101
|
Liu X, Wei Y, Li W, Li B, Liu L. Cytoskeleton induced the changes of microvilli and mechanical properties in living cells by atomic force microscopy. J Cell Physiol 2020; 236:3725-3733. [PMID: 33169846 DOI: 10.1002/jcp.30110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/08/2020] [Accepted: 10/05/2020] [Indexed: 01/05/2023]
Abstract
The cytoskeleton acts as a scaffold for membrane protrusion, such as microvilli. However, the relationship between the characteristics of microvilli and cytoskeleton remains poorly understood under the physiological state. To investigate the role of the cytoskeleton in regulating microvilli and cellular mechanical properties, atomic force microscopy (AFM) was used to detect the dynamic characteristics of microvillus morphology and elastic modulus of living HeLa cells. First, HeLa and MCF-7 cell lines were stained with Fluor-488-phalloidin and microtubules antibody. Then, the microvilli morphology was analyzed by high-resolution images of AFM in situ. Furthermore, changes in elastic modulus were investigated by the force curve of AFM. Fluorescence microscopy and AFM results revealed that destroyed microfilaments led to a smaller microvilli size, whereas the increase in the aggregation and number of microfilaments led to a larger microvilli size. The destruction and aggregation of microfilaments remarkably affected the mechanical properties of HeLa cells. Microtubule-related drugs induced the change of microtubule, but we failed to note significant differences in microvilli morphology and mechanical properties of cells. In summary, our results unraveled the relationship between microfilaments and the structure of microvilli and Young's modulus in living HeLa cells, which would contribute to the further understanding of the physiological function of the cytoskeleton in vivo.
Collapse
Affiliation(s)
- Xueyan Liu
- Key Laboratory of Medicine, School of Laboratory Medicine and Life Sciences, Ministry of Education of China, Wenzhou Medical University, Wenzhou, China
| | - Yuhui Wei
- Division of Physical Biology and Bioimaging Centre, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Chinese Academy of Sciences, Shanghai Institute of Applied Physics, Shanghai, China.,Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Wei Li
- Key Laboratory of Medicine, School of Laboratory Medicine and Life Sciences, Ministry of Education of China, Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Medical Genetics, Wenzhou Medical University, Wenzhou, China
| | - Bin Li
- Division of Physical Biology and Bioimaging Centre, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Chinese Academy of Sciences, Shanghai Institute of Applied Physics, Shanghai, China.,Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Lin Liu
- Key Laboratory of Medicine, School of Laboratory Medicine and Life Sciences, Ministry of Education of China, Wenzhou Medical University, Wenzhou, China.,Division of Physical Biology and Bioimaging Centre, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Chinese Academy of Sciences, Shanghai Institute of Applied Physics, Shanghai, China.,Zhejiang Provincial Key Laboratory of Medical Genetics, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
102
|
Mastrogiovanni M, Juzans M, Alcover A, Di Bartolo V. Coordinating Cytoskeleton and Molecular Traffic in T Cell Migration, Activation, and Effector Functions. Front Cell Dev Biol 2020; 8:591348. [PMID: 33195256 PMCID: PMC7609836 DOI: 10.3389/fcell.2020.591348] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/24/2020] [Indexed: 12/28/2022] Open
Abstract
Dynamic localization of receptors and signaling molecules at the plasma membrane and within intracellular vesicular compartments is crucial for T lymphocyte sensing environmental cues, triggering membrane receptors, recruiting signaling molecules, and fine-tuning of intracellular signals. The orchestrated action of actin and microtubule cytoskeleton and intracellular vesicle traffic plays a key role in all these events that together ensure important steps in T cell physiology. These include extravasation and migration through lymphoid and peripheral tissues, T cell interactions with antigen-presenting cells, T cell receptor (TCR) triggering by cognate antigen-major histocompatibility complex (MHC) complexes, immunological synapse formation, cell activation, and effector functions. Cytoskeletal and vesicle traffic dynamics and their interplay are coordinated by a variety of regulatory molecules. Among them, polarity regulators and membrane-cytoskeleton linkers are master controllers of this interplay. Here, we review the various ways the T cell plasma membrane, receptors, and their signaling machinery interplay with the actin and microtubule cytoskeleton and with intracellular vesicular compartments. We highlight the importance of this fine-tuned crosstalk in three key stages of T cell biology involving cell polarization: T cell migration in response to chemokines, immunological synapse formation in response to antigen cues, and effector functions. Finally, we discuss two examples of perturbation of this interplay in pathological settings, such as HIV-1 infection and mutation of the polarity regulator and tumor suppressor adenomatous polyposis coli (Apc) that leads to familial polyposis and colorectal cancer.
Collapse
Affiliation(s)
- Marta Mastrogiovanni
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
- Collège Doctoral, Sorbonne Université, Paris, France
| | - Marie Juzans
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Andrés Alcover
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Vincenzo Di Bartolo
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
| |
Collapse
|
103
|
Mørch AM, Bálint Š, Santos AM, Davis SJ, Dustin ML. Coreceptors and TCR Signaling - the Strong and the Weak of It. Front Cell Dev Biol 2020; 8:597627. [PMID: 33178706 PMCID: PMC7596257 DOI: 10.3389/fcell.2020.597627] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 09/28/2020] [Indexed: 12/02/2022] Open
Abstract
The T-cell coreceptors CD4 and CD8 have well-characterized and essential roles in thymic development, but how they contribute to immune responses in the periphery is unclear. Coreceptors strengthen T-cell responses by many orders of magnitude - beyond a million-fold according to some estimates - but the mechanisms underlying these effects are still debated. T-cell receptor (TCR) triggering is initiated by the binding of the TCR to peptide-loaded major histocompatibility complex (pMHC) molecules on the surfaces of other cells. CD4 and CD8 are the only T-cell proteins that bind to the same pMHC ligand as the TCR, and can directly associate with the TCR-phosphorylating kinase Lck. At least three mechanisms have been proposed to explain how coreceptors so profoundly amplify TCR signaling: (1) the Lck recruitment model and (2) the pseudodimer model, both invoked to explain receptor triggering per se, and (3) two-step coreceptor recruitment to partially triggered TCRs leading to signal amplification. More recently it has been suggested that, in addition to initiating or augmenting TCR signaling, coreceptors effect antigen discrimination. But how can any of this be reconciled with TCR signaling occurring in the absence of CD4 or CD8, and with their interactions with pMHC being among the weakest specific protein-protein interactions ever described? Here, we review each theory of coreceptor function in light of the latest structural, biochemical, and functional data. We conclude that the oldest ideas are probably still the best, i.e., that their weak binding to MHC proteins and efficient association with Lck allow coreceptors to amplify weak incipient triggering of the TCR, without comprising TCR specificity.
Collapse
Affiliation(s)
- Alexander M. Mørch
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Štefan Bálint
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Ana Mafalda Santos
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Simon J. Davis
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Michael L. Dustin
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
104
|
Demetriou P, Abu-Shah E, Valvo S, McCuaig S, Mayya V, Kvalvaag A, Starkey T, Korobchevskaya K, Lee LYW, Friedrich M, Mann E, Kutuzov MA, Morotti M, Wietek N, Rada H, Yusuf S, Afrose J, Siokis A, Meyer-Hermann M, Ahmed AA, Depoil D, Dustin ML. A dynamic CD2-rich compartment at the outer edge of the immunological synapse boosts and integrates signals. Nat Immunol 2020; 21:1232-1243. [PMID: 32929275 PMCID: PMC7611174 DOI: 10.1038/s41590-020-0770-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 07/28/2020] [Indexed: 12/18/2022]
Abstract
The CD2-CD58 recognition system promotes adhesion and signaling and counters exhaustion in human T cells. We found that CD2 localized to the outer edge of the mature immunological synapse, with cellular or artificial APC, in a pattern we refer to as a 'CD2 corolla'. The corolla captured engaged CD28, ICOS, CD226 and SLAM-F1 co-stimulators. The corolla amplified active phosphorylated Src-family kinases (pSFK), LAT and PLC-γ over T cell receptor (TCR) alone. CD2-CD58 interactions in the corolla boosted signaling by 77% as compared with central CD2-CD58 interactions. Engaged PD-1 invaded the CD2 corolla and buffered CD2-mediated amplification of TCR signaling. CD2 numbers and motifs in its cytoplasmic tail controlled corolla formation. CD8+ tumor-infiltrating lymphocytes displayed low expression of CD2 in the majority of people with colorectal, endometrial or ovarian cancer. CD2 downregulation may attenuate antitumor T cell responses, with implications for checkpoint immunotherapies.
Collapse
Affiliation(s)
| | - Enas Abu-Shah
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Salvatore Valvo
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Sarah McCuaig
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Viveka Mayya
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Skirball Institute of Biomolecular Medicine, New York University of School of Medicine, New York, NY, USA
| | - Audun Kvalvaag
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Thomas Starkey
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | | | - Lennard Y W Lee
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | | | - Elizabeth Mann
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Mikhail A Kutuzov
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Matteo Morotti
- Ovarian Cancer Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Nina Wietek
- Ovarian Cancer Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Heather Rada
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Shamsideen Yusuf
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Jehan Afrose
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Skirball Institute of Biomolecular Medicine, New York University of School of Medicine, New York, NY, USA
- Department of Medical Laboratory Sciences, CUNY Hunter College, New York, NY, USA
| | - Anastasios Siokis
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology (BRICS), Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology (BRICS), Helmholtz Centre for Infection Research, Braunschweig, Germany
- Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Ahmed Ashour Ahmed
- Ovarian Cancer Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - David Depoil
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Skirball Institute of Biomolecular Medicine, New York University of School of Medicine, New York, NY, USA
- Immunocore Ltd, Abingdon, Oxford, UK
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
- Skirball Institute of Biomolecular Medicine, New York University of School of Medicine, New York, NY, USA.
| |
Collapse
|
105
|
Razvag Y, Neve-Oz Y, Sajman J, Yakovian O, Reches M, Sherman E. T Cell Activation through Isolated Tight Contacts. Cell Rep 2020; 29:3506-3521.e6. [PMID: 31825832 DOI: 10.1016/j.celrep.2019.11.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 08/12/2019] [Accepted: 11/06/2019] [Indexed: 01/01/2023] Open
Abstract
T cells engage antigen-presenting cells in search for cognate antigens via dynamic cell protrusions before forming a tight immune synapse. The spatiotemporal events that may lead to rapid TCR triggering and signal amplification in microvilli-driven isolated contacts, and in subsequent, more uniform contacts, remain poorly understood. Here, we combined interference-reflectance microscopy and single-molecule localization microscopy in live cells to resolve TCR-dependent signaling at tight cell contacts. We show that early contacts are sufficient for robust TCR triggering and ZAP-70 recruitment. With cell spreading, TCR activation and ZAP-70 recruitment increase and shift to the edges of the growing tight contacts. CD45 segregates from TCR at tight contacts and is enriched at high local curvature membrane. Surprisingly, cortical actin and LFA localized at contact regions of intermediate tightness. Our results show in molecular detail the roles of early and tight T cell contacts in T cell activation, as both sensing and decision-making entities.
