s)
- Jorge Escribano
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Michelle B. Chen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Bioengineering, Stanford University, Stanford, California, United States of America
| | - Emad Moeendarbary
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Mechanical Engineering, University College London, London, United Kingdom
| | - Xuan Cao
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Vivek Shenoy
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | | | - Roger D. Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- BioSystems and Micromechanics (BioSyM), Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Fabian Spill
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- School of Mathematics, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
40
|
Neutrophil transendothelial migration: updates and new perspectives. Blood 2019; 133:2149-2158. [PMID: 30898863 DOI: 10.1182/blood-2018-12-844605] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 03/14/2019] [Indexed: 02/06/2023] Open
Abstract
Neutrophils represent the first line of cellular defense against invading microorganism by rapidly moving across the blood-endothelial cell (EC) barrier and exerting effector cell functions. The neutrophil recruitment cascade to inflamed tissues involves elements of neutrophil rolling, firm adhesion, and crawling onto the EC surface before extravasating by breaching the EC barrier. The interaction between neutrophils and ECs occurs via various adhesive modules and is a critical event determining the mode of neutrophil transmigration, either at the EC junction (paracellular) or directly through the EC body (transcellular). Once thought to be a homogenous entity, new evidence clearly points to the plasticity of neutrophil functions. This review will focus on recent advances in our understanding of the mechanism of the neutrophil transmigration process. It will discuss how neutrophil-EC interactions and the subsequent mode of diapedesis, junctional or nonjunctional, can be context dependent and how this plasticity may be exploited clinically.
Collapse
|
41
|
Mossu A, Rosito M, Khire T, Li Chung H, Nishihara H, Gruber I, Luke E, Dehouck L, Sallusto F, Gosselet F, McGrath JL, Engelhardt B. A silicon nanomembrane platform for the visualization of immune cell trafficking across the human blood-brain barrier under flow. J Cereb Blood Flow Metab 2019; 39:395-410. [PMID: 30565961 PMCID: PMC6421249 DOI: 10.1177/0271678x18820584] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Here we report on the development of a breakthrough microfluidic human in vitro cerebrovascular barrier (CVB) model featuring stem cell-derived brain-like endothelial cells (BLECs) and nanoporous silicon nitride (NPN) membranes (µSiM-CVB). The nanoscale thinness of NPN membranes combined with their high permeability and optical transparency makes them an ideal scaffold for the assembly of an in vitro microfluidic model of the blood-brain barrier (BBB) featuring cellular elements of the neurovascular unit (NVU). Dual-chamber devices divided by NPN membranes yield tight barrier properties in BLECs and allow an abluminal pericyte-co-culture to be replaced with pericyte-conditioned media. With the benefit of physiological flow and superior imaging quality, the µSiM-CVB platform captures each phase of the multi-step T-cell migration across the BBB in live cell imaging. The small volume of <100 µL of the µSiM-CVB will enable in vitro investigations of rare patient-derived immune cells with the human BBB. The µSiM-CVB is a breakthrough in vitro human BBB model to enable live and high-quality imaging of human immune cell interactions with the BBB under physiological flow. We expect it to become a valuable new tool for the study of cerebrovascular pathologies ranging from neuroinflammation to metastatic cancer.
