1
|
Mittelheisser V, Gensbittel V, Bonati L, Li W, Tang L, Goetz JG. Evidence and therapeutic implications of biomechanically regulated immunosurveillance in cancer and other diseases. NATURE NANOTECHNOLOGY 2024; 19:281-297. [PMID: 38286876 DOI: 10.1038/s41565-023-01535-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 09/26/2023] [Indexed: 01/31/2024]
Abstract
Disease progression is usually accompanied by changes in the biochemical composition of cells and tissues and their biophysical properties. For instance, hallmarks of cancer include the stiffening of tissues caused by extracellular matrix remodelling and the softening of individual cancer cells. In this context, accumulating evidence has shown that immune cells sense and respond to mechanical signals from the environment. However, the mechanisms regulating these mechanical aspects of immune surveillance remain partially understood. The growing appreciation for the 'mechano-immunology' field has urged researchers to investigate how immune cells sense and respond to mechanical cues in various disease settings, paving the way for the development of novel engineering strategies that aim at mechanically modulating and potentiating immune cells for enhanced immunotherapies. Recent pioneer developments in this direction have laid the foundations for leveraging 'mechanical immunoengineering' strategies to treat various diseases. This Review first outlines the mechanical changes occurring during pathological progression in several diseases, including cancer, fibrosis and infection. We next highlight the mechanosensitive nature of immune cells and how mechanical forces govern the immune responses in different diseases. Finally, we discuss how targeting the biomechanical features of the disease milieu and immune cells is a promising strategy for manipulating therapeutic outcomes.
Collapse
Affiliation(s)
- Vincent Mittelheisser
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Valentin Gensbittel
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Lucia Bonati
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Weilin Li
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Li Tang
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- Institute of Materials Science and Engineering, EPFL, Lausanne, Switzerland.
| | - Jacky G Goetz
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France.
- Université de Strasbourg, Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France.
| |
Collapse
|
2
|
Liu GY, Chen S, Lee G, Shaiv K, Chen P, Cheng H, Hong S, Yang W, Huang S, Chang Y, Wang H, Kao C, Sun P, Chao M, Lee Y, Tang M, Lin Y. Precise control of microtubule disassembly in living cells. EMBO J 2022; 41:e110472. [PMID: 35686621 PMCID: PMC9340485 DOI: 10.15252/embj.2021110472] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/15/2022] [Accepted: 05/05/2022] [Indexed: 12/28/2022] Open
Abstract
Microtubules tightly regulate various cellular activities. Our understanding of microtubules is largely based on experiments using microtubule-targeting agents, which, however, are insufficient to dissect the dynamic mechanisms of specific microtubule populations, due to their slow effects on the entire pool of microtubules. To overcome this technological limitation, we have used chemo and optogenetics to disassemble specific microtubule subtypes, including tyrosinated microtubules, primary cilia, mitotic spindles, and intercellular bridges, by rapidly recruiting engineered microtubule-cleaving enzymes onto target microtubules in a reversible manner. Using this approach, we show that acute microtubule disassembly swiftly halts vesicular trafficking and lysosomal dynamics. It also immediately triggers Golgi and ER reorganization and slows the fusion/fission of mitochondria without affecting mitochondrial membrane potential. In addition, cell rigidity is increased after microtubule disruption owing to increased contractile stress fibers. Microtubule disruption furthermore prevents cell division, but does not cause cell death during interphase. Overall, the reported tools facilitate detailed analysis of how microtubules precisely regulate cellular architecture and functions.
Collapse
Affiliation(s)
- Grace Y Liu
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Shiau‐Chi Chen
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Gang‐Hui Lee
- Department of Physiology, College of MedicineNational Cheng Kung UniversityTainanTaiwan
- International Center for Wound Repair and RegenerationNational Cheng Kung UniversityTainanTaiwan
| | - Kritika Shaiv
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Pin‐Yu Chen
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Hsuan Cheng
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Shi‐Rong Hong
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Wen‐Ting Yang
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Shih‐Han Huang
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Ya‐Chu Chang
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Hsien‐Chu Wang
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Ching‐Lin Kao
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Pin‐Chiao Sun
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Ming‐Hong Chao
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Yian‐Ying Lee
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Ming‐Jer Tang
- Department of Physiology, College of MedicineNational Cheng Kung UniversityTainanTaiwan
- International Center for Wound Repair and RegenerationNational Cheng Kung UniversityTainanTaiwan
| | - Yu‐Chun Lin
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
- Department of Medical ScienceNational Tsing Hua UniversityHsinchuTaiwan
| |
Collapse
|
3
|
Engineering T cells to enhance 3D migration through structurally and mechanically complex tumor microenvironments. Nat Commun 2021; 12:2815. [PMID: 33990566 PMCID: PMC8121808 DOI: 10.1038/s41467-021-22985-5] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 04/07/2021] [Indexed: 12/18/2022] Open
Abstract
Defining the principles of T cell migration in structurally and mechanically complex tumor microenvironments is critical to understanding escape from antitumor immunity and optimizing T cell-related therapeutic strategies. Here, we engineered nanotextured elastic platforms to study and enhance T cell migration through complex microenvironments and define how the balance between contractility localization-dependent T cell phenotypes influences migration in response to tumor-mimetic structural and mechanical cues. Using these platforms, we characterize a mechanical optimum for migration that can be perturbed by manipulating an axis between microtubule stability and force generation. In 3D environments and live tumors, we demonstrate that microtubule instability, leading to increased Rho pathway-dependent cortical contractility, promotes migration whereas clinically used microtubule-stabilizing chemotherapies profoundly decrease effective migration. We show that rational manipulation of the microtubule-contractility axis, either pharmacologically or through genome engineering, results in engineered T cells that more effectively move through and interrogate 3D matrix and tumor volumes. Thus, engineering cells to better navigate through 3D microenvironments could be part of an effective strategy to enhance efficacy of immune therapeutics. The mechanics of the migration of T cells into tumours is an important aspect of tumour immunity. Here the authors engineer complex 3D environments to explore functions of microtubules and cell contractility as strategies to enhance T cell migration in tumour microenvironments.
Collapse
|
4
|
Franchini DM, Lanvin O, Tosolini M, Patras de Campaigno E, Cammas A, Péricart S, Scarlata CM, Lebras M, Rossi C, Ligat L, Pont F, Arimondo PB, Laurent C, Ayyoub M, Despas F, Lapeyre-Mestre M, Millevoi S, Fournié JJ. Microtubule-Driven Stress Granule Dynamics Regulate Inhibitory Immune Checkpoint Expression in T Cells. Cell Rep 2020; 26:94-107.e7. [PMID: 30605689 DOI: 10.1016/j.celrep.2018.12.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 11/02/2018] [Accepted: 12/03/2018] [Indexed: 12/31/2022] Open
Abstract
Despite the clinical success of blocking inhibitory immune checkpoint receptors such as programmed cell death-1 (PD-1) in cancer, the mechanisms controlling the expression of these receptors have not been fully elucidated. Here, we identify a post-transcriptional mechanism regulating PD-1 expression in T cells. Upon activation, the PDCD1 mRNA and ribonucleoprotein complexes coalesce into stress granules that require microtubules and the kinesin 1 molecular motor to proceed to translation. Hence, PD-1 expression is highly sensitive to microtubule or stress granule inhibitors targeting this pathway. Evidence from healthy donors and cancer patients reveals a common regulation for the translation of CTLA4, LAG3, TIM3, TIGIT, and BTLA but not of the stimulatory co-receptors OX40, GITR, and 4-1BB mRNAs. In patients, disproportionality analysis of immune-related adverse events for currently used microtubule drugs unveils a significantly higher risk of autoimmunity. Our findings reveal a fundamental mechanism of immunoregulation with great importance in cancer immunotherapy.
Collapse
Affiliation(s)
- Don-Marc Franchini
- Cancer Research Centre of Toulouse, INSERM UMR 1037, 31037 Toulouse, France; Université Toulouse III Paul Sabatier, 31330 Toulouse, France; ERL 5294, CNRS, 31037 Toulouse, France; Institut Universitaire du Cancer de Toulouse-Oncopole, 31100 Toulouse, France; Laboratoire d'Excellence "TOUCAN," Toulouse, France; Programme Hospitalo-Universitaire en Cancérologie CAPTOR, 31059 Toulouse, France; Institut Carnot Lymphome CALYM, 69495 Pierre-Benite, France.
| | - Olivia Lanvin
- Cancer Research Centre of Toulouse, INSERM UMR 1037, 31037 Toulouse, France; Université Toulouse III Paul Sabatier, 31330 Toulouse, France; ERL 5294, CNRS, 31037 Toulouse, France; Institut Universitaire du Cancer de Toulouse-Oncopole, 31100 Toulouse, France; Laboratoire d'Excellence "TOUCAN," Toulouse, France; Programme Hospitalo-Universitaire en Cancérologie CAPTOR, 31059 Toulouse, France; Institut Carnot Lymphome CALYM, 69495 Pierre-Benite, France
| | - Marie Tosolini
- Cancer Research Centre of Toulouse, INSERM UMR 1037, 31037 Toulouse, France; Université Toulouse III Paul Sabatier, 31330 Toulouse, France; ERL 5294, CNRS, 31037 Toulouse, France; Institut Universitaire du Cancer de Toulouse-Oncopole, 31100 Toulouse, France; Laboratoire d'Excellence "TOUCAN," Toulouse, France; Programme Hospitalo-Universitaire en Cancérologie CAPTOR, 31059 Toulouse, France; Institut Carnot Lymphome CALYM, 69495 Pierre-Benite, France
| | - Emilie Patras de Campaigno
- Medical and Clinical Pharmacology Unit, CHU Toulouse University Hospital, 31000 Toulouse, France; Medical and Pharmacoepidemiology Research Unit, INSERM 1027, 31000 Toulouse, France; Centre d'Investigations Cliniques, CIC 1436, Toulouse University Hospital, 31000 Toulouse, France
| | - Anne Cammas
- Cancer Research Centre of Toulouse, INSERM UMR 1037, 31037 Toulouse, France; Université Toulouse III Paul Sabatier, 31330 Toulouse, France; Laboratoire d'Excellence "TOUCAN," Toulouse, France
| | - Sarah Péricart
- Cancer Research Centre of Toulouse, INSERM UMR 1037, 31037 Toulouse, France; Université Toulouse III Paul Sabatier, 31330 Toulouse, France; ERL 5294, CNRS, 31037 Toulouse, France; Institut Universitaire du Cancer de Toulouse-Oncopole, 31100 Toulouse, France; Laboratoire d'Excellence "TOUCAN," Toulouse, France; Programme Hospitalo-Universitaire en Cancérologie CAPTOR, 31059 Toulouse, France; Institut Carnot Lymphome CALYM, 69495 Pierre-Benite, France
| | - Clara-Maria Scarlata
- Cancer Research Centre of Toulouse, INSERM UMR 1037, 31037 Toulouse, France; Université Toulouse III Paul Sabatier, 31330 Toulouse, France; Institut Universitaire du Cancer de Toulouse-Oncopole, 31100 Toulouse, France
| | - Morgane Lebras
- Cancer Research Centre of Toulouse, INSERM UMR 1037, 31037 Toulouse, France; Université Toulouse III Paul Sabatier, 31330 Toulouse, France; Laboratoire d'Excellence "TOUCAN," Toulouse, France
| | - Cédric Rossi
- Cancer Research Centre of Toulouse, INSERM UMR 1037, 31037 Toulouse, France; Université Toulouse III Paul Sabatier, 31330 Toulouse, France; ERL 5294, CNRS, 31037 Toulouse, France; Institut Universitaire du Cancer de Toulouse-Oncopole, 31100 Toulouse, France; Laboratoire d'Excellence "TOUCAN," Toulouse, France; Programme Hospitalo-Universitaire en Cancérologie CAPTOR, 31059 Toulouse, France; Institut Carnot Lymphome CALYM, 69495 Pierre-Benite, France
| | - Laetitia Ligat
- Cancer Research Centre of Toulouse, INSERM UMR 1037, 31037 Toulouse, France; Université Toulouse III Paul Sabatier, 31330 Toulouse, France
| | - Fréderic Pont
- Cancer Research Centre of Toulouse, INSERM UMR 1037, 31037 Toulouse, France; Université Toulouse III Paul Sabatier, 31330 Toulouse, France
| | - Paola B Arimondo
- Programme Hospitalo-Universitaire en Cancérologie CAPTOR, 31059 Toulouse, France; Epigenetic Targeting of Cancer, FRE3600 CNRS, 31035 Toulouse, France
| | - Camille Laurent
- Cancer Research Centre of Toulouse, INSERM UMR 1037, 31037 Toulouse, France; Université Toulouse III Paul Sabatier, 31330 Toulouse, France; ERL 5294, CNRS, 31037 Toulouse, France; Institut Universitaire du Cancer de Toulouse-Oncopole, 31100 Toulouse, France; Laboratoire d'Excellence "TOUCAN," Toulouse, France; Programme Hospitalo-Universitaire en Cancérologie CAPTOR, 31059 Toulouse, France; Institut Carnot Lymphome CALYM, 69495 Pierre-Benite, France
| | - Maha Ayyoub
- Cancer Research Centre of Toulouse, INSERM UMR 1037, 31037 Toulouse, France; Université Toulouse III Paul Sabatier, 31330 Toulouse, France; Institut Universitaire du Cancer de Toulouse-Oncopole, 31100 Toulouse, France
| | - Fabien Despas
- Medical and Clinical Pharmacology Unit, CHU Toulouse University Hospital, 31000 Toulouse, France; Medical and Pharmacoepidemiology Research Unit, INSERM 1027, 31000 Toulouse, France; Centre d'Investigations Cliniques, CIC 1436, Toulouse University Hospital, 31000 Toulouse, France
| | - Maryse Lapeyre-Mestre
- Medical and Clinical Pharmacology Unit, CHU Toulouse University Hospital, 31000 Toulouse, France; Medical and Pharmacoepidemiology Research Unit, INSERM 1027, 31000 Toulouse, France; Centre d'Investigations Cliniques, CIC 1436, Toulouse University Hospital, 31000 Toulouse, France
| | - Stefania Millevoi
- Cancer Research Centre of Toulouse, INSERM UMR 1037, 31037 Toulouse, France; Université Toulouse III Paul Sabatier, 31330 Toulouse, France; Laboratoire d'Excellence "TOUCAN," Toulouse, France.
| | - Jean-Jacques Fournié
- Cancer Research Centre of Toulouse, INSERM UMR 1037, 31037 Toulouse, France; Université Toulouse III Paul Sabatier, 31330 Toulouse, France; ERL 5294, CNRS, 31037 Toulouse, France; Institut Universitaire du Cancer de Toulouse-Oncopole, 31100 Toulouse, France; Laboratoire d'Excellence "TOUCAN," Toulouse, France; Programme Hospitalo-Universitaire en Cancérologie CAPTOR, 31059 Toulouse, France; Institut Carnot Lymphome CALYM, 69495 Pierre-Benite, France.
| |
Collapse
|
5
|
Balagopalan L, Yi J, Nguyen T, McIntire KM, Harned AS, Narayan K, Samelson LE. Plasma membrane LAT activation precedes vesicular recruitment defining two phases of early T-cell activation. Nat Commun 2018; 9:2013. [PMID: 29789604 PMCID: PMC5964120 DOI: 10.1038/s41467-018-04419-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 04/24/2018] [Indexed: 12/20/2022] Open
Abstract
The relative importance of plasma membrane-localized LAT versus vesicular LAT for microcluster formation and T-cell receptor (TCR) activation is unclear. Here, we show the sequence of events in LAT microcluster formation and vesicle delivery, using lattice light sheet microscopy to image a T cell from the earliest point of activation. A kinetic lag occurs between LAT microcluster formation and vesicular pool recruitment to the synapse. Correlative 3D light and electron microscopy show an absence of vesicles at microclusters at early times, but an abundance of vesicles as activation proceeds. Using TIRF-SIM to look at the activated T-cell surface with high resolution, we capture directed vesicle movement between microclusters on microtubules. We propose a model in which cell surface LAT is recruited rapidly and phosphorylated at sites of T-cell activation, while the vesicular pool is subsequently recruited and dynamically interacts with microclusters.
Collapse
Affiliation(s)
- Lakshmi Balagopalan
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD, 20892, USA.
| | - Jason Yi
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD, 20892, USA
| | - Tiffany Nguyen
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD, 20892, USA
| | - Katherine M McIntire
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD, 20892, USA
| | - Adam S Harned
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 8560 Progress Drive, Frederick, MD, 21701, USA
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Kedar Narayan
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 8560 Progress Drive, Frederick, MD, 21701, USA
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Lawrence E Samelson
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD, 20892, USA.
| |
Collapse
|
6
|
Abstract
T lymphocytes use surface [Formula: see text] T-cell receptors (TCRs) to recognize peptides bound to MHC molecules (pMHCs) on antigen-presenting cells (APCs). How the exquisite specificity of high-avidity T cells is achieved is unknown but essential, given the paucity of foreign pMHC ligands relative to the ubiquitous self-pMHC array on an APC. Using optical traps, we determine physicochemical triggering thresholds based on load and force direction. Strikingly, chemical thresholds in the absence of external load require orders of magnitude higher pMHC numbers than observed physiologically. In contrast, force applied in the shear direction ([Formula: see text]10 pN per TCR molecule) triggers T-cell Ca2+ flux with as few as two pMHC molecules at the interacting surface interface with rapid positional relaxation associated with similarly directed motor-dependent transport via [Formula: see text]8-nm steps, behaviors inconsistent with serial engagement during initial TCR triggering. These synergistic directional forces generated during cell motility are essential for adaptive T-cell immunity against infectious pathogens and cancers.
Collapse
|
7
|
Dynamic microtubules regulate cellular contractility during T-cell activation. Proc Natl Acad Sci U S A 2017; 114:E4175-E4183. [PMID: 28490501 DOI: 10.1073/pnas.1614291114] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
T-cell receptor (TCR) triggering and subsequent T-cell activation are essential for the adaptive immune response. Recently, multiple lines of evidence have shown that force transduction across the TCR complex is involved during TCR triggering, and that the T cell might use its force-generation machinery to probe the mechanical properties of the opposing antigen-presenting cell, giving rise to different signaling and physiological responses. Mechanistically, actin polymerization and turnover have been shown to be essential for force generation by T cells, but how these actin dynamics are regulated spatiotemporally remains poorly understood. Here, we report that traction forces generated by T cells are regulated by dynamic microtubules (MTs) at the interface. These MTs suppress Rho activation, nonmuscle myosin II bipolar filament assembly, and actin retrograde flow at the T-cell-substrate interface. Our results suggest a novel role of the MT cytoskeleton in regulating force generation during T-cell activation.
Collapse
|
8
|
Guedj C, Abraham N, Jullié D, Randriamampita C. T cell adhesion triggers an early signaling pole distal to the immune synapse. J Cell Sci 2016; 129:2526-37. [PMID: 27185862 DOI: 10.1242/jcs.182311] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 05/09/2016] [Indexed: 01/09/2023] Open
Abstract
The immunological synapse forms at the interface between a T cell and an antigen-presenting cell after foreign antigen recognition. The immunological synapse is considered to be the site where the signaling cascade leading to T lymphocyte activation is triggered. Here, we show that another signaling region can be detected before formation of the synapse at the opposite pole of the T cell. This structure appears during the first minute after the contact forms, is transient and contains all the classic components that have been previously described at the immunological synapse. Its formation is independent of antigen recognition but is driven by adhesion itself. It constitutes a reservoir of signaling molecules that are potentially ready to be sent to the immunological synapse through a microtubule-dependent pathway. The antisynapse can thus be considered as a pre-synapse that is triggered independently of antigen recognition.
Collapse
Affiliation(s)
- Chloé Guedj
- INSERM, U1016, Institut Cochin, Infection, Immunity and Inflammation Department, 22 rue Méćhain, Paris 75014, France CNRS, UMR8104, Paris 75014, France Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France
| | - Nicolas Abraham
- INSERM, U1016, Institut Cochin, Infection, Immunity and Inflammation Department, 22 rue Méćhain, Paris 75014, France CNRS, UMR8104, Paris 75014, France Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France
| | - Damien Jullié
- INSERM, U1016, Institut Cochin, Infection, Immunity and Inflammation Department, 22 rue Méćhain, Paris 75014, France CNRS, UMR8104, Paris 75014, France Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France
| | - Clotilde Randriamampita
- INSERM, U1016, Institut Cochin, Infection, Immunity and Inflammation Department, 22 rue Méćhain, Paris 75014, France CNRS, UMR8104, Paris 75014, France Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France
| |
Collapse
|
9
|
Type II PtdIns 4-kinase β associates with CD4-p56lck complex and is involved in CD4 receptor signaling. Mol Cell Biochem 2014; 395:231-9. [PMID: 24972704 DOI: 10.1007/s11010-014-2129-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 06/17/2014] [Indexed: 11/27/2022]
Abstract
Type II phosphatidylinositol (PtdIns) 4-kinases are involved in the synthesis of PtdIns 4-phosphates and modulate various cell functions like, intracellular signaling, cytoskeletal rearrangements, vesicular trafficking, and pathogen invasion. In CD3 receptor activated T cells, a type II PtdIns 4-kinase β is recruited to CD3 receptor zeta and plays a role in intracellular calcium release and probably in actin cytoskeleton reorganization. T cell receptor mediated activation is supported by CD4 receptor. The role of type II PtdIns 4-kinase β in CD4 receptor-mediated signaling was addressed in the present manuscript. Crosslinking of CD4 receptors with monoclonal antibodies showed an increase in CD4-associated PtdIns 4-kinase activity and requires p56(lck) activity. Biochemical characterization suggests that it belongs to type II PtdIns 4-kinase family. shRNA mediated knockdown of type II PtdIns 4-kinase β showed abrogation of CD4 receptor induced intracellular calcium release. These results suggest that type II PtdIns 4-kinase β plays an integral part in CD4 receptor-mediated signaling.
Collapse
|
10
|
Wan Z, Zhang S, Fan Y, Liu K, Du F, Davey AM, Zhang H, Han W, Xiong C, Liu W. B Cell Activation Is Regulated by the Stiffness Properties of the Substrate Presenting the Antigens. THE JOURNAL OF IMMUNOLOGY 2013; 190:4661-75. [DOI: 10.4049/jimmunol.1202976] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
11
|
Filbert EL, Le Borgne M, Lin J, Heuser JE, Shaw AS. Stathmin regulates microtubule dynamics and microtubule organizing center polarization in activated T cells. THE JOURNAL OF IMMUNOLOGY 2012; 188:5421-7. [PMID: 22529300 DOI: 10.4049/jimmunol.1200242] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Polarization of T cells involves reorientation of the microtubule organizing center (MTOC). Because activated ERK is localized at the immunological synapse, we investigated its role by showing that ERK activation is important for MTOC polarization. Suspecting that ERK phosphorylates a regulator of microtubules, we next focused on stathmin, a known ERK substrate. Our work indicates that during T cell activation, ERK is recruited to the synapse, allowing it to phosphorylate stathmin molecules near the immunological synapse. Supporting an important role of stathmin phosphorylation in T cell activation, we showed that T cell activation results in increased microtubule growth rate dependent on the presence of stathmin. The significance of this finding was demonstrated by results showing that CTLs from stathmin(-/-) mice displayed defective MTOC polarization and defective target cell cytolysis. These data implicate stathmin as a regulator of the microtubule network during T cell activation.
Collapse
Affiliation(s)
- Erin L Filbert
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
12
|
Hashimoto-Tane A, Yokosuka T, Sakata-Sogawa K, Sakuma M, Ishihara C, Tokunaga M, Saito T. Dynein-driven transport of T cell receptor microclusters regulates immune synapse formation and T cell activation. Immunity 2011; 34:919-31. [PMID: 21703543 DOI: 10.1016/j.immuni.2011.05.012] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 02/11/2011] [Accepted: 05/26/2011] [Indexed: 11/27/2022]
Abstract
When T cells recognize a peptide-major histocompatibility complex on antigen-presenting cells (APCs), T cell receptor microclusters (TCR-MCs) are generated and move to the center of the T cell-APC interface to form the central supramolecular activation cluster (cSMAC). cSMAC formation depends on stimulation strength and regulates T cell activation. We demonstrate that the dynein motor complex colocalized and coimmunoprecipitated with the TCR complex and that TCR-MCs moved along microtubules (MTs) toward the center of the immune synapse in a dynein-dependent manner to form cSMAC. MTs are located in close proximity to the plasma membrane at the activation site. TCR-MC velocity and cSMAC formation were impaired by dynein or MT inhibitors or by ablation of dynein expression. T cells with impaired cSMAC formation exhibited enhanced cellular activation including protein phosphorylation and interleukin-2 production. These results indicate that cSMAC formation by TCR-MC movement depends on dynein and MTs, and the movement regulates T cell activation.
Collapse
Affiliation(s)
- Akiko Hashimoto-Tane
- Laboratory for Cell Signaling, RIKEN Research Center for Allergy and Immunology, Yokohama, Kanagawa 230-0045, Japan
| | | | | | | | | | | | | |
Collapse
|
13
|
Jung DW, Williams DR. Novel chemically defined approach to produce multipotent cells from terminally differentiated tissue syncytia. ACS Chem Biol 2011; 6:553-62. [PMID: 21322636 DOI: 10.1021/cb2000154] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In urodele amphibians, a critical step in limb regeneration is the cellularization and dedifferentiation of skeletal muscle. In contrast, mammalian skeletal muscle does not undergo this response to injury. We have developed a novel simple, stepwise chemical method to induce dedifferentiation and multipotency in mammalian skeletal muscle. Optimal muscle fiber cellularization was induced by the trisubstituted purine small molecule, myoseverin, compared to colchicine, nocodazole, or myoseverin B. The induction of a proliferative response in the cellulate was found to be a crucial step in the dedifferentiation process. This was achieved by down-regulation of the cyclin-dependent kinase inhibitor, p21 (CDKN 1A, CIP1). p21 was found to be a key regulator of this process, because down-regulation of the cyclin-dependent kinase inhibitors p27 (CDKN1B/KIP1) or p57 (CDKN1C/KIP2) or the tumor suppressor p53 (TP53/LFS1) failed to induce proliferation and subsequent dedifferentiation. Treatment with the small molecule reversine (2-(4-morpholinoanilino)-6-cyclohexylaminopurine) during this proliferative "window" induced the muscle cellulate to differentiate into non-muscle cell types. This lineage switching was assessed using a relatively stringent approach, based on comparative functional and phenotypic assays of cell-type specific properties. This showed that our chemical method allowed the derivation of adipogenic and osteogenic cells that possessed a degree of functionality. This is the first demonstration that mammalian muscle culture can be induced to undergo cellularization, proliferation, and dedifferentiation, which is grossly similar to the key early steps in urodele limb regeneration. These results, based solely on the use of simple chemical approaches, have implications for both regenerative medicine and stem cell biology.
Collapse
Affiliation(s)
- Da-Woon Jung
- Small Molecule Regulators and Biosystems Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, 1 Oryong-Dong, Buk-Gu, Gwangju 500-712, Republic of Korea
| | - Darren R. Williams
- Small Molecule Regulators and Biosystems Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, 1 Oryong-Dong, Buk-Gu, Gwangju 500-712, Republic of Korea
| |
Collapse
|
14
|
Effects of microtubule modulators on HIV-1 infection of transformed and resting CD4 T cells. J Virol 2011; 85:3020-4. [PMID: 21209111 DOI: 10.1128/jvi.02462-10] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Previous studies have observed fluorescently labeled HIV particles tracking along microtubule networks for nuclear localization. To provide direct evidence for the involvement of microtubules in early steps of HIV infection of human CD4 T cells, we used multiple microtubule modulators such as paclitaxel (originally called taxol; 1 μM), vinblastine (1 and 10 μM), colchicine (10 and 100 μM), and nocodazole (10 and 100 μM) to disturb microtubule networks in transformed and resting CD4 T cells. Although these drugs disrupted microtubule integrity, almost no inhibition of HIV-1 infection was observed. Our results do not appear to support an essential role for microtubules in the initiation of HIV infection of CD4 T cells.
Collapse
|
15
|
Liu W, Meckel T, Tolar P, Sohn HW, Pierce SK. Antigen affinity discrimination is an intrinsic function of the B cell receptor. ACTA ACUST UNITED AC 2010; 207:1095-111. [PMID: 20404102 PMCID: PMC2867278 DOI: 10.1084/jem.20092123] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Antibody affinity maturation, a hallmark of adaptive immune responses, results from the selection of B cells expressing somatically hypermutated B cell receptors (BCRs) with increased affinity for antigens. Despite the central role of affinity maturation in antibody responses, the molecular mechanisms by which the increased affinity of a B cell for antigen is translated into a selective advantage for that B cell in immune responses is incompletely understood. We use high resolution live-cell imaging to provide evidence that the earliest BCR-intrinsic events that follow within seconds of BCR-antigen binding are highly sensitive to the affinity of the BCR for antigen. High affinity BCRs readily form oligomers and the resulting microclusters grow rapidly, resulting in enhanced recruitment of Syk kinase and calcium fluxes. Thus, B cells are able to read the affinity of antigen by BCR-intrinsic mechanisms during the earliest phases of BCR clustering, leading to the initiation of B cell responses.
Collapse
Affiliation(s)
- Wanli Liu
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | | | | | | | | |
Collapse
|
16
|
Thomas EC, Zhe Y, Molero JC, Schmitz-Peiffer C, Ramm G, James DE, Whitehead JP. The subcellular fractionation properties and function of insulin receptor substrate-1 (IRS-1) are independent of cytoskeletal integrity. Int J Biochem Cell Biol 2006; 38:1686-99. [PMID: 16702017 DOI: 10.1016/j.biocel.2006.03.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2006] [Revised: 03/15/2006] [Accepted: 03/20/2006] [Indexed: 10/24/2022]
Abstract
Efficient insulin action requires spatial and temporal coordination of signaling cascades. The prototypical insulin receptor substrate, IRS-1 plays a central role in insulin signaling. By subcellular fractionation IRS-1 is enriched in a particulate fraction, termed the high speed pellet (HSP), and its redistribution from this fraction is associated with signal attenuation and insulin resistance. Anecdotal evidence suggests the cytoskeleton may underpin the localization of IRS-1 to the HSP. In the present study we have taken a systematic approach to examine whether the cytoskeleton contributes to the subcellular fractionation properties and function of IRS-1. By standard microscopy or immunoprecipitation we were unable to detect evidence to support a specific interaction between IRS-1 and the major cytoskeletal components actin (microfilaments), vimentin (intermediate filaments), and tubulin (microtubules) in 3T3-L1 adipocytes or in CHO.IR.IRS-1 cells. Pharmacological disruption of microfilaments and microtubules, individually or in combination, was without effect on the subcellular distribution of IRS-1 or insulin-stimulated tyrosine phosphorylation in either cell type. Phosphorylation of Akt was modestly reduced (20-35%) in 3T3-L1 adipocytes but not in CHO.IR.IRS-1 cells. In cells lacking intermediate filaments (Vim(-/-)) IRS-1 expression, distribution and insulin-stimulated phosphorylation appeared normal. Even after depolymerisation of microfilaments and microtubules, insulin-stimulated phosphorylation of IRS-1 and Akt were maintained in Vim(-/-) cells. Taken together these data indicate that the characteristic subcellular fractionation properties and function of IRS-1 are unlikely to be mediated by cytoskeletal networks and that proximal insulin signaling does not require an intact cytoskeleton.
Collapse
Affiliation(s)
- Elaine C Thomas
- Centre for Diabetes and Endocrine Research, Princess Alexandra Hospital, University of Queensland, Brisbane, Qld 4102, Australia
| | | | | | | | | | | | | |
Collapse
|
17
|
Nishimura SY, Vrljic M, Klein LO, McConnell HM, Moerner WE. Cholesterol depletion induces solid-like regions in the plasma membrane. Biophys J 2005; 90:927-38. [PMID: 16272447 PMCID: PMC1367117 DOI: 10.1529/biophysj.105.070524] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Glycosylphosphatidylinositol-linked and transmembrane major histocompatibility complex (MHC) class II I-E(k) proteins, as well as N-(6-tetramethylrhodaminethiocarbamoyl)-1,2-dihexadecanoyl-sn-glycero-3-phosphoethanolamine (Tritc-DHPE), are used as probes to determine the effect of cholesterol concentration on the organization of the plasma membrane at temperatures in the range 22 degrees C-42 degrees C. Cholesterol depletion caused a decrease in the diffusion coefficients for the MHC II proteins and also for a slow fraction of the Tritc-DHPE population. At 37 degrees C, reduction of the total cell cholesterol concentration results in a smaller suppression of the translational diffusion for I-E(k) proteins (twofold) than was observed in earlier work at 22 degrees C (five sevenfold) Vrljic, M., S. Y. Nishimura, W. E. Moerner, and H. M. McConnell. 2005. Biophys. J. 88:334-347. At 37 degrees C, the diffusion of both I-E(k) proteins is Brownian (0.9 < alpha-parameter < 1.1). More than 99% of the protein population diffuses homogeneously when imaged at 65 frames per s. As the temperature is raised from 22 degrees C to 42 degrees C, a change in activation energy is seen at approximately 35 degrees C in the Arrhenius plots. Cytoskeletal effects appear to be minimal. These results are consistent with a previously described model of solid-like domain formation in the plasma membrane.
Collapse
Affiliation(s)
- Stefanie Y Nishimura
- Department of Chemistry, Molecular and Cellular Physiology, and Biophysics Program, Stanford University, Stanford, California 94305-5080, USA.
| | | | | | | | | |
Collapse
|
18
|
Groves JT. Molekulare Organisation und Signaltransduktion an Kontaktstellen zwischen Membranen. Angew Chem Int Ed Engl 2005. [DOI: 10.1002/ange.200461014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
19
|
Groves JT. Molecular Organization and Signal Transduction at Intermembrane Junctions. Angew Chem Int Ed Engl 2005; 44:3524-38. [PMID: 15844101 DOI: 10.1002/anie.200461014] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Surfaces create an environment in which multiple forces conspire together to yield a wealth of complex chemical processes. This is especially true of cell membranes, whose fluidity and flexibility enables responsive feedback with surface chemical interactions in ways not generally seen with inorganic materials. Spatial pattern formation of cell-surface proteins at intermembrane junctions provides many beautiful examples of these phenomena, and is also emerging as a functional aspect of intercellular signaling. Correspondingly, the study of interactions of cell-membrane surfaces is attracting significant attention from cell biologists and physical chemists alike. This convergence is fueled be recent, exquisite observations of protein pattern formation events within living immunological synapses along with parallel advances in membrane reconstitution, manipulation, and imaging technologies.
Collapse
Affiliation(s)
- Jay T Groves
- Department of Chemistry, University of California Berkeley, USA.
| |
Collapse
|
20
|
Vrljic M, Nishimura SY, Moerner WE, McConnell HM. Cholesterol depletion suppresses the translational diffusion of class II major histocompatibility complex proteins in the plasma membrane. Biophys J 2004; 88:334-47. [PMID: 15516525 PMCID: PMC1305010 DOI: 10.1529/biophysj.104.045989] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Glycosylphosphatidylinositol (GPI)-linked and native major histocompatibility complex class II I-E(k) were used as probes to determine the effect of varying cholesterol concentration on the mobility of proteins in the plasma membrane. These proteins were imaged in Chinese hamster ovary cells using single-molecule fluorescence microscopy. Observed diffusion coefficients of both native and GPI-linked I-E(k) proteins were found to depend on cholesterol concentration. As the cholesterol concentration decreases the diffusion coefficients decrease by up to a factor of 7 for native and 5 for GPI-linked I-E(k). At low cholesterol concentrations, after sphingomyelinase treatment, the diffusion coefficients are reduced by up to a factor of 60 for native and 190 for GPI-linked I-E(k). The effect is reversible on cholesterol reintroduction. Diffusion at all studied cholesterol concentrations, for both proteins, appears to be predominantly Brownian for time lags up to 2.5 s when imaged at 10 Hz. A decrease in diffusion coefficients is observed for other membrane proteins and lipid probes, DiIC12 and DiIC18. Fluorescence recovery after photobleaching measurements shows that the fraction of immobile lipid probe increases from 8 to approximately 40% after cholesterol extraction. These results are consistent with the previous work on cholesterol-phospholipid interactions. That is, cholesterol extraction destroys liquid cholesterol-phospholipid complexes, leaving solid-like high melting phospholipid domains that inhibit the lateral diffusion of membrane components.
Collapse
Affiliation(s)
- Marija Vrljic
- Biophysics Program, Stanford University, Stanford, California 94305-5080, USA.
| | | | | | | |
Collapse
|
21
|
Wong WSF, Leong KP. Tyrosine kinase inhibitors: a new approach for asthma. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2004; 1697:53-69. [PMID: 15023350 DOI: 10.1016/j.bbapap.2003.11.013] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/25/2003] [Accepted: 11/12/2003] [Indexed: 01/21/2023]
Abstract
The pathogenesis of allergic asthma involves the interplay of inflammatory cells and airway-resident cells, and of their secreted mediators including cytokines, chemokines, growth factors and inflammatory mediators. Receptor tyrosine kinases are important for the pathogenesis of airway remodeling. Activation of epidermal growth factor (EGF) receptor kinase and platelet-derived growth factor (PDGF) receptor kinase leads to hyperplasia of airway smooth muscle cells, epithelial cells and goblet cells. Stimulation of non-receptor tyrosine kinases (e.g. Lyn, Lck, Syk, ZAP-70, Fyn, Btk, Itk) is the earliest detectable signaling response upon antigen-induced immunoreceptor activation in inflammatory cells. Cytokine receptor dimerization upon ligand stimulation induces activation of Janus tyrosine kinases (JAKs), leading to recruitment and phosphorylation of signal transducer and activator of transcription (STAT) for selective gene expression regulation. Activation of chemokine receptors can trigger JAK-STAT pathway, Lck, Fyn, Lyn, Fgr, and Syk/Zap-70 to induce chemotaxis of inflammatory cells. Inhibitors of tyrosine kinases have been shown in vitro to block growth factor-induced hyperplasia of airway-resident cells; antigen-induced inflammatory cell activation and cytokine synthesis; cytokine-mediated pro-inflammatory gene expression in inflammatory and airway cells; and chemokine-induced chemotaxis of inflammatory cells. Recently, anti-inflammatory effects of tyrosine kinase inhibitors (e.g. genistein, tyrphostin AG213, piceatannol, tyrphostin AG490, WHI-P97, WHI-P131, Syk antisense) in animal models of allergic asthma have been reported. Therefore, development of inhibitors of tyrosine kinases can be a very attractive strategy for the treatment of asthma.
Collapse
Affiliation(s)
- W S Fred Wong
- Department of Pharmacology, Faculty of Medicine, National University of Singapore, MD2 18 Medical Drive, Singapore 117597, Singapore.
| | | |
Collapse
|
22
|
Roose JP, Diehn M, Tomlinson MG, Lin J, Alizadeh AA, Botstein D, Brown PO, Weiss A. T cell receptor-independent basal signaling via Erk and Abl kinases suppresses RAG gene expression. PLoS Biol 2003; 1:E53. [PMID: 14624253 PMCID: PMC261890 DOI: 10.1371/journal.pbio.0000053] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2003] [Accepted: 09/17/2003] [Indexed: 02/07/2023] Open
Abstract
Signal transduction pathways guided by cellular receptors commonly exhibit low-level constitutive signaling in a continuous, ligand-independent manner. The dynamic equilibrium of positive and negative regulators establishes such a tonic signal. Ligand-independent signaling by the precursors of mature antigen receptors regulates development of B and T lymphocytes. Here we describe a basal signal that controls gene expression profiles in the Jurkat T cell line and mouse thymocytes. Using DNA microarrays and Northern blots to analyze unstimulated cells, we demonstrate that expression of a cluster of genes, including RAG-1 and RAG-2, is repressed by constitutive signals requiring the adapter molecules LAT and SLP-76. This TCR-like pathway results in constitutive low-level activity of Erk and Abl kinases. Inhibition of Abl by the drug STI-571 or inhibition of signaling events upstream of Erk increases RAG-1 expression. Our data suggest that physiologic gene expression programs depend upon tonic activity of signaling pathways independent of receptor ligation. In the absence of basal signaling, RAG activity is high at a time during T cell development when it is otherwise normally suppressed
Collapse
Affiliation(s)
- Jeroen P Roose
- 1Department of Medicine, University of CaliforniaSan Francisco, San Francisco, CaliforniaUnited States of America
- 2Department of Microbiology and Immunology, University of CaliforniaSan Francisco, San Francisco, CaliforniaUnited States of America
| | - Maximilian Diehn
- 3Department of Biochemistry, Stanford University School of MedicineStanford, CaliforniaUnited States of America
| | - Michael G Tomlinson
- 1Department of Medicine, University of CaliforniaSan Francisco, San Francisco, CaliforniaUnited States of America
- 2Department of Microbiology and Immunology, University of CaliforniaSan Francisco, San Francisco, CaliforniaUnited States of America
| | - Joseph Lin
- 1Department of Medicine, University of CaliforniaSan Francisco, San Francisco, CaliforniaUnited States of America
- 2Department of Microbiology and Immunology, University of CaliforniaSan Francisco, San Francisco, CaliforniaUnited States of America
| | - Ash A Alizadeh
- 3Department of Biochemistry, Stanford University School of MedicineStanford, CaliforniaUnited States of America
| | - David Botstein
- 4Department of Genetics, Stanford University School of MedicineStanford, CaliforniaUnited States of America
| | - Patrick O Brown
- 3Department of Biochemistry, Stanford University School of MedicineStanford, CaliforniaUnited States of America
- 5Howard Hughes Medical Institute, Stanford University School of MedicineStanford, CaliforniaUnited States of America
| | - Arthur Weiss
- 1Department of Medicine, University of CaliforniaSan Francisco, San Francisco, CaliforniaUnited States of America
- 2Department of Microbiology and Immunology, University of CaliforniaSan Francisco, San Francisco, CaliforniaUnited States of America
- 6Howard Hughes Medical Institute, University of CaliforniaSan Francisco, San Francisco, CaliforniaUnited States of America
- 7Rosalind Russell Medical Research Center for Arthritis, University of CaliforniaSan Francisco, San Francisco, CaliforniaUnited States of America
| |
Collapse
|
23
|
Abstract
In the past two decades, an immense amount of information has been generated on the mechanism of T cell receptor (TCR) signaling (also called signal 1). This overview describes the major signalling pathways in the TCR signal transduction cascade and focuses on proximal events in TCR signaling. The review also discusses some of the strategies that target proximal TCR signaling, which are used for preventing graft rejection.
Collapse
Affiliation(s)
- Majed M Hamawy
- Department of Surgery, University of Wisconsin, Madison, 53792, USA.
| |
Collapse
|
24
|
Fuller CL, Braciale VL, Samelson LE. All roads lead to actin: the intimate relationship between TCR signaling and the cytoskeleton. Immunol Rev 2003; 191:220-36. [PMID: 12614363 DOI: 10.1034/j.1600-065x.2003.00004.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Regardless of cell type, the regulation of the actin cytoskeleton is tightly linked to vital biological properties such as polarity, motility, cell-cell contact, exocytosis and proliferation. In the immune system, where rapid and efficient response to antigen-provoked stimuli is crucial, an overwhelming amount of data implicate the actin cytoskeleton and its regulators as central to immune function. Increasingly, the cytoskeleton is considered an essential amplification step in T cell receptor (TCR)-, costimulatory-, and integrin-mediated signaling. Advances in genetic manipulation and confocal imaging have led to a keener appreciation of the importance of TCR signal integration by the actin cytoskeleton. This review outlines recent advances in elucidating the regulation of T cell function through the actin cytoskeleton. We also examine intriguing parallels between the immune system and other models of cytoskeletal regulation.
Collapse
Affiliation(s)
- Claudette L Fuller
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892-4255, USA
| | | | | |
Collapse
|
25
|
Vicente-Manzanares M, Sancho D, Yáñez-Mó M, Sánchez-Madrid F. The leukocyte cytoskeleton in cell migration and immune interactions. INTERNATIONAL REVIEW OF CYTOLOGY 2002; 216:233-89. [PMID: 12049209 DOI: 10.1016/s0074-7696(02)16007-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Leukocyte migration is crucial during the development of the immune system and in the responses to infection, inflammation, and tumor rejection. The migratory behavior of leukocytes under physiological and pathological conditions as well as the extracellular cues and intracellular machinery that control and guide migration have been studied thoroughly. The cytoskeleton of leukocytes is extremely versatile, bearing characteristic features that enable these cells to migrate under conditions of flow through narrow spaces and onto target tissues. What makes the cytoskeleton machinery so extraordinary is not so much its molecular composition, but its flexibility which allows it to display a unique combination of responses to the extracellular medium and a rapid regulation of the architecture of its components. This review focuses on the cytoskeleton of the leukocyte. Its molecular components and the regulation of their assembly and organization are discussed. Furthermore, it highlights aspects of the regulation of the leukocyte cytoskeleton that confer flexibility to these cells in order to perform their specific tasks. Finally, different subcellular structures such as the immunological synapse, the uropod of migrating leukocytes, and the phagosome displayed by phagocytic cells are discussed in detail. The relationship of the leukocyte with its environment occurs through different kinds of receptors that interact with ligands that are soluble, fixed on the membrane of other cells, or immobilized on the extracellular matrix. The impact of receptor-ligand binding on the functional responses and the rearrangement of the cytoskeleton is also examined.
Collapse
Affiliation(s)
- Miguel Vicente-Manzanares
- Servicio de Inmunología, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid, Spain
| | | | | | | |
Collapse
|
26
|
Sancho D, Vicente-Manzanares M, Mittelbrunn M, Montoya MC, Gordón-Alonso M, Serrador JM, Sánchez-Madrid F. Regulation of microtubule-organizing center orientation and actomyosin cytoskeleton rearrangement during immune interactions. Immunol Rev 2002; 189:84-97. [PMID: 12445267 DOI: 10.1034/j.1600-065x.2002.18908.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The reorganization of membrane, cytoskeletal and signaling molecules during immune interactions is critical for the generation of immune response. At the initiation of the T cell-antigen presenting cell (APC) interaction, antigen-independent weak adhesion forces allow the scanning of the APC surface by the T cell receptor for specific antigens. The stabilization of T cell-APC conjugates involves the segregation of membrane and intracellular signaling proteins, driven by reorganization of membrane microdomains and cytoskeletal changes. In early T cell-APC cognate interactions, the microtubular cytoskeleton undergoes drastic changes that lead to microtubule-organizing center (MTOC) reorientation to the vicinity of the cell-cell contact area. Recent data on the dynamics of MTOC redistribution and its influence in T cell-APC conjugate stabilization, together with the description of an increasing number of signaling molecules associated to this complex, underscore the key role of MTOC translocation in the T cell response. We focus on the mechanisms that control the early MTOC reorientation during T cell-APC interaction and the relevance of this process to T cell activation.
Collapse
Affiliation(s)
- David Sancho
- Servicio de Inmunología, Hospital de la Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
27
|
Vrljic M, Nishimura SY, Brasselet S, Moerner WE, McConnell HM. Translational diffusion of individual class II MHC membrane proteins in cells. Biophys J 2002; 83:2681-92. [PMID: 12414700 PMCID: PMC1302352 DOI: 10.1016/s0006-3495(02)75277-6] [Citation(s) in RCA: 211] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Single-molecule epifluorescence microscopy was used to observe the translational motion of GPI-linked and native I-E(k) class II MHC membrane proteins in the plasma membrane of CHO cells. The purpose of the study was to look for deviations from Brownian diffusion that might arise from barriers to this motion. Detergent extraction had suggested that these proteins may be confined to lipid microdomains in the plasma membrane. The individual I-E(k) proteins were visualized with a Cy5-labeled peptide that binds to a specific extracytoplasmic site common to both proteins. Single-molecule trajectories were used to compute a radial distribution of displacements, yielding average diffusion coefficients equal to 0.22 (GPI-linked I-E(k)) and 0.18 microm(2)/s (native I-E(k)). The relative diffusion of pairs of proteins was also studied for intermolecular separations in the range 0.3-1.0 microm, to distinguish between free diffusion of a protein molecule and diffusion of proteins restricted to a rapidly diffusing small domain. Both analyses show that motion is predominantly Brownian. This study finds no strong evidence for significant confinement of either GPI-linked or native I-E(k) in the plasma membrane of CHO cells.
Collapse
Affiliation(s)
- Marija Vrljic
- Biophysics Program, Stanford University, Stanford, CA 94305-5080, USA
| | | | | | | | | |
Collapse
|
28
|
Kesavan KP, Isaacson CC, Ashendel CL, Geahlen RL, Harrison ML. Characterization of the in vivo sites of serine phosphorylation on Lck identifying serine 59 as a site of mitotic phosphorylation. J Biol Chem 2002; 277:14666-73. [PMID: 11847223 DOI: 10.1074/jbc.m111911200] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The lymphocyte-specific protein-tyrosine kinase Lck plays a critical role in T cell activation. In response to T cell antigen receptor binding Lck undergoes phosphorylation on serine residues that include serines 59 and 194. Serine 59 is phosphorylated by ERK mitogen-activated protein kinase. Recently, we showed that in mitotic T cells Lck becomes hyper-phosphorylated on serine residues. In this report, using one-dimensional phosphopeptide mapping analysis, we identify serine 59 as a site of in vivo mitotic phosphorylation in Lck. The mitotic phosphorylation of serine 59 did not require either the catalytic activity or functional SH2 or SH3 domains of Lck. In addition, the presence of ZAP-70 also was dispensable for the phosphorylation of serine 59. Although previous studies demonstrated that serine 59 is a substrate for the ERK MAPK pathway, inhibitors of this pathway did not block the mitotic phosphorylation of serine 59. These results identify serine 59 as a site of mitotic phosphorylation in Lck and suggest that a pathway distinct from that induced by antigen receptor signaling is responsible for its phosphorylation. Thus, the phosphorylation of serine 59 is the result of two distinct signaling pathways, differentially activated in response to the physiological state of the T cell.
Collapse
Affiliation(s)
- Kamala P Kesavan
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907, USA
| | | | | | | | | |
Collapse
|
29
|
Abstract
Lipid domains or rafts are currently embraced by immunologists as critical participants in receptor-mediated signaling events occurring at the plasma membrane. This view of membrane heterogeneity and its functional importance is supported by many years of different experimental approaches. We can now refine our investigations, moving beyond the simple models to ask more detailed questions about structural properties and mechanistic interactions. As highlighted for the IgE receptor (Fc(epsilon)RI), new information about initial engagement with src family kinases, cytoskeletal regulation, and coupling with downstream signaling is beginning to emerge.
Collapse
Affiliation(s)
- D Holowka
- Department of Chemistry and Chemical Biology, Baker Laboratory, Cornell University, Ithaca, NY 14853-1301, USA
| | | |
Collapse
|
30
|
Krawczyk C, Penninger JM. Molecular motors involved in T cell receptor clusterings. J Leukoc Biol 2001. [DOI: 10.1189/jlb.69.3.317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Connie Krawczyk
- Amgen Institute/Ontario Cancer Institute, Departments of Medical Biophysics and Immunology, University of Toronto, Ontario, Canada
| | - Josef M. Penninger
- Amgen Institute/Ontario Cancer Institute, Departments of Medical Biophysics and Immunology, University of Toronto, Ontario, Canada
| |
Collapse
|
31
|
Carey KD, Dillon TJ, Schmitt JM, Baird AM, Holdorf AD, Straus DB, Shaw AS, Stork PJ. CD28 and the tyrosine kinase lck stimulate mitogen-activated protein kinase activity in T cells via inhibition of the small G protein Rap1. Mol Cell Biol 2000; 20:8409-19. [PMID: 11046138 PMCID: PMC102148 DOI: 10.1128/mcb.20.22.8409-8419.2000] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Proliferation of T cells via activation of the T-cell receptor (TCR) requires concurrent engagement of accessory costimulatory molecules to achieve full activation. The best-studied costimulatory molecule, CD28, achieves these effects, in part, by augmenting signals from the TCR to the mitogen-activated protein (MAP) kinase cascade. We show here that TCR-mediated stimulation of MAP kinase extracellular-signal-regulated kinases (ERKs) is limited by activation of the Ras antagonist Rap1. CD28 increases ERK signaling by blocking Rap1 action. CD28 inhibits Rap1 activation because it selectively stimulates an extrinsic Rap1 GTPase activity. The ability of CD28 to stimulate Rap1 GTPase activity was dependent on the tyrosine kinase Lck. Our results suggest that CD28-mediated Rap1 GTPase-activating protein activation can help explain the augmentation of ERKs during CD28 costimulation.
Collapse
Affiliation(s)
- K D Carey
- Vollum Institute, Oregon Health Sciences University, Portland, Oregon 97201, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Beveridge M, Park YW, Hermes J, Marenghi A, Brophy G, Santos A. Detection of p56(lck) kinase activity using scintillation proximity assay in 384-well format and imaging proximity assay in 384- and 1536-well format. JOURNAL OF BIOMOLECULAR SCREENING 2000; 5:205-12. [PMID: 10992041 DOI: 10.1177/108705710000500403] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
p56(lck) is a lymphocyte-specific tyrosine kinase that plays an important role in both T-cell maturation and activation. We have developed a homogeneous assay in which p56(lck) catalyzes the transfer of the gamma-phosphate group from [gamma-(33)P]ATP to a biotinylated peptide substrate. The labeled peptide is then captured on a streptavidin-coated scintillation proximity assay (SPA) bead or imaging proximity bead. The SPA is counted in a microplate scintillation counter and the imaging proximity assay is counted in a charge-coupled device-based imaging system called LEADseekertrade mark, recently launched as a homogeneous imaging system by Amersham Pharmacia Biotech. We show, via time-dependence assays and inhibitor studies, that this assay can be performed in 1536-well microplate format using imaging proximity as the method of detection. The results compare favorably with the same assay performed in 384-well microplate format using both SPA and imaging proximity as the detection methods. From this study, we conclude that a kinase assay can be performed in 384- and 1536-well format using imaging as the detection method, with significant time savings over standard scintillation counting. In addition, we show cost saving advantages of 1536- over 384-well format in terms of reagent usage, higher throughput, and waste disposal.
Collapse
Affiliation(s)
- M Beveridge
- Amersham Pharmacia Biotech UK Limited, Little Chalfont, Buckinghamshire, UK
| | | | | | | | | | | |
Collapse
|
33
|
Gringhuis SI, Leow A, Papendrecht-Van Der Voort EA, Remans PH, Breedveld FC, Verweij CL. Displacement of linker for activation of T cells from the plasma membrane due to redox balance alterations results in hyporesponsiveness of synovial fluid T lymphocytes in rheumatoid arthritis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:2170-9. [PMID: 10657671 DOI: 10.4049/jimmunol.164.4.2170] [Citation(s) in RCA: 106] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The T lymphocytes that reside in the synovium of the inflamed joints in patients with rheumatoid arthritis display severe hyporesponsiveness upon antigenic stimulation, which is probably due to their constant subjection to high levels of oxidative stress. Here we report that the synovial fluid T lymphocytes exert severely impaired phosphorylation of the adaptor protein linker for activation of T cells (LAT), a crucial component of the TCR-mediated signaling pathways. In healthy T lymphocytes, LAT is a membrane-bound protein and becomes phosphorylated by zeta-associated protein of 70 kDa (ZAP-70) upon TCR engagement. The molecular basis underlying the deficient phosphorylation of LAT and consequently the hyporesponsiveness of the synovial fluid T lymphocytes lies in the membrane displacement of LAT. We demonstrate that the subcellular localization of LAT is sensitive to changes in the intracellular levels of the antioxidant glutathione. The membrane anchorage of LAT, and consequently the phosphorylation of LAT and the cellular activation of the synovial fluid T lymphocytes upon TCR engagement, is restored in synovial fluid T lymphocytes after supplementation of the intracellular glutathione levels with N-acetyl-l -cysteine. These data suggest a role for the membrane displacement of LAT in the hyporesponsiveness of the synovial fluid T lymphocytes as a consequence of oxidative stress.
Collapse
Affiliation(s)
- S I Gringhuis
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | |
Collapse
|
34
|
Wienands J. The B-cell antigen receptor: formation of signaling complexes and the function of adaptor proteins. Curr Top Microbiol Immunol 1999; 245:53-76. [PMID: 10533310 DOI: 10.1007/978-3-642-57066-7_2] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- J Wienands
- Department for Molecular Immunology, Biology III, University of Freiburg, Germany.
| |
Collapse
|
35
|
Abstract
The role of lipid rafts in T cell antigen receptor (TCR) signaling was investigated using fluorescence microscopy. Lipid rafts labeled with cholera toxin B subunit (CT-B) and cross-linked into patches displayed characteristics of rafts isolated biochemically, including detergent resistance and colocalization with raft-associated proteins. LCK, LAT, and the TCR all colocalized with lipid patches, although TCR association was sensitive to nonionic detergent. Aggregation of the TCR by anti-CD3 mAb cross-linking also caused coaggregation of raft-associated proteins. However, the protein tyrosine phosphatase CD45 did not colocalize to either CT-B or CD3 patches. Cross-linking of either CD3 or CT-B strongly induced tyrosine phosphorylation and recruitment of a ZAP-70(SH2)(2)-green fluorescent protein (GFP) fusion protein to the lipid patches. Also, CT-B patching induced signaling events analagous to TCR stimulation, with the same dependence on expression of key TCR signaling molecules. Targeting of LCK to rafts was necessary for these events, as a nonraft- associated transmembrane LCK chimera, which did not colocalize with TCR patches, could not reconstitute CT-B-induced signaling. Thus, our results indicate a mechanism whereby TCR engagement promotes aggregation of lipid rafts, which facilitates colocalization of LCK, LAT, and the TCR whilst excluding CD45, thereby triggering protein tyrosine phosphorylation.
Collapse
Affiliation(s)
- Peter W. Janes
- Division of Membrane Biology, National Institute for Medical Research, London NW7 1AA, United Kingdom
- Division of Cellular Immunology, National Institute for Medical Research, London NW7 1AA, United Kingdom
| | - Steven C. Ley
- Division of Cellular Immunology, National Institute for Medical Research, London NW7 1AA, United Kingdom
| | - Anthony I. Magee
- Division of Membrane Biology, National Institute for Medical Research, London NW7 1AA, United Kingdom
| |
Collapse
|
36
|
Abstract
We have used site-directed mutagenesis to explore the mechanisms underlying Raf-1 activation in mitosis, and we have excluded most previously characterized activating interactions. Our results indicate that the primary locus of activation lies in the carboxyl-half of the molecule, although the extent of activation can be influenced by the amino-proximal region, particularly by the Raf-1 zinc finger. We also found that Raf-1 is hyperphosphorylated in mitosis at multiple sites within residues 283-302 and that these hyperphosphorylations are not required for activation. In addition, neither Mek1 nor Mek2 are stably activated in coordination with Raf-1 in nocodazole-arrested cells. Overall, the data suggest that the mechanism(s) responsible for activating Raf-1 during mitosis, and the subsequent downstream effects, are distinct from those involved in growth factor stimulation.
Collapse
Affiliation(s)
- A D Laird
- Section of Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, New York 14853, USA
| | | | | |
Collapse
|
37
|
Krumenacker JS, Montgomery DW, Buckley DJ, Gout PW, Buckley AR. Prolactin receptor signaling: shared components with the T-cell antigen receptor in Nb2 lymphoma cells. Endocrine 1998; 9:313-20. [PMID: 10221598 DOI: 10.1385/endo:9:3:313] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/1998] [Revised: 10/13/1998] [Accepted: 10/13/1998] [Indexed: 02/06/2023]
Abstract
Previously, we reported that activation of the human prolactin receptor (PRLR) produced a protein phosphorylation pattern strikingly similar to that provoked by Concanavalin A (Con A), an activator of the T-cell antigen receptor (TCR). These results suggested that certain signaling components of the TCR may be shared by the activated PRLR. Additional studies here assessed the levels of TCR expression following PRLR stimulation and the effect of TCR activation on PRL-stimulated proliferation in lactogen-dependent pre-T Nb2-11 lymphoma cells. The results indicated that the TCR was expressed on the surface of approx 4% of exponentially proliferating and prolactin- (PRL) treated cells. In contrast, approx 45% of quiescent cells, cultured in the absence of PRL for 24 h, expressed the TCR at the cell surface, suggesting that lactogen withdrawal may up-regulate TCR cell-surface expression. Moreover, TCR activation with anti-CD3 antibodies attenuated PRL-stimulated Nb2-11 cell proliferation in a concentration-dependent manner. In other experiments, immunoprecipitation and immunoblotting of Nb2-11 lysates revealed that activation of the PRLR resulted in rapid tyrosyl phosphorylation of ZAP-70, a critical TCR-associated tyrosine kinase. In addition, ZAP-70 was found to associate transiently with the putative guanine nucleotide exchange factor and substrate, Vav, in PRL-treated cells. ZAP-70 was also found to associate constitutively with the PRLR; PRL stimulation provoked the transient recruitment of Vav to the complex. These observations suggest that PRL signaling reflects the transient formation of a PRLR-ZAP-70-Vav complex and its immunomodulatory actions involve diverse interactions that affect TCR expression and signaling mechanisms.
Collapse
Affiliation(s)
- J S Krumenacker
- Department of Pharmacology and Toxicology, University of North Dakota School of Medicine and Health Sciences, Grand Forks, USA
| | | | | | | | | |
Collapse
|
38
|
Sloan-Lancaster J, Presley J, Ellenberg J, Yamazaki T, Lippincott-Schwartz J, Samelson LE. ZAP-70 association with T cell receptor zeta (TCRzeta): fluorescence imaging of dynamic changes upon cellular stimulation. J Cell Biol 1998; 143:613-24. [PMID: 9813084 PMCID: PMC2148150 DOI: 10.1083/jcb.143.3.613] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/1998] [Revised: 09/11/1998] [Indexed: 11/22/2022] Open
Abstract
The nonreceptor protein tyrosine kinase ZAP-70 is a critical enzyme required for successful T lymphocyte activation. After antigenic stimulation, ZAP-70 rapidly associates with T cell receptor (TCR) subunits. The kinetics of its translocation to the cell surface, the properties of its specific interaction with the TCRzeta chain expressed as a chimeric protein (TTzeta and Tzetazeta), and its mobility in different intracellular compartments were studied in individual live HeLa cells, using ZAP-70 and Tzetazeta fused to green fluorescent protein (ZAP-70 GFP and Tzetazeta-GFP, respectively). Time-lapse imaging using confocal microscopy indicated that the activation-induced redistribution of ZAP-70 to the plasma membrane, after a delayed onset, is of long duration. The presence of the TCRzeta chain is critical for the redistribution, which is enhanced when an active form of the protein tyrosine kinase Lck is coexpressed. Binding specificity to TTzeta was indicated using mutant ZAP-70 GFPs and a truncated zeta chimera. Photobleaching techniques revealed that ZAP-70 GFP has decreased mobility at the plasma membrane, in contrast to its rapid mobility in the cytosol and nucleus. Tzetazeta- GFP is relatively immobile, while peripherally located ZAP-70 in stimulated cells is less mobile than cytosolic ZAP-70 in unstimulated cells, a phenotype confirmed by determining the respective diffusion constants. Examination of the specific molecular association of signaling proteins using these approaches has provided new insights into the TCRzeta-ZAP-70 interaction and will be a powerful tool for continuing studies of lymphocyte activation.
Collapse
Affiliation(s)
- J Sloan-Lancaster
- The Section on Lymphocyte Signaling, Cell Biology and Metabolism Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | |
Collapse
|