1
|
Su Z, Almo SC, Wu Y. Computational simulations of bispecific T cell engagers by a multiscale model. Biophys J 2024; 123:235-247. [PMID: 38102828 PMCID: PMC10808035 DOI: 10.1016/j.bpj.2023.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 11/04/2023] [Accepted: 12/12/2023] [Indexed: 12/17/2023] Open
Abstract
The use of bispecific antibodies as T cell engagers can bypass the normal T cell receptor-major histocompatibility class interaction, redirect the cytotoxic activity of T cells, and lead to highly efficient tumor cell killing. However, this immunotherapy also causes significant on-target off-tumor toxicologic effects, especially when it is used to treat solid tumors. To avoid these adverse events, it is necessary to understand the fundamental mechanisms involved in the physical process of T cell engagement. We developed a multiscale computational framework to reach this goal. The framework combines simulations on the intercellular and multicellular levels. On the intercellular level, we simulated the spatial-temporal dynamics of three-body interactions among bispecific antibodies, CD3 and tumor-associated antigens (TAAs). The derived number of intercellular bonds formed between CD3 and TAAs was further transferred to the multicellular simulations as the input parameter of adhesive density between cells. Through the simulations under various molecular and cellular conditions, we were able to gain new insights into how to adopt the most appropriate strategy to maximize the drug efficacy and avoid the off-target effect. For instance, we discovered that the low antibody-binding affinity resulted in the formation of large clusters at the cell-cell interface, which could be important to control the downstream signaling pathways. We also tested different molecular architectures of the bispecific antibody and suggested the existence of an optimal length in regulating the T cell engagement. Overall, the current multiscale simulations serve as a proof-of-concept study to help in the future design of new biological therapeutics.
Collapse
Affiliation(s)
- Zhaoqian Su
- Data Science Institute, Vanderbilt University, Nashville, Tennessee
| | - Steven C Almo
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York; Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, New York
| | - Yinghao Wu
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
2
|
Junghans V, Chouliara M, Santos AM, Hatherley D, Petersen J, Dam T, Svensson LM, Rossjohn J, Davis SJ, Jönsson P. Effects of a local auxiliary protein on the two-dimensional affinity of a TCR-peptide MHC interaction. J Cell Sci 2020; 133:jcs245985. [PMID: 32591485 DOI: 10.1242/jcs.245985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/11/2020] [Indexed: 12/20/2022] Open
Abstract
The affinity of T-cell receptors (TCRs) for major histocompatibility complex molecules (MHCs) presenting cognate antigens likely determines whether T cells initiate immune responses, or not. There exist few measurements of two-dimensional (2D) TCR-MHC interactions, and the effect of auxiliary proteins on binding is unexplored. Here, Jurkat T-cells expressing the MHC molecule HLA-DQ8-glia-α1 and the ligand of an adhesion protein (rat CD2) were allowed to bind supported lipid bilayers (SLBs) presenting fluorescently labelled L3-12 TCR and rat CD2, allowing measurements of binding unconfounded by cell signaling effects or co-receptor binding. The 2D Kd for L3-12 TCR binding to HLA-DQ8-glia-α1, of 14±5 molecules/μm2 (mean±s.d.), was only marginally influenced by including CD2 up to ∼200 bound molecules/μm2 but higher CD2 densities reduced the affinity up to 1.9-fold. Cell-SLB contact size increased steadily with ligand density without affecting binding for contacts at up to ∼20% of total cell area, but beyond this lamellipodia appeared, giving an apparent increase in bound receptors of up to 50%. Our findings show how parameters other than the specific protein-protein interaction can influence binding behavior at cell-cell contacts.