Collapse
Affiliation(s)
- Adrien Mossu
- 1 Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Maria Rosito
- 1 Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Tejas Khire
- 2 Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Hung Li Chung
- 2 Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | | | - Isabelle Gruber
- 1 Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Emma Luke
- 2 Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Lucie Dehouck
- 3 Blood Brain Barrier Laboratory, University of Artois, Lens, France
| | - Federica Sallusto
- 4 Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,5 Institute for Microbiology, ETH Zurich, Zurich, Switzerland
| | - Fabien Gosselet
- 3 Blood Brain Barrier Laboratory, University of Artois, Lens, France
| | - James L McGrath
- 2 Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | | |
Collapse
|
42
|
Hörner M, Raute K, Hummel B, Madl J, Creusen G, Thomas OS, Christen EH, Hotz N, Gübeli RJ, Engesser R, Rebmann B, Lauer J, Rolauffs B, Timmer J, Schamel WWA, Pruszak J, Römer W, Zurbriggen MD, Friedrich C, Walther A, Minguet S, Sawarkar R, Weber W. Phytochrome-Based Extracellular Matrix with Reversibly Tunable Mechanical Properties. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1806727. [PMID: 30687975 DOI: 10.1002/adma.201806727] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/04/2019] [Indexed: 06/09/2023]
Abstract
Interrogation and control of cellular fate and function using optogenetics is providing revolutionary insights into biology. Optogenetic control of cells is achieved by coupling genetically encoded photoreceptors to cellular effectors and enables unprecedented spatiotemporal control of signaling processes. Here, a fast and reversibly switchable photoreceptor is used to tune the mechanical properties of polymer materials in a fully reversible, wavelength-specific, and dose- and space-controlled manner. By integrating engineered cyanobacterial phytochrome 1 into a poly(ethylene glycol) matrix, hydrogel materials responsive to light in the cell-compatible red/far-red spectrum are synthesized. These materials are applied to study in human mesenchymal stem cells how different mechanosignaling pathways respond to changing mechanical environments and to control the migration of primary immune cells in 3D. This optogenetics-inspired matrix allows fundamental questions of how cells react to dynamic mechanical environments to be addressed. Further, remote control of such matrices can create new opportunities for tissue engineering or provide a basis for optically stimulated drug depots.
Collapse
Affiliation(s)
- Maximilian Hörner
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104, Freiburg, Germany
| | - Katrin Raute
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104, Freiburg, Germany
| | - Barbara Hummel
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- Max Planck Institute of Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Josef Madl
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Guido Creusen
- Institute for Macromolecular Chemistry, FMF Freiburg Materials Research Center, University of Freiburg, 79104, Freiburg, Germany
- Freiburg Center for Interactive Materials and Bioinspired Technology (FIT), University of Freiburg, 79110, Freiburg, Germany
| | - Oliver S Thomas
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104, Freiburg, Germany
| | - Erik H Christen
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Natascha Hotz
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104, Freiburg, Germany
| | - Raphael J Gübeli
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104, Freiburg, Germany
| | - Raphael Engesser
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104, Freiburg, Germany
- Institute of Physics, University of Freiburg, 79104, Freiburg, Germany
| | - Balder Rebmann
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Jasmin Lauer
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- G.E.R.N. Tissue Replacement, Regeneration and Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center, Faculty of Medicine, University of Freiburg, 79085, Freiburg, Germany
| | - Bernd Rolauffs
- G.E.R.N. Tissue Replacement, Regeneration and Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center, Faculty of Medicine, University of Freiburg, 79085, Freiburg, Germany
| | - Jens Timmer
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104, Freiburg, Germany
- Institute of Physics, University of Freiburg, 79104, Freiburg, Germany
| | - Wolfgang W A Schamel
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, 79104, Freiburg, Germany
| | - Jan Pruszak
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104, Freiburg, Germany
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Winfried Römer
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104, Freiburg, Germany
- Freiburg Center for Interactive Materials and Bioinspired Technology (FIT), University of Freiburg, 79110, Freiburg, Germany
| | - Matias D Zurbriggen
- Institute of Synthetic Biology and CEPLAS, Heinrich Heine University Düsseldorf, 40204, Düsseldorf, Germany
| | - Christian Friedrich
- Institute for Macromolecular Chemistry, FMF Freiburg Materials Research Center, University of Freiburg, 79104, Freiburg, Germany
| | - Andreas Walther
- Institute for Macromolecular Chemistry, FMF Freiburg Materials Research Center, University of Freiburg, 79104, Freiburg, Germany
- Freiburg Center for Interactive Materials and Bioinspired Technology (FIT), University of Freiburg, 79110, Freiburg, Germany
- Cluster of Excellence Living, Adaptive and Energy-Autonomous Materials Systems (livMatS), University of Freiburg, 79110, Freiburg, Germany
| | - Susana Minguet
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, 79104, Freiburg, Germany
| | - Ritwick Sawarkar
- Max Planck Institute of Immunobiology and Epigenetics, 79108, Freiburg, Germany
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany
| | - Wilfried Weber
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104, Freiburg, Germany
| |
Collapse
|
43
|
Roy NH, MacKay JL, Robertson TF, Hammer DA, Burkhardt JK. Crk adaptor proteins mediate actin-dependent T cell migration and mechanosensing induced by the integrin LFA-1. Sci Signal 2018; 11:eaat3178. [PMID: 30538176 PMCID: PMC6333317 DOI: 10.1126/scisignal.aat3178] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
T cell entry into inflamed tissue involves firm adhesion, spreading, and migration of the T cells across endothelial barriers. These events depend on "outside-in" signals through which engaged integrins direct cytoskeletal reorganization. We investigated the molecular events that mediate this process and found that T cells from mice lacking expression of the adaptor protein Crk exhibited defects in phenotypes induced by the integrin lymphocyte function-associated antigen 1 (LFA-1), namely, actin polymerization, leading edge formation, and two-dimensional cell migration. Crk protein was an essential mediator of LFA-1 signaling-induced phosphorylation of the E3 ubiquitin ligase c-Cbl and its subsequent interaction with the phosphatidylinositol 3-kinase (PI3K) subunit p85, thus promoting PI3K activity and cytoskeletal remodeling. In addition, we found that Crk proteins were required for T cells to respond to changes in substrate stiffness, as measured by alterations in cell spreading and differential phosphorylation of the force-sensitive protein CasL. These findings identify Crk proteins as key intermediates coupling LFA-1 signals to actin remodeling and provide mechanistic insights into how T cells sense and respond to substrate stiffness.
Collapse
Affiliation(s)
- Nathan H Roy
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joanna L MacKay
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Tanner F Robertson
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel A Hammer
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Janis K Burkhardt
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA.
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
44
|
Ludewig P, Winneberger J, Magnus T. The cerebral endothelial cell as a key regulator of inflammatory processes in sterile inflammation. J Neuroimmunol 2018; 326:38-44. [PMID: 30472304 DOI: 10.1016/j.jneuroim.2018.10.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 09/17/2018] [Accepted: 10/24/2018] [Indexed: 11/18/2022]
Abstract
Cerebral endothelial cells accomplish numerous tasks connected to the maintenance of homeostasis of the central nervous system. They create a barrier between the central nervous system and peripheral blood and regulate mechanotransduction, vascular permeability, rheology, thrombogenesis, and leukocyte adhesion. In pathophysiological conditions (e.g., stroke or ischemia-reperfusion injury) the endothelial functions are impaired, leading to increased vascular permeability, vascular inflammation, leukocyte-endothelium interactions, and transendothelial migration, driving CNS inflammation and neuronal destruction. This review describes the current knowledge on the regulatory roles of endothelial cells in neuroinflammatory processes.
Collapse
Affiliation(s)
- Peter Ludewig
- Department of Neurology at the University Medical Center Hamburg- Eppendorf, Hamburg, Germany.
| | - Jack Winneberger
- Department of Neurology at the University Medical Center Hamburg- Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Department of Neurology at the University Medical Center Hamburg- Eppendorf, Hamburg, Germany
| |
Collapse
|
45
|
Jenkins E, Santos AM, O'Brien-Ball C, Felce JH, Wilcock MJ, Hatherley D, Dustin ML, Davis SJ, Eggeling C, Sezgin E. Reconstitution of immune cell interactions in free-standing membranes. J Cell Sci 2018; 132:jcs219709. [PMID: 30209137 PMCID: PMC6398472 DOI: 10.1242/jcs.219709] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 09/04/2018] [Indexed: 12/28/2022] Open
Abstract
The spatiotemporal regulation of signalling proteins at the contacts formed between immune cells and their targets determines how and when immune responses begin and end. Therapeutic control of immune responses therefore relies on thorough elucidation of the molecular processes occurring at these interfaces. However, the detailed investigation of each component's contribution to the formation and regulation of the contact is hampered by the complexities of cell composition and architecture. Moreover, the transient nature of these interactions creates additional challenges, especially in the use of advanced imaging technology. One approach that circumvents these problems is to establish in vitro systems that faithfully mimic immune cell interactions, but allow complexity to be 'dialled-in' as needed. Here, we present an in vitro system that makes use of synthetic vesicles that mimic important aspects of immune cell surfaces. Using this system, we began to explore the spatial distribution of signalling molecules (receptors, kinases and phosphatases) and how this changes during the initiation of signalling. The GUV/cell system presented here is expected to be widely applicable.
Collapse
Affiliation(s)
- Edward Jenkins
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Ana Mafalda Santos
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Caitlin O'Brien-Ball
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - James H Felce
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, UK
| | - Martin J Wilcock
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Deborah Hatherley
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, UK
| | - Simon J Davis
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Christian Eggeling
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
- Institute of Applied Optics Friedrich-Schiller-University Jena, Max-Wien Platz 4, 07743 Jena, Germany
- Leibniz Institute of Photonic Technology e.V., Albert-Einstein-Straße 9, 07745 Jena, Germany
| | - Erdinc Sezgin
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| |
Collapse
|
46
|
Roy NH, Burkhardt JK. The Actin Cytoskeleton: A Mechanical Intermediate for Signal Integration at the Immunological Synapse. Front Cell Dev Biol 2018; 6:116. [PMID: 30283780 PMCID: PMC6156151 DOI: 10.3389/fcell.2018.00116] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 08/27/2018] [Indexed: 12/22/2022] Open
Abstract
The immunological synapse (IS) is a specialized structure that serves as a platform for cell-cell communication between a T cell and an antigen-presenting cell (APC). Engagement of the T cell receptor (TCR) with cognate peptide-MHC complexes on the APC activates the T cell and instructs its differentiation. Proper T cell activation also requires engagement of additional receptor-ligand pairs, which promote sustained adhesion and deliver costimulatory signals. These events are orchestrated by T cell actin dynamics, which organize IS components and facilitate their signaling. The actin network flows from the edge of the cell inward, driving the centralization of TCR microclusters and providing the force to activate the integrin LFA-1. We recently showed that engagement of LFA-1 slows actin flow, and that this affects TCR signaling. This study highlights the physical nature of the IS, and contributes to a growing appreciation in the field that mechanosensing and mechanotransduction are essential for IS function. Additionally, it is becoming clear that there are multiple types of actin structures at the IS that promote signaling in distinct ways. How the different actin structures contribute to force production and mechanotransduction is just beginning to be explored. In this Perspective, we will feature recent work from our lab and others, that collectively points toward a model in which actin dynamics drive mechanical signaling and receptor crosstalk during T cell activation.
Collapse
Affiliation(s)
- Nathan H Roy
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, United States
| | - Janis K Burkhardt
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, United States.,Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
47
|
Caveolin1 Is Required for Th1 Cell Infiltration, but Not Tight Junction Remodeling, at the Blood-Brain Barrier in Autoimmune Neuroinflammation. Cell Rep 2018; 21:2104-2117. [PMID: 29166603 DOI: 10.1016/j.celrep.2017.10.094] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 09/16/2017] [Accepted: 10/25/2017] [Indexed: 01/24/2023] Open
Abstract
Lymphocytes cross vascular boundaries via either disrupted tight junctions (TJs) or caveolae to induce tissue inflammation. In the CNS, Th17 lymphocytes cross the blood-brain barrier (BBB) before Th1 cells; yet this differential crossing is poorly understood. We have used intravital two-photon imaging of the spinal cord in wild-type and caveolae-deficient mice with fluorescently labeled endothelial tight junctions to determine how tight junction remodeling and caveolae regulate CNS entry of lymphocytes during the experimental autoimmune encephalomyelitis (EAE) model for multiple sclerosis. We find that dynamic tight junction remodeling occurs early in EAE but does not depend upon caveolar transport. Moreover, Th1, but not Th17, lymphocytes are significantly reduced in the inflamed CNS of mice lacking caveolae. Therefore, tight junction remodeling facilitates Th17 migration across the BBB, whereas caveolae promote Th1 entry into the CNS. Moreover, therapies that target both tight junction degradation and caveolar transcytosis may limit lymphocyte infiltration during inflammation.
Collapse
|
48
|
Bharadwaj VN, Rowe RK, Harrison J, Wu C, Anderson TR, Lifshitz J, Adelson PD, Kodibagkar VD, Stabenfeldt SE. Blood-brainbarrier disruption dictates nanoparticle accumulation following experimental brain injury. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:2155-2166. [PMID: 29933022 DOI: 10.1016/j.nano.2018.06.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 05/09/2018] [Accepted: 06/11/2018] [Indexed: 01/03/2023]
Abstract
Clinically, traumatic brain injury (TBI) results in complex heterogeneous pathology that cannot be recapitulated in single pre-clinical animal model. Therefore, we focused on evaluating utility of nanoparticle (NP)-based therapeutics following three diffuse-TBI models: mildclosed-head injury (mCHI), repetitive-mCHI and midline-fluid percussion injury (FPI). We hypothesized that NP accumulation after diffuse TBI correlates directly with blood-brainbarrier permeability. Mice received PEGylated-NP cocktail (20-500 nm) (intravenously) after single- or repetitive-(1 impact/day, 5 consecutive days) CHI (immediately) and midline-FPI (1 h, 3 h and 6 h). NPs circulated for 1 h before perfusion/brain extraction. NP accumulation was analyzed using fluorescent microscopy in brain regions vulnerable to neuropathology. Minimal/no NP accumulation after mCHI/RmCHI was observed. In contrast, midlineFPI resulted in significant peak accumulation of up to 500 nm NP at 3 h post-injury compared to sham, 1 h, and 6 h groups in the cortex. Therefore, our study provides the groundwork for feasibility of NP-delivery based on NPinjection time and NPsize after mCHI/RmCHI and midline-FPI.
Collapse
Affiliation(s)
- Vimala N Bharadwaj
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ
| | - Rachel K Rowe
- Department of Child Health, University of Arizona, College of Medicine, Phoenix, AZ; BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ; Phoenix Veteran Affairs Healthcare System, Phoenix, AZ
| | - Jordan Harrison
- Interdisciplinary Graduate Program in Neuroscience, Arizona State University, Tempe, AZ
| | - Chen Wu
- Department of Child Health, University of Arizona, College of Medicine, Phoenix, AZ
| | - Trent R Anderson
- Basic Medical Sciences, University of Arizona, College of Medicine, Phoenix, AZ
| | - Jonathan Lifshitz
- Department of Child Health, University of Arizona, College of Medicine, Phoenix, AZ; BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ; Phoenix Veteran Affairs Healthcare System, Phoenix, AZ; Interdisciplinary Graduate Program in Neuroscience, Arizona State University, Tempe, AZ
| | - P David Adelson
- Department of Child Health, University of Arizona, College of Medicine, Phoenix, AZ; BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ
| | - Vikram D Kodibagkar
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ
| | - Sarah E Stabenfeldt
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ.
| |
Collapse
|
49
|
Le Master E, Fancher IS, Lee J, Levitan I. Comparative analysis of endothelial cell and sub-endothelial cell elastic moduli in young and aged mice: Role of CD36. J Biomech 2018; 76:263-268. [PMID: 29954596 DOI: 10.1016/j.jbiomech.2018.06.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 05/22/2018] [Accepted: 06/09/2018] [Indexed: 10/14/2022]
Abstract
OBJECTIVE To perform comparative analysis of the role of scavenger receptor CD36 on endothelial vs. sub-endothelial elastic modulus (stiffness) in the aortas of young and aged mice. APPROACHES AND RESULTS Elastic moduli of endothelial and sub-endothelial layers of freshly isolated mouse aortas were quantified using atomic force microscopy. In young mice (4-6 months old), we found that while endothelial stiffness is markedly reduced in aortas of CD36-/-mice, as compared to WT controls, no difference between CD36-/- and WT aortas is observed in the stiffness of the sub-endothelial layer in denuded arteries. Additionally, inhibition of myosin phosphorylation also decreases the elastic modulus in the EC, but not the sub-EC layer in WT mice. Moreover, inhibiting CD36 mediated uptake of oxLDL in intact WT aortas abrogated oxLDL-induced endothelial stiffening. Further analysis of aged mice (22-25 months) revealed that aging resulted not only in significant stiffening of the denuded arteries, as was previously known, but also a comparable increase in the elastic modulus of the endothelial layer. Most significantly, this stiffening in the EC layer is dependent on CD36, whereas the denuded layer is not affected. CONCLUSIONS Our results show that the role CD36 in stiffening of cellular components of intact aortas is endothelial-specific and that genetic deficiency of CD36 protects against endothelial stiffening in aged mice. Moreover, these data suggest that endothelial stiffness in intact mouse aortas depends more on the expression of CD36 than on the stiffness of the sub-endothelial layer.
Collapse
Affiliation(s)
- Elizabeth Le Master
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, United States; Bioengineering, University of Illinois at Chicago, United States
| | - Ibra S Fancher
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, United States
| | - James Lee
- Bioengineering, University of Illinois at Chicago, United States
| | - Irena Levitan
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, United States; Bioengineering, University of Illinois at Chicago, United States.
| |
Collapse
|
50
|
Small GTPase-dependent regulation of leukocyte-endothelial interactions in inflammation. Biochem Soc Trans 2018; 46:649-658. [PMID: 29743277 DOI: 10.1042/bst20170530] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/12/2018] [Accepted: 04/16/2018] [Indexed: 12/19/2022]
Abstract
Inflammation is a complex biological response that serves to protect the body's tissues following harmful stimuli such as infection, irritation or injury and initiates tissue repair. At the start of an inflammatory response, pro-inflammatory mediators induce changes in the endothelial lining of the blood vessels and in leukocytes. This results in increased vascular permeability and increased expression of adhesion proteins, and promotes adhesion of leukocytes, especially neutrophils to the endothelium. Adhesion is a prerequisite for neutrophil extravasation and chemoattractant-stimulated recruitment to inflammatory sites, where neutrophils phagocytose and kill microbes, release inflammatory mediators and cross-talk with other immune cells to co-ordinate the immune response in preparation for tissue repair. Many signalling proteins are critically involved in the complex signalling processes that underpin the inflammatory response and cross-talk between endothelium and leukocytes. As key regulators of cell-cell and cell-substratum adhesion, small GTPases (guanosine triphosphatases) act as important controls of neutrophil-endothelial cell interactions as well as neutrophil recruitment to sites of inflammation. Here, we summarise key processes that are dependent upon small GTPases in leukocytes during these early inflammatory events. We place a particular focus on the regulation of integrin-dependent events and their control by Rho and Rap family GTPases as well as their regulators during neutrophil adhesion, chemotaxis and recruitment.
Collapse
|