Collapse
Affiliation(s)
- Yair Razvag
- Racah Institute of Physics, The Hebrew University, Jerusalem 9190401, Israel; Institute of Chemistry and The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Yair Neve-Oz
- Racah Institute of Physics, The Hebrew University, Jerusalem 9190401, Israel
| | - Julia Sajman
- Racah Institute of Physics, The Hebrew University, Jerusalem 9190401, Israel
| | - Oren Yakovian
- Racah Institute of Physics, The Hebrew University, Jerusalem 9190401, Israel
| | - Meital Reches
- Institute of Chemistry and The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Eilon Sherman
- Racah Institute of Physics, The Hebrew University, Jerusalem 9190401, Israel.
| |
Collapse
|
106
|
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: 31] [Impact Index Per Article: 7.8] [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
|
107
|
Gesper A, Wennmalm S, Hagemann P, Eriksson SG, Happel P, Parmryd I. Variations in Plasma Membrane Topography Can Explain Heterogenous Diffusion Coefficients Obtained by Fluorescence Correlation Spectroscopy. Front Cell Dev Biol 2020; 8:767. [PMID: 32903922 PMCID: PMC7443568 DOI: 10.3389/fcell.2020.00767] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/21/2020] [Indexed: 01/18/2023] Open
Abstract
Fluorescence correlation spectroscopy (FCS) is frequently used to study diffusion in cell membranes, primarily the plasma membrane. The diffusion coefficients reported in the plasma membrane of the same cell type and even within single cells typically display a large spread. We have investigated whether this spread can be explained by variations in membrane topography throughout the cell surface, that changes the amount of membrane in the FCS focal volume at different locations. Using FCS, we found that diffusion of the membrane dye DiI in the apical plasma membrane was consistently faster above the nucleus than above the cytoplasm. Using live cell scanning ion conductance microscopy (SICM) to obtain a topography map of the cell surface, we demonstrate that cell surface roughness is unevenly distributed with the plasma membrane above the nucleus being the smoothest, suggesting that the difference in diffusion observed in FCS is related to membrane topography. FCS modeled on simulated diffusion in cell surfaces obtained by SICM was consistent with the FCS data from live cells and demonstrated that topography variations can cause the appearance of anomalous diffusion in FCS measurements. Furthermore, we found that variations in the amount of the membrane marker DiD, a proxy for the membrane, but not the transmembrane protein TCRζ or the lipid-anchored protein Lck, in the FCS focal volume were related to variations in diffusion times at different positions in the plasma membrane. This relationship was seen at different positions both at the apical cell and basal cell sides. We conclude that it is crucial to consider variations in topography in the interpretation of FCS results from membranes.
Collapse
Affiliation(s)
| | - Stefan Wennmalm
- SciLifeLab, Royal Institute of Technology, Stockholm, Sweden
| | | | | | | | - Ingela Parmryd
- Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
108
|
Chaudhuri PK, Wang MS, Black CT, Huse M, Kam LC. Modulating T Cell Activation Using Depth Sensing Topographic Cues. ACTA ACUST UNITED AC 2020; 4:e2000143. [PMID: 32744809 DOI: 10.1002/adbi.202000143] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/16/2020] [Indexed: 12/11/2022]
Abstract
This report examines how sensing of substrate topography can be used to modulate T cell activation, a key coordinating step in the adaptive immune response. Inspired by the native T cell-antigen presenting cell interface, micrometer scale pits with varying depth are fabricated into planar substrates. Primary CD4+ T cells extend actin-rich protrusions into the micropits. T cell activation, reflected in secretion of cytokines interleukin-2 and interferon gamma, is sensitive to the micropit depth. Surprisingly, arrays of micropits with 4 μm depth enhance activation compared to flat substrates but deeper micropits are less effective at increasing cell response, revealing a biphasic dependence in activation as a function of feature dimensions. Inhibition of cell contractility abrogates the enhanced activation associated with the micropits. In conclusion, this report demonstrates that the 3D, microscale topography can be used to enhance T cell activation, an ability that most directly can be used to improve production of these cells for immunotherapy.
Collapse
Affiliation(s)
| | - Mitchell S Wang
- Pharmacology Graduate Program, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Charles T Black
- Center for Functional Nanomaterials, Brookhaven National Laboratory, Upton, NY, 11973, USA
| | - Morgan Huse
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Lance C Kam
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| |
Collapse
|
109
|
Wang Y, Xiang Y, Xin VW, Wang XW, Peng XC, Liu XQ, Wang D, Li N, Cheng JT, Lyv YN, Cui SZ, Ma Z, Zhang Q, Xin HW. Dendritic cell biology and its role in tumor immunotherapy. J Hematol Oncol 2020. [PMID: 32746880 DOI: 10.1186/s13045-020-00939-6.pmid:32746880;pmcid:pmc7397618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
As crucial antigen presenting cells, dendritic cells (DCs) play a vital role in tumor immunotherapy. Taking into account the many recent advances in DC biology, we discuss how DCs (1) recognize pathogenic antigens with pattern recognition receptors through specific phagocytosis and through non-specific micropinocytosis, (2) process antigens into small peptides with proper sizes and sequences, and (3) present MHC-peptides to CD4+ and CD8+ T cells to initiate immune responses against invading microbes and aberrant host cells. During anti-tumor immune responses, DC-derived exosomes were discovered to participate in antigen presentation. T cell microvillar dynamics and TCR conformational changes were demonstrated upon DC antigen presentation. Caspase-11-driven hyperactive DCs were recently reported to convert effectors into memory T cells. DCs were also reported to crosstalk with NK cells. Additionally, DCs are the most important sentinel cells for immune surveillance in the tumor microenvironment. Alongside DC biology, we review the latest developments for DC-based tumor immunotherapy in preclinical studies and clinical trials. Personalized DC vaccine-induced T cell immunity, which targets tumor-specific antigens, has been demonstrated to be a promising form of tumor immunotherapy in patients with melanoma. Importantly, allogeneic-IgG-loaded and HLA-restricted neoantigen DC vaccines were discovered to have robust anti-tumor effects in mice. Our comprehensive review of DC biology and its role in tumor immunotherapy aids in the understanding of DCs as the mentors of T cells and as novel tumor immunotherapy cells with immense potential.
Collapse
Affiliation(s)
- Yingying Wang
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
- Department of Gynaecology, Comprehensive Cancer Center, Hannover Medical School, 30625, Hannover, Germany
| | - Ying Xiang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | | | - Xian-Wang Wang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China
- Department of Laboratory Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China
| | - Xiao-Chun Peng
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China
- Department of Pathophysiology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Xiao-Qin Liu
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
- Department of Medical Imaging, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Dong Wang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Na Li
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| | - Jun-Ting Cheng
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Yan-Ning Lyv
- Institute for Infectious Diseases and Endemic Diseases Prevention and Control, Beijing Center for Diseases Prevention and Control, Beijing, 100013, China
| | - Shu-Zhong Cui
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Zhaowu Ma
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China.
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China.
| | - Qing Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China.
- Institute of Sun Yat-sen University in Shenzhen, Shenzhen, China.
| | - Hong-Wu Xin
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China.
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China.
- People's Hospital of Lianjiang, Lianjiang, 524400, Guangdong, China.
| |
Collapse
|
110
|
Wang Y, Xiang Y, Xin VW, Wang XW, Peng XC, Liu XQ, Wang D, Li N, Cheng JT, Lyv YN, Cui SZ, Ma Z, Zhang Q, Xin HW. Dendritic cell biology and its role in tumor immunotherapy. J Hematol Oncol 2020; 13:107. [PMID: 32746880 PMCID: PMC7397618 DOI: 10.1186/s13045-020-00939-6] [Citation(s) in RCA: 231] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/20/2020] [Indexed: 12/11/2022] Open
Abstract
As crucial antigen presenting cells, dendritic cells (DCs) play a vital role in tumor immunotherapy. Taking into account the many recent advances in DC biology, we discuss how DCs (1) recognize pathogenic antigens with pattern recognition receptors through specific phagocytosis and through non-specific micropinocytosis, (2) process antigens into small peptides with proper sizes and sequences, and (3) present MHC-peptides to CD4+ and CD8+ T cells to initiate immune responses against invading microbes and aberrant host cells. During anti-tumor immune responses, DC-derived exosomes were discovered to participate in antigen presentation. T cell microvillar dynamics and TCR conformational changes were demonstrated upon DC antigen presentation. Caspase-11-driven hyperactive DCs were recently reported to convert effectors into memory T cells. DCs were also reported to crosstalk with NK cells. Additionally, DCs are the most important sentinel cells for immune surveillance in the tumor microenvironment. Alongside DC biology, we review the latest developments for DC-based tumor immunotherapy in preclinical studies and clinical trials. Personalized DC vaccine-induced T cell immunity, which targets tumor-specific antigens, has been demonstrated to be a promising form of tumor immunotherapy in patients with melanoma. Importantly, allogeneic-IgG-loaded and HLA-restricted neoantigen DC vaccines were discovered to have robust anti-tumor effects in mice. Our comprehensive review of DC biology and its role in tumor immunotherapy aids in the understanding of DCs as the mentors of T cells and as novel tumor immunotherapy cells with immense potential.
Collapse
Affiliation(s)
- Yingying Wang
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China.,Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China.,Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China.,Department of Gynaecology, Comprehensive Cancer Center, Hannover Medical School, 30625, Hannover, Germany
| | - Ying Xiang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China.,Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | | | - Xian-Wang Wang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China.,Department of Laboratory Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China
| | - Xiao-Chun Peng
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China.,Department of Pathophysiology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Xiao-Qin Liu
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China.,Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China.,Department of Medical Imaging, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Dong Wang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China.,Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Na Li
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| | - Jun-Ting Cheng
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China.,Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Yan-Ning Lyv
- Institute for Infectious Diseases and Endemic Diseases Prevention and Control, Beijing Center for Diseases Prevention and Control, Beijing, 100013, China
| | - Shu-Zhong Cui
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Zhaowu Ma
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China. .,Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China.
| | - Qing Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China. .,Institute of Sun Yat-sen University in Shenzhen, Shenzhen, China.
| | - Hong-Wu Xin
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China. .,Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023, Hubei, China. .,People's Hospital of Lianjiang, Lianjiang, 524400, Guangdong, China.
| |
Collapse
|
111
|
Farrell MV, Webster S, Gaus K, Goyette J. T Cell Membrane Heterogeneity Aids Antigen Recognition and T Cell Activation. Front Cell Dev Biol 2020; 8:609. [PMID: 32850786 PMCID: PMC7399036 DOI: 10.3389/fcell.2020.00609] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/19/2020] [Indexed: 12/21/2022] Open
Abstract
T cells are critical for co-ordinating the immune response. T cells are activated when their surface T cell receptors (TCRs) engage cognate antigens in the form of peptide-major histocompatibility complexes (pMHC) presented on the surface of antigen presenting cells (APCs). Large changes in the contact interface between T cells and APCs occur over the course of tens of minutes from the initial contact to the formation of a large-scale junction between the two cells. The mature junction between a T cell and APC is known as the immunological synapse, and this specialized plasma membrane structure is the major platform for TCR signaling. It has long been known that the complex organization of signaling molecules at the synapse is critical for appropriate activation of T cells, but within the last decade advances in microscopy have opened up investigation into the dynamics of T cell surface topology in the immune synapse. From mechanisms mediating the initial contact between T cells and APCs to roles in the organization of molecules in the mature synapse, these studies have made it increasingly clear that local membrane topology has a large impact on signaling processes. This review focuses on the functional consequences of the T cells' highly dynamic and heterogeneous membrane, in particular, how membrane topology leads to the reorganization of membrane proteins on the T cell surface.
Collapse
Affiliation(s)
- Megan V Farrell
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Samantha Webster
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Katharina Gaus
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Jesse Goyette
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
112
|
Ruhland MK, Roberts EW, Cai E, Mujal AM, Marchuk K, Beppler C, Nam D, Serwas NK, Binnewies M, Krummel MF. Visualizing Synaptic Transfer of Tumor Antigens among Dendritic Cells. Cancer Cell 2020; 37:786-799.e5. [PMID: 32516589 PMCID: PMC7671443 DOI: 10.1016/j.ccell.2020.05.002] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 02/07/2020] [Accepted: 04/30/2020] [Indexed: 10/24/2022]
Abstract
Generation of tumor-infiltrating lymphocytes begins when tumor antigens reach the lymph node (LN) to stimulate T cells, yet we know little of how tumor material is disseminated among the large variety of antigen-presenting dendritic cell (DC) subsets in the LN. Here, we demonstrate that tumor proteins are carried to the LN within discrete vesicles inside DCs and are then transferred among DC subsets. A synapse is formed between interacting DCs and vesicle transfer takes place in the absence of free exosomes. DCs -containing vesicles can uniquely activate T cells, whereas DCs lacking them do not. Understanding this restricted sharing of tumor identity provides substantial room for engineering better anti-tumor immunity.
Collapse
Affiliation(s)
- Megan K Ruhland
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Edward W Roberts
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA
| | - En Cai
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Adriana M Mujal
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Kyle Marchuk
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA; Biological Imaging Development CoLab, University of California, San Francisco, CA 94143, USA
| | - Casey Beppler
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA
| | - David Nam
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Nina K Serwas
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Mikhail Binnewies
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Matthew F Krummel
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
113
|
Garcia E, Ismail S. Spatiotemporal Regulation of Signaling: Focus on T Cell Activation and the Immunological Synapse. Int J Mol Sci 2020; 21:E3283. [PMID: 32384769 PMCID: PMC7247333 DOI: 10.3390/ijms21093283] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/01/2020] [Accepted: 05/03/2020] [Indexed: 01/22/2023] Open
Abstract
In a signaling network, not only the functions of molecules are important but when (temporal) and where (spatial) those functions are exerted and orchestrated is what defines the signaling output. To temporally and spatially modulate signaling events, cells generate specialized functional domains with variable lifetime and size that concentrate signaling molecules, enhancing their transduction potential. The plasma membrane is a key in this regulation, as it constitutes a primary signaling hub that integrates signals within and across the membrane. Here, we examine some of the mechanisms that cells exhibit to spatiotemporally regulate signal transduction, focusing on the early events of T cell activation from triggering of T cell receptor to formation and maturation of the immunological synapse.
Collapse
Affiliation(s)
- Esther Garcia
- CR-UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Shehab Ismail
- CR-UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| |
Collapse
|
114
|
Chabaud M, Paillon N, Gaus K, Hivroz C. Mechanobiology of antigen‐induced T cell arrest. Biol Cell 2020; 112:196-212. [DOI: 10.1111/boc.201900093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/19/2020] [Accepted: 03/29/2020] [Indexed: 12/21/2022]
Affiliation(s)
- Mélanie Chabaud
- Institut Curie‐PSL Research University INSERM U932 Paris France
- EMBL Australia Node in Single Molecule Science, School of Medical SciencesUniversity of New South Wales Sydney NSW Australia
- ARC Centre of Excellence in Advanced Molecular ImagingUniversity of New South Wales Sydney NSW Australia
| | - Noémie Paillon
- Institut Curie‐PSL Research University INSERM U932 Paris France
| | - Katharina Gaus
- EMBL Australia Node in Single Molecule Science, School of Medical SciencesUniversity of New South Wales Sydney NSW Australia
- ARC Centre of Excellence in Advanced Molecular ImagingUniversity of New South Wales Sydney NSW Australia
| | - Claire Hivroz
- Institut Curie‐PSL Research University INSERM U932 Paris France
| |
Collapse
|
115
|
Tamzalit F, Wang MS, Jin W, Tello-Lafoz M, Boyko V, Heddleston JM, Black CT, Kam LC, Huse M. Interfacial actin protrusions mechanically enhance killing by cytotoxic T cells. Sci Immunol 2020; 4:4/33/eaav5445. [PMID: 30902904 DOI: 10.1126/sciimmunol.aav5445] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 02/05/2019] [Indexed: 12/30/2022]
Abstract
Cytotoxic T lymphocytes (CTLs) kill by forming immunological synapses with target cells and secreting toxic proteases and the pore-forming protein perforin into the intercellular space. Immunological synapses are highly dynamic structures that boost perforin activity by applying mechanical force against the target cell. Here, we used high-resolution imaging and microfabrication to investigate how CTLs exert synaptic forces and coordinate their mechanical output with perforin secretion. Using micropatterned stimulatory substrates that enable synapse growth in three dimensions, we found that perforin release occurs at the base of actin-rich protrusions that extend from central and intermediate locations within the synapse. These protrusions, which depended on the cytoskeletal regulator WASP and the Arp2/3 actin nucleation complex, were required for synaptic force exertion and efficient killing. They also mediated physical deformation of the target cell surface during CTL-target cell interactions. Our results reveal the mechanical basis of cellular cytotoxicity and highlight the functional importance of dynamic, three-dimensional architecture in immune cell-cell interfaces.
Collapse
Affiliation(s)
- Fella Tamzalit
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mitchell S Wang
- Pharmacology Graduate Program, Weill Cornell Medical College, New York, NY, USA
| | - Weiyang Jin
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Maria Tello-Lafoz
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vitaly Boyko
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John M Heddleston
- Advanced Imaging Center, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Charles T Black
- Center for Functional Nanomaterials, Brookhaven National Laboratory, Upton, NY, USA
| | - Lance C Kam
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Morgan Huse
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
116
|
Kulenkampff K, Lippert AH, McColl J, Santos AM, Ponjavic A, Jenkins E, Humphrey J, Winkel A, Franze K, Lee SF, Davis SJ, Klenerman D. The Costs of Close Contacts: Visualizing the Energy Landscape of Cell Contacts at the Nanoscale. Biophys J 2020; 118:1261-1269. [PMID: 32075748 PMCID: PMC7091464 DOI: 10.1016/j.bpj.2020.01.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 12/27/2022] Open
Abstract
Cell-cell contacts often underpin signaling between cells. For immunology, the binding of a T cell receptor to an antigen-presenting pMHC initiates downstream signaling and an immune response. Although this contact is mediated by proteins on both cells creating interfaces with gap sizes typically around 14 nm, many, often contradictory observations have been made regarding the influence of the contact on parameters such as the binding kinetics, spatial distribution, and diffusion of signaling proteins within the contact. Understanding the basic physical constraints on probes inside this crowded environment will help inform studies on binding kinetics and dynamics of signaling of relevant proteins in the synapse. By tracking quantum dots of different dimensions for extended periods of time, we have shown that it is possible to obtain the probability of a molecule entering the contact, the change in its diffusion upon entry, and the impact of spatial heterogeneity of adhesion protein density in the contact. By analyzing the contacts formed by a T cell interacting with adhesion proteins anchored to a supported lipid bilayer, we find that probes are excluded from contact entry in a size-dependent manner for gap-to-probe differences of 4.1 nm. We also observed probes being trapped inside the contact and a decrease in diffusion of up to 85% in dense adhesion protein contacts. This approach provides new, to our knowledge, insights into the nature of cell-cell contacts, revealing that cell contacts are highly heterogeneous because of topography- and protein-density-related processes. These effects are likely to profoundly influence signaling between cells.
Collapse
Affiliation(s)
- Klara Kulenkampff
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Anna H Lippert
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - James McColl
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Ana Mafalda Santos
- Radcliffe Department of Medicine and MRC Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Aleks Ponjavic
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Edward Jenkins
- Radcliffe Department of Medicine and MRC Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Jane Humphrey
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Alexander Winkel
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Kristian Franze
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Steven F Lee
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Simon J Davis
- Radcliffe Department of Medicine and MRC Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - David Klenerman
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
117
|
Moreau HD, Lennon-Duménil AM, Pierobon P. “If you please… draw me a cell”. Insights from immune cells. J Cell Sci 2020; 133:133/5/jcs244806. [DOI: 10.1242/jcs.244806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
ABSTRACT
Studies in recent years have shed light on the particular features of cytoskeleton dynamics in immune cells, challenging the classical picture drawn from typical adherent cell lines. New mechanisms linking the dynamics of the membrane–cytoskeleton interface to the mechanical properties of immune cells have been uncovered and shown to be essential for immune surveillance functions. In this Essay, we discuss these features, and propose immune cells as a new playground for cell biologists who try to understand how cells adapt to different microenvironments to fulfil their functions efficiently.
Collapse
Affiliation(s)
- Hélène D. Moreau
- INSERM U932, Institut Curie, ANR-10-IDEX-0001-02 PSL and ANR-11-LABX-0043, 26 rue d'Ulm, 75248 Paris, Cedex 05, France
| | - Ana-Maria Lennon-Duménil
- INSERM U932, Institut Curie, ANR-10-IDEX-0001-02 PSL and ANR-11-LABX-0043, 26 rue d'Ulm, 75248 Paris, Cedex 05, France
| | - Paolo Pierobon
- INSERM U932, Institut Curie, ANR-10-IDEX-0001-02 PSL and ANR-11-LABX-0043, 26 rue d'Ulm, 75248 Paris, Cedex 05, France
| |
Collapse
|
118
|
Schamel WW, Alarcon B, Minguet S. The TCR is an allosterically regulated macromolecular machinery changing its conformation while working. Immunol Rev 2020; 291:8-25. [PMID: 31402501 DOI: 10.1111/imr.12788] [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: 05/22/2019] [Accepted: 05/30/2019] [Indexed: 12/13/2022]
Abstract
The αβ T-cell receptor (TCR) is a multiprotein complex controlling the activation of T cells. Although the structure of the complete TCR is not known, cumulative evidence supports that the TCR cycles between different conformational states that are promoted either by thermal motion or by force. These structural transitions determine whether the TCR engages intracellular effectors or not, regulating TCR phosphorylation and signaling. As for other membrane receptors, ligand binding selects and stabilizes the TCR in active conformations, and/or switches the TCR to activating states that were not visited before ligand engagement. Here we review the main models of TCR allostery, that is, ligand binding at TCRαβ changes the structure at CD3 and ζ. (a) The ITAM and proline-rich sequence exposure model, in which the TCR's cytoplasmic tails shield each other and ligand binding exposes them for phosphorylation. (b) The membrane-ITAM model, in which the CD3ε and ζ tails are sequestered inside the membrane and again ligand binding exposes them. (c) The mechanosensor model in which ligand binding exerts force on the TCR, inducing structural changes that allow signaling. Since these models are complementary rather than competing, we propose a unified model that aims to incorporate all existing data.
Collapse
Affiliation(s)
- Wolfgang W Schamel
- Department of Immunology, Faculty of Biology, Albert-Ludwigs-University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Center of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Freiburg, Germany
| | - Balbino Alarcon
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain
| | - Susana Minguet
- Department of Immunology, Faculty of Biology, Albert-Ludwigs-University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Center of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Freiburg, Germany
| |
Collapse
|
119
|
Rushdi M, Li K, Yuan Z, Travaglino S, Grakoui A, Zhu C. Mechanotransduction in T Cell Development, Differentiation and Function. Cells 2020; 9:E364. [PMID: 32033255 PMCID: PMC7072571 DOI: 10.3390/cells9020364] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 02/07/2023] Open
Abstract
Cells in the body are actively engaging with their environments that include both biochemical and biophysical aspects. The process by which cells convert mechanical stimuli from their environment to intracellular biochemical signals is known as mechanotransduction. Exemplifying the reliance on mechanotransduction for their development, differentiation and function are T cells, which are central to adaptive immune responses. T cell mechanoimmunology is an emerging field that studies how T cells sense, respond and adapt to the mechanical cues that they encounter throughout their life cycle. Here we review different stages of the T cell's life cycle where existing studies have shown important effects of mechanical force or matrix stiffness on a T cell as sensed through its surface molecules, including modulating receptor-ligand interactions, inducing protein conformational changes, triggering signal transduction, amplifying antigen discrimination and ensuring directed targeted cell killing. We suggest that including mechanical considerations in the immunological studies of T cells would inform a more holistic understanding of their development, differentiation and function.
Collapse
Affiliation(s)
- Muaz Rushdi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; (M.R.); (K.L.); (S.T.)
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA;
| | - Kaitao Li
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; (M.R.); (K.L.); (S.T.)
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA;
| | - Zhou Yuan
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA;
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30313, USA
| | - Stefano Travaglino
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; (M.R.); (K.L.); (S.T.)
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA;
| | - Arash Grakoui
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes Research Primate Center, Emory University School of Medicine, Atlanta, GA 30329, USA;
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Cheng Zhu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; (M.R.); (K.L.); (S.T.)
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA;
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30313, USA
| |
Collapse
|
120
|
Blumenthal D, Burkhardt JK. Multiple actin networks coordinate mechanotransduction at the immunological synapse. J Cell Biol 2020; 219:e201911058. [PMID: 31977034 PMCID: PMC7041673 DOI: 10.1083/jcb.201911058] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/31/2019] [Accepted: 01/02/2020] [Indexed: 12/26/2022] Open
Abstract
Activation of naive T cells by antigen-presenting cells (APCs) is an essential step in mounting an adaptive immune response. It is known that antigen recognition and T cell receptor (TCR) signaling depend on forces applied by the T cell actin cytoskeleton, but until recently, the underlying mechanisms have been poorly defined. Here, we review recent advances in the field, which show that specific actin-dependent structures contribute to the process in distinct ways. In essence, T cell priming involves a tug-of-war between the cytoskeletons of the T cell and the APC, where the actin cytoskeleton serves as a mechanical intermediate that integrates force-dependent signals. We consider each of the relevant actin-rich T cell structures separately and address how they work together at the topologically and temporally complex cell-cell interface. In addition, we address how this mechanobiology can be incorporated into canonical immunological models to improve how these models explain T cell sensitivity and antigenic specificity.
Collapse
Affiliation(s)
| | - Janis K. Burkhardt
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
121
|
Rosenberg J, Huang J. Visualizing Surface T-Cell Receptor Dynamics Four-Dimensionally Using Lattice Light-Sheet Microscopy. J Vis Exp 2020. [PMID: 32065118 DOI: 10.3791/59914] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The signaling and function of a cell are dictated by the dynamic structures and interactions of its surface receptors. To truly understand the structure-function relationship of these receptors in situ, we need to visualize and track them on the live cell surface with enough spatiotemporal resolution. Here we show how to use recently developed Lattice Light-Sheet Microscopy (LLSM) to image T-cell receptors (TCRs) four-dimensionally (4D, space and time) at the live cell membrane. T cells are one of the main effector cells of the adaptive immune system, and here we used T cells as an example to show that the signaling and function of these cells are driven by the dynamics and interactions of the TCRs. LLSM allows for 4D imaging with unprecedented spatiotemporal resolution. This microscopy technique therefore can be generally applied to a wide array of surface or intracellular molecules of different cells in biology.
Collapse
Affiliation(s)
| | - Jun Huang
- Committee on Cancer Biology, The University of Chicago; Pritzker School of Molecular Engineering, The University of Chicago;
| |
Collapse
|
122
|
Kennedy PR, Barthen C, Williamson DJ, Pitkeathly WTE, Hazime KS, Cumming J, Stacey KB, Hilton HG, Carrington M, Parham P, Davis DM. Genetic diversity affects the nanoscale membrane organization and signaling of natural killer cell receptors. Sci Signal 2019; 12:eaaw9252. [PMID: 31848320 PMCID: PMC6944503 DOI: 10.1126/scisignal.aaw9252] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Genetic diversity in human natural killer (NK) cell receptors is linked to resistance and susceptibility to many diseases. Here, we tested the effect of this diversity on the nanoscale organization of killer cell immunoglobulin-like receptors (KIRs). Using superresolution microscopy, we found that inhibitory KIRs encoded by different genes and alleles were organized differently at the surface of primary human NK cells. KIRs that were found at low abundance assembled into smaller clusters than those formed by KIRs that were more highly abundant, and at low abundance, there was a greater proportion of KIRs in clusters. Upon receptor triggering, a structured interface called the immune synapse assembles, which facilitates signal integration and controls NK cell responses. Here, triggering of low-abundance receptors resulted in less phosphorylation of the downstream phosphatase SHP-1 but more phosphorylation of the adaptor protein Crk than did triggering of high-abundance receptors. In cells with greater KIR abundance, SHP-1 dephosphorylated Crk, which potentiated NK cell spreading during activation. Thus, genetic variation modulates both the abundance and nanoscale organization of inhibitory KIRs. That is, as well as the number of receptors at the cell surface varying with genotype, the way in which these receptors are organized in the membrane also varies. Essentially, a change in the average surface abundance of a protein at the cell surface is a coarse descriptor entwined with changes in local nanoscale clustering. Together, our data indicate that genetic diversity in inhibitory KIRs affects membrane-proximal signaling and, unexpectedly, the formation of activating immune synapses.
Collapse
Affiliation(s)
- Philippa R Kennedy
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| | - Charlotte Barthen
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| | - David J Williamson
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| | - William T E Pitkeathly
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| | - Khodor S Hazime
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| | - Joshua Cumming
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| | - Kevin B Stacey
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| | - Hugo G Hilton
- Department of Structural Biology, Stanford University School of Medicine, D159, Sherman Fairchild Science Building, 299 Campus Drive West, Stanford, CA 94305, USA
| | - Mary Carrington
- Basic Science Program, Frederick National Laboratory for Cancer Research, Building 560, Room 21-89, Frederick, MD 21702, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Peter Parham
- Department of Structural Biology, Stanford University School of Medicine, D159, Sherman Fairchild Science Building, 299 Campus Drive West, Stanford, CA 94305, USA
| | - Daniel M Davis
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK.
| |
Collapse
|
123
|
Siokis A, Robert PA, Demetriou P, Dustin ML, Meyer-Hermann M. F-Actin-Driven CD28-CD80 Localization in the Immune Synapse. Cell Rep 2019; 24:1151-1162. [PMID: 30067972 DOI: 10.1016/j.celrep.2018.06.114] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 04/18/2018] [Accepted: 06/28/2018] [Indexed: 12/27/2022] Open
Abstract
During immunological synapse (IS) formation, T cell receptor (TCR) signaling complexes, integrins, and costimulatory molecules exhibit a particular spatial localization. Here, we develop an agent-based model for the IS formation based on TCR peptide-bound major histocompatibility complex (pMHC) and leukocyte-function-associated antigen 1 (LFA-1) intracellular activation molecule 1 (ICAM-1) dynamics, including CD28 binding to a costimulatory ligand, coupling of molecules to the centripetal actin flow, and size-based segregation (SBS). A radial gradient of LFA-1 in the peripheral supramolecular activation cluster (pSMAC) toward the central supramolecular activation cluster (cSMAC) emerged as a combined consequence of actin binding and diffusion and modified the positioning of other molecules. The simulations predict a mechanism of CD28 movement, according to which CD28-CD80 complexes passively follow TCR-pMHC microclusters. However, the characteristic CD28-CD80 localization in a ring pattern around the cSMAC only emerges with a particular CD28-actin coupling strength that induces a centripetal motion. These results have implications for the understanding of T cell activation and fate decisions.
Collapse
Affiliation(s)
- Anastasios Siokis
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig 38106, Germany
| | - Philippe A Robert
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig 38106, Germany.
| | - Philippos Demetriou
- Kennedy Institute, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, Oxford OX3 7FY, UK
| | - Michael L Dustin
- Kennedy Institute, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, Oxford OX3 7FY, UK; Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig 38106, Germany; Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig 38106, Germany.
| |
Collapse
|
124
|
Korolj A, Wu HT, Radisic M. A healthy dose of chaos: Using fractal frameworks for engineering higher-fidelity biomedical systems. Biomaterials 2019; 219:119363. [PMID: 31376747 PMCID: PMC6759375 DOI: 10.1016/j.biomaterials.2019.119363] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 07/09/2019] [Accepted: 07/14/2019] [Indexed: 12/18/2022]
Abstract
Optimal levels of chaos and fractality are distinctly associated with physiological health and function in natural systems. Chaos is a type of nonlinear dynamics that tends to exhibit seemingly random structures, whereas fractality is a measure of the extent of organization underlying such structures. Growing bodies of work are demonstrating both the importance of chaotic dynamics for proper function of natural systems, as well as the suitability of fractal mathematics for characterizing these systems. Here, we review how measures of fractality that quantify the dose of chaos may reflect the state of health across various biological systems, including: brain, skeletal muscle, eyes and vision, lungs, kidneys, tumours, cell regulation, skin and wound repair, bone, vasculature, and the heart. We compare how reports of either too little or too much chaos and fractal complexity can be damaging to normal biological function, and suggest that aiming for the healthy dose of chaos may be an effective strategy for various biomedical applications. We also discuss rising examples of the implementation of fractal theory in designing novel materials, biomedical devices, diagnostics, and clinical therapies. Finally, we explain important mathematical concepts of fractals and chaos, such as fractal dimension, criticality, bifurcation, and iteration, and how they are related to biology. Overall, we promote the effectiveness of fractals in characterizing natural systems, and suggest moving towards using fractal frameworks as a basis for the research and development of better tools for the future of biomedical engineering.
Collapse
Affiliation(s)
- Anastasia Korolj
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Canada
| | - Hau-Tieng Wu
- Department of Statistical Science, Duke University, Durham, NC, USA; Department of Mathematics, Duke University, Durham, NC, USA; Mathematics Division, National Center for Theoretical Sciences, Taipei, Taiwan
| | - Milica Radisic
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Canada; Toronto General Research Institute, University Health Network, Toronto, Canada; The Heart and Stroke/Richard Lewar Center of Excellence, Toronto, Canada.
| |
Collapse
|
125
|
Liu L, Wei Y, Liu J, Wang K, Zhang J, Zhang P, Zhou Y, Li B. Spatial high resolution of actin filament organization by PeakForce atomic force microscopy. Cell Prolif 2019; 53:e12670. [PMID: 31568631 PMCID: PMC6985672 DOI: 10.1111/cpr.12670] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/08/2019] [Accepted: 05/16/2019] [Indexed: 12/12/2022] Open
Abstract
Objectives To investigate the heterogeneous feature of actin filaments (ACFs) associated with the cellular membrane in HeLa and HCT‐116 cells at the nanoscale level. Materials and Methods Fluorescence microscopy coupled with atomic force microscopy (AFM) was used to identify and characterize ACFs of cells. The distribution of ACFs was detected by Fluor‐488‐phalloidin–labelled actin. The morphology of the ACFs was probed by AFM images. The spatial correlation of the microvilli and ACFs was explored with different forces of AFM loading on cells. Results Intricate but ordered structures of the actin cytoskeletons associated with cellular membrane were characterized and revealed. Two different layers of ACFs with distinct structural organizations were directly observed in HCT‐116 and HeLa cells. Bundle‐shaped ACFs protruding the cellular membrane forming the microvilli, and the network ACFs underneath the cellular membrane were resolved with high resolution under near‐physiological conditions. Approximately 14 nm lateral resolution was achieved when imaging single ACF beneath the cellular membrane. On the basis of the observed spatial distribution of the ultrastructure of the ACF organization, a model for this organization of ACFs was proposed. Conclusions We revealed the two layers of the ACF organization in Hela and HCT‐116 cells. The resolved heterogeneous structures at the nanoscale level provide a spatial view of the ACFs, which would contribute to the understanding of the essential biological functions of the actin cytoskeleton.
Collapse
Affiliation(s)
- Lin Liu
- Division of Physical Biology & Bioimaging Centre, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yuhui Wei
- Division of Physical Biology & Bioimaging Centre, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China
| | - Jingyuan Liu
- Fourth Military Medical University, Xi'an, China
| | - Kaizhe Wang
- Division of Physical Biology & Bioimaging Centre, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jinjin Zhang
- Division of Physical Biology & Bioimaging Centre, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China
| | - Ping Zhang
- Division of Physical Biology & Bioimaging Centre, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yi Zhou
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bin Li
- Division of Physical Biology & Bioimaging Centre, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
126
|
Abstract
The immune response is orchestrated by a variety of immune cells. The function of each cell is determined by the collective signals from various immunoreceptors, whose expression and activity depend on the developmental stages of the cell and its environmental context. Recent studies have highlighted the presence of mechanical force on several immunoreceptor-ligand pairs and the important role of force in regulating their interaction and function. In this Perspective, we use the T cell antigen receptor as an example with which to review the current understanding of the mechanosensing properties of immunoreceptors. We discuss the types of forces that immunoreceptors may encounter and the effects of force on ligand bonding, conformational change and the triggering of immunoreceptors, as well as the effects of force on the downstream signal transduction, cell-fate decisions and effector function of immune cells.
Collapse
|
127
|
Saliba DG, Céspedes-Donoso PF, Bálint Š, Compeer EB, Korobchevskaya K, Valvo S, Mayya V, Kvalvaag A, Peng Y, Dong T, Tognoli ML, O'Neill E, Bonham S, Fischer R, Kessler BM, Dustin ML. Composition and structure of synaptic ectosomes exporting antigen receptor linked to functional CD40 ligand from helper T cells. eLife 2019; 8:e47528. [PMID: 31469364 PMCID: PMC6748831 DOI: 10.7554/elife.47528] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/28/2019] [Indexed: 12/21/2022] Open
Abstract
Planar supported lipid bilayers (PSLB) presenting T cell receptor (TCR) ligands and ICAM-1 induce budding of extracellular microvesicles enriched in functional TCR, defined here as synaptic ectosomes (SE), from helper T cells. SE bind peptide-MHC directly exporting TCR into the synaptic cleft, but incorporation of other effectors is unknown. Here, we utilized bead supported lipid bilayers (BSLB) to capture SE from single immunological synapses (IS), determined SE composition by immunofluorescence flow cytometry and enriched SE for proteomic analysis by particle sorting. We demonstrate selective enrichment of CD40L and ICOS in SE in response to addition of CD40 and ICOSL, respectively, to SLB presenting TCR ligands and ICAM-1. SE are enriched in tetraspanins, BST-2, TCR signaling and ESCRT proteins. Super-resolution microscopy demonstrated that CD40L is present in microclusters within CD81 defined SE that are spatially segregated from TCR/ICOS/BST-2. CD40L+ SE retain the capacity to induce dendritic cell maturation and cytokine production.
Collapse
Affiliation(s)
- David G Saliba
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal SciencesUniversity of OxfordOxfordUnited Kingdom
- Department of Applied Biomedical Science, Faculty of Health ScienceUniversity of MaltaMsidaMalta
| | - Pablo F Céspedes-Donoso
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal SciencesUniversity of OxfordOxfordUnited Kingdom
| | - Štefan Bálint
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal SciencesUniversity of OxfordOxfordUnited Kingdom
| | - Ewoud B Compeer
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal SciencesUniversity of OxfordOxfordUnited Kingdom
| | - Kseniya Korobchevskaya
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal SciencesUniversity of OxfordOxfordUnited Kingdom
| | - Salvatore Valvo
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal SciencesUniversity of OxfordOxfordUnited Kingdom
| | - Viveka Mayya
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal SciencesUniversity of OxfordOxfordUnited Kingdom
| | - Audun Kvalvaag
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal SciencesUniversity of OxfordOxfordUnited Kingdom
| | - Yanchun Peng
- MRC Human Immunology Unit, Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUnited Kingdom
- Nuffield Department of Medicine, Chinese Academy of Medical Science Oxford InstituteUniversity of OxfordOxfordUnited Kingdom
| | - Tao Dong
- MRC Human Immunology Unit, Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUnited Kingdom
- Nuffield Department of Medicine, Chinese Academy of Medical Science Oxford InstituteUniversity of OxfordOxfordUnited Kingdom
| | | | - Eric O'Neill
- Department of OncologyUniversity of OxfordOxfordUnited Kingdom
| | - Sarah Bonham
- Discovery Proteomics Facility, Target Discovery Institute, Nuffield Department of MedicineUniversity of OxfordOxfordUnited Kingdom
| | - Roman Fischer
- Discovery Proteomics Facility, Target Discovery Institute, Nuffield Department of MedicineUniversity of OxfordOxfordUnited Kingdom
| | - Benedikt M Kessler
- Discovery Proteomics Facility, Target Discovery Institute, Nuffield Department of MedicineUniversity of OxfordOxfordUnited Kingdom
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal SciencesUniversity of OxfordOxfordUnited Kingdom
| |
Collapse
|
128
|
Saveanu L, Zucchetti AE, Evnouchidou I, Ardouin L, Hivroz C. Is there a place and role for endocyticTCRsignaling? Immunol Rev 2019; 291:57-74. [DOI: 10.1111/imr.12764] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 04/02/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Loredana Saveanu
- National French Institute of Health and Medical Research (INSERM) 1149 Center of Research on Inflammation Paris France
- National French Center of Scientific Research (CNRS) ERL8252 Paris France
- Laboratory of Inflamex Excellency Faculty of Medicine Xavier Bichat Site Paris France
- Paris Diderot UniversitySorbonne Paris Cité Paris France
| | - Andres E. Zucchetti
- Institut Curie PSL Research UniversityINSERMU932 “Integrative analysis of T cell activation” team Paris France
| | - Irini Evnouchidou
- National French Institute of Health and Medical Research (INSERM) 1149 Center of Research on Inflammation Paris France
- National French Center of Scientific Research (CNRS) ERL8252 Paris France
- Laboratory of Inflamex Excellency Faculty of Medicine Xavier Bichat Site Paris France
- Paris Diderot UniversitySorbonne Paris Cité Paris France
- Inovarion Paris France
| | - Laurence Ardouin
- Institut Curie PSL Research UniversityINSERMU932 “Integrative analysis of T cell activation” team Paris France
| | - Claire Hivroz
- Institut Curie PSL Research UniversityINSERMU932 “Integrative analysis of T cell activation” team Paris France
| |
Collapse
|
129
|
DNA probes that store mechanical information reveal transient piconewton forces applied by T cells. Proc Natl Acad Sci U S A 2019; 116:16949-16954. [PMID: 31391300 DOI: 10.1073/pnas.1904034116] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The advent of molecular tension probes for real-time mapping of piconewton forces in living systems has had a major impact on mechanobiology. For example, DNA-based tension probes have revealed roles for mechanics in platelet, B cell, T cell, and fibroblast function. Nonetheless, imaging short-lived forces transmitted by low-abundance receptors remains a challenge. This is a particular problem for mechanoimmunology where ligand-receptor bindings are short lived, and a few antigens are sufficient for cell triggering. Herein, we present a mechanoselection strategy that uses locking oligonucleotides to preferentially and irreversibly bind DNA probes that are mechanically strained over probes at rest. Thus, infrequent and short-lived mechanical events are tagged. This strategy allows for integration and storage of mechanical information into a map of molecular tension history. Upon addition of unlocking oligonucleotides that drive toehold-mediated strand displacement, the probes reset to the real-time state, thereby erasing stored mechanical information. As a proof of concept, we applied this strategy to study OT-1 T cells, revealing that the T cell receptor (TCR) mechanically samples antigens carrying single amino acid mutations. Such events are not detectable using conventional tension probes. Each mutant peptide ligand displayed a different level of mechanical sampling and spatial scanning by the TCR that strongly correlated with its functional potency. Finally, we show evidence that T cells transmit pN forces through the programmed cell death receptor-1 (PD1), a major target in cancer immunotherapy. We anticipate that mechanical information storage will be broadly useful in studying the mechanobiology of the immune system.
Collapse
|
130
|
Kwak K, Quizon N, Sohn H, Saniee A, Manzella-Lapeira J, Holla P, Brzostowski J, Lu J, Xie H, Xu C, Spillane KM, Tolar P, Pierce SK. Intrinsic properties of human germinal center B cells set antigen affinity thresholds. Sci Immunol 2019; 3:3/29/eaau6598. [PMID: 30504208 DOI: 10.1126/sciimmunol.aau6598] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 10/26/2018] [Indexed: 12/17/2022]
Abstract
Protective antibody responses to vaccination or infection depend on affinity maturation, a process by which high-affinity germinal center (GC) B cells are selected on the basis of their ability to bind, gather, and present antigen to T follicular helper (Tfh) cells. Here, we show that human GC B cells have intrinsically higher-affinity thresholds for both B cell antigen receptor (BCR) signaling and antigen gathering as compared with naïve B cells and that these functions are mediated by distinct cellular structures and pathways that ultimately lead to antigen affinity- and Tfh cell-dependent differentiation to plasma cells. GC B cells bound antigen through highly dynamic, actin- and ezrin-rich pod-like structures that concentrated BCRs. The behavior of these structures was dictated by the intrinsic antigen affinity thresholds of GC B cells. Low-affinity antigens triggered continuous engagement and disengagement of membrane-associated antigens, whereas high-affinity antigens induced stable synapse formation. The pod-like structures also mediated affinity-dependent antigen internalization by unconventional pathways distinct from those of naïve B cells. Thus, intrinsic properties of human GC B cells set thresholds for affinity selection.
Collapse
Affiliation(s)
- Kihyuck Kwak
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Nicolas Quizon
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Haewon Sohn
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Avva Saniee
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Javier Manzella-Lapeira
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Prasida Holla
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Joseph Brzostowski
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Jinghua Lu
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - HengYi Xie
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Chenguang Xu
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Katelyn M Spillane
- Department of Physics, King's College London, London WC2R 2LS, UK.,Immune Receptor Activation Laboratory, Francis Crick Institute, London NW1 1AT, UK.,Division of Immunology and Inflammation, Imperial College London, London SW7 2AZ, UK
| | - Pavel Tolar
- Immune Receptor Activation Laboratory, Francis Crick Institute, London NW1 1AT, UK.,Division of Immunology and Inflammation, Imperial College London, London SW7 2AZ, UK
| | - Susan K Pierce
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA.
| |
Collapse
|
131
|
Abstract
Communication between cells is essential for multicellular life. During cognate immune interactions, T cells communicate with antigen-presenting cells (APC) via direct cell-cell contact or the release of molecules and vesicles containing T cell messages. A wide variety of mechanisms have been reported and among them a process called "trogocytosis" has traditionally been thought to be the fastest way to directly transfer membrane portions containing intact proteins from one cell to another; however, the mechanism is unverified. Trogocytosis has been distinguished from the generation of extracellular vesicles (EVs), a term that encompasses exosomes and microvesicles, as EVs are released via a contact-independent manner and are suggested to potentially send molecular messages over a distance. However, some previous reports regarding EVs in T cells may be misleading in terms of explaining their cellular origins. In addition, there is little evidence on how EVs are generated from T cells in vivo and function to regulate complex immune responses. A recent work demonstrated that T cell microvilli-thin and finger-like membrane protrusions-are highly fragile and easily separated as membrane particles by trogocytosis, forming a new class of EVs. Surprisingly, released T cell microvilli-derived particles act as vectors, transmitting T cell messages to cognate APCs. This review focuses on how T cell microvilli vesicles are connected with immune regulation mechanisms discovered previously.
Collapse
Affiliation(s)
- Hye-Ran Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Chang-Duk Jun
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology, Gwangju, South Korea
| |
Collapse
|
132
|
Cassioli C, Baldari CT. A Ciliary View of the Immunological Synapse. Cells 2019; 8:E789. [PMID: 31362462 PMCID: PMC6721628 DOI: 10.3390/cells8080789] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/15/2019] [Accepted: 07/25/2019] [Indexed: 12/28/2022] Open
Abstract
The primary cilium has gone from being a vestigial organelle to a crucial signaling hub of growing interest given the association between a group of human disorders, collectively known as ciliopathies, and defects in its structure or function. In recent years many ciliogenesis proteins have been observed at extraciliary sites in cells and likely perform cilium-independent functions ranging from regulation of the cytoskeleton to vesicular trafficking. Perhaps the most striking example is the non-ciliated T lymphocyte, in which components of the ciliary machinery are repurposed for the assembly and function of the immunological synapse even in the absence of a primary cilium. Furthermore, the specialization traits described at the immunological synapse are similar to those seen in the primary cilium. Here, we review common regulators and features shared by the immunological synapse and the primary cilium that document the remarkable homology between these structures.
Collapse
Affiliation(s)
- Chiara Cassioli
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Cosima T Baldari
- Department of Life Sciences, University of Siena, 53100 Siena, Italy.
| |
Collapse
|
133
|
Membrane Organization and Physical Regulation of Lymphocyte Antigen Receptors: A Biophysicist's Perspective. J Membr Biol 2019; 252:397-412. [PMID: 31352492 DOI: 10.1007/s00232-019-00085-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 07/18/2019] [Indexed: 12/19/2022]
Abstract
Receptors at the membrane of immune cells are the central players of innate and adaptative immunity, providing effective defence mechanisms against pathogens or cancer cells. Their function is intimately linked to their position at and within the membrane which provides accessibility, mobility as well as membrane proximal cytoskeleton anchoring, all of these elements playing important roles in the final function and links to cellular actions. Understanding how immune cells integrate the specific signals received at their membrane to take a decision remains an immense challenge and a very active field of fundamental and applied research. Recent progress in imaging and micromanipulation techniques have led to an unprecedented refinement in the description of molecular structures and supramolecular assemblies at the immune cell membrane, and provided a glimpse into their dynamics and regulation by force. Several key elements have been scrutinized such as the roles of relative sizes of molecules, lateral organisation, motion in the membrane of the receptors, but also physical cues such as forces, mediated by cellular substrates of different rigidities or applied by the cell itself, in conjunction with its partner cell. We review here these recent discoveries associated with a description of the biophysical methods used. While a conclusive picture integrating all of these components is still lacking, mainly due to the implication of diverse and different mechanisms and spatio-temporal scales involved, the amount of quantitative data available opens the way for physical modelling and numerical simulations and new avenues for experimental research.
Collapse
|
134
|
Cai G, Pan S, Feng N, Zou H, Gu J, Yuan Y, Liu X, Liu Z, Bian J. Zearalenone inhibits T cell chemotaxis by inhibiting cell adhesion and migration related proteins. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 175:263-271. [PMID: 30903882 DOI: 10.1016/j.ecoenv.2019.03.045] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/10/2019] [Accepted: 03/12/2019] [Indexed: 06/09/2023]
Abstract
Zearalenone (ZEA) is a phenolic resorcylic acid lactone mycotoxin produced by several Fusarium species that grow on temperate and tropical crops. The number of reports documenting the immunotoxic effects of ZEA is increasing, but the underlying mechanism is not clear. The purpose of this study was to investigate the effects of ZEA on T cell chemotaxis and evaluate changes in adhesion and migration proteins associated with this process. Specifically, T cells were isolated from BALB/C mouse splenic lymphocytes, activated by concanavalin A (Con A), and then exposed to different concentrations of ZEA. Transmission electron microscopy (TEM) and scanning electron microscopy (SEM) were used observe the ultrastructural changes inside the cell and on the cell surface, respectively. The transwell migration assay was used to evaluate the effect of ZEA on T cell chemotaxis in the presence of CCL19 or CCL21. A confocal 3D laser was used to capture the morphology of perforated cells and western blot was used to detect the expression of proteins associated with cell migration and adhesion. Additionally, we used flow cytometry to examine the expression of chemokine receptors on T cells. Finally, the chemokine (RANTES and MIP-1α) levels secreted by T cells were assessed using cytometric bead array. Overall, our data showed that treatment with ZEA caused ultrastructural damage on the surface as well as inside of T cells. Moreover, ZEA inhibited T cell chemotaxis which was mediated by CCL19 or CCL21 and disrupted the balance of T cell subtypes. The expression of T cell adhesion and migration proteins was also inhibited by ZEA. The expression of T cell chemokine receptor as well as secretion of RANTES and MIP-1α by T cells was suppressed after ZEA treatment. In summary, our results indicate that ZEA reduced the chemotactic effect of T cells mediated by chemokines, which was likely linked to the inhibition of T cell motility and accompanied by decreased expression of adhesion and migration proteins.
Collapse
Affiliation(s)
- Guodong Cai
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; .Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Shunye Pan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; China Animal Husbandry Group, Beijing, 100000, China
| | - Nannan Feng
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; .Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; .Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China
| | - Jianhong Gu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; .Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China
| | - Yan Yuan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; .Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China
| | - Xuezhong Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; .Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; .Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; .Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, Jiangsu, China.
| |
Collapse
|
135
|
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: 47] [Impact Index Per Article: 9.4] [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
|
136
|
Starvation effect on the morphology of microvilli in HeLa cells. Biochem Biophys Res Commun 2019; 514:1238-1243. [DOI: 10.1016/j.bbrc.2019.05.073] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 05/09/2019] [Indexed: 01/03/2023]
|
137
|
Möckl L, Pedram K, Roy AR, Krishnan V, Gustavsson AK, Dorigo O, Bertozzi CR, Moerner WE. Quantitative Super-Resolution Microscopy of the Mammalian Glycocalyx. Dev Cell 2019; 50:57-72.e6. [PMID: 31105009 PMCID: PMC6675415 DOI: 10.1016/j.devcel.2019.04.035] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 01/11/2019] [Accepted: 04/19/2019] [Indexed: 12/13/2022]
Abstract
The mammalian glycocalyx is a heavily glycosylated extramembrane compartment found on nearly every cell. Despite its relevance in both health and disease, studies of the glycocalyx remain hampered by a paucity of methods to spatially classify its components. We combine metabolic labeling, bioorthogonal chemistry, and super-resolution localization microscopy to image two constituents of cell-surface glycans, N-acetylgalactosamine (GalNAc) and sialic acid, with 10-20 nm precision in 2D and 3D. This approach enables two measurements: glycocalyx height and the distribution of individual sugars distal from the membrane. These measurements show that the glycocalyx exhibits nanoscale organization on both cell lines and primary human tumor cells. Additionally, we observe enhanced glycocalyx height in response to epithelial-to-mesenchymal transition and to oncogenic KRAS activation. In the latter case, we trace increased height to an effector gene, GALNT7. These data highlight the power of advanced imaging methods to provide molecular and functional insights into glycocalyx biology.
Collapse
Affiliation(s)
- Leonhard Möckl
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Kayvon Pedram
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Anish R Roy
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Venkatesh Krishnan
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Anna-Karin Gustavsson
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA; Department of Biosciences and Nutrition, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Oliver Dorigo
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Carolyn R Bertozzi
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford, CA 94305, USA.
| | - W E Moerner
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
138
|
Ilan-Ber T, Ilan Y. The role of microtubules in the immune system and as potential targets for gut-based immunotherapy. Mol Immunol 2019; 111:73-82. [DOI: 10.1016/j.molimm.2019.04.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/11/2019] [Accepted: 04/23/2019] [Indexed: 12/18/2022]
|
139
|
Kumari A, Pineau J, Sáez PJ, Maurin M, Lankar D, San Roman M, Hennig K, Boura VF, Voituriez R, Karlsson MCI, Balland M, Lennon Dumenil AM, Pierobon P. Actomyosin-driven force patterning controls endocytosis at the immune synapse. Nat Commun 2019; 10:2870. [PMID: 31253773 PMCID: PMC6599028 DOI: 10.1038/s41467-019-10751-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 05/30/2019] [Indexed: 11/30/2022] Open
Abstract
An important channel of cell-to-cell communication is direct contact. The immune synapse is a paradigmatic example of such type of interaction: it forms upon engagement of antigen receptors in lymphocytes by antigen-presenting cells and allows the local exchange of molecules and information. Although mechanics has been shown to play an important role in this process, how forces organize and impact on synapse function is unknown. We find that mechanical forces are spatio-temporally patterned at the immune synapse: global pulsatile myosin II-driven tangential forces are observed at the synapse periphery while localised forces generated by invadosome-like F-actin protrusions are detected at its centre. Noticeably, we observe that these force-producing actin protrusions constitute the main site of antigen extraction and endocytosis and require myosin II contractility to form. The interplay between global and local forces dictated by the organization of the actomyosin cytoskeleton therefore controls endocytosis at the immune synapse. The immune synapse promotes cellular information exchange but the role of biophysical forces in synapse function is unclear. Here, the authors show that B cells exert two types of forces, a centripetal myosin II-driven force and a central actin protrusive force at the site of antigen extraction.
Collapse
Affiliation(s)
- Anita Kumari
- Institut Curie, PSL Research University, INSERM U932, 26 rue d'Ulm, 75248, Paris, Cedex 05, France.,Université Paris Descartes, Paris, 75006, France
| | - Judith Pineau
- Institut Curie, PSL Research University, INSERM U932, 26 rue d'Ulm, 75248, Paris, Cedex 05, France.,Université Paris Descartes, Paris, 75006, France
| | - Pablo J Sáez
- Institut Curie, PSL Research University, INSERM U932, 26 rue d'Ulm, 75248, Paris, Cedex 05, France
| | - Mathieu Maurin
- Institut Curie, PSL Research University, INSERM U932, 26 rue d'Ulm, 75248, Paris, Cedex 05, France
| | - Danielle Lankar
- Institut Curie, PSL Research University, INSERM U932, 26 rue d'Ulm, 75248, Paris, Cedex 05, France
| | - Mabel San Roman
- Institut Curie, PSL Research University, INSERM U932, 26 rue d'Ulm, 75248, Paris, Cedex 05, France
| | - Katharina Hennig
- Laboratoire Interdisciplinaire de Physique, Université Joseph Fourier (Grenoble 1), 38402, Saint, Martin d'Hères Cedex 9, France
| | - Vanessa F Boura
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Raphael Voituriez
- Laboratoire de Physique Théorique de la Matière Condensée, UMR 7600 CNRS /UPMC and Laboratoire Jean Perrin, UMR 8237 CNRS /UPMC, 4 Place Jussieu, 75255, Paris, Cedex 05, France
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Martial Balland
- Laboratoire Interdisciplinaire de Physique, Université Joseph Fourier (Grenoble 1), 38402, Saint, Martin d'Hères Cedex 9, France
| | - Ana-Maria Lennon Dumenil
- Institut Curie, PSL Research University, INSERM U932, 26 rue d'Ulm, 75248, Paris, Cedex 05, France.
| | - Paolo Pierobon
- Institut Curie, PSL Research University, INSERM U932, 26 rue d'Ulm, 75248, Paris, Cedex 05, France.
| |
Collapse
|
140
|
Abstract
T cells initiate and regulate adaptive immune responses that can clear infections. To do this, they use their T cell receptors (TCRs) to continually scan the surfaces of other cells for cognate peptide antigens presented on major histocompatibility complexes (pMHCs). Experimental work has established that as few 1-10 pMHCs are sufficient to activate T cells. This sensitivity is remarkable in light of a number of factors, including the observation that the TCR and pMHC are short molecules relative to highly abundant long surface molecules, such as CD45, that can hinder initial binding, and moreover, the TCR/pMHC interaction is of weak affinity with solution lifetimes of approximately 1 second. Here, we review experimental and mathematical work that has contributed to uncovering molecular mechanisms of T cell sensitivity. We organize the mechanisms by where they act in the pathway to activate T cells, namely mechanisms that (a) promote TCR/pMHC binding, (b) induce rapid TCR signaling, and (c) amplify TCR signaling. We discuss work showing that high sensitivity reduces antigen specificity unless molecular feedbacks are invoked. We conclude by summarizing a number of open questions.
Collapse
Affiliation(s)
| | - Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| |
Collapse
|
141
|
Aramesh M, Stoycheva D, Raaz L, Klotzsch E. Engineering T-cell activation for immunotherapy by mechanical forces. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2019. [DOI: 10.1016/j.cobme.2019.05.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
142
|
Pullen RH, Abel SM. Mechanical feedback enables catch bonds to selectively stabilize scanning microvilli at T-cell surfaces. Mol Biol Cell 2019; 30:2087-2095. [PMID: 31116687 PMCID: PMC6727777 DOI: 10.1091/mbc.e19-01-0048] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
T-cells use microvilli to search the surfaces of antigen-presenting cells for antigenic ligands. The active motion of scanning microvilli provides a force-generating mechanism that is intriguing in light of single-molecule experiments showing that applied forces increase the lifetimes of stimulatory receptor–ligand bonds (catch-bond behavior). In this work, we introduce a theoretical framework to explore the motion of a microvillar tip above an antigen-presenting surface when receptors on the tip stochastically bind to ligands on the surface and dissociate from them in a force-dependent manner. Forces on receptor-ligand bonds impact the motion of the microvillus, leading to feedback between binding and microvillar motion. We use computer simulations to show that the average microvillar velocity varies in a ligand-dependent manner; that catch bonds generate responses in which some microvilli almost completely stop, while others move with a broad distribution of velocities; and that the frequency of stopping depends on the concentration of stimulatory ligands. Typically, a small number of catch bonds initially immobilize the microvillus, after which additional bonds accumulate and increase the cumulative receptor-engagement time. Our results demonstrate that catch bonds can selectively slow and stabilize scanning microvilli, suggesting a physical mechanism that may contribute to antigen discrimination by T-cells.
Collapse
Affiliation(s)
- Robert H Pullen
- Department of Chemical and Biomolecular Engineering, National Institute for Mathematical and Biological Synthesis, University of Tennessee, Knoxville, TN 37996
| | - Steven M Abel
- Department of Chemical and Biomolecular Engineering, National Institute for Mathematical and Biological Synthesis, University of Tennessee, Knoxville, TN 37996
| |
Collapse
|
143
|
Wan Z, Shaheen S, Chau A, Zeng Y, Liu W. Imaging: Gear up for mechano-immunology. Cell Immunol 2019; 350:103926. [PMID: 31151736 DOI: 10.1016/j.cellimm.2019.103926] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 04/15/2019] [Accepted: 05/15/2019] [Indexed: 12/17/2022]
Abstract
Immune cells including B and T lymphocytes have a remarkable ability to sense the physical perturbations through their surface expressed receptors. At the advent of modern imaging technologies paired with biophysical methods, we have gained the understanding of mechanical forces exerted by immune cells to perform their functions. This review will go over the imaging techniques already being used to study mechanical forces in immune cells. We will also discuss the dire need for new modern technologies for future work.
Collapse
Affiliation(s)
- Zhengpeng Wan
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing 100084, China; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Samina Shaheen
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Alicia Chau
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Yingyue Zeng
- School of Life Science, Liaoning University, Shenyang 110036, China
| | - Wanli Liu
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing 100084, China; Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing 100084, China.
| |
Collapse
|
144
|
Simeonov S, Schäffer TE. High-speed scanning ion conductance microscopy for sub-second topography imaging of live cells. NANOSCALE 2019; 11:8579-8587. [PMID: 30994121 DOI: 10.1039/c8nr10162k] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Scanning ion conductance microscopy (SICM) is an emerging tool for non-invasive and high-resolution topography imaging of live cells. However, the imaging speed of conventional SICM setups is slow, requiring several seconds or even minutes per image, thereby making it difficult to study cellular dynamics. Here, we describe a high-speed SICM (HS-SICM) setup for topography imaging in the hopping mode with a pixel rate of 11.0 kHz, which is 15 times faster than what was reported before. In combination with a "turn step" procedure for rapid pipette retraction, we image the ultra-fast morphodynamics of live human platelets, A6 cells, and U2OS cells at a rate as fast as 0.6 s per frame. The results show that HS-SICM provides a useful platform for investigating the dynamics of cell morphology on a sub-second timescale.
Collapse
Affiliation(s)
- Stefan Simeonov
- Institute of Applied Physics, University of Tübingen, Auf der Morgenstelle 10, 72076 Tübingen, Germany.
| | | |
Collapse
|
145
|
Torralba D, Martín-Cófreces NB, Sanchez-Madrid F. Mechanisms of polarized cell-cell communication of T lymphocytes. Immunol Lett 2019; 209:11-20. [PMID: 30954509 DOI: 10.1016/j.imlet.2019.03.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/13/2019] [Accepted: 03/17/2019] [Indexed: 01/07/2023]
Abstract
Cell-cell communication comprises a variety of molecular mechanisms that immune cells use to respond appropriately to diverse pathogenic stimuli. T lymphocytes polarize in response to different stimuli, such as cytokines, adhesion to specific ligands and cognate antigens presented in the context of MHC. Polarization takes different shapes, from migratory front-back polarization to the formation of immune synapses (IS). The formation of IS between a T cell and an antigen-presenting cell involves early events of receptor-ligand interaction leading to the reorganization of the plasma membrane and the cytoskeleton to orchestrate vesicular and endosomal traffic and directed secretion of several types of mediators, including cytokines and nanovesicles. Cell polarization involves the repositioning of many subcellular organelles, including the endosomal compartment, which becomes an effective platform for the shuttling of molecules as vesicular cargoes that lately will be secreted to transfer information to antigen-presenting cells. Overall, the polarized interaction between a T cell and APC modifies the recipient cell in different ways that are likely lineage-dependent, e.g. dendritic cells, B cells or even other T cells. In this review, we will discuss the mechanisms that mediate the polarization of different membrane receptors, cytoskeletal components and organelles in T cells in a variety of immune contexts.
Collapse
Affiliation(s)
- D Torralba
- Servicio de Inmunología, Hospital Universitario de la Princesa, UAM, IIS-IP, 28006 Madrid, Spain; Area of Vascular Pathophysiology, Laboratory of Intercellular Communication Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - N B Martín-Cófreces
- Servicio de Inmunología, Hospital Universitario de la Princesa, UAM, IIS-IP, 28006 Madrid, Spain; Area of Vascular Pathophysiology, Laboratory of Intercellular Communication Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - F Sanchez-Madrid
- Servicio de Inmunología, Hospital Universitario de la Princesa, UAM, IIS-IP, 28006 Madrid, Spain; Area of Vascular Pathophysiology, Laboratory of Intercellular Communication Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain.
| |
Collapse
|
146
|
Glatzová D, Cebecauer M. Dual Role of CD4 in Peripheral T Lymphocytes. Front Immunol 2019; 10:618. [PMID: 31001252 PMCID: PMC6454155 DOI: 10.3389/fimmu.2019.00618] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 03/08/2019] [Indexed: 01/07/2023] Open
Abstract
The interaction of T-cell receptors (TCRs) with self- and non-self-peptides in the major histocompatibility complex (MHC) stimulates crucial signaling events, which in turn can activate T lymphocytes. A variety of accessory molecules further modulate T-cell signaling. Of these, the CD4 and CD8 coreceptors make the most critical contributions to T cell sensitivity in vivo. Whereas, CD4 function in T cell development is well-characterized, its role in peripheral T cells remains incompletely understood. It was originally suggested that CD4 stabilizes weak interactions between TCRs and peptides in the MHC and delivers Lck kinases to that complex. The results of numerous experiments support the latter role, indicating that the CD4-Lck complex accelerates TCR-triggered signaling and controls the availability of the kinase for TCR in the absence of the ligand. On the other hand, extremely low affinity of CD4 for MHC rules out its ability to stabilize the receptor-ligand complex. In this review, we summarize the current knowledge on CD4 in T cells, with a special emphasis on the spatio-temporal organization of early signaling events and the relevance for CD4 function. We further highlight the capacity of CD4 to interact with the MHC in the absence of TCR. It drives the adhesion of T cells to the cells that express the MHC. This process is facilitated by the CD4 accumulation in the tips of microvilli on the surface of unstimulated T cells. Based on these observations, we suggest an alternative model of CD4 role in T-cell activation.
Collapse
Affiliation(s)
- Daniela Glatzová
- Department of Biophysical Chemistry, J. Heyrovsky Institute of Physical Chemistry of the Czech Academy of Sciences, Prague, Czechia
- Laboratory of Leukocyte Signaling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Marek Cebecauer
- Department of Biophysical Chemistry, J. Heyrovsky Institute of Physical Chemistry of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
147
|
Pageon SV, Govendir MA, Kempe D, Biro M. Mechanoimmunology: molecular-scale forces govern immune cell functions. Mol Biol Cell 2019; 29:1919-1926. [PMID: 30088799 PMCID: PMC6232972 DOI: 10.1091/mbc.e18-02-0120] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Immune cell recognition of antigens is a pivotal process in initiating immune responses against injury, pathogens, and cancers. Breakthroughs over the past decade support a major role for mechanical forces in immune responses, laying the foundation for the emerging field of mechanoimmunology. In this Perspective, we discuss the mechanical forces acting at the level of ligand–receptor interactions and how they underpin receptor triggering, signal initiation, and immune cell activation. We also highlight the novel biophysical tools and advanced imaging techniques that have afforded us the recent progress in our understanding of the role of forces in immune cell functions.
Collapse
Affiliation(s)
- Sophie V Pageon
- EMBL Australia, Single Molecule Science Node, School of Medical Sciences, and
| | - Matt A Govendir
- EMBL Australia, Single Molecule Science Node, School of Medical Sciences, and
| | - Daryan Kempe
- EMBL Australia, Single Molecule Science Node, School of Medical Sciences, and
| | - Maté Biro
- EMBL Australia, Single Molecule Science Node, School of Medical Sciences, and.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
148
|
Liu Z, Liu J, Sun Z, Zhang Z, Yuan Y, Fang X, Wang F, Qin W, Wu C. Cooperative Blinking from Dye Ensemble Activated by Energy Transfer for Super-resolution Cellular Imaging. Anal Chem 2019; 91:4179-4185. [DOI: 10.1021/acs.analchem.9b00279] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Zhihe Liu
- State Key Laboratory of Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun 130012, China
| | - Jie Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 510855, China
| | - Zezhou Sun
- State Key Laboratory of Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun 130012, China
| | - Zhe Zhang
- State Key Laboratory of Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun 130012, China
| | - Ye Yuan
- State Key Laboratory of Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun 130012, China
| | - Xiaofeng Fang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 510855, China
| | - Fei Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 510855, China
| | - Weiping Qin
- State Key Laboratory of Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun 130012, China
| | - Changfeng Wu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 510855, China
| |
Collapse
|
149
|
Goyette J, Nieves DJ, Ma Y, Gaus K. How does T cell receptor clustering impact on signal transduction? J Cell Sci 2019; 132:132/4/jcs226423. [DOI: 10.1242/jcs.226423] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
ABSTRACT
The essential function of the T cell receptor (TCR) is to translate the engagement of peptides on the major histocompatibility complex (pMHC) into appropriate intracellular signals through the associated cluster of differentiation 3 (CD3) complex. The spatial organization of the TCR–CD3 complex in the membrane is thought to be a key regulatory element of signal transduction, raising the question of how receptor clustering impacts on TCR triggering. How signal transduction at the TCR–CD3 complex encodes the quality and quantity of pMHC molecules is not fully understood. This question can be approached by reconstituting T cell signaling in model and cell membranes and addressed by single-molecule imaging of endogenous proteins in T cells. We highlight such methods and further discuss how TCR clustering could affect pMHC rebinding rates, the local balance between kinase and phosphatase activity and/or the lipid environment to regulate the signal efficiency of the TCR–CD3 complex. We also examine whether clustering could affect the conformation of cytoplasmic CD3 tails through a biophysical mechanism. Taken together, we highlight how the spatial organization of the TCR–CD3 complex – addressed by reconstitution approaches – has emerged as a key regulatory element in signal transduction of this archetypal immune receptor.
Collapse
Affiliation(s)
- Jesse Goyette
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney 2052, Australia
- ARC Centre of Excellence in Advanced Molecular imaging, University of New South Wales, Sydney 2052, Australia
| | - Daniel J. Nieves
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney 2052, Australia
- ARC Centre of Excellence in Advanced Molecular imaging, University of New South Wales, Sydney 2052, Australia
| | - Yuanqing Ma
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney 2052, Australia
- ARC Centre of Excellence in Advanced Molecular imaging, University of New South Wales, Sydney 2052, Australia
| | - Katharina Gaus
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney 2052, Australia
- ARC Centre of Excellence in Advanced Molecular imaging, University of New South Wales, Sydney 2052, Australia
| |
Collapse
|
150
|
Brockman JM, Salaita K. Mechanical Proofreading: A General Mechanism to Enhance the Fidelity of Information Transfer Between Cells. FRONTIERS IN PHYSICS 2019; 7:14. [PMID: 31328129 PMCID: PMC6641563 DOI: 10.3389/fphy.2019.00014] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The cells and receptors of the immune system are mechanically active. Single molecule force spectroscopy, traction force microscopy, and molecular tension probe measurements all point to the importance of piconewton (pN) molecular forces in immune function. For example, forces enhance the ability of a T cell to discriminate between nearly identical antigens. The role of molecular forces at these critical immune recognition junctions is puzzling because mechanical forces generally facilitate bond dissociation, potentially increasing the difficulty for a receptor to recognize its cognate antigen. The advantage molecular forces confer in the process of immune recognition is not clear. Why would cells expend energy to exert force on the critical, but tenuous bonds that mediate immune surveillance? Do molecular forces provide some advantage to the immune system? The premise of this review is that molecular forces provide a specificity advantage to immune cells. Inspired by the recent discovery that receptor forces regulate immune signaling in T cells and B cells, we dub this notion "mechanical proofreading," akin to more classic kinetic proofreading models. During the process of mechanical proofreading, cells exert pN receptor forces on receptor-ligand interactions, deliberately increasing the energy cost of the immune recognition process in exchange for increased specificity of signaling. Here, we review the role of molecular forces in the immune system and suggest how these forces may facilitate mechanical proofreading to increase the specificity of the immune response.
Collapse
Affiliation(s)
- Joshua M Brockman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Khalid Salaita
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
- Department of Chemistry, Emory University, Atlanta, Georgia, USA
| |
Collapse
|