Collapse
Affiliation(s)
| | - Manto Chouliara
- Department of Chemistry, Lund University, 221 00 Lund, Sweden
| | - Ana Mafalda Santos
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford; and Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, UK
| | - Deborah Hatherley
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford; and Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, UK
| | - Jan Petersen
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Tommy Dam
- Department of Chemistry, Lund University, 221 00 Lund, Sweden
| | - Lena M Svensson
- Department of Experimental Medical Science, Lund University, 221 00 Lund, Sweden; and School of Medical Sciences, Örebro University, 701 82 Örebro, Sweden
| | - Jamie Rossjohn
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
- Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Simon J Davis
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford; and Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, OX3 9DS Oxford, UK
| | - Peter Jönsson
- Department of Chemistry, Lund University, 221 00 Lund, Sweden
| |
Collapse
|
3
|
Xu H, Shaw DE. A Simple Model of Multivalent Adhesion and Its Application to Influenza Infection. Biophys J 2016; 110:218-33. [PMID: 26745425 PMCID: PMC4805874 DOI: 10.1016/j.bpj.2015.10.045] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 10/12/2015] [Accepted: 10/29/2015] [Indexed: 01/06/2023] Open
Abstract
Adhesion between biological surfaces, which is typically the result of molecular binding between receptors on one surface and ligands on another, plays a fundamental role in biology and is key to the infection mechanisms of certain viruses, including influenza. The physiological outcome of adhesion depends on both the number of bound cells (or viruses, or other biological particles) and the properties of the adhesion interface that is formed, including the equilibrium number of receptor-ligand connections. Here, we introduce a quantitative model for biological adhesion by adapting thermodynamic models developed for the related problem of multivalent molecular binding. In our model, adhesion affinity is approximated by a simple, analytical expression involving the numbers of ligands and receptors at the interface. Our model contains only two fitting parameters and is simple to interpret. When applied to the adhesion between the hemagglutinin ligands on influenza viruses and the sialic acid receptors on biosensors or on host cells, our model generates adhesion affinities consistent with experimental measurements performed over a range of numbers of receptors, and provides a semiquantitative estimate of the affinity range of the hemagglutinin-sialic acid interaction necessary for the influenza virus to successfully infect host cells. The model also provides a quantitative explanation for the experimental finding that a mutant avian virus gained transmissibility in mammals despite the mutations conferring only a less than twofold increase in the affinity of its hemagglutinin for mammalian receptors: the model predicts an order-of-magnitude improvement in adhesion to mammalian cells. We also extend our model to describe the competitive inhibition of adhesion: the model predicts that hemagglutinin inhibitors of relatively modest affinity can dramatically reduce influenza virus adhesion to host cells, suggesting that such inhibitors, if discovered, may be viable therapeutic agents against influenza.
Collapse
Affiliation(s)
- Huafeng Xu
- D. E. Shaw Research, New York, New York.
| | - David E Shaw
- D. E. Shaw Research, New York, New York; Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York.
| |
Collapse
|
4
|
Dustin ML, Choudhuri K. Signaling and Polarized Communication Across the T Cell Immunological Synapse. Annu Rev Cell Dev Biol 2016; 32:303-325. [PMID: 27501450 DOI: 10.1146/annurev-cellbio-100814-125330] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
T cells express a somatically recombined antigen receptor (αβTCR) that is calibrated during development to respond to changes in peptides displayed by major histocompatibility complex proteins (pMHC) on the surface of antigen-presenting cells (APC). A key characteristic of pMHC for adaptive immunity is the ability to sample internal states of cells and tissues to sensitively detect changes associated with infection, cell derangement, or tissue injury. Physical T cell-APC contact sets up an axis for polarization of TCR, adhesion molecules, kinases, cytoskeletal elements, and organelles inherent in this mode of juxtacrine signaling. The discovery of further lateral organization of the TCR and adhesion molecules into radially symmetric compartments, the immunological synapse, revealed an intersecting plane of symmetry and potential for regulated symmetry breaking to control duration of T cell-APC interactions. In addition to organizing signaling machinery, the immunological synapse directs the polarized transport and secretion of cytokines and cytolytic agents across the synaptic cleft and is a site for the generation and exocytic release of bioactive microvesicles that can functionally affect recipient APC and other cells in the environment. This machinery is coopted by retroviruses, and human immune deficiency virus-1 may even use antigen-specific synapses for infection of healthy T cells. Here, we discuss recent advances in the molecular and cell biological mechanisms of immunological synapse assembly and signaling and its role in intercellular communication across the synaptic cleft.
Collapse
Affiliation(s)
- Michael L Dustin
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, United Kingdom;
| | - Kaushik Choudhuri
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, Michigan 48109-5620;
| |
Collapse
|
5
|
Depoil D, Dustin ML. Force and affinity in ligand discrimination by the TCR. Trends Immunol 2014; 35:597-603. [PMID: 25466309 DOI: 10.1016/j.it.2014.10.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/24/2014] [Accepted: 10/24/2014] [Indexed: 01/30/2023]
Abstract
T cell recognition of antigen is a physical process that requires formation of a cell-cell junction that is rich in active force generation. Recently a biomolecular force probe was used to examine how the T cell receptor (TCR)-pMHC interaction responds to force and the consequences of force-dependent interactions for T cell activation. While adhesion and costimulatory molecules in the immunological synapse impact upon the overall force of the interaction, these results suggest that the TCR uses a force-dependent bond - a catch bond - and that it may therefore be important to consider the TCR-pMHC interaction in isolation in the early phases of the decision process. We discuss here these findings in the context of other work on the impact of forces on the TCR and the quantification of interaction in interfaces.
Collapse
Affiliation(s)
- David Depoil
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopedics and Musculosceletal Sciences, The University of Oxford, Roosevelt Drive, Headington, OX3 7FY, United Kingdom; Helene and Martin Kimmel Center for Biology and Medicine of the Skirball Institute of Biomolecular Medicine, Department of Pathology, New York University School of Medicine, 540 First Avenue, New York, NY 10012, USA
| | - Michael L Dustin
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopedics and Musculosceletal Sciences, The University of Oxford, Roosevelt Drive, Headington, OX3 7FY, United Kingdom; Helene and Martin Kimmel Center for Biology and Medicine of the Skirball Institute of Biomolecular Medicine, Department of Pathology, New York University School of Medicine, 540 First Avenue, New York, NY 10012, USA.
| |
Collapse
|
6
|
Huang X, Zheng F, Zhan CG. Binding structures and energies of the human neonatal Fc receptor with human Fc and its mutants by molecular modeling and dynamics simulations. MOLECULAR BIOSYSTEMS 2013; 9:3047-58. [PMID: 24057047 DOI: 10.1039/c3mb70231f] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Homology modeling and molecular dynamics simulations have been carried out to model the detailed structures of the human neonatal Fc receptor (FcRn) binding with the wild-type Fc of human immunoglobulin G1 (IgG1) and its various mutants. Based on the modeled human FcRn-Fc binding structures, it has been proposed that the protein-protein binding interface is composed of three subsites. The first subsite is a hydrophobic core where residue I39 of human Fc can be accommodated very well, and the other two subsites are all composed of critical salt bridges between human FcRn and human Fc. All of the modeled structures and the calculated binding energies are qualitatively consistent with the available experimental data, suggesting that the modeled human FcRn-Fc binding structures are reasonable. The modeled human FcRn-Fc binding structure may be valuable for future rational design of novel mutants of human Fc and Fc-fused therapeutic proteins with a potentially higher binding affinity for human FcRn and, thus, a longer in vivo half-life in humans.
Collapse
Affiliation(s)
- Xiaoqin Huang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, Kentucky 40536, USA.
| | | | | |
Collapse
|
7
|
Pharmacokinetics, pharmacodynamics and physiologically-based pharmacokinetic modelling of monoclonal antibodies. Clin Pharmacokinet 2013; 52:83-124. [PMID: 23299465 DOI: 10.1007/s40262-012-0027-4] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Development of monoclonal antibodies (mAbs) and their functional derivatives represents a growing segment of the development pipeline in the pharmaceutical industry. More than 25 mAbs and derivatives have been approved for a variety of therapeutic applications. In addition, around 500 mAbs and derivatives are currently in different stages of development. mAbs are considered to be large molecule therapeutics (in general, they are 2-3 orders of magnitude larger than small chemical molecule therapeutics), but they are not just big chemicals. These compounds demonstrate much more complex pharmacokinetic and pharmacodynamic behaviour than small molecules. Because of their large size and relatively poor membrane permeability and instability in the conditions of the gastrointestinal tract, parenteral administration is the most usual route of administration. The rate and extent of mAb distribution is very slow and depends on extravasation in tissue, distribution within the particular tissue, and degradation. Elimination primarily happens via catabolism to peptides and amino acids. Although not definitive, work has been published to define the human tissues mainly involved in the elimination of mAbs, and it seems that many cells throughout the body are involved. mAbs can be targeted against many soluble or membrane-bound targets, thus these compounds may act by a variety of mechanisms to achieve their pharmacological effect. mAbs targeting soluble antigen generally exhibit linear elimination, whereas those targeting membrane-bound antigen often exhibit non-linear elimination, mainly due to target-mediated drug disposition (TMDD). The high-affinity interaction of mAbs and their derivatives with the pharmacological target can often result in non-linear pharmacokinetics. Because of species differences (particularly due to differences in target affinity and abundance) in the pharmacokinetics and pharmacodynamics of mAbs, pharmacokinetic/pharmacodynamic modelling of mAbs has been used routinely to expedite the development of mAbs and their derivatives and has been utilized to help in the selection of appropriate dose regimens. Although modelling approaches have helped to explain variability in both pharmacokinetic and pharmacodynamic properties of these drugs, there is a clear need for more complex models to improve understanding of pharmacokinetic processes and pharmacodynamic interactions of mAbs with the immune system. There are different approaches applied to physiologically based pharmacokinetic (PBPK) modelling of mAbs and important differences between the models developed. Some key additional features that need to be accounted for in PBPK models of mAbs are neonatal Fc receptor (FcRn; an important salvage mechanism for antibodies) binding, TMDD and lymph flow. Several models have been described incorporating some or all of these features and the use of PBPK models are expected to expand over the next few years.
Collapse
|
8
|
Affiliation(s)
- Facundo D Batista
- Lymphocyte Interaction Group, London Research Institute, Cancer Research UK, London, UK
| | | |
Collapse
|
9
|
Zhu C, Jiang N, Huang J, Zarnitsyna VI, Evavold BD. Insights from in situ analysis of TCR-pMHC recognition: response of an interaction network. Immunol Rev 2013; 251:49-64. [PMID: 23278740 PMCID: PMC3539230 DOI: 10.1111/imr.12016] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Recognition of peptide presented by the major histocompatibility complex (pMHC) molecule by the T-cell receptor (TCR) determines T-cell selection, development, differentiation, fate, and function. Despite intensive studies on the structures, thermodynamic properties, kinetic rates, and affinities of TCR-pMHC interactions in the past two decades, questions regarding the functional outcome of these interactions, i.e. how binding of the αβ TCR heterodimer with distinct pMHCs triggers different intracellular signals via the adjacent CD3 components to produce different T-cell responses, remain unclear. Most kinetic measurements have used surface plasmon resonance, a three-dimensional (3D) technique in which fluid-phase receptors and ligands are removed from their cellular environment. Recently, several two-dimensional (2D) techniques have been developed to analyze molecular interactions on live T cells with pMHCs presented by surrogate antigen-presenting cells or supported planar lipid bilayers. The insights from these in situ analyses have provided a sharp contrast of the 2D network biology approach to the 3D reductionist approach and prompted rethinking of our current views of T-cell triggering. Based on these insights, we propose a mechanochemical coupled triggering hypothesis to explain why the in situ kinetic parameters differ so much from their 3D counterparts, yet correlate so much better with T-cell functional responses.
Collapse
Affiliation(s)
- Cheng Zhu
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0363, USA.
| | | | | | | | | |
Collapse
|
10
|
Burroughs NJ, Köhler K, Miloserdov V, Dustin ML, van der Merwe PA, Davis DM. Boltzmann energy-based image analysis demonstrates that extracellular domain size differences explain protein segregation at immune synapses. PLoS Comput Biol 2011; 7:e1002076. [PMID: 21829338 PMCID: PMC3150282 DOI: 10.1371/journal.pcbi.1002076] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Accepted: 04/19/2011] [Indexed: 01/08/2023] Open
Abstract
Immune synapses formed by T and NK cells both show segregation of the integrin ICAM1 from other proteins such as CD2 (T cell) or KIR (NK cell). However, the mechanism by which these proteins segregate remains unclear; one key hypothesis is a redistribution based on protein size. Simulations of this mechanism qualitatively reproduce observed segregation patterns, but only in certain parameter regimes. Verifying that these parameter constraints in fact hold has not been possible to date, this requiring a quantitative coupling of theory to experimental data. Here, we address this challenge, developing a new methodology for analysing and quantifying image data and its integration with biophysical models. Specifically we fit a binding kinetics model to 2 colour fluorescence data for cytoskeleton independent synapses (2 and 3D) and test whether the observed inverse correlation between fluorophores conforms to size dependent exclusion, and further, whether patterned states are predicted when model parameters are estimated on individual synapses. All synapses analysed satisfy these conditions demonstrating that the mechanisms of protein redistribution have identifiable signatures in their spatial patterns. We conclude that energy processes implicit in protein size based segregation can drive the patternation observed in individual synapses, at least for the specific examples tested, such that no additional processes need to be invoked. This implies that biophysical processes within the membrane interface have a crucial impact on cell:cell communication and cell signalling, governing protein interactions and protein aggregation.
Collapse
Affiliation(s)
- Nigel J Burroughs
- Systems Biology Centre, University of Warwick, Coventry, United Kingdom.
| | | | | | | | | | | |
Collapse
|
11
|
Gutenkunst RN, Coombs D, Starr T, Dustin ML, Goldstein B. A biophysical model of cell adhesion mediated by immunoadhesin drugs and antibodies. PLoS One 2011; 6:e19701. [PMID: 21629715 PMCID: PMC3100730 DOI: 10.1371/journal.pone.0019701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 04/07/2011] [Indexed: 12/29/2022] Open
Abstract
A promising direction in drug development is to exploit the ability of natural killer cells to kill antibody-labeled target cells. Monoclonal antibodies and drugs designed to elicit this effect typically bind cell-surface epitopes that are overexpressed on target cells but also present on other cells. Thus it is important to understand adhesion of cells by antibodies and similar molecules. We present an equilibrium model of such adhesion, incorporating heterogeneity in target cell epitope density, nonspecific adhesion forces, and epitope immobility. We compare with experiments on the adhesion of Jurkat T cells to bilayers containing the relevant natural killer cell receptor, with adhesion mediated by the drug alefacept. We show that a model in which all target cell epitopes are mobile and available is inconsistent with the data, suggesting that more complex mechanisms are at work. We hypothesize that the immobile epitope fraction may change with cell adhesion, and we find that such a model is more consistent with the data, although discrepancies remain. We also quantitatively describe the parameter space in which binding occurs. Our model elaborates substantially on previous work, and our results offer guidance for the refinement of therapeutic immunoadhesins. Furthermore, our comparison with data from Jurkat T cells also points toward mechanisms relating epitope immobility to cell adhesion.
Collapse
Affiliation(s)
- Ryan N. Gutenkunst
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona, United States of America
- * E-mail:
| | - Daniel Coombs
- Department of Mathematics and Institute of Applied Mathematics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Toby Starr
- Department of Pathology, New York University School of Medicine and Program in Molecular Pathogenesis, Skirball Institute of Biomolecular Medicine, New York, New York, United States of America
| | - Michael L. Dustin
- Department of Pathology, New York University School of Medicine and Program in Molecular Pathogenesis, Skirball Institute of Biomolecular Medicine, New York, New York, United States of America
| | - Byron Goldstein
- Theoretical Biology and Biophysics Group, Theoretical Division, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| |
Collapse
|
12
|
Hierarchical organization of the plasma membrane: investigations by single-molecule tracking vs. fluorescence correlation spectroscopy. FEBS Lett 2010; 584:1814-23. [PMID: 20178787 DOI: 10.1016/j.febslet.2010.02.047] [Citation(s) in RCA: 131] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Revised: 02/15/2010] [Accepted: 02/15/2010] [Indexed: 11/20/2022]
Abstract
Single-molecule tracking and fluorescence correlation spectroscopy (FCS) applied to the plasma membrane in living cells have allowed a number of unprecedented observations, thus fostering a new basic understanding of molecular diffusion, interaction, and signal transduction in the plasma membrane. It is becoming clear that the plasma membrane is a heterogeneous entity, containing diverse structures on nano-meso-scales (2-200 nm) with a variety of lifetimes, where certain membrane molecules stay together for limited durations. Molecular interactions occur in the time-dependent inhomogeneous two-dimensional liquid of the plasma membrane, which might be a key for plasma membrane functions.
Collapse
|
13
|
Dustin ML. Insights into function of the immunological synapse from studies with supported planar bilayers. Curr Top Microbiol Immunol 2010; 340:1-24. [PMID: 19960306 DOI: 10.1007/978-3-642-03858-7_1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Innate and adaptive immunity is dependent upon reliable cell-cell communication mediated by direct interactions of cell surface receptors with ligands integrated into the surface of apposing cells or bound directly to the surface as in complement deposition or antibody mediated recognition through Fc receptors. Supported lipid bilayers formed on glass surfaces offer a useful model system in which to explore some basic features of molecular interactions in immunological relevant contacts, which include signal integration and effector functions through immunological synapses and kinapses. We have exploited that lateral mobility of molecules in the supported planar bilayers and fluorescence microscopy to develop a system for measurement of two-dimensional affinities and kinetic rates in the contact area, which is of immunological interest. Affinity measurements are based on a modified Scatchard analysis. Measurements of kinetic rates are based on fluorescence photo bleaching after recovery at the level of the entire contact area. This has been coupled to a reaction-diffusion equation that allows calculation of on- and off-rates. We have found that mixtures of ligands in supported planar bilayers can effectively activate T lymphocytes and simultaneously allow monitoring of the immunological synapse. Recent studies in planar bilayers have provided additional insights into organization principles of cell-cell interfaces. Perennial problems in understanding cell-cell communication are yielding quantitative measurements based on planar bilayers in areas of ligand-driven receptor clustering and the role of the actin cytoskeleton in immune cell activation. A major goal for the field is determining quantitative rules involved in signaling complex formation by innate and adaptive receptor systems.
Collapse
Affiliation(s)
- Michael L Dustin
- Helen L. and Martin S. Kimmel Center for Biology and Medicine in the Skirball Institute for Biomolecular Medicine and Department of Pathology, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
14
|
Dustin ML. Supported bilayers at the vanguard of immune cell activation studies. J Struct Biol 2009; 168:152-60. [PMID: 19500675 DOI: 10.1016/j.jsb.2009.05.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Revised: 05/23/2009] [Accepted: 05/27/2009] [Indexed: 12/27/2022]
Abstract
Biological adhesion between cells is critical for development of multicellular organisms and for the function of the adaptive immune system of vertebrates. A gap in understanding of adhesion systems arises from the difficulty of collecting quantitative data on the molecular interactions underlying adhesion, which is typically studied by population statistics such as percent adhesion in the presence of empirically defined forces to separate less adherent cells. Supported lipid bilayers formed on glass surfaces offer a useful model system in which to explore some basic features of molecular interactions in adhesive contacts. We have exploited the lateral mobility of molecules in the supported planar bilayers and fluorescence microscopy to develop a system for measurement of two-dimensional affinities and kinetic rates in contact areas. Affinity measurements are based on a modified Scatchard analysis. Measurements of kinetic rates are based on fluorescence photobleaching after recovery at the level of the entire contact area. This has been coupled to a reaction-diffusion equation that allows calculation of on- and off-rates. We have found that mixtures of ligands in supported planar bilayers can effectively activate T lymphocytes and simultaneously allow monitoring of the immunological synapse. Recent studies in planar bilayers have provided additional insights into organization principles of cell-cell interfaces. Perennial problems in understanding cell-cell communication are yielding to quantitative measurements based on planar bilayers in areas of ligand driven receptor clustering and the role of the actin cytoskeleton in immune cell activation. A major goal for the field is determining quantitative rules involved in signaling complex formation.
Collapse
Affiliation(s)
- Michael L Dustin
- Helen L. and Martin S. Kimmel Center for Biology and Medicine in the Skirball Institute for Biomolecular Medicine and Department of Pathology, NYU School of Medicine, New York, 10016, USA.
| |
Collapse
|
15
|
Zheng X, Wang Y, Wei H, Sun R, Tian Z. LFA-1 and CD2 synergize for the Erk1/2 activation in the Natural Killer (NK) cell immunological synapse. J Biol Chem 2009; 284:21280-7. [PMID: 19502238 DOI: 10.1074/jbc.m807053200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Natural killer (NK) cell recognition and formation of a conjugate with target cells, followed by intracellular signal pathway activation and degradation of cytolytic granules, are essential for NK cell cytotoxicity. In this study, NK92 cells were used to investigate synapse formation and subsequent signaling after binding to the target cell. The binding rate of the NK92-target cell was associated with NK92 cell cytotoxicity. Confocal results showed that adhesion molecules, LFA-1 (CD11a) and CD2, accumulated at the interface of the NK92-K562 contact. Ligation with K562 cells activated the Erk1/2 signal pathway of NK92 cells. The blocking of the NK-target conjugate by EDTA or anti-CD11a or/and anti-CD2 antibody decreased the phosphorylation of Erk1/2 and NK cell cytotoxicity. Inhibition of Erk1/2 phosphorylation by the chemical inhibitor U0126 suppressed the cytolytic activity of NK92 cells, but had no effect on NK-target conjugate formation. Thus, conjugate formation of the NK92-target cell was prerequisite to NK cell activation, and subsequent signal transduction was also required for NK cell cytotoxicity.
Collapse
Affiliation(s)
- Xiaodong Zheng
- Institute of Immunology, Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | | | | | | | | |
Collapse
|
16
|
Dustin ML. Multiscale analysis of T cell activation: correlating in vitro and in vivo analysis of the immunological synapse. Curr Top Microbiol Immunol 2009; 334:47-70. [PMID: 19521681 DOI: 10.1007/978-3-540-93864-4_3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Recently implemented fluorescence imaging techniques, such as total internal reflection fluorescence microscopy and two-photon laser scanning microscopy, have made possible multiscale analysis of the immune response from single molecules in an interface to cells moving in lymphoid tissues and tumors. In this review, we consider components of T cell sensitivity: the immunological synapse, the coordination of migration, and antigen recognition in vivo. Potency, dose, and detection threshold for peptide-MHC determine T cell sensitivity. The immunological synapse incorporates T cell receptor microclusters that initiate and sustain signaling, and it also determines the positional stability of the T cells through symmetry and symmetry breaking. In vivo decisions by T cells on stopping or migration are based on antigen stop signals and environmental go signals that can sometimes prevent arrest of T cells altogether, and thus can change the outcome of antigen encounters.
Collapse
Affiliation(s)
- Michael L Dustin
- Department of Pathology, Program of Molecular Pathogenesis, Skirball Institute of BioMolecular Medicine, NYU School of Medicine, 540 First Avenue, New York, NY 10016, USA
| |
Collapse
|
17
|
Kim N, Stiegler AL, Cameron TO, Hallock PT, Gomez AM, Huang JH, Hubbard SR, Dustin ML, Burden SJ. Lrp4 is a receptor for Agrin and forms a complex with MuSK. Cell 2008; 135:334-42. [PMID: 18848351 DOI: 10.1016/j.cell.2008.10.002] [Citation(s) in RCA: 497] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2008] [Revised: 09/12/2008] [Accepted: 10/01/2008] [Indexed: 12/14/2022]
Abstract
Neuromuscular synapse formation requires a complex exchange of signals between motor neurons and skeletal muscle fibers, leading to the accumulation of postsynaptic proteins, including acetylcholine receptors in the muscle membrane and specialized release sites, or active zones in the presynaptic nerve terminal. MuSK, a receptor tyrosine kinase that is expressed in skeletal muscle, and Agrin, a motor neuron-derived ligand that stimulates MuSK phosphorylation, play critical roles in synaptic differentiation, as synapses do not form in their absence, and mutations in MuSK or downstream effectors are a major cause of a group of neuromuscular disorders, termed congenital myasthenic syndromes (CMS). How Agrin activates MuSK and stimulates synaptic differentiation is not known and remains a fundamental gap in our understanding of signaling at neuromuscular synapses. Here, we report that Lrp4, a member of the LDLR family, is a receptor for Agrin, forms a complex with MuSK, and mediates MuSK activation by Agrin.
Collapse
Affiliation(s)
- Natalie Kim
- Molecular Neurobiology Program, Skirball Institute of Biomolecular Medicine, Helen and Martin Kimmel Center for Biology and Medicine, NYU Medical School, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
|
19
|
Abstract
Routine quantitative analysis of biomolecule surface density by fluorescence microscopy has been limited by the difficulty of preparing appropriate calibration standards that relate measured fluorescence intensity to actual surface concentration. Supported lipid bilayers are planar fluid films of uniform density and composition which can incorporate a variety of lipidated fluorophores and work well as fluorescence standards. Here, we outline a straightforward strategy to calibrate digital micrographs of fluorescent surfaces such as planar cellular junctions for comparison to supported bilayer standards. It can be implemented with standard microscopy equipment. To illustrate the advantages of this approach, we quantify cell- and bilayer-side protein density patterns in a hybrid immunological synapse between a T-cell and a supported bilayer.
Collapse
|