1
|
McArthur HCW, Bajur AT, Iliopoulou M, Spillane KM. Antigen mobility regulates the dynamics and precision of antigen capture in the B cell immune synapse. Proc Natl Acad Sci U S A 2025; 122:e2422528122. [PMID: 40354540 DOI: 10.1073/pnas.2422528122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 04/03/2025] [Indexed: 05/14/2025] Open
Abstract
B cells discriminate antigens in immune synapses by capturing them from antigen-presenting cells (APCs). This discrimination relies on the application of mechanical force to B cell receptor (BCR)-antigen bonds, allowing B cells to selectively disrupt low-affinity interactions while internalizing high-affinity antigens. Using DNA-based tension sensors combined with high-resolution imaging, we demonstrate that the magnitude, location, and timing of forces within the immune synapse are influenced by the fluidity of the antigen-presenting membrane. Transitioning antigens from a high-mobility to a low-mobility substrate significantly increases the probability and speed of antigen extraction while also improving affinity discrimination. This shift in antigen mobility also reshapes the synapse architecture, altering spatial patterns of antigen uptake. Despite these adaptations, B cells maintain consistent levels of proximal and downstream signaling pathway activation regardless of antigen mobility. They also efficiently transport internalized antigens to major histocompatibility complex class II (MHCII)-positive compartments for processing. These results demonstrate that B cells mount effective responses to antigens across diverse physical environments, though the characteristics of that environment may influence the speed and accuracy of B cell adaptation during an immune response.
Collapse
Affiliation(s)
- Hannah C W McArthur
- Department of Physics, King's College London, London WC2R 2LS, United Kingdom
| | - Anna T Bajur
- Department of Physics, King's College London, London WC2R 2LS, United Kingdom
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, United Kingdom
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Maro Iliopoulou
- Department of Physics, King's College London, London WC2R 2LS, United Kingdom
| | - Katelyn M Spillane
- Department of Physics, King's College London, London WC2R 2LS, United Kingdom
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, United Kingdom
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
2
|
Warren J, Corti A, Meyer CA, Hayenga HN. Bridging hemodynamics, tissue mechanics, and pathophysiology in coronary artery disease: A new agent-based model with tetrahedral mesh integration. J Biomech 2025; 183:112631. [PMID: 40132244 DOI: 10.1016/j.jbiomech.2025.112631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 01/20/2025] [Accepted: 03/09/2025] [Indexed: 03/27/2025]
Abstract
We introduce a new multi-physics, multi-scale modeling approach to understand plaque progression during coronary artery disease. Prior works have coupled agent-based models (ABMs) with finite element analysis (FEA) or computational fluid dynamics (CFD) to study the individual contributions of tissue mechanics or hemodynamics to plaque growth. However, these approaches could not simultaneously capture the dynamic interplay between all three domains that drive plaque development. This study aims to present a novel method that merges hemodynamics via CFD, biological processes via ABM, and biomechanics via FEA into a single multi-scale, multi-physics simulation (CAFe). A description of the mechanisms and modeling approaches utilized in the CAFe model is provided, as well as preliminary exploration of the model's capabilities in idealized healthy and stenosed coronary artery models. A volumetric 3D tetrahedral mesh of the artery is shared between CFD, ABM, and FEA to simulate geometrical and biological changes with continuity and consistency. The CFD and FEA modules, implemented with FEBio, calculate the wall shear stress and structural stress and strain, respectively. These biomechanical values are passed to the ABM, implemented in MATLAB, which simulates vascular remodeling using molecular diffusion, cell migration, equations for cellular processes, and volumetric growth to update the geometry. Initial results using CAFe suggest atherosclerotic arteries seek to maintain a hemodynamic threshold through preferential growth and remodeling downstream of a stenosis. The innovative approach described herein marks a significant step forward in predictive modeling of CAD progression and paves the way for powerful coupling of the spatiotemporal-dependent remodeling paradigms exhibited by the disease.
Collapse
Affiliation(s)
- Jeremy Warren
- Department of Bioengineering, University of Texas at Dallas, Richardson TX 75080, USA
| | - Anna Corti
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Clark A Meyer
- Department of Bioengineering, University of Texas at Dallas, Richardson TX 75080, USA
| | - Heather N Hayenga
- Department of Bioengineering, University of Texas at Dallas, Richardson TX 75080, USA.
| |
Collapse
|
3
|
Chen J, Zou X, Spencer DC, Morgan H. Single-cell electro-mechanical shear flow deformability cytometry. MICROSYSTEMS & NANOENGINEERING 2024; 10:173. [PMID: 39572527 PMCID: PMC11582679 DOI: 10.1038/s41378-024-00810-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/13/2024] [Accepted: 08/20/2024] [Indexed: 11/24/2024]
Abstract
The complex structural and molecular features of a cell lead to a set of specific dielectric and mechanical properties which can serve as intrinsic phenotypic markers that enable different cell populations to be characterised and distinguished. We have developed a microfluidic technique that exploits non-contact shear flow deformability cytometry to simultaneously characterise both the electrical and mechanical properties of single cells at high speed. Cells flow along a microchannel and are deformed (elongated) to different degrees by the shear force created by a viscoelastic fluid and channel wall. The electrical impedance of each cell is measured using sets of integrated microelectrodes along two orthogonal axes to determine the shape change and thus the electrical deformability, together with cell dielectric properties. The system performance was evaluated by measuring the electro-mechanical properties of cells treated in different ways, including osmotic shock, glutaraldehyde cross-linking and cytoskeletal disruption with Cytochalasin D and Latrunculin B. To confirm the accuracy of the system images of deformed cells were also captured using a camera. Correlation between the optical deformability and the electrical deformability is excellent. This novel cytometer has a throughput of ~100 cells s-1 is simple, does not use sheath flow or require high speed optical imaging.
Collapse
Affiliation(s)
- Junyu Chen
- School of Electronics and Computer Science, and Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Xueping Zou
- School of Electronics and Computer Science, and Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Daniel C Spencer
- School of Electronics and Computer Science, and Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Hywel Morgan
- School of Electronics and Computer Science, and Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
| |
Collapse
|
4
|
Riobó I, Yuseff MI. B cell mechanosensing regulates ER remodeling at the immune synapse. Front Immunol 2024; 15:1464000. [PMID: 39434873 PMCID: PMC11491372 DOI: 10.3389/fimmu.2024.1464000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/16/2024] [Indexed: 10/23/2024] Open
Abstract
Introduction Engagement of the B-cell receptor with immobilized antigens triggers the formation of an immune synapse (IS), a complex cellular platform where B-cells recruit signaling molecules and reposition lysosomes to promote antigen uptake and processing. Calcium efflux from the endoplasmic reticulum (ER) released upon BCR stimulation is necessary to promote B-cell survival and differentiation. Whether the spatial organization of the ER within the B-cell synapse can tune IS function and B-cell activation remains unaddressed. Here, we characterized ER structure and interaction with the microtubule network during BCR activation and evaluated how mechanical cues arising from antigen presenting surfaces affect this process. Methods B-cells were cultured on surfaces of varying stiffness coated with BCR ligands, fixed, and stained for the ER and microtubule network. Imaging analysis was used to assess the distribution of the ER and microtubules at the IS. Results Upon BCR activation, the ER is redistributed towards the IS independently of peripheral microtubules and accumulates around the microtubule-organization center. Furthermore, this remodeling is also dependent on substrate stiffness, where greater stiffness triggers enhanced redistribution of the ER. Discussion Our results highlight how spatial reorganization of the ER is coupled to the context of antigen recognition and could tune B-cell responses. Additionally, we provide novel evidence that the structural maturation of the ER in plasma cells is initiated during early activation of B-cells.
Collapse
Affiliation(s)
| | - María Isabel Yuseff
- Immune Cell Biology Lab, Pontificia Universidad Católica de Chile, Facultad de Ciencias Biológicas, Santiago, Chile
| |
Collapse
|
5
|
Settle AH, Winer BY, de Jesus MM, Seeman L, Wang Z, Chan E, Romin Y, Li Z, Miele MM, Hendrickson RC, Vorselen D, Perry JSA, Huse M. β2 integrins impose a mechanical checkpoint on macrophage phagocytosis. Nat Commun 2024; 15:8182. [PMID: 39294148 PMCID: PMC11411054 DOI: 10.1038/s41467-024-52453-9] [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: 07/08/2024] [Accepted: 09/06/2024] [Indexed: 09/20/2024] Open
Abstract
Phagocytosis is an intensely physical process that depends on the mechanical properties of both the phagocytic cell and its chosen target. Here, we employed differentially deformable hydrogel microparticles to examine the role of cargo rigidity in the regulation of phagocytosis by macrophages. Whereas stiff cargos elicited canonical phagocytic cup formation and rapid engulfment, soft cargos induced an architecturally distinct response, characterized by filamentous actin protrusions at the center of the contact site, slower cup advancement, and frequent phagocytic stalling. Using phosphoproteomics, we identified β2 integrins as critical mediators of this mechanically regulated phagocytic switch. Macrophages lacking β2 integrins or their downstream effectors, Talin1 and Vinculin, exhibited specific defects in phagocytic cup architecture and selective suppression of stiff cargo uptake. We conclude that integrin signaling serves as a mechanical checkpoint during phagocytosis to pair cargo rigidity to the appropriate mode of engulfment.
Collapse
Affiliation(s)
- Alexander H Settle
- Louis V. Gerstner, Jr., Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Benjamin Y Winer
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Miguel M de Jesus
- Louis V. Gerstner, Jr., Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lauren Seeman
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zhaoquan Wang
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology & Molecular Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Eric Chan
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yevgeniy Romin
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zhuoning Li
- Proteomics Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew M Miele
- Proteomics Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ronald C Hendrickson
- Proteomics Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, Miami, FL, USA
| | - Daan Vorselen
- Cell Biology and Immunology, Wageningen University & Research, Wageningen, The Netherlands
| | - Justin S A Perry
- Louis V. Gerstner, Jr., Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology & Molecular Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Morgan Huse
- Louis V. Gerstner, Jr., Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Immunology & Molecular Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
6
|
Pathni A, Wagh K, Rey-Suarez I, Upadhyaya A. Mechanical regulation of lymphocyte activation and function. J Cell Sci 2024; 137:jcs219030. [PMID: 38995113 PMCID: PMC11267459 DOI: 10.1242/jcs.219030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024] Open
Abstract
Mechanosensing, or how cells sense and respond to the physical environment, is crucial for many aspects of biological function, ranging from cell movement during development to cancer metastasis, the immune response and gene expression driving cell fate determination. Relevant physical stimuli include the stiffness of the extracellular matrix, contractile forces, shear flows in blood vessels, complex topography of the cellular microenvironment and membrane protein mobility. Although mechanosensing has been more widely studied in non-immune cells, it has become increasingly clear that physical cues profoundly affect the signaling function of cells of the immune system. In this Review, we summarize recent studies on mechanical regulation of immune cells, specifically lymphocytes, and explore how the force-generating cytoskeletal machinery might mediate mechanosensing. We discuss general principles governing mechanical regulation of lymphocyte function, spanning from the molecular scale of receptor activation to cellular responses to mechanical stimuli.
Collapse
Affiliation(s)
- Aashli Pathni
- Biological Sciences Graduate Program, University of Maryland, College Park, MD 20742, USA
| | - Kaustubh Wagh
- Department of Physics, University of Maryland, College Park, MD 20742, USA
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ivan Rey-Suarez
- Insitute for Physical Science and Technology, University of Maryland, College Park, MD 20742, USA
- Microcore, Universidad de Los Andes, Bogota, DC 111711, USA
| | - Arpita Upadhyaya
- Biological Sciences Graduate Program, University of Maryland, College Park, MD 20742, USA
- Department of Physics, University of Maryland, College Park, MD 20742, USA
- Insitute for Physical Science and Technology, University of Maryland, College Park, MD 20742, USA
- Biophysics Program, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
7
|
Urbanska M, Guck J. Single-Cell Mechanics: Structural Determinants and Functional Relevance. Annu Rev Biophys 2024; 53:367-395. [PMID: 38382116 DOI: 10.1146/annurev-biophys-030822-030629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
The mechanical phenotype of a cell determines its ability to deform under force and is therefore relevant to cellular functions that require changes in cell shape, such as migration or circulation through the microvasculature. On the practical level, the mechanical phenotype can be used as a global readout of the cell's functional state, a marker for disease diagnostics, or an input for tissue modeling. We focus our review on the current knowledge of structural components that contribute to the determination of the cellular mechanical properties and highlight the physiological processes in which the mechanical phenotype of the cells is of critical relevance. The ongoing efforts to understand how to efficiently measure and control the mechanical properties of cells will define the progress in the field and drive mechanical phenotyping toward clinical applications.
Collapse
Affiliation(s)
- Marta Urbanska
- Max Planck Institute for the Science of Light, Erlangen, Germany; ,
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Jochen Guck
- Max Planck Institute for the Science of Light, Erlangen, Germany; ,
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
8
|
Pouraria H, Houston JP. Elasticity of Carrier Fluid: A Key Factor Affecting Mechanical Phenotyping in Deformability Cytometry. MICROMACHINES 2024; 15:822. [PMID: 39064333 PMCID: PMC11278870 DOI: 10.3390/mi15070822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 07/28/2024]
Abstract
Recently, microfluidics deformability cytometry has emerged as a powerful tool for high-throughput mechanical phenotyping of large populations of cells. These methods characterize cells by their mechanical fingerprints by exerting hydrodynamic forces and monitoring the resulting deformation. These devices have shown great promise for label-free cytometry, yet there is a critical need to improve their accuracy and reconcile any discrepancies with other methods, such as atomic force microscopy. In this study, we employ computational fluid dynamics simulations and uncover how the elasticity of frequently used carrier fluids, such as methylcellulose dissolved in phosphate-buffered saline, is significantly influential to the resulting cellular deformation. We conducted CFD simulations conventionally used within the deformability cytometry field, which neglect fluid elasticity. Subsequently, we incorporated a more comprehensive model that simulates the viscoelastic nature of the carrier fluid. A comparison of the predicted stresses between these two approaches underscores the significance of the emerging elastic stresses in addition to the well-recognized viscous stresses along the channel. Furthermore, we utilize a two-phase flow model to predict the deformation of a promyelocyte (i.e., HL-60 cell type) within a hydrodynamic constriction channel. The obtained results highlight a substantial impact of the elasticity of carrier fluid on cellular deformation and raise questions about the accuracy of mechanical property estimates derived by neglecting elastic stresses.
Collapse
Affiliation(s)
| | - Jessica P. Houston
- Department of Chemical and Materials Engineering, New Mexico State University, Las Cruces, NM 88003, USA;
| |
Collapse
|
9
|
Bergamaschi G, Taris KKH, Biebricher AS, Seymonson XMR, Witt H, Peterman EJG, Wuite GJL. Viscoelasticity of diverse biological samples quantified by Acoustic Force Microrheology (AFMR). Commun Biol 2024; 7:683. [PMID: 38834871 PMCID: PMC11150513 DOI: 10.1038/s42003-024-06367-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 05/21/2024] [Indexed: 06/06/2024] Open
Abstract
In the context of soft matter and cellular mechanics, microrheology - the use of micron-sized particles to probe the frequency-dependent viscoelastic response of materials - is widely used to shed light onto the mechanics and dynamics of molecular structures. Here we present the implementation of active microrheology in an Acoustic Force Spectroscopy setup (AFMR), which combines multiplexing with the possibility of probing a wide range of forces ( ~ pN to ~nN) and frequencies (0.01-100 Hz). To demonstrate the potential of this approach, we perform active microrheology on biological samples of increasing complexity and stiffness: collagen gels, red blood cells (RBCs), and human fibroblasts, spanning a viscoelastic modulus range of five orders of magnitude. We show that AFMR can successfully quantify viscoelastic properties by probing many beads with high single-particle precision and reproducibility. Finally, we demonstrate that AFMR to map local sample heterogeneities as well as detect cellular responses to drugs.
Collapse
Affiliation(s)
- Giulia Bergamaschi
- Department of Physics and Astronomy and LaserLaB Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Kees-Karel H Taris
- Department of Physics and Astronomy and LaserLaB Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Andreas S Biebricher
- Department of Physics and Astronomy and LaserLaB Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Xamanie M R Seymonson
- Department of Physics and Astronomy and LaserLaB Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Hannes Witt
- Department of Physics and Astronomy and LaserLaB Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Erwin J G Peterman
- Department of Physics and Astronomy and LaserLaB Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Gijs J L Wuite
- Department of Physics and Astronomy and LaserLaB Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
10
|
Lith SC, Evers TMJ, Freire BM, van Tiel CM, Vos WG, Mashaghi A, de Vries CJM. Nuclear receptor Nur77 regulates immunomechanics of macrophages. Eur J Cell Biol 2024; 103:151419. [PMID: 38763048 DOI: 10.1016/j.ejcb.2024.151419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/17/2024] [Accepted: 05/08/2024] [Indexed: 05/21/2024] Open
Abstract
Nuclear receptor Nur77 plays a pivotal role in immune regulation across various tissues, influencing pro-inflammatory signaling pathways and cellular metabolism. While cellular mechanics have been implicated in inflammation, the contribution of Nur77 to these mechanical processes remains elusive. Macrophages exhibit remarkable plasticity in their morphology and mechanics, enabling them to adapt and execute essential inflammatory functions, such as navigating through inflamed tissue and pathogen engulfment. However, the precise regulatory mechanisms governing these dynamic changes in macrophage mechanics during inflammation remain poorly understood. To establish the potential correlation of Nur77 with cellular mechanics, we compared bone marrow-derived macrophages (BMDMs) from wild-type (WT) and Nur77-deficient (Nur77-KO) mice and employed cytoskeletal imaging, single-cell acoustic force spectroscopy (AFS), migration and phagocytosis assays, and RNA-sequencing. Our findings reveal that Nur77-KO BMDMs exhibit changes to their actin networks compared to WT BMDMs, which is associated with a stiffer and more rigid phenotype. Subsequent in vitro experiments validated our observations, showcasing that Nur77 deficiency leads to enhanced migration, reduced adhesion, and increased phagocytic activity. The transcriptomics data confirmed altered mechanics-related pathways in Nur77-deficient macrophage that are accompanied by a robust pro-inflammatory phenotype. Utilizing previously obtained ChIP-data, we revealed that Nur77 directly targets differentially expressed genes associated with cellular mechanics. In conclusion, while Nur77 is recognized for its role in reducing inflammation of macrophages by inhibiting the expression of pro-inflammatory genes, our study identifies a novel regulatory mechanism where Nur77 governs macrophage inflammation through the modulation of expression of genes involved in cellular mechanics. Our findings suggest that immune regulation by Nur77 may be partially mediated through alterations in cellular mechanics, highlighting a potential avenue for therapeutic targeting.
Collapse
Affiliation(s)
- Sanne C Lith
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, University of Amsterdam, The Netherlands; Amsterdam Institute for Immunology and Infectious diseases, University of Amsterdam, The Netherlands
| | - Tom M J Evers
- Medical Systems Biophysics and Bioengineering, Leiden Academic Centre for Drug Research, Faculty of Mathematics and Natural Sciences, Leiden University, Leiden, The Netherlands
| | - Beatriz M Freire
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, University of Amsterdam, The Netherlands; Amsterdam Institute for Immunology and Infectious diseases, University of Amsterdam, The Netherlands
| | - Claudia M van Tiel
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, University of Amsterdam, The Netherlands; Amsterdam Institute for Immunology and Infectious diseases, University of Amsterdam, The Netherlands
| | - Winnie G Vos
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, University of Amsterdam, The Netherlands; Amsterdam Institute for Immunology and Infectious diseases, University of Amsterdam, The Netherlands
| | - Alireza Mashaghi
- Medical Systems Biophysics and Bioengineering, Leiden Academic Centre for Drug Research, Faculty of Mathematics and Natural Sciences, Leiden University, Leiden, The Netherlands.
| | - Carlie J M de Vries
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, University of Amsterdam, The Netherlands; Amsterdam Institute for Immunology and Infectious diseases, University of Amsterdam, The Netherlands.
| |
Collapse
|
11
|
Ayyanar C, Rakshit S, Sarkar K, Pramanik S. Unprecedented Approach of Fabrication and Analysis of a Bioactive PDMS/Hydroxyapatite/Graphene Nanocomposite Scaffold with a Vascular Channel to Combat Carcinogenesis. ACS APPLIED BIO MATERIALS 2024; 7:3388-3402. [PMID: 38660938 DOI: 10.1021/acsabm.4c00299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
In the present investigation, natural bone-derived hydroxyapatite (HA, 2 wt %) and/or exfoliated graphene (Gr, 0.1 wt %)-embedded polydimethylsiloxane (PDMS) elastomeric films were prepared using a vascular method. The morphology, mechanical properties, crystallinity, and chemical structure of the composite films were evaluated. The in vitro biodegradation kinetics of the films indicates their adequate physiological stability. Most of the results favored PDMS/HA/Gr as a best composite scaffold having more than 703% elongation. A simulation study of the microfluidic vascular channel of the PDMS/HA/Gr scaffold suggests that the pressure drop at the outlet became greater (from 1.19 to 0.067 Pa) unlike velocity output (from 0.071 to 0.089 m/s), suggesting a turbulence-free laminar flow. Our bioactive scaffold material, PDMS/HA/Gr, showed highest cytotoxicity toward the lung cancer and breast cancer cells through Runx3 protein-mediated cytotoxic T lymphocyte (CTL) generation. Our data and predicted mechanism also suggested that the PDMS/HA/Gr-supported peripheral blood mononuclear cells (PBMCs) not only increased the generation of CTL but also upregulated the expression of RUNX3. Since the PDMS/HA/Gr scaffold-supported Runx3 induced CTL generation caused maximum cell cytotoxicity of breast cancer (MCF-7) and lung cancer (A549) cells, PDMS/HA/Gr can be treated as an excellent potential candidate for CTL-mediated cancer therapy.
Collapse
Affiliation(s)
- Chellaiah Ayyanar
- Functional and Biomaterials Engineering Lab, Department of Mechanical Engineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamil Nadu, India
| | - Sudeshna Rakshit
- Cancer Immunology and Gene Editing Technology Lab, Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamil Nadu, India
| | - Koustav Sarkar
- Cancer Immunology and Gene Editing Technology Lab, Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamil Nadu, India
| | - Sumit Pramanik
- Functional and Biomaterials Engineering Lab, Department of Mechanical Engineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamil Nadu, India
| |
Collapse
|
12
|
Ali FEM, Ibrahim IM, Althagafy HS, Hassanein EHM. Role of immunotherapies and stem cell therapy in the management of liver cancer: A comprehensive review. Int Immunopharmacol 2024; 132:112011. [PMID: 38581991 DOI: 10.1016/j.intimp.2024.112011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/08/2024]
Abstract
Liver cancer (LC) is the sixth most common disease and the third most common cause of cancer-related mortality. The WHO predicts that more than 1 million deaths will occur from LC by 2030. Hepatocellular carcinoma (HCC) is a common form of primary LC. Today, the management of LC involves multiple disciplines, and multimodal therapy is typically selected on an individual basis, considering the intricate interactions between the patient's overall health, the stage of the tumor, and the degree of underlying liver disease. Currently, the treatment of cancers, including LC, has undergone a paradigm shift in the last ten years because of immuno-oncology. To treat HCC, immune therapy approaches have been developed to enhance or cause the body's natural immune response to specifically target tumor cells. In this context, immune checkpoint pathway inhibitors, engineered cytokines, adoptive cell therapy, immune cells modified with chimeric antigen receptors, and therapeutic cancer vaccines have advanced to clinical trials and offered new hope to cancer patients. The outcomes of these treatments are encouraging. Additionally, treatment using stem cells is a new approach for restoring deteriorated tissues because of their strong differentiation potential and capacity to release cytokines that encourage cell division and the formation of blood vessels. Although there is no proof that stem cell therapy works for many types of cancer, preclinical research on stem cells has shown promise in treating HCC. This review provides a recent update regarding the impact of immunotherapy and stem cells in HCC and promising outcomes.
Collapse
Affiliation(s)
- Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, 71524, Egypt; Michael Sayegh, Faculty of Pharmacy, Aqaba University of Technology, Aqaba 77110, Jordan.
| | - Islam M Ibrahim
- Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, 71524, Egypt
| |
Collapse
|
13
|
Hou F, Guo Z, Ho MT, Hui Y, Zhao CX. Particle-Based Artificial Antigen-Presenting Cell Systems for T Cell Activation in Adoptive T Cell Therapy. ACS NANO 2024; 18:8571-8599. [PMID: 38483840 DOI: 10.1021/acsnano.3c10180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
T cell-based adoptive cell therapy (ACT) has emerged as a promising treatment for various diseases, particularly cancers. Unlike other immunotherapy modalities, ACT involves directly transferring engineered T cells into patients to eradicate diseased cells; hence, it necessitates methods for effectively activating and expanding T cells in vitro. Artificial antigen-presenting cells (aAPCs) have been widely developed based on biomaterials, particularly micro- and nanoparticles, and functionalized with T cell stimulatory antibodies to closely mimic the natural T cell-APC interactions. Due to their vast clinical utility, aAPCs have been employed as an off-the-shelf technology for T cell activation in FDA-approved ACTs, and the development of aAPCs is constantly advancing with the emergence of aAPCs with more sophisticated designs and additