1
|
Ramseier NT, Jing H, Anderson J, Hu YS. Superresolution Imaging Reveals the Spatial Organization of CD81 Microdomains in Regulating Membrane Signaling on Jurkat T Cell Microvilli. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.07.627345. [PMID: 39677771 PMCID: PMC11643289 DOI: 10.1101/2024.12.07.627345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Tetraspanin proteins are closely associated with high-curvature membrane structures and play key roles in organizing membrane domains and regulating membrane signaling in immune cells. However, their specific roles in regulating T cell membrane signaling, particularly within the microvilli often characteristic of these cells, remain poorly understood. Here, we used Jurkat T cells as a model system and investigated CD81 as a member of the tetraspanin family. Using total internal reflection fluorescence (TIRF) microscopy and structured illumination microscopy (SIM), we identified an enrichment of the tetraspanin CD81 microdomains along the actin-rich membrane microvilli. At the distal end of the microvilli, SIM images revealed the spatial colocalization of CD81 with T cell receptors (TCR) and CD63, implying a potential role for CD81 in regulating TCR signaling in conjunction with CD63. Spatial analysis of CD81 and CD63 microdomains from the dual-color SIM data revealed their preference for associating with each other. Cluster analysis of direct stochastic optical reconstruction microscopy (dSTORM) data revealed that in vitro T cell activation results in reduced domain sizes and increased domain separation of CD81. These findings provide visual evidence of the spatial organization and rearrangement of CD81 on the T cell microvilli, highlighting its potential role in signal regulation on specialized membrane protrusions.
Collapse
Affiliation(s)
- Neal T. Ramseier
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois Chicago, Chicago, IL 60607, USA
| | - Haoran Jing
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois Chicago, Chicago, IL 60607, USA
| | - Jesse Anderson
- Department of Chemical Engineering, College of Engineering, University of Illinois Chicago, Chicago, IL 60607, USA
| | - Ying S. Hu
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois Chicago, Chicago, IL 60607, USA
| |
Collapse
|
2
|
Abstract
T cell activation is initiated by the recognition of specific antigenic peptides and subsequently accomplished by complex signaling cascades. These aspects have been extensively studied for decades as pivotal factors in the establishment of adaptive immunity. However, how receptors or signaling molecules are organized in the resting state prior to encountering antigens has received less attention. Recent advancements in super-resolution microscopy techniques have revealed topographically controlled pre-formed organization of key molecules involved in antigen recognition and signal transduction on microvillar projections of T cells before activation and substantial effort has been dedicated to characterizing the topological structure of resting T cells over the past decade. This review will summarize our current understanding of how key surface receptors are pre-organized on the T-cell plasma membrane and discuss the potential role of these receptors, which are preassembled prior to ligand binding in the early activation events of T cells.
Collapse
Affiliation(s)
- Yunmin Jung
- Department of Nano-Biomedical Engineering, Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science, Seoul, Republic of Korea
| |
Collapse
|
3
|
Ovadia S, Özcan A, Hidalgo A. The circadian neutrophil, inside-out. J Leukoc Biol 2023; 113:555-566. [PMID: 36999376 PMCID: PMC10583762 DOI: 10.1093/jleuko/qiad038] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 04/01/2023] Open
Abstract
The circadian clock has sway on a myriad of physiological targets, among which the immune and inflammatory systems are particularly prominent. In this review, we discuss how neutrophils, the wildcard of the immune system, are regulated by circadian oscillations. We describe cell-intrinsic and extrinsic diurnal mechanisms governing the general physiology and function of these cells, from purely immune to homeostatic. Repurposing the concepts discovered in other cell types, we then speculate on various uncharted avenues of neutrophil-circadian relationships, such as topology, metabolism, and the regulation of tissue clocks, with the hope of identifying exciting new avenues of work in the context of circadian immunity.
Collapse
Affiliation(s)
- Samuel Ovadia
- Department of Immunobiology and Program of Vascular Biology and Therapeutics, Yale University, 10 Amistad Street, New Haven, CT 06519, United States
| | - Alaz Özcan
- Department of Immunobiology and Program of Vascular Biology and Therapeutics, Yale University, 10 Amistad Street, New Haven, CT 06519, United States
| | - Andrés Hidalgo
- Department of Immunobiology and Program of Vascular Biology and Therapeutics, Yale University, 10 Amistad Street, New Haven, CT 06519, United States
- Program of Cardiovascular Regeneration, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Calle Melchor Fernandez Almagro 3, Madrid 28029, Spain
| |
Collapse
|
4
|
Toffali L, D'Ulivo B, Giagulli C, Montresor A, Zenaro E, Delledonne M, Rossato M, Iadarola B, Sbarbati A, Bernardi P, Angelini G, Rossi B, Lopez N, Linke WA, Unger A, Di Silvestre D, Benazzi L, De Palma A, Motta S, Constantin G, Mauri P, Laudanna C. An isoform of the giant protein titin is a master regulator of human T lymphocyte trafficking. Cell Rep 2023; 42:112516. [PMID: 37204926 DOI: 10.1016/j.celrep.2023.112516] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/24/2023] [Accepted: 05/01/2023] [Indexed: 05/21/2023] Open
Abstract
Response to multiple microenvironmental cues and resilience to mechanical stress are essential features of trafficking leukocytes. Here, we describe unexpected role of titin (TTN), the largest protein encoded by the human genome, in the regulation of mechanisms of lymphocyte trafficking. Human T and B lymphocytes express five TTN isoforms, exhibiting cell-specific expression, distinct localization to plasma membrane microdomains, and different distribution to cytosolic versus nuclear compartments. In T lymphocytes, the LTTN1 isoform governs the morphogenesis of plasma membrane microvilli independently of ERM protein phosphorylation status, thus allowing selectin-mediated capturing and rolling adhesions. Likewise, LTTN1 controls chemokine-triggered integrin activation. Accordingly, LTTN1 mediates rho and rap small GTPases activation, but not actin polymerization. In contrast, chemotaxis is facilitated by LTTN1 degradation. Finally, LTTN1 controls resilience to passive cell deformation and ensures T lymphocyte survival in the blood stream. LTTN1 is, thus, a critical and versatile housekeeping regulator of T lymphocyte trafficking.
Collapse
Affiliation(s)
- Lara Toffali
- Department of Medicine, Division of General Pathology, Laboratory of Cell Trafficking and Signal Transduction, University of Verona; 37134 Verona, Veneto, Italy
| | - Beatrice D'Ulivo
- Department of Medicine, Division of General Pathology, Laboratory of Cell Trafficking and Signal Transduction, University of Verona; 37134 Verona, Veneto, Italy
| | - Cinzia Giagulli
- Department of Molecular and Translational Medicine, University of Brescia; 25123 Brescia, Lombardia, Italy
| | - Alessio Montresor
- Department of Medicine, Division of General Pathology, Laboratory of Cell Trafficking and Signal Transduction, University of Verona; 37134 Verona, Veneto, Italy; The Center for Biomedical Computing (CBMC), University of Verona; 37134 Verona, Veneto, Italy
| | - Elena Zenaro
- Department of Medicine, Division of General Pathology, Laboratory of Cell Trafficking and Signal Transduction, University of Verona; 37134 Verona, Veneto, Italy
| | - Massimo Delledonne
- Department of Biotechnology, University of Verona; 37134 Verona, Veneto, Italy
| | - Marzia Rossato
- Department of Biotechnology, University of Verona; 37134 Verona, Veneto, Italy
| | - Barbara Iadarola
- Department of Biotechnology, University of Verona; 37134 Verona, Veneto, Italy
| | - Andrea Sbarbati
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona; 37134 Verona, Veneto, Italy
| | - Paolo Bernardi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona; 37134 Verona, Veneto, Italy
| | - Gabriele Angelini
- Department of Medicine, Division of General Pathology, Laboratory of Cell Trafficking and Signal Transduction, University of Verona; 37134 Verona, Veneto, Italy
| | - Barbara Rossi
- Department of Medicine, Division of General Pathology, Laboratory of Cell Trafficking and Signal Transduction, University of Verona; 37134 Verona, Veneto, Italy
| | - Nicola Lopez
- Department of Medicine, Division of General Pathology, Laboratory of Cell Trafficking and Signal Transduction, University of Verona; 37134 Verona, Veneto, Italy
| | - Wolfgang A Linke
- Institute of Physiology II, University of Muenster, and Heart Center, University Medicine; 37075 Göttingen, Germany
| | - Andreas Unger
- Institute of Physiology II, University of Muenster, and Heart Center, University Medicine; 37075 Göttingen, Germany
| | - Dario Di Silvestre
- Institute of Biomedical Technologies (ITB) CNR; 20090 Milan, Lombardia, Italy
| | - Louise Benazzi
- Institute of Biomedical Technologies (ITB) CNR; 20090 Milan, Lombardia, Italy
| | - Antonella De Palma
- Institute of Biomedical Technologies (ITB) CNR; 20090 Milan, Lombardia, Italy
| | - Sara Motta
- Institute of Biomedical Technologies (ITB) CNR; 20090 Milan, Lombardia, Italy
| | - Gabriela Constantin
- Department of Medicine, Division of General Pathology, Laboratory of Cell Trafficking and Signal Transduction, University of Verona; 37134 Verona, Veneto, Italy; The Center for Biomedical Computing (CBMC), University of Verona; 37134 Verona, Veneto, Italy
| | - Pierluigi Mauri
- Institute of Biomedical Technologies (ITB) CNR; 20090 Milan, Lombardia, Italy
| | - Carlo Laudanna
- Department of Medicine, Division of General Pathology, Laboratory of Cell Trafficking and Signal Transduction, University of Verona; 37134 Verona, Veneto, Italy; The Center for Biomedical Computing (CBMC), University of Verona; 37134 Verona, Veneto, Italy.
| |
Collapse
|
5
|
Kim HR, Park JS, Soh WC, Kim NY, Moon HY, Lee JS, Jun CD. T Cell Microvilli: Finger-Shaped External Structures Linked to the Fate of T Cells. Immune Netw 2023; 23:e3. [PMID: 36911802 PMCID: PMC9995986 DOI: 10.4110/in.2023.23.e3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/11/2023] [Accepted: 02/11/2023] [Indexed: 03/07/2023] Open
Abstract
Microvilli are outer membrane organelles that contain cross-linked filamentous actin. Unlike well-characterized epithelial microvilli, T-cell microvilli are dynamic similar to those of filopodia, which grow and shrink intermittently via the alternate actin-assembly and -disassembly. T-cell microvilli are specialized for sensing Ags on the surface of Ag-presenting cells (APCs). Thus, these finger-shaped microprotrusions contain many signaling-related proteins and can serve as a signaling platforms that induce intracellular signals. However, they are not limited to sensing external information but can provide sites for parts of the cell-body to tear away from the cell. Cells are known to produce many types of extracellular vesicles (EVs), such as exosomes, microvesicles, and membrane particles. T cells also produce EVs, but little is known about under what conditions T cells generate EVs and which types of EVs are released. We discovered that T cells produce few exosomes but release large amounsts of microvilli-derived particles during physical interaction with APCs. Although much is unanswered as to why T cells use the same organelles to sense Ags or to produce EVs, these events can significantly affect T cell fate, including clonal expansion and death. Since TCRs are localized at microvilli tips, this membrane event also raises a new question regarding long-standing paradigm in T cell biology; i.e., surface TCR downmodulation following T cell activation. Since T-cell microvilli particles carry T-cell message to their cognate partner, these particles are termed T-cell immunological synaptosomes (TISs). We discuss the potential physiological role of TISs and their application to immunotherapies.
Collapse
Affiliation(s)
- Hye-Ran Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Division of Rare and Refractory Cancer, Tumor Immunology, Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Jeong-Su Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Won-Chang Soh
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Na-Young Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Hyun-Yoong Moon
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Ji-Su Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Chang-Duk Jun
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| |
Collapse
|
6
|
Ghosh S, Alcover A, Haran G. Microvillar Cartography: A Super-Resolution Single-Molecule Imaging Method to Map the Positions of Membrane Proteins with Respect to Cellular Surface Topography. Methods Mol Biol 2023; 2654:169-199. [PMID: 37106183 DOI: 10.1007/978-1-0716-3135-5_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
We describe microvillar cartography (MC), a method to map proteins on cellular surfaces with respect to the membrane topography. The surfaces of many cells are not smooth, but are rather covered with various protrusions such as microvilli. These protrusions may play key roles in multiple cellular functions, due to their ability to control the distribution of specific protein assemblies on the cell surface. Thus, for example, we have shown that the T-cell receptor and several of its proximal signaling proteins reside on microvilli, while others are excluded from these projections. These results have indicated that microvilli can function as key signaling hubs for the initiation of the immune response. MC has facilitated our observations of particular surface proteins and their specialized distribution on microvillar and non-microvillar compartments. MC combines membrane topography imaging, using variable-angle total internal microscopy, with stochastic localization nanoscopy, which generates deep sub-diffraction maps of protein distribution. Since the method is based on light microscopy, it avoids some of the pitfalls inherent to electron-microscopy-based techniques, such as dehydration, the need for carbon coating, and immunogold clustering, and is amenable to future developments involving, for example, live-cell imaging. This protocol details the procedures we developed for MC, which can be readily adopted to study a broad range of cell-surface molecules and dissect their distribution within distinct surface assemblies under multiple cell activation states.
Collapse
Affiliation(s)
- Shirsendu Ghosh
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot, Israel
| | - Andres Alcover
- Institut Pasteur, Université Paris Cité, INSERM U1224, Unité Biologie Cellulaire des Lymphocytes, Paris, France
| | - Gilad Haran
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
7
|
Inaba M, Ridwan SM, Antel M. Removal of cellular protrusions. Semin Cell Dev Biol 2022; 129:126-134. [PMID: 35260295 PMCID: PMC9378436 DOI: 10.1016/j.semcdb.2022.02.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 02/08/2023]
Abstract
Cell-cell communications are central to a variety of physiological and pathological processes in multicellular organisms. Cells often rely on cellular protrusions to communicate with one another, which enable highly selective and efficient signaling within complex tissues. Owing to significant improvements in imaging techniques, identification of signaling protrusions has increased in recent years. These protrusions are structurally specialized for signaling and facilitate interactions between cells. Therefore, physical regulation of these structures must be key for the appropriate strength and pattern of signaling outcomes. However, the typical approaches for understanding signaling regulation tend to focus solely on changes in signaling molecules, such as gene expression, protein-protein interaction, and degradation. In this short review, we summarize the studies proposing the removal of different types of signaling protrusions-including cilia, neurites, MT (microtubule based)-nanotubes and microvilli-and discuss their mechanisms and significance in signaling regulation.
Collapse
Affiliation(s)
- Mayu Inaba
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - Sharif M Ridwan
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Matthew Antel
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
| |
Collapse
|
8
|
Nayak L, Sweet DR, Thomas A, Lapping SD, Kalikasingh K, Madera A, Vinayachandran V, Padmanabhan R, Vasudevan NT, Myers JT, Huang AY, Schmaier A, Mackman N, Liao X, Maiseyeu A, Jain MK. A targetable pathway in neutrophils mitigates both arterial and venous thrombosis. Sci Transl Med 2022; 14:eabj7465. [PMID: 36044595 DOI: 10.1126/scitranslmed.abj7465] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Arterial and venous thrombosis constitutes a major source of morbidity and mortality worldwide. Long considered as distinct entities, accumulating evidence indicates that arterial and venous thrombosis can occur in the same populations, suggesting that common mechanisms are likely operative. Although hyperactivation of the immune system is a common forerunner to the genesis of thrombotic events in both vascular systems, the key molecular control points remain poorly understood. Consequently, antithrombotic therapies targeting the immune system for therapeutics gain are lacking. Here, we show that neutrophils are key effectors of both arterial and venous thrombosis and can be targeted through immunoregulatory nanoparticles. Using antiphospholipid antibody syndrome (APS) as a model for arterial and venous thrombosis, we identified the transcription factor Krüppel-like factor 2 (KLF2) as a key regulator of neutrophil activation. Upon activation through genetic loss of KLF2 or administration of antiphospholipid antibodies, neutrophils clustered P-selectin glycoprotein ligand 1 (PSGL-1) by cortical actin remodeling, thereby increasing adhesion potential at sites of thrombosis. Targeting clustered PSGL-1 using nanoparticles attenuated neutrophil-mediated thrombosis in APS and KLF2 knockout models, illustrating the importance and feasibility of targeting activated neutrophils to prevent pathological thrombosis. Together, our results demonstrate a role for activated neutrophils in both arterial and venous thrombosis and identify key molecular events that serve as potential targets for therapeutics against diverse causes of immunothrombosis.
Collapse
Affiliation(s)
- Lalitha Nayak
- Division of Hematology and Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - David R Sweet
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA.,Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA.,Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Asha Thomas
- Division of Hematology and Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Stephanie D Lapping
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA.,Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Kenneth Kalikasingh
- Division of Hematology and Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Annmarie Madera
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA.,Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Vinesh Vinayachandran
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA.,Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Roshan Padmanabhan
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA.,Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Neelakantan T Vasudevan
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA.,Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Jay T Myers
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Alex Y Huang
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Alvin Schmaier
- Division of Hematology and Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Nigel Mackman
- Division of Hematology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Xudong Liao
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Andrei Maiseyeu
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA.,Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Mukesh K Jain
- Warren Alpert Medical School of Brown University, Providence, R1 02903
| |
Collapse
|
9
|
Cai E, Beppler C, Eichorst J, Marchuk K, Eastman SW, Krummel MF. T cells use distinct topographical and membrane receptor scanning strategies that individually coalesce during receptor recognition. Proc Natl Acad Sci U S A 2022; 119:e2203247119. [PMID: 35914144 PMCID: PMC9372542 DOI: 10.1073/pnas.2203247119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/22/2022] [Indexed: 02/03/2023] Open
Abstract
During immune surveillance, CD8 T cells scan the surface of antigen-presenting cells using dynamic microvillar palpation and movements as well as by having their receptors preconcentrated into patches. Here, we use real-time lattice light-sheet microscopy to demonstrate the independence of microvillar and membrane receptor patch scanning. While T cell receptor (TCR) patches can distribute to microvilli, they do so stochastically and not preferentially as for other receptors such as CD62L. The distinctness of TCR patch movement from microvillar movement extends to many other receptors that form patches that also scan independent of the TCR. An exception to this is the CD8 coreceptor which largely comigrates in patches that overlap with or are closely adjacent to those containing TCRs. Microvilli that assemble into a synapse contain various arrays of the engaged patches, notably of TCRs and the inhibitory receptor PD-1, creating a pastiche of occupancies that vary from microvillar contact to contact. In summary, this work demonstrates that localization of receptor patches within the membrane and on microvillar projections is random prior to antigen detection and that such random variation may play into the generation of many individually composed receptor patch compositions at a single synapse.
Collapse
Affiliation(s)
- En Cai
- Department of Pathology, University of California, San Francisco, CA 94143-0511
| | - Casey Beppler
- Department of Pathology, University of California, San Francisco, CA 94143-0511
| | - John Eichorst
- Department of Pathology, University of California, San Francisco, CA 94143-0511
- Biological Imaging Development CoLab, University of California, San Francisco, CA 94143-0511
| | - Kyle Marchuk
- Department of Pathology, University of California, San Francisco, CA 94143-0511
- Biological Imaging Development CoLab, University of California, San Francisco, CA 94143-0511
- ImmunoX Initiative, University of California, San Francisco, CA 94143-0511
| | - Scott W. Eastman
- Lilly Research Laboratories, Eli Lilly and Company, New York, NY 10016
| | - Matthew F. Krummel
- Department of Pathology, University of California, San Francisco, CA 94143-0511
- ImmunoX Initiative, University of California, San Francisco, CA 94143-0511
| |
Collapse
|
10
|
Zeng Q, Gao Y, Yu H, Zhu W, Wang Q, Long Q, Fan Z, Xiao B. Stick, stretch, and scan imaging method for DNA and filaments. RSC Adv 2021; 11:36060-36065. [PMID: 35492749 PMCID: PMC9043539 DOI: 10.1039/d1ra07067c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 10/29/2021] [Indexed: 11/21/2022] Open
Abstract
Biomolecules and organelles usually undergo changes to their structure or form as a result of mechanical stretching or stimulation. It is critical to be able to observe these changes and responses, which trigger mechano-chemical coupling or signal transduction. Advanced techniques have been developed to observe structure and form during manipulation; however, these require sophisticated methods. We have developed a simple approach to observe fine structure after stretching without fluorophore labeling. DNAs or molecules on the cell surface were bound to magnetic microbeads, followed by stretching with a magnetic field. After fixing, staining, and drying, the samples were examined by scanning electron microscopy with no need to build a functional surface with complex processes. Straight DNAs were observed rather than random-walk-like loose polymers. In our cellular experiment, the magnetic beads were bound to a Jurkat cell and formed a rosette which was later stuck to the substrate. A 41.3 μm filament on the base of a filopodium was pulled out via integrin from a cell. Therefore, our method can reveal long structures up to hundreds of micrometers at nanometer resolution after stretching or twisting. Our approach could have wide applications in structure–function studies of biomolecules, and in mechanobiology and cell biology when diffraction cannot used. Magnetic force was applied to stretch single DNAs and cells which were stuck to magnetic beads and substrates via simple conjugation methods. Scanning electron microscopy images show that the filopodia of cells were pulled to extraordinary length.![]()
Collapse
Affiliation(s)
- Qiuling Zeng
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, School of Biology and Biological Engineering, South China University of Technology Guangzhou 510006 China
| | - Yuanyuan Gao
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, School of Biology and Biological Engineering, South China University of Technology Guangzhou 510006 China
| | - Hong Yu
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, School of Biology and Biological Engineering, South China University of Technology Guangzhou 510006 China
| | - Wei Zhu
- Joint International Research Laboratory of Synthetic Biology and Medicine, School of Biology and Biological Engineering, South China University of Technology Guangzhou 510006 China
| | - Qi Wang
- Artemisinin Research Center, Guangzhou University of Chinese Medicine Guangzhou Guangdong 510405 China.,Institute of Science and Technology, Guangzhou University of Chinese Medicine Guangzhou Guangdong 510006 China
| | - Quan Long
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, School of Biology and Biological Engineering, South China University of Technology Guangzhou 510006 China
| | - Zhuo Fan
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, School of Biology and Biological Engineering, South China University of Technology Guangzhou 510006 China
| | - Botao Xiao
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, School of Biology and Biological Engineering, South China University of Technology Guangzhou 510006 China .,Joint International Research Laboratory of Synthetic Biology and Medicine, School of Biology and Biological Engineering, South China University of Technology Guangzhou 510006 China
| |
Collapse
|
11
|
Ghosh S, Feigelson SW, Montresor A, Shimoni E, Roncato F, Legler DF, Laudanna C, Haran G, Alon R. CCR7 signalosomes are preassembled on tips of lymphocyte microvilli in proximity to LFA-1. Biophys J 2021; 120:4002-4012. [PMID: 34411577 DOI: 10.1016/j.bpj.2021.08.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/27/2021] [Accepted: 08/11/2021] [Indexed: 10/20/2022] Open
Abstract
Leukocyte microvilli are elastic actin-rich projections implicated in rapid sensing and penetration across glycocalyx barriers. Microvilli are critical for the capture and arrest of flowing lymphocytes by high endothelial venules, the main lymph node portal vessels. T lymphocyte arrest involves subsecond activation of the integrin LFA-1 by the G-protein-coupled receptor CCR7 and its endothelial-displayed ligands, the chemokines CCL21 and CCL19. The topographical distribution of CCR7 and of LFA-1 in relation to lymphocyte microvilli has never been elucidated. We applied the recently developed microvillar cartography imaging technique to determine the topographical distribution of CCR7 and LFA-1 with respect to microvilli on peripheral blood T lymphocytes. We found that CCR7 is clustered on the tips of T cell microvilli. The vast majority of LFA-1 molecules were found on the cell body, likely assembled in macroclusters, but a subset of LFA-1, 5% of the total, were found scattered within 20 nm from the CCR7 clusters, implicating these LFA-1 molecules as targets for inside-out activation signals transmitted within a fraction of a second by chemokine-bound CCR7. Indeed, RhoA, the key GTPase involved in rapid LFA-1 affinity triggering by CCR7, was also found to be clustered near CCR7. In addition, we observed that the tyrosine kinase JAK2 controls CCR7-mediated LFA-1 affinity triggering and is also highly enriched on tips of microvilli. We propose that tips of lymphocyte microvilli are novel signalosomes for subsecond CCR7-mediated inside-out signaling to neighboring LFA-1 molecules, a critical checkpoint in LFA-1-mediated lymphocyte arrest on high endothelial venules.
Collapse
Affiliation(s)
- Shirsendu Ghosh
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot, Israel
| | - Sara W Feigelson
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Eyal Shimoni
- Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel
| | - Francesco Roncato
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Daniel F Legler
- Biotechnology Institute Thurgau, University of Konstanz, Kreuzlingen, Switzerland
| | - Carlo Laudanna
- Department of Medicine, University of Verona, Verona, Italy
| | - Gilad Haran
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot, Israel.
| | - Ronen Alon
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
12
|
Jung Y, Wen L, Altman A, Ley K. CD45 pre-exclusion from the tips of T cell microvilli prior to antigen recognition. Nat Commun 2021; 12:3872. [PMID: 34162836 PMCID: PMC8222282 DOI: 10.1038/s41467-021-23792-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 05/11/2021] [Indexed: 12/18/2022] Open
Abstract
The tyrosine phosphatase CD45 is a major gatekeeper for restraining T cell activation. Its exclusion from the immunological synapse (IS) is crucial for T cell receptor (TCR) signal transduction. Here, we use expansion super-resolution microscopy to reveal that CD45 is mostly pre-excluded from the tips of microvilli (MV) on primary T cells prior to antigen encounter. This pre-exclusion is diminished by depleting cholesterol or by engineering the transmembrane domain of CD45 to increase its membrane integration length, but is independent of the CD45 extracellular domain. We further show that brief MV-mediated contacts can induce Ca2+ influx in mouse antigen-specific T cells engaged by antigen-pulsed antigen presenting cells (APC). We propose that the scarcity of CD45 phosphatase activity at the tips of MV enables or facilitates TCR triggering from brief T cell-APC contacts before formation of a stable IS, and that these MV-mediated contacts represent the earliest step in the initiation of a T cell adaptive immune response.
Collapse
Affiliation(s)
- Yunmin Jung
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, USA.
| | - Lai Wen
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Amnon Altman
- Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Klaus Ley
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
13
|
Neutrophils lacking ERM proteins polarize and crawl directionally but have decreased adhesion strength. Blood Adv 2021; 4:3559-3571. [PMID: 32761234 DOI: 10.1182/bloodadvances.2020002423] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 06/28/2020] [Indexed: 12/19/2022] Open
Abstract
Ezrin/radixin/moesin (ERM) proteins are adaptors that link the actin cytoskeleton to the cytoplasmic domains of membrane proteins. Leukocytes express mostly moesin with lower levels of ezrin but no radixin. When leukocytes are activated, ERMs are postulated to redistribute membrane proteins from microvilli into uropods during polarization and to transduce signals that influence adhesion and other responses. However, these functions have not been tested in leukocytes lacking all ERMs. We used knockout (KO) mice with neutrophils lacking ezrin, moesin, or both proteins (double knockout [DKO]) to probe how ERMs modulate cell shape, adhesion, and signaling in vitro and in vivo. Surprisingly, chemokine-stimulated DKO neutrophils still polarized and redistributed ERM-binding proteins such as PSGL-1 and CD44 to the uropods. Selectin binding to PSGL-1 on moesin KO or DKO neutrophils activated kinases that enable integrin-dependent slow rolling but not those that generate neutrophil extracellular traps. Flowing neutrophils of all genotypes rolled normally on selectins and, upon chemokine stimulation, arrested on integrin ligands. However, moesin KO and DKO neutrophils exhibited defective integrin outside-in signaling and reduced adhesion strength. In vivo, DKO neutrophils displayed normal directional crawling toward a chemotactic gradient, but premature detachment markedly reduced migration from venules into inflamed tissues. Our results demonstrate that stimulated neutrophils do not require ERMs to polarize or to move membrane proteins into uropods. They also reveal an unexpected contribution of moesin to integrin outside-in signaling and adhesion strengthening.
Collapse
|
14
|
Ghosh S, Di Bartolo V, Tubul L, Shimoni E, Kartvelishvily E, Dadosh T, Feigelson SW, Alon R, Alcover A, Haran G. ERM-Dependent Assembly of T Cell Receptor Signaling and Co-stimulatory Molecules on Microvilli prior to Activation. Cell Rep 2021; 30:3434-3447.e6. [PMID: 32160548 DOI: 10.1016/j.celrep.2020.02.069] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/16/2019] [Accepted: 02/18/2020] [Indexed: 01/25/2023] Open
Abstract
T cell surfaces are covered with microvilli, actin-rich and flexible protrusions. We use super-resolution microscopy to show that ≥90% of T cell receptor (TCR) complex molecules TCRαβ and TCRζ, as well as the co-receptor CD4 (cluster of differentiation 4) and the co-stimulatory molecule CD2, reside on microvilli of resting human T cells. Furthermore, TCR proximal signaling molecules involved in the initial stages of the immune response, including the protein tyrosine kinase Lck (lymphocyte-specific protein tyrosine kinase) and the key adaptor LAT (linker for activation of T cells), are also enriched on microvilli. Notably, phosphorylated proteins of the ERM (ezrin, radixin, and moesin) family colocalize with TCRαβ as well as with actin filaments, implying a role for one or more ERMs in linking the TCR complex to the actin cytoskeleton within microvilli. Our results establish microvilli as key signaling hubs, in which the TCR complex and its proximal signaling molecules and adaptors are preassembled prior to activation in an ERM-dependent manner, facilitating initial antigen sensing.
Collapse
Affiliation(s)
- Shirsendu Ghosh
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot 7610001, Israel.
| | - Vincenzo Di Bartolo
- Lymphocyte Cell Biology Unit, INSERM U1221, Department of Immunology, Institut Pasteur, Paris 75015, France
| | - Liron Tubul
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Eyal Shimoni
- Chemical Research Support, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Elena Kartvelishvily
- Chemical Research Support, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Tali Dadosh
- Chemical Research Support, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Sara W Feigelson
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ronen Alon
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Andres Alcover
- Lymphocyte Cell Biology Unit, INSERM U1221, Department of Immunology, Institut Pasteur, Paris 75015, France
| | - Gilad Haran
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
15
|
Grégory Franck. Role of mechanical stress and neutrophils in the pathogenesis of plaque erosion. Atherosclerosis 2020; 318:60-69. [PMID: 33190807 DOI: 10.1016/j.atherosclerosis.2020.11.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/05/2020] [Accepted: 11/03/2020] [Indexed: 02/05/2023]
Abstract
Mechanical stress is a well-recognized driver of plaque rupture. Likewise, investigating the role of mechanical forces in plaque erosion has recently begun to provide some important insights, yet the knowledge is by far less advanced. The most significant example is that of shear stress, which has early been proposed as a possible driver for focal endothelial death and denudation. Recent findings using optical coherence tomography, computational sciences and mechanical models show that plaque erosion occurs most likely around atheromatous plaque throats with specific stress pattern. In parallel, we have recently shown that neutrophil-dependent inflammation promotes plaque erosion, possibly through a noxious action on ECs. Most importantly, spontaneous thrombosis - associated or not with EC denudation - can be impacted by hemodynamics, and it is now established that neutrophils promote thrombosis and platelet activation, highlighting a potential relationship between, mechanical stress, inflammation, and EC loss in the setting of coronary plaque erosion. Here, we review our current knowledge regarding the implication of both mechanical stress and neutrophils, and we discuss their implication in the promotion of plaque erosion via EC loss and thrombosis.
Collapse
Affiliation(s)
- Grégory Franck
- Inserm LVTS U1148. CHU Bichat, 46 Rue Henri Huchard, 75018, Paris, France.
| |
Collapse
|
16
|
Orbach R, Su X. Surfing on Membrane Waves: Microvilli, Curved Membranes, and Immune Signaling. Front Immunol 2020; 11:2187. [PMID: 33013920 PMCID: PMC7516127 DOI: 10.3389/fimmu.2020.02187] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/11/2020] [Indexed: 01/22/2023] Open
Abstract
Microvilli are finger-like membrane protrusions, supported by the actin cytoskeleton, and found on almost all cell types. A growing body of evidence suggests that the dynamic lymphocyte microvilli, with their highly curved membranes, play an important role in signal transduction leading to immune responses. Nevertheless, challenges in modulating local membrane curvature and monitoring the high dynamicity of microvilli hampered the investigation of the curvature-generation mechanism and its functional consequences in signaling. These technical barriers have been partially overcome by recent advancements in adapted super-resolution microscopy. Here, we review the up-to-date progress in understanding the mechanisms and functional consequences of microvillus formation in T cell signaling. We discuss how the deformation of local membranes could potentially affect the organization of signaling proteins and their biochemical activities. We propose that curved membranes, together with the underlying cytoskeleton, shape microvilli into a unique compartment that sense and process signals leading to lymphocyte activation.
Collapse
Affiliation(s)
- Ron Orbach
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, United States
| | - Xiaolei Su
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, United States
- Yale Cancer Center, Yale University, New Haven, CT, United States
| |
Collapse
|
17
|
Hughes MR, Canals Hernaez D, Cait J, Refaeli I, Lo BC, Roskelley CD, McNagny KM. A sticky wicket: Defining molecular functions for CD34 in hematopoietic cells. Exp Hematol 2020; 86:1-14. [PMID: 32422232 DOI: 10.1016/j.exphem.2020.05.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/05/2020] [Accepted: 05/09/2020] [Indexed: 02/06/2023]
Abstract
The CD34 cell surface antigen is widely expressed in tissues on cells with progenitor-like properties and on mature vascular endothelia. In adult human bone marrow, CD34 marks hematopoietic stem and progenitor cells (HSPCs) starting from the bulk of hematopoietic stem cells with long-term repopulating potential (LT-HSCs) throughout expansion and differentiation of oligopotent and unipotent progenitors. CD34 protein surface expression is typically lost as cells mature into terminal effectors. Because of this expression pattern of HSPCs, CD34 has had a central role in the evaluation or selection of donor graft tissue in HSC transplant (HSCT). Given its clinical importance, it is surprising that the biological functions of CD34 are still poorly understood. This enigma is due, in part, to CD34's context-specific role as both a pro-adhesive and anti-adhesive molecule and its potential functional redundancy with other sialomucins. Moreover, there are also critical differences in the regulation of CD34 expression on HSPCs in humans and experimental mice. In this review, we highlight some of the more well-defined functions of CD34 in HSPCs with a focus on proposed functions most relevant to HSCT biology.
Collapse
Affiliation(s)
- Michael R Hughes
- The Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Diana Canals Hernaez
- The Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Jessica Cait
- The Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - Ido Refaeli
- The Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Bernard C Lo
- The Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - Calvin D Roskelley
- Life Sciences Institute, Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Kelly M McNagny
- The Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
18
|
Phan TKT, Shahbazzadeh F, Kihara T. Alpha-mangostin reduces mechanical stiffness of various cells. Hum Cell 2020; 33:347-355. [PMID: 32078151 DOI: 10.1007/s13577-020-00330-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/07/2020] [Indexed: 02/07/2023]
Abstract
Alpha-mangostin (α-mangostin) has been identified as a naturally occurring compound with potential anticancer properties. It can induce apoptosis and inhibit the growth and metastasis of cancer cells. Moreover, α-mangostin reduces the mechanical stiffness of lung cancer cells. The objective of this study was to determine the effect of α-mangostin on the mechanical stiffness of various cells, as well as cell viability. The following cell types were examined: human fibroblast TIG-1 cells, human cancerous HeLa cells, human embryonic kidney HEK293 cells, mouse macrophage RAW 264.7 cells, and human myeloblasts KG-1 cells. Cells were treated with α-mangostin, and then examined for cell viability, actin cytoskeletal structures, and surface mechanical stiffness using atomic force microscopy. α-Mangostin demonstrated cytotoxicity against TIG-1, HeLa, HEK293, and KG-1 cells, but not against RAW 264.7 cells. The cytotoxic effect of α-mangostin varies according to cell type. On the other hand, α-mangostin reduced the mechanical stiffness of all cell types, including RAW 264.7 cells. Upon treatment with α-mangostin, F-actin was slightly reduced but the actin cytoskeletal structures were little altered in these cells. Thus, reducing mechanical stiffness of animal cells is an inherent effect of α-mangostin. Our results show that α-mangostin is a naturally occurring compound with potential to change the actin cytoskeletal micro-structures and reduce the surface stiffness of various cells.
Collapse
Affiliation(s)
- Thi Kieu Trang Phan
- Department of Life and Environment Engineering, Faculty of Environmental Engineering, The University of Kitakyushu, 1-1 Hibikino, Wakamatsu, Kitakyushu, Fukuoka, 808-0135, Japan
| | - Fahimeh Shahbazzadeh
- Department of Life and Environment Engineering, Faculty of Environmental Engineering, The University of Kitakyushu, 1-1 Hibikino, Wakamatsu, Kitakyushu, Fukuoka, 808-0135, Japan
| | - Takanori Kihara
- Department of Life and Environment Engineering, Faculty of Environmental Engineering, The University of Kitakyushu, 1-1 Hibikino, Wakamatsu, Kitakyushu, Fukuoka, 808-0135, Japan.
| |
Collapse
|
19
|
Fernandes RA, Ganzinger KA, Tzou JC, Jönsson P, Lee SF, Palayret M, Santos AM, Carr AR, Ponjavic A, Chang VT, Macleod C, Lagerholm BC, Lindsay AE, Dushek O, Tilevik A, Davis SJ, Klenerman D. A cell topography-based mechanism for ligand discrimination by the T cell receptor. Proc Natl Acad Sci U S A 2019; 116:14002-14010. [PMID: 31221762 PMCID: PMC6628812 DOI: 10.1073/pnas.1817255116] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The T cell receptor (TCR) initiates the elimination of pathogens and tumors by T cells. To avoid damage to the host, the receptor must be capable of discriminating between wild-type and mutated self and nonself peptide ligands presented by host cells. Exactly how the TCR does this is unknown. In resting T cells, the TCR is largely unphosphorylated due to the dominance of phosphatases over the kinases expressed at the cell surface. However, when agonist peptides are presented to the TCR by major histocompatibility complex proteins expressed by antigen-presenting cells (APCs), very fast receptor triggering, i.e., TCR phosphorylation, occurs. Recent work suggests that this depends on the local exclusion of the phosphatases from regions of contact of the T cells with the APCs. Here, we developed and tested a quantitative treatment of receptor triggering reliant only on TCR dwell time in phosphatase-depleted cell contacts constrained in area by cell topography. Using the model and experimentally derived parameters, we found that ligand discrimination likely depends crucially on individual contacts being ∼200 nm in radius, matching the dimensions of the surface protrusions used by T cells to interrogate their targets. The model not only correctly predicted the relative signaling potencies of known agonists and nonagonists but also achieved this in the absence of kinetic proofreading. Our work provides a simple, quantitative, and predictive molecular framework for understanding why TCR triggering is so selective and fast and reveals that, for some receptors, cell topography likely influences signaling outcomes.
Collapse
Affiliation(s)
- Ricardo A Fernandes
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, United Kingdom
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, United Kingdom
| | - Kristina A Ganzinger
- Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
| | - Justin C Tzou
- Department of Applied & Computational Mathematics & Statistics, University of Notre Dame, Notre Dame, IN 46556
| | - Peter Jönsson
- Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
| | - Steven F Lee
- Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
| | - Matthieu Palayret
- Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
| | - Ana Mafalda Santos
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, United Kingdom
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, United Kingdom
| | - Alexander R Carr
- Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
| | - Aleks Ponjavic
- Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
| | - Veronica T Chang
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, United Kingdom
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, United Kingdom
| | - Charlotte Macleod
- Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
| | - B Christoffer Lagerholm
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, United Kingdom
| | - Alan E Lindsay
- Mathematics Department, University of British Columbia, Vancouver, BC V6T 1Z2, Canada
| | - Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE Oxford, United Kingdom
- Wolfson Centre for Mathematical Biology, University of Oxford, OX1 3RE Oxford, United Kingdom
| | - Andreas Tilevik
- School of Bioscience, University of Skövde, 541 28 Skövde, Sweden
| | - Simon J Davis
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, United Kingdom;
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, United Kingdom
| | - David Klenerman
- Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom;
| |
Collapse
|
20
|
Molecular Dynamics of the Association of L-Selectin and FERM Regulated by PIP2. Biophys J 2019; 114:1858-1868. [PMID: 29694864 DOI: 10.1016/j.bpj.2018.02.034] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/05/2018] [Accepted: 02/20/2018] [Indexed: 11/22/2022] Open
Abstract
Phosphatidylinositol 4,5-bisphosphate (PIP2) acts as a signaling lipid, mediating membrane trafficking and recruitment of proteins to membranes. A key example is the PIP2-dependent regulation of the adhesion of L-selectin to the cytoskeleton adaptors of the N-terminal subdomain of ezrin-radixin-moesin (FERM). The molecular details of the mediating behavior of multivalent anionic PIP2 lipids in this process, however, remain unclear. Here, we use coarse-grained molecular dynamics simulation to explore the mechanistic details of PIP2 in the transformation, translocation, and association of the FERM/L-selectin complex. We compare membranes of different compositions and find that anionic phospholipids are necessary for both FERM and the cytoplasmic domain of L-selectin to absorb on the membrane surface. The subsequent formation of the FERM/L-selectin complex is strongly favored by the presence of PIP2, which clusters around both proteins and triggers a conformational transition in the cytoplasmic domain of L-selectin. We are able to quantify the effect of PIP2 on the association free energy of the complex by means of a potential of mean force. We conclude that PIP2 behaves as an adhesive agent to enhance the stability of the FERM/L-selectin complex and identify key residues involved. The molecular information revealed in this study highlights the specific role of membrane lipids such as PIP2 in protein translocation and potential signaling.
Collapse
|
21
|
Adrover JM, Del Fresno C, Crainiciuc G, Cuartero MI, Casanova-Acebes M, Weiss LA, Huerga-Encabo H, Silvestre-Roig C, Rossaint J, Cossío I, Lechuga-Vieco AV, García-Prieto J, Gómez-Parrizas M, Quintana JA, Ballesteros I, Martin-Salamanca S, Aroca-Crevillen A, Chong SZ, Evrard M, Balabanian K, López J, Bidzhekov K, Bachelerie F, Abad-Santos F, Muñoz-Calleja C, Zarbock A, Soehnlein O, Weber C, Ng LG, Lopez-Rodriguez C, Sancho D, Moro MA, Ibáñez B, Hidalgo A. A Neutrophil Timer Coordinates Immune Defense and Vascular Protection. Immunity 2019; 50:390-402.e10. [PMID: 30709741 DOI: 10.1016/j.immuni.2019.01.002] [Citation(s) in RCA: 263] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 11/23/2018] [Accepted: 01/02/2019] [Indexed: 12/23/2022]
Abstract
Neutrophils eliminate pathogens efficiently but can inflict severe damage to the host if they over-activate within blood vessels. It is unclear how immunity solves the dilemma of mounting an efficient anti-microbial defense while preserving vascular health. Here, we identify a neutrophil-intrinsic program that enabled both. The gene Bmal1 regulated expression of the chemokine CXCL2 to induce chemokine receptor CXCR2-dependent diurnal changes in the transcriptional and migratory properties of circulating neutrophils. These diurnal alterations, referred to as neutrophil aging, were antagonized by CXCR4 (C-X-C chemokine receptor type 4) and regulated the outer topology of neutrophils to favor homeostatic egress from blood vessels at night, resulting in boosted anti-microbial activity in tissues. Mice engineered for constitutive neutrophil aging became resistant to infection, but the persistence of intravascular aged neutrophils predisposed them to thrombo-inflammation and death. Thus, diurnal compartmentalization of neutrophils, driven by an internal timer, coordinates immune defense and vascular protection.
Collapse
Affiliation(s)
- José M Adrover
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Carlos Del Fresno
- Area of Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Georgiana Crainiciuc
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Maria Isabel Cuartero
- Unidad de Investigación Neurovascular, Department of Pharmacology, Faculty of Medicine, Universidad Complutense; Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - María Casanova-Acebes
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Present address: Tisch Cancer Institute, Mount Sinai School of Medicine, New York City, New York, USA
| | - Linnea A Weiss
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Present address: Centro Nacional de Biotecnología, Madrid, Spain
| | - Hector Huerga-Encabo
- Immunology Unit, Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona
| | - Carlos Silvestre-Roig
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität München; German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Jan Rossaint
- Department of Anesthesiology, Intensive Care, and Pain Medicine, University of Münster, Germany
| | - Itziar Cossío
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Ana V Lechuga-Vieco
- Area of Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Jaime García-Prieto
- Area of Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Mónica Gómez-Parrizas
- Area of Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Juan A Quintana
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Ivan Ballesteros
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Sandra Martin-Salamanca
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Alejandra Aroca-Crevillen
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Shu Zhen Chong
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Biopolis, Singapore
| | - Maximilien Evrard
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Biopolis, Singapore
| | - Karl Balabanian
- Inserm Unité Mixte de Recherche (UMR) S996, Université Paris-Sud, Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Clamart, France
| | - Jorge López
- Department of Immunology, Instituto de Investigación Sanitaria Princesa, Hospital Universitario de La Princesa, Madrid, Spain
| | - Kiril Bidzhekov
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität München
| | - Françoise Bachelerie
- Inserm Unité Mixte de Recherche (UMR) S996, Université Paris-Sud, Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Clamart, France
| | - Francisco Abad-Santos
- Department of Clinical Pharmacology, Instituto Teófilo Hernando, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
| | - Cecilia Muñoz-Calleja
- Department of Immunology, Instituto de Investigación Sanitaria Princesa, Hospital Universitario de La Princesa, Madrid, Spain
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care, and Pain Medicine, University of Münster, Germany
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität München; German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität München; Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Lai Guan Ng
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Biopolis, Singapore
| | - Cristina Lopez-Rodriguez
- Immunology Unit, Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona
| | - David Sancho
- Area of Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - María A Moro
- Unidad de Investigación Neurovascular, Department of Pharmacology, Faculty of Medicine, Universidad Complutense; Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Borja Ibáñez
- Area of Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain; Department of Cardiology, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Madrid, Spain
| | - Andrés Hidalgo
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität München.
| |
Collapse
|
22
|
Rossy J, Laufer JM, Legler DF. Role of Mechanotransduction and Tension in T Cell Function. Front Immunol 2018; 9:2638. [PMID: 30519239 PMCID: PMC6251326 DOI: 10.3389/fimmu.2018.02638] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/26/2018] [Indexed: 12/23/2022] Open
Abstract
T cell migration from blood to, and within lymphoid organs and tissue, as well as, T cell activation rely on complex biochemical signaling events. But T cell migration and activation also take place in distinct mechanical environments and lead to drastic morphological changes and reorganization of the acto-myosin cytoskeleton. In this review we discuss how adhesion proteins and the T cell receptor act as mechanosensors to translate these mechanical contexts into signaling events. We further discuss how cell tension could bring a significant contribution to the regulation of T cell signaling and function.
Collapse
Affiliation(s)
- Jérémie Rossy
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, Switzerland.,Department of Biology, University of Konstanz, Konstanz, Germany
| | - Julia M Laufer
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, Switzerland
| | - Daniel F Legler
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, Switzerland.,Department of Biology, University of Konstanz, Konstanz, Germany
| |
Collapse
|
23
|
Pettmann J, Santos AM, Dushek O, Davis SJ. Membrane Ultrastructure and T Cell Activation. Front Immunol 2018; 9:2152. [PMID: 30319617 PMCID: PMC6167458 DOI: 10.3389/fimmu.2018.02152] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/31/2018] [Indexed: 11/13/2022] Open
Abstract
The immune system serves as a crucial line of defense from infection and cancer, while also contributing to tissue homeostasis. Communication between immune cells is mediated by small soluble factors called cytokines, and also by direct cellular interactions. Cell-cell interactions are particularly important for T cell activation. T cells direct the adaptive immune response and therefore need to distinguish between self and foreign antigens. Even though decades have passed since the discovery of T cells, exactly why and how they are able to recognize and discriminate between antigens is still not fully understood. Early imaging of T cells was very successful in capturing the early stages of conjugate formation of T cells with antigen-presenting cells upon recognition of peptide-loaded major histocompatibility complexes by the T cell receptor (TCR). These studies lead to the discovery of a “supramolecular activation cluster” now known as the immunological synapse, followed by the identification of microclusters of TCRs formed upon receptor triggering, that eventually coalesce at the center of the synapse. New developments in light microscopy have since allowed attention to turn to the very earliest stages of T cell activation, and to resting cells, at high resolution. This includes single-molecule localization microscopy, which has been applied to the question of whether TCRs are pre-clustered on resting T cells, and lattice light-sheet microscopy that has enabled imaging of whole cells interacting with antigen-presenting cells. The utilization of lattice light-sheet microscopy has yielded important insights into structures called microvilli, which are small membrane protrusions on T cells that seem likely to have a large impact on T cell recognition and activation. Here we consider how imaging has shaped our thinking about T cell activation. We summarize recent findings obtained by applying more advanced microscopy techniques and discuss some of the limitations of these methods.
Collapse
Affiliation(s)
- Johannes Pettmann
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom.,Radcliffe Department of Medicine, Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Ana Mafalda Santos
- Radcliffe Department of Medicine, Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Simon J Davis
- Radcliffe Department of Medicine, Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
24
|
T cell microvilli constitute immunological synaptosomes that carry messages to antigen-presenting cells. Nat Commun 2018; 9:3630. [PMID: 30194420 PMCID: PMC6128830 DOI: 10.1038/s41467-018-06090-8] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 08/17/2018] [Indexed: 12/22/2022] Open
Abstract
Microvilli on T cells have been proposed to survey surfaces of antigen-presenting cells (APC) or facilitate adhesion under flow; however, whether they serve essential functions during T cell activation remains unclear. Here we show that antigen-specific T cells deposit membrane particles derived from microvilli onto the surface of cognate antigen-bearing APCs. Microvilli carry T cell receptors (TCR) at all stages of T cell activation and are released as large TCR-enriched, T cell microvilli particles (TMP) in a process of trogocytosis. These microvilli exclusively contain protein arrestin-domain-containing protein 1, which is directly involved in membrane budding and, in combination with vacuolar protein-sorting-associated protein 4, transforms large TMPs into smaller, exosome-sized TMPs. Notably, TMPs from CD4+ T cells are enriched with LFA-2/CD2 and various cytokines involved in activating dendritic cells. Collectively, these results demonstrate that T cell microvilli constitute “immunological synaptosomes” that carry T cell messages to APCs. Microvilli can participate in adhesion or migration of T cells, but whether they are involved in function regulation is unclear. Here the authors show that T cell microvilli form budding vesicles containing T cell signalling components for deposition onto antigen presenting cells (APC) and modulation of APC functions.
Collapse
|
25
|
Lamb CA, O'Byrne S, Keir ME, Butcher EC. Gut-Selective Integrin-Targeted Therapies for Inflammatory Bowel Disease. J Crohns Colitis 2018; 12:S653-S668. [PMID: 29767705 DOI: 10.1093/ecco-jcc/jjy060] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Integrins are cell surface receptors with bidirectional signalling capabilities that can bind to adhesion molecules in order to mediate homing of leukocytes to peripheral tissues. Gut-selective leukocyte homing is facilitated by interactions between α4β7 and its ligand, mucosal addressin cellular adhesion molecule-1 [MAdCAM-1], while retention of lymphocytes in mucosal tissues is mediated by αEβ7 binding to its ligand E-cadherin. Therapies targeting gut-selective trafficking have shown efficacy in inflammatory bowel disease [IBD], confirming the importance of leukocyte trafficking in disease pathobiology. This review will provide an overview of integrin structure, function and signalling, and highlight the role that these molecules play in leukocyte homing and retention. Anti-integrin therapeutics, including gut-selective antibodies against the β7 integrin subunit [etrolizumab] and the α4β7 integrin heterodimer [vedolizumab and abrilumab], and the non-gut selective anti-α4 integrin [natalizumab], will be discussed, as well as novel targeting approaches using small molecules.
Collapse
Affiliation(s)
- Christopher A Lamb
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK.,Department of Gastroenterology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Sharon O'Byrne
- Global Medical Affairs, Takeda Pharmaceuticals International AG, Zurich, Switzerland
| | - Mary E Keir
- Genentech Research & Early Development, South San Francisco, CA, USA
| | - Eugene C Butcher
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.,Veterans Affairs Palo Alto Health Care System and The Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| |
Collapse
|
26
|
AbuZineh K, Joudeh LI, Al Alwan B, Hamdan SM, Merzaban JS, Habuchi S. Microfluidics-based super-resolution microscopy enables nanoscopic characterization of blood stem cell rolling. SCIENCE ADVANCES 2018; 4:eaat5304. [PMID: 30035228 PMCID: PMC6051739 DOI: 10.1126/sciadv.aat5304] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/04/2018] [Indexed: 05/06/2023]
Abstract
Hematopoietic stem/progenitor cell (HSPC) homing occurs via cell adhesion mediated by spatiotemporally organized ligand-receptor interactions. Although molecules and biological processes involved in this multistep cellular interaction with endothelium have been studied extensively, molecular mechanisms of this process, in particular the nanoscale spatiotemporal behavior of ligand-receptor interactions and their role in the cellular interaction, remain elusive. We introduce a microfluidics-based super-resolution fluorescence imaging platform and apply the method to investigate the initial essential step in the homing, tethering, and rolling of HSPCs under external shear stress that is mediated by selectins, expressed on endothelium, with selectin ligands (that is, CD44) expressed on HSPCs. Our new method reveals transient nanoscale reorganization of CD44 clusters during cell rolling on E-selectin. We demonstrate that this mechanical force-induced reorganization is accompanied by a large structural reorganization of actin cytoskeleton. The CD44 clusters were partly disrupted by disrupting lipid rafts. The spatial reorganization of CD44 and actin cytoskeleton was not observed for the lipid raft-disrupted cells, demonstrating the essential role of the spatial clustering of CD44 on its reorganization during cell rolling. The lipid raft disruption causes faster and unstable cell rolling on E-selectin compared with the intact cells. Together, our results demonstrate that the spatial reorganization of CD44 and actin cytoskeleton is the result of concerted effect of E-selectin-ligand interactions, external shear stress, and spatial clustering of the selectin ligands, and has significant effect on the tethering/rolling step in HSPC homing. Our new experimental platform provides a foundation for characterizing complicated HSPC homing.
Collapse
Affiliation(s)
- Karmen AbuZineh
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Sciences and Engineering Division, Thuwal 23955-6900, Saudi Arabia
| | - Luay I. Joudeh
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Sciences and Engineering Division, Thuwal 23955-6900, Saudi Arabia
| | - Bader Al Alwan
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Sciences and Engineering Division, Thuwal 23955-6900, Saudi Arabia
| | - Samir M. Hamdan
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Sciences and Engineering Division, Thuwal 23955-6900, Saudi Arabia
| | | | - Satoshi Habuchi
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Sciences and Engineering Division, Thuwal 23955-6900, Saudi Arabia
| |
Collapse
|
27
|
Moreau HD, Piel M, Voituriez R, Lennon-Duménil AM. Integrating Physical and Molecular Insights on Immune Cell Migration. Trends Immunol 2018; 39:632-643. [PMID: 29779848 DOI: 10.1016/j.it.2018.04.007] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 04/18/2018] [Accepted: 04/18/2018] [Indexed: 01/09/2023]
Abstract
The function of most immune cells depends on their ability to migrate through complex microenvironments, either randomly to patrol for the presence of antigens or directionally to reach their next site of action. The actin cytoskeleton and its partners are key conductors of immune cell migration as they control the intrinsic migratory properties of leukocytes as well as their capacity to respond to cues present in their environment. In this review we focus on the latest discoveries regarding the role of the actomyosin cytoskeleton in optimizing immune cell migration in complex environments, with a special focus on recent insights provided by physical modeling.
Collapse
Affiliation(s)
- Hélène D Moreau
- INSERM U932, Institut Curie, ANR-10-IDEX-0001-02 PSL* and ANR-11-LABX-0043, Paris, France.
| | - Matthieu Piel
- Institut Curie, PSL Research University, CNRS, UMR 144, F-75005 Paris, France; Institut Pierre-Gilles de Gennes, PSL Research University, F-75005 Paris, France
| | - Raphaël Voituriez
- Laboratoire Jean Perrin, UM 8237 CNRS/UPMC, 4 place Jussieu, 75005 Paris, France
| | | |
Collapse
|
28
|
Cai E, Marchuk K, Beemiller P, Beppler C, Rubashkin MG, Weaver VM, Gérard A, Liu TL, Chen BC, Betzig E, Bartumeus F, Krummel MF. Visualizing dynamic microvillar search and stabilization during ligand detection by T cells. Science 2018; 356:356/6338/eaal3118. [PMID: 28495700 DOI: 10.1126/science.aal3118] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 03/17/2017] [Indexed: 12/11/2022]
Abstract
During immune surveillance, T cells survey the surface of antigen-presenting cells. In searching for peptide-loaded major histocompatibility complexes (pMHCs), they must solve a classic trade-off between speed and sensitivity. It has long been supposed that microvilli on T cells act as sensory organs to enable search, but their strategy has been unknown. We used lattice light-sheet and quantum dot-enabled synaptic contact mapping microscopy to show that anomalous diffusion and fractal organization of microvilli survey the majority of opposing surfaces within 1 minute. Individual dwell times were long enough to discriminate pMHC half-lives and T cell receptor (TCR) accumulation selectively stabilized microvilli. Stabilization was independent of tyrosine kinase signaling and the actin cytoskeleton, suggesting selection for avid TCR microclusters. This work defines the efficient cellular search process against which ligand detection takes place.
Collapse
Affiliation(s)
- En Cai
- Department of Pathology, University of California, San Francisco, CA 94143, USA
| | - Kyle Marchuk
- Department of Pathology, University of California, San Francisco, CA 94143, USA.,Biological Imaging Development Center, University of California, San Francisco, CA 94143, USA
| | - Peter Beemiller
- Department of Pathology, University of California, San Francisco, CA 94143, USA
| | - Casey Beppler
- Department of Pathology, University of California, San Francisco, CA 94143, USA
| | - Matthew G Rubashkin
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, CA 94143, USA
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, CA 94143, USA
| | - Audrey Gérard
- Department of Pathology, University of California, San Francisco, CA 94143, USA
| | - Tsung-Li Liu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Bi-Chang Chen
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Eric Betzig
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Frederic Bartumeus
- Center for Advanced Studies of Blanes (CEAB-CSIC), 17300 Girona, Spain.,Ecological and Forestry Applications Research Center (CREAF), 08193 Barcelona, Spain.,Institut Català de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, CA 94143, USA. .,Biological Imaging Development Center, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
29
|
Ivetic A. A head-to-tail view of L-selectin and its impact on neutrophil behaviour. Cell Tissue Res 2018; 371:437-453. [PMID: 29353325 PMCID: PMC5820395 DOI: 10.1007/s00441-017-2774-x] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 12/05/2017] [Indexed: 01/04/2023]
Abstract
L-selectin is a type I transmembrane cell adhesion molecule expressed on most circulating leukocytes, including neutrophils. Engagement of L-selectin with endothelial-derived ligands initiates neutrophil tethering and rolling behaviour along luminal walls of post-capillary venules, constituting the first step of the multi-step adhesion cascade. There is a large body of evidence to suggest that signalling downstream of L-selectin can influence neutrophil behaviour: adhesion, migration and priming. This review will cover aspects of L-selectin form and function and introduce the “triad of L-selectin regulation”, highlighting the inextricable links between adhesion, signalling and ectodomain shedding and also highlighting the cytosolic proteins that interconnect them. Recent advances in how L-selectin impacts priming, transendothelial migration (TEM) and cell polarity will also be discussed.
Collapse
Affiliation(s)
- Aleksandar Ivetic
- BHF Centre for Research Excellence, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, James Black Centre 125, Coldharbour Lane, London, SE5 9NU, UK.
| |
Collapse
|
30
|
Begandt D, Thome S, Sperandio M, Walzog B. How neutrophils resist shear stress at blood vessel walls: molecular mechanisms, subcellular structures, and cell-cell interactions. J Leukoc Biol 2017; 102:699-709. [PMID: 28619950 DOI: 10.1189/jlb.3mr0117-026rr] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 04/27/2017] [Accepted: 04/28/2017] [Indexed: 12/22/2022] Open
Abstract
Neutrophils are the first cells arriving at sites of tissue injury or infection to combat invading pathogens. Successful neutrophil recruitment to sites of inflammation highly depends on specific molecular mechanisms, fine-tuning the received information into signaling pathways and converting them into well-described recruitment steps. This review highlights the impact of vascular flow conditions on neutrophil recruitment and the multitude of mechanisms developed to enable this sophisticated process under wall shear stress conditions. The recruitment process underlies a complex interplay between adhesion and signaling molecules, as well as chemokines, in which neutrophils developed specific mechanisms to travel to sites of lesion in low and high shear stress conditions. Rolling, as the first step in the recruitment process, highly depends on endothelial selectins and their ligands on neutrophils, inducting of intracellular signaling and subsequently activating β2 integrins, enabling adhesion and postadhesion events. In addition, subcellular structures, such as microvilli, tethers, and slings allow the cell to arrest, even under high wall shear stress. Thereby, microvilli that are pulled out from the cell body form tethers that develop into slings upon their detachment from the substrate. In addition to the above-described primary capture, secondary capture of neutrophils via neutrophil-neutrophil or neutrophil-platelet interaction promotes the process of neutrophil recruitment to sites of lesion. Thus, precise mechanisms based on a complex molecular interplay, subcellular structures, and cell-cell interactions turn the delicate process of neutrophil trafficking during flow into a robust response allowing effective neutrophil accumulation at sites of injury.
Collapse
Affiliation(s)
- Daniela Begandt
- Walter Brendel Centre of Experimental Medicine, Department of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Sarah Thome
- Walter Brendel Centre of Experimental Medicine, Department of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Markus Sperandio
- Walter Brendel Centre of Experimental Medicine, Department of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Barbara Walzog
- Walter Brendel Centre of Experimental Medicine, Department of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany.
| |
Collapse
|
31
|
Dib K, Tikhonova IG, Ivetic A, Schu P. The cytoplasmic tail of L-selectin interacts with the adaptor-protein complex AP-1 subunit μ1A via a novel basic binding motif. J Biol Chem 2017; 292:6703-6714. [PMID: 28235798 DOI: 10.1074/jbc.m116.768598] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/30/2017] [Indexed: 01/02/2023] Open
Abstract
L-selectin regulates leukocyte adhesion and rolling along the endothelium. Proteins binding to the cytoplasmic tail of L-selectin regulate L-selectin functions. We used L-selectin cytoplasmic tail peptide pulldown assays combined with high sensitivity liquid chromatography/mass spectrometry to identify novel L-selectin tail-binding proteins. Incubation of the L-selectin tail with cell extracts from phorbol 12-myristate 13-acetate-stimulated Raw 264.7 macrophages resulted in the binding of μ1A of the clathrin-coated vesicle AP-1 complex. Furthermore, full-length GST-μ1A and the GST-μ1A C-terminal domain, but not the GST-μ1A N-terminal domain, bind to L-selectin tail peptide, and the intracellular pool of L-selectin colocalizes with AP-1 at the trans-Golgi network. We identified a novel basic protein motif consisting of a cluster of three dibasic residues (356RR357, 359KK360, and 362KK363) in the membrane-proximal domain of the L-selectin tail as well as a doublet of aspartic acid residues (369DD370) in the membrane-distal end of the L-selectin tail involved in μ1A binding. Stimulation of Raw 264.7 macrophages with PMA augmented the amount of μ1A associated with anti-L-selectin immunoprecipitates. However, full-length GST-μ1A did not bind to the phospho-L-selectin tail or phospho-mimetic S364D L-selectin tail. Accordingly, we propose that phosphorylation of μ1A is required for interaction with the L-selectin tail and that L-selectin tail phosphorylation may regulate this interaction in vivo Molecular docking of the L-selectin tail to μ1A was used to identify the μ1A surface domain binding the L-selectin tail and to explain how phosphorylation of the L-selectin tail abrogates μ1A interaction. Our findings indicate that L-selectin is transported constitutively by the AP-1 complex, leading to the formation of a trans-Golgi network reserve pool and that phosphorylation of the L-selectin tail blocks AP-1-dependent retrograde transport of L-selectin.
Collapse
Affiliation(s)
- Karim Dib
- From the Max Planck Institute for Biochemistry, Department of Molecular Medicine, 82152 Martinsried, Germany, .,the Center for Experimental Medicine, Queen's University Belfast, Belfast BT9 7BL, Northern Ireland, United Kingdom
| | - Irina G Tikhonova
- the School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, Northern Ireland, United Kingdom
| | - Aleksandar Ivetic
- the BHF Center for Research Excellence, Cardiovascular Division, Faculty of Life Sciences and Medicine, King's College London, London SE5 9NU, United Kingdom, and
| | - Peter Schu
- University Medical Center Göttingen, Department of Cellular Biochemistry, Georg-August University Göttingen, D-37073 Göttingen, Germany
| |
Collapse
|
32
|
Jung Y, Riven I, Feigelson SW, Kartvelishvily E, Tohya K, Miyasaka M, Alon R, Haran G. Three-dimensional localization of T-cell receptors in relation to microvilli using a combination of superresolution microscopies. Proc Natl Acad Sci U S A 2016; 113:E5916-E5924. [PMID: 27647916 PMCID: PMC5056101 DOI: 10.1073/pnas.1605399113] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Leukocyte microvilli are flexible projections enriched with adhesion molecules. The role of these cellular projections in the ability of T cells to probe antigen-presenting cells has been elusive. In this study, we probe the spatial relation of microvilli and T-cell receptors (TCRs), the major molecules responsible for antigen recognition on the T-cell membrane. To this end, an effective and robust methodology for mapping membrane protein distribution in relation to the 3D surface structure of cells is introduced, based on two complementary superresolution microscopies. Strikingly, TCRs are found to be highly localized on microvilli, in both peripheral blood human T cells and differentiated effector T cells, and are barely found on the cell body. This is a decisive demonstration that different types of T cells universally localize their TCRs to microvilli, immediately pointing to these surface projections as effective sensors for antigenic moieties. This finding also suggests how previously reported membrane clusters might form, with microvilli serving as anchors for specific T-cell surface molecules.
Collapse
Affiliation(s)
- Yunmin Jung
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Inbal Riven
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Sara W Feigelson
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | - Kazuo Tohya
- Department of Anatomy, Kansai University of Health Sciences, Kumatori, Osaka 590-0482, Japan
| | - Masayuki Miyasaka
- Institute for Academic Initiatives, Osaka University, Suita, Osaka 565-0871, Japan; MediCity Research Laboratory, University of Turku, FI-20521, Turku, Finland
| | - Ronen Alon
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Gilad Haran
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot 76100, Israel;
| |
Collapse
|
33
|
|
34
|
Hardy CL. Specificity of Hematopoietic Stem and Progenitor Cell Homing to Bone Marrow: A Perspective. Hematology 2016; 5:391-401. [DOI: 10.1080/10245332.2000.11746535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Cheryl L. Hardy
- G.V. (Sonny) Montgomery Department of Veterans Affairs Medical Center, Department of Medicine, University of Mississippi School of Medicine, Jackson, MS
| |
Collapse
|
35
|
Riese SB, Buscher K, Enders S, Kuehne C, Tauber R, Dernedde J. Structural requirements of mono- and multivalent L-selectin blocking aptamers for enhanced receptor inhibition in vitro and in vivo. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:901-908. [PMID: 26772426 DOI: 10.1016/j.nano.2015.12.379] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 08/28/2015] [Accepted: 12/24/2015] [Indexed: 01/08/2023]
Abstract
UNLABELLED L-selectin mediates extravasation of leukocytes from blood into the surrounding tissue during inflammation and is therefore a therapeutical target in certain overwhelming immune reactions. In this study, we characterized an L-selectin specific blocking DNA aptamer with respect to nucleotide composition and target binding. Introduction of deletions and nucleotide exchanges resulted in an optimized DNA sequence but preservation of the IC50 in the low nanomolar range. The inhibitory potential was significantly increased when the aptamer was displayed as a di- and trimer connected via appropriate linker length. Similar to monoclonal antibodies, trimer yielded picomolar IC50 values in a competitive binding assay. In comparison to the monovalent aptamer, the trivalent assembly reduced PBMC interactions to L-selectin ligands 90-fold under shear and exerted superior inhibition of PBMC rolling in vivo. In conclusion, our work demonstrates the feasibility of optimizing aptamer sequences and shows that multivalent ligand presentation enables superior adhesion receptor targeting. FROM THE CLINICAL EDITOR During inflammation, leukocytes extravasate from blood vessels under chemotaxic signals. The presence of L-selectin on endothelium acts as a mediator for the extravasation process. In this study, the authors investigated an L-selectin specific blocking DNA aptamer in various forms, as inhibitors to leukocyte binding and extravasation. This new approach confirmed the potential use of aptamers in clinical setting.
Collapse
Affiliation(s)
- Sebastian B Riese
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Konrad Buscher
- Institute for Physiological Chemistry Pathobiochemistry, University of Muenster, Muenster, Germany; Department of Nephrology and Rheumatology, University Hospital Muenster, Muenster, Germany.
| | - Sven Enders
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Christian Kuehne
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Rudolf Tauber
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Jens Dernedde
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
36
|
Cohen M. Notable Aspects of Glycan-Protein Interactions. Biomolecules 2015; 5:2056-72. [PMID: 26340640 PMCID: PMC4598788 DOI: 10.3390/biom5032056] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 08/27/2015] [Accepted: 08/27/2015] [Indexed: 01/01/2023] Open
Abstract
This mini review highlights several interesting aspects of glycan-mediated interactions that are common between cells, bacteria, and viruses. Glycans are ubiquitously found on all living cells, and in the extracellular milieu of multicellular organisms. They are known to mediate initial binding and recognition events of both immune cells and pathogens with their target cells or tissues. The host target tissues are hidden under a layer of secreted glycosylated decoy targets. In addition, pathogens can utilize and display host glycans to prevent identification as foreign by the host’s immune system (molecular mimicry). Both the host and pathogens continually evolve. The host evolves to prevent infection and the pathogens evolve to evade host defenses. Many pathogens express both glycan-binding proteins and glycosidases. Interestingly, these proteins are often located at the tip of elongated protrusions in bacteria, or in the leading edge of the cell. Glycan-protein interactions have low affinity and, as a result, multivalent interactions are often required to achieve biologically relevant binding. These enable dynamic forms of adhesion mechanisms, reviewed here, and include rolling (cells), stick and roll (bacteria) or surfacing (viruses).
Collapse
Affiliation(s)
- Miriam Cohen
- Depatment of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, BRF2 MC 0687, La Jolla, CA 92093-0687, USA.
| |
Collapse
|
37
|
Arita-Okubo S, Kim-Kaneyama JR, Lei XF, Fu WG, Ohnishi K, Takeya M, Miyauchi A, Honda H, Itabe H, Miyazaki T, Miyazaki A. Role of Hic-5 in the formation of microvilli-like structures and the monocyte-endothelial interaction that accelerates atherosclerosis. Cardiovasc Res 2015; 105:361-71. [PMID: 25587044 DOI: 10.1093/cvr/cvv003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
AIMS The adhesion of circulating monocytes to endothelial cells (ECs) is an early and critical event in the formation of atherosclerotic plaques. Hydrogen peroxide-inducible clone 5 (Hic-5) serves as an adaptor molecule in cell adhesion complexes. However, the role of endothelial Hic-5 in monocyte-EC interaction and atherogenesis remains unclear. We examined the roles of endothelial Hic-5 in monocyte-EC interaction and atherogenesis using mouse models of atherosclerosis and cultured human umbilical vein endothelial cells (HUVECs). METHODS AND RESULTS Hic-5 was expressed in ECs, but not in monocytes/macrophages. An ex vivo monocyte adhesion assay revealed that adhesion of THP-1 monocytes to aortas isolated from Apoe(-/-) and LDLR(-/-) mice stimulated by TNF-α or oxidized LDL was suppressed by Hic-5 deficiency. Scanning electron microscopic observations of aortas harvested from Apoe(-/-) mice revealed that TNF-α- or oxidized LDL-induced microvilli-like structures were markedly suppressed by Hic-5 deficiency. Relative Hic-5 deficiency suppressed 60% of the atherosclerotic lesions in aortas from Apoe(-/-) and LDLR(-/-) mice. In contrast, overexpression of Hic-5 in HUVECs promoted induction of microvilli-like structures and adherence of THP-1 cells in an adhesion receptor such as intercellular adhesion molecule-1- and vascular cell adhesion molecule-1-dependent manner. CONCLUSION Hic-5 in ECs plays an important role in the formation of microvilli-like structures and in the interaction between ECs and monocytes, leading to monocyte recruitment and subsequent development of atherosclerosis.
Collapse
Affiliation(s)
- Shigeko Arita-Okubo
- Department of Biochemistry, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Joo-Ri Kim-Kaneyama
- Department of Biochemistry, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Xiao-Feng Lei
- Department of Biochemistry, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Wen-Guang Fu
- Department of Biochemistry, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Koji Ohnishi
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Motohiro Takeya
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Aya Miyauchi
- Department of Biochemistry, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Hirokazu Honda
- Division of Nephrology, Department of Medicine, Showa University Koto Toyosu Hospital, Tokyo, Japan
| | - Hiroyuki Itabe
- Division of Biological Chemistry, Department of Molecular Biology, Showa University School of Pharmacy, Tokyo, Japan
| | - Takuro Miyazaki
- Department of Biochemistry, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Akira Miyazaki
- Department of Biochemistry, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| |
Collapse
|
38
|
Wang Y, Yago T, Zhang N, Abdisalaam S, Alexandrakis G, Rodgers W, McEver RP. Cytoskeletal regulation of CD44 membrane organization and interactions with E-selectin. J Biol Chem 2014; 289:35159-71. [PMID: 25359776 DOI: 10.1074/jbc.m114.600767] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Interactions of CD44 on neutrophils with E-selectin on activated endothelial cells mediate rolling under flow, a prerequisite for neutrophil arrest and migration into perivascular tissues. How CD44 functions as a rolling ligand despite its weak affinity for E-selectin is unknown. We examined the nanometer scale organization of CD44 on intact cells. CD44 on leukocytes and transfected K562 cells was cross-linked within a 1.14-nm spacer. Depolymerizing actin with latrunculin B reduced cross-linking. Fluorescence resonance energy transfer (FRET) revealed tight co-clustering between CD44 fused to yellow fluorescent protein (YFP) and CD44 fused to cyan fluorescent protein on K562 cells. Latrunculin B reduced FRET-reported co-clustering. Number and brightness analysis confirmed actin-dependent CD44-YFP clusters on living cells. CD44 lacking binding sites for ankyrin and for ezrin/radixin/moesin (ERM) proteins on its cytoplasmic domain (ΔANKΔERM) did not cluster. Unexpectedly, CD44 lacking only the ankyrin-binding site (ΔANK) formed larger but looser clusters. Fluorescence recovery after photobleaching demonstrated increased CD44 mobility by latrunculin B treatment or by deleting the cytoplasmic domain. ΔANKΔERM mobility increased only modestly, suggesting that the cytoplasmic domain engages the cytoskeleton by an additional mechanism. Ex vivo differentiated CD44-deficient neutrophils expressing exogenous CD44 rolled on E-selectin and activated Src kinases after binding anti-CD44 antibody. In contrast, differentiated neutrophils expressing ΔANK had impaired rolling and kinase activation. These data demonstrate that spectrin and actin networks regulate CD44 clustering and suggest that ankyrin enhances CD44-mediated neutrophil rolling and signaling.
Collapse
Affiliation(s)
- Ying Wang
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104 and
| | - Tadayuki Yago
- From the Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation and
| | - Nan Zhang
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104 and
| | - Salim Abdisalaam
- Department of Biomedical Engineering, University of Texas, Arlington, Texas 76010
| | - George Alexandrakis
- Department of Biomedical Engineering, University of Texas, Arlington, Texas 76010
| | - William Rodgers
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104 and
| | - Rodger P McEver
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104 and From the Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation and
| |
Collapse
|
39
|
Abeynaike LD, Deane JA, Westhorpe CLV, Chow Z, Alikhan MA, Kitching AR, Issekutz A, Hickey MJ. Regulatory T cells dynamically regulate selectin ligand function during multiple challenge contact hypersensitivity. THE JOURNAL OF IMMUNOLOGY 2014; 193:4934-44. [PMID: 25274531 DOI: 10.4049/jimmunol.1400641] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Regulatory T cells (Tregs) play critical roles in restricting T cell-mediated inflammation. In the skin, this is dependent on expression of selectin ligands required for leukocyte rolling in dermal microvessels. However, whether there are differences in the molecules used by Tregs and proinflammatory T cells to undergo rolling in the skin remains unclear. In this study, we used spinning disk confocal microscopy in Foxp3-GFP mice to visualize rolling of endogenous Tregs in dermal postcapillary venules. Tregs underwent consistent but low-frequency rolling interactions under resting and inflamed conditions. At the early stage of the response, Treg adhesion was minimal. However, at the peak of inflammation, Tregs made up 40% of the adherent CD4(+) T cell population. In a multiple challenge model of contact hypersensitivity, rolling of Tregs and conventional CD4(+) T cells was mostly dependent on overlapping contributions of P- and E-selectin. However, after a second challenge, rolling of Tregs but not conventional CD4(+) T cells became P-selectin independent, and Tregs showed reduced capacity to bind P-selectin. Moreover, inhibition of E-selectin at this time point resulted in exacerbation of inflammation. These findings demonstrate that in this multiple challenge model of inflammation, Treg selectin binding capacity and the molecular basis of Treg rolling can be regulated dynamically.
Collapse
Affiliation(s)
- Latasha D Abeynaike
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria 3168, Australia
| | - James A Deane
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria 3168, Australia
| | - Clare L V Westhorpe
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria 3168, Australia
| | - Zachary Chow
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria 3168, Australia
| | - Maliha A Alikhan
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria 3168, Australia
| | - A Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria 3168, Australia; Department of Nephrology, Monash Medical Centre, Clayton, Victoria 3168, Australia; Department of Pediatric Nephrology, Monash Medical Centre, Clayton, Victoria 3168, Australia
| | - Andrew Issekutz
- Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada B3K 6R8; Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4R2; and Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4R2
| | - Michael J Hickey
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria 3168, Australia;
| |
Collapse
|
40
|
CD147 (EMMPRIN/Basigin) in kidney diseases: from an inflammation and immune system viewpoint. Nephrol Dial Transplant 2014; 30:1097-103. [DOI: 10.1093/ndt/gfu302] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 08/18/2014] [Indexed: 11/15/2022] Open
|
41
|
Gjurich BN, Taghavie-Moghadam PL, Ley K, Galkina EV. L-selectin deficiency decreases aortic B1a and Breg subsets and promotes atherosclerosis. Thromb Haemost 2014; 112:803-11. [PMID: 24989887 DOI: 10.1160/th13-10-0865] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 04/29/2014] [Indexed: 12/18/2022]
Abstract
There is a significant recruitment of leucocytes into aortas during atherogenesis. L-selectin regulates leucocyte migration into secondary lymphoid and peripheral tissues and was proposed to play a role in leucocyte homing into aortas. Here, we determine the role of L-selectin in atherosclerosis. L-selectin-deficient Apoe-/- (Sell-/-Apoe-/-) mice had a 74% increase in plaque burden compared to Apoe-/- mice fed a chow diet for 50 weeks. Elevated atherosclerosis was accompanied by increased aortic leucocyte content, but a 50% reduction in aortic B cells despite elevated B cell counts in the blood. Follicular B cells represented 65%, whereas B1a and regulatory B cells (Breg) comprised 5% of aortic B cells. B1a and Breg cell subsets were reduced in Sell-/-Apoe-/- aortas with accompanied two-fold decrease in aortic T15 antibody and 1.2-fold decrease of interleukin-10 (IL-10) levels. L-selectin was required for B1 cell homing to the atherosclerotic aorta, as demonstrated by a 1.5-fold decrease in the migration of Sell-/-Apoe-/- vs Apoe-/- cells. Notably, we found a 1.6-fold increase in CD68hi macrophages in Sell-/-Apoe-/- compared to Apoe-/- aortas, despite comparable blood monocyte numbers and L-selectin-dependent aortic homing. L-selectin had no effect on neutrophil migration into aorta, but led to elevated blood neutrophil numbers, suggesting a potential involvement of neutrophils in atherogenesis of Sell-/-Apoe-/- mice. Thus, L-selectin deficiency increases peripheral blood neutrophil and lymphocyte numbers, decreases aortic B1a and Breg populations, T15 antibody and IL-10 levels, and increases aortic macrophage content of Sell-/-Apoe-/- mice. Altogether, these data provide evidence for an overall atheroprotective role of L-selectin.
Collapse
Affiliation(s)
| | | | | | - Elena V Galkina
- Elena V. Galkina, PhD, Associate Professor, Dept. Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Road, Norfolk, VA 23507-1696, USA, Tel.: +1 757 446 5019, Fax: +1 757 624 2255, E-mail:
| |
Collapse
|
42
|
Luo J, Li C, Xu T, Liu W, Ba X, Wang X, Zeng X. PI3K is involved in β1 integrin clustering by PSGL-1 and promotes β1 integrin-mediated Jurkat cell adhesion to fibronectin. Mol Cell Biochem 2014; 385:287-95. [PMID: 24122451 DOI: 10.1007/s11010-013-1837-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 09/26/2013] [Indexed: 10/26/2022]
Abstract
P-selectin glycoprotein ligand-1 (PSGL-1) is involved in the initial step of lymphocyte homing by interacting with P- or E-selectins expressed on activated endothelium cells. Besides, it also functions as a receptor to induce signals that increase integrin affinity to ligands and mediate cell adhesion to endothelium. Integrin is required for the second step of lymphocyte homing, whose activation has been reported tightly regulated by inside-out signals triggered by chemokines or the shear-stress generated during lymphocyte rolling on endothelium. However, the relationship between PSGL-1-triggered signals and integrin activation is not clear. In this study, we demonstrated that PSGL-1 ligation induces β1 integrin-mediated adhesion to fibronectin via regulation of both β1 subunit clustering and conformation changes. Phosphoinositide 3-kinase (PI3K) is required for PSGL-1-induced β1 integrin clustering which ultimately regulates β1 integrin-mediated Jurkat cell adhesion to fibronectin. However, PI3K is not involved in the conformation changes or increases in the total expression of β1 integrin. Taken together, we found a novel signal pathway, PSGL-1-PI3K-β1 integrin, demonstrating the cooperation between initial adhesion and subsequent arrest and stable adhesion.
Collapse
|
43
|
Riese SB, Kuehne C, Tedder TF, Hallmann R, Hohenester E, Buscher K. Heterotropic modulation of selectin affinity by allosteric antibodies affects leukocyte rolling. THE JOURNAL OF IMMUNOLOGY 2014; 192:1862-9. [PMID: 24431230 DOI: 10.4049/jimmunol.1302147] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Selectins are a family of adhesion receptors designed for efficient leukocyte tethering to the endothelium under shear. As a key property to resist premature bond disruption, selectin adhesiveness is enhanced by tensile forces that promote the conversion of a bent into an extended conformation of the N-terminal lectin and epidermal growth factor-like domains. Conformation-specific Abs have been invaluable in deciphering the activation mechanism of integrins, but similar reagents are not available for selectins. In this study, we show that the anti-human L-selectin mAbs DREG-55 and LAM1-5 but not DREG-56, DREG-200, or LAM1-1 heterotropically modulate adhesion presumably by stabilizing the extended receptor conformation. Force-free affinity assays, flow chamber, and microkinetic studies reveal a ligand-specific modulation of L-selectin affinity by DREG-55 mAb, resulting in a dramatic decrease of rolling velocity under flow. Furthermore, secondary tethering of polymorphonuclear cells was blocked by DREG-200 but significantly boosted by DREG-55 mAb. The results emphasize the need for a new classification for selectin Abs and introduce the new concept of heterotropic modulation of receptor function.
Collapse
Affiliation(s)
- Sebastian B Riese
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-University of Medicine Berlin, 10117 Berlin, Germany
| | | | | | | | | | | |
Collapse
|
44
|
Selectins and their ligands are required for homing and engraftment of BCR-ABL1+ leukemic stem cells in the bone marrow niche. Blood 2014; 123:1361-71. [PMID: 24394666 DOI: 10.1182/blood-2013-11-538694] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
We investigated adhesion pathways that contribute to engraftment of breakpoint cluster region-Abelson murine leukemia viral oncogene homolog 1 (BCR-ABL1)-induced chronic myelogenous leukemia (CML)-like myeloproliferative neoplasia in a mouse retroviral transduction/transplantation model. Compared with normal stem/progenitor cells, BCR-ABL1(+) progenitors had similar expression of very late antigen-4 (VLA4), VLA5, leukocyte functional antigen-1, and CXCR4 but lower expression of P-selectin glycoprotein ligand-1 (PSGL-1) and of L-selectin. Whereas vascular cell adhesion molecule-1 and P-selectin were not required, deficiency of E-selectin in the recipient bone marrow endothelium significantly reduced engraftment by BCR-ABL1-expressing stem cells following intravenous injection, with leukemogenesis restored by direct intrafemoral injection. BCR-ABL1-expressing cells deficient for PSGL-1 or the selectin ligand-synthesizing enzymes core-2 β1,6-N-acetylglucosaminyltransferase or fucosyltransferases IV/VII were impaired for engraftment, and destruction of selectin ligands on leukemic progenitors by neuraminidase reduced engraftment. BCR-ABL1-expressing L-selectin-deficient progenitors were also defective in homing and engraftment, with leukemogenesis rescued by coexpression of chimeric E/L-selectin. Antibody to L-selectin decreased the engraftment of BCR-ABL1-transduced stem cells. These results establish that BCR-ABL1(+) leukemic stem cells rely to a greater extent on selectins and their ligands for homing and engraftment than do normal stem cells. Selectin blockade is a novel strategy to exploit differences between normal and leukemic stem cells that may be beneficial in autologous transplantation for CML and perhaps other leukemias.
Collapse
|
45
|
Kawashima H. Analysis of L-selectin-mediated cellular interactions under flow conditions. Methods Mol Biol 2014; 1200:401-12. [PMID: 25117254 DOI: 10.1007/978-1-4939-1292-6_35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Lymphocyte homing is mediated by a specific interaction between L-selectin expressed on lymphocytes and its ligands expressed on high endothelial venules (HEVs) in lymph nodes under physiological flow conditions. In this chapter, two methods for detecting L-selectin-mediated cellular interactions under shear stress mimicking physiological flow conditions are described. First, a modified Stamper-Woodruff cell-binding assay using leukocytes labeled with a fluorescent orange dye, CMTMR, is introduced. In this method, leukocytes are allowed to bind to frozen lymph node sections under shear stress and their binding to HEVs can be clearly visualized by fluorescence microscopy. Second, a parallel flow chamber assay is described. In this assay, leukocytes are allowed to roll on L-selectin ligand-expressing cells under various levels of shear stress and their adhesive interactions are recorded by a video camera equipped with an inverted microscope. These methods can be applied to determine the effects of various agents that might affect L-selectin-mediated lymphocyte homing and recruitment.
Collapse
Affiliation(s)
- Hiroto Kawashima
- Department of Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, 142-8501, Tokyo, Japan,
| |
Collapse
|
46
|
Dimasi D, Sun WY, Bonder CS. Neutrophil interactions with the vascular endothelium. Int Immunopharmacol 2013; 17:1167-75. [DOI: 10.1016/j.intimp.2013.05.034] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 05/31/2013] [Indexed: 01/13/2023]
|
47
|
Liu B, Shao JY. Tangential tether extraction and spontaneous tether retraction of human neutrophils. Biophys J 2013; 103:2257-64. [PMID: 23283224 DOI: 10.1016/j.bpj.2012.10.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 10/11/2012] [Accepted: 10/17/2012] [Indexed: 12/26/2022] Open
Abstract
Membrane tethers are extracted when neutrophils roll on the endothelium to initiate their transendothelial migration. Tether extraction from both neutrophils and endothelial cells stabilizes neutrophil rolling, so it has been studied extensively and the force-velocity relationship for tether extraction is of great interest. Due to limitations of the techniques used in previous studies, this relationship has been obtained only from tethers perpendicular to the cell surface. Here, with the microcantilever technique, where latex beads affixed on silicon cantilevers were used as the force transducer, we extracted tethers either perpendicular or tangential to the neutrophil surface. We found that the force-velocity relationship was not sensitive to tether pulling direction. Little movement of the tether-cell junction was observed during tangential tether extraction, and no coalescence was observed during multiple tether extraction. Following adhesion rupture, spontaneous tether retraction was visualized by membrane staining, which revealed two phases: one was fast and exponential, whereas the other was slow and linear. Both phases can be reproduced with a mechanical model. These results show for the first time, to our knowledge, how neutrophil tethers shorten upon instantaneous force removal, and furthermore, they illustrate how membrane tethers contribute to neutrophil rolling stability during the inflammatory response.
Collapse
Affiliation(s)
- Baoyu Liu
- Department of Biomedical Engineering, Washington University, Saint Louis, MO, USA
| | | |
Collapse
|
48
|
Luo ZY, Wang SQ, He L, Lu TJ, Xu F, Bai BF. Front tracking simulation of cell detachment dynamic mechanism in microfluidics. Chem Eng Sci 2013. [DOI: 10.1016/j.ces.2013.04.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
49
|
Murai T, Sato C, Sato M, Nishiyama H, Suga M, Mio K, Kawashima H. Membrane cholesterol modulates the hyaluronan-binding ability of CD44 in T lymphocytes and controls rolling under shear flow. J Cell Sci 2013; 126:3284-94. [PMID: 23729731 DOI: 10.1242/jcs.120014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The adhesion of circulating lymphocytes to the surface of vascular endothelial cells is important for their recruitment from blood to secondary lymphoid organs and to inflammatory sites. CD44 is a key adhesion molecule for this interaction and its ligand-binding ability is tightly regulated. Here we show that the hyaluronan-binding ability of CD44 in T cells is upregulated by the depletion of membrane cholesterol with methyl-β-cyclodextrin (MβCD), which disintegrates lipid rafts, i.e. cholesterol- and sphingolipid-enriched membrane microdomains. Increasing concentrations of MβCD led to a dose-dependent decrease in cellular cholesterol content and to upregulation of hyaluronan binding. Additionally, a cholesterol-binding agent filipin also increased hyaluronan binding. Cholesterol depletion caused CD44 to be dispersed from cholesterol-enriched membrane microdomains. Cholesterol depletion also increased the number of cells undergoing rolling adhesion under physiological flow conditions. Our results suggest that the ligand-binding ability of CD44 is governed by its cholesterol-dependent allocation to membrane microdomains at the cell surface. These findings provide novel insight into the regulation of T cell adhesion under blood flow.
Collapse
Affiliation(s)
- Toshiyuki Murai
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan.
| | | | | | | | | | | | | |
Collapse
|
50
|
Zhang Y, Jiang N, Zarnitsyna VI, Klopocki AG, McEver RP, Zhu C. P-selectin glycoprotein ligand-1 forms dimeric interactions with E-selectin but monomeric interactions with L-selectin on cell surfaces. PLoS One 2013; 8:e57202. [PMID: 23451187 PMCID: PMC3581448 DOI: 10.1371/journal.pone.0057202] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 01/18/2013] [Indexed: 11/19/2022] Open
Abstract
Interactions of selectins with cell surface glycoconjugates mediate the first step of the adhesion and signaling cascade that recruits circulating leukocytes to sites of infection or injury. P-selectin dimerizes on the surface of endothelial cells and forms dimeric bonds with P-selectin glycoprotein ligand-1 (PSGL-1), a homodimeric sialomucin on leukocytes. It is not known whether leukocyte L-selectin or endothelial cell E-selectin are monomeric or oligomeric. Here we used the micropipette technique to analyze two-dimensional binding of monomeric or dimeric L- and E-selectin with monomeric or dimeric PSGL-1. Adhesion frequency analysis demonstrated that E-selectin on human aortic endothelial cells supported dimeric interactions with dimeric PSGL-1 and monomeric interactions with monomeric PSGL-1. In contrast, L-selectin on human neutrophils supported monomeric interactions with dimeric or monomeric PSGL-1. Our work provides a new method to analyze oligomeric cross-junctional molecular binding at the interface of two interacting cells.
Collapse
Affiliation(s)
- Yan Zhang
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | | | | | | | | | | |
Collapse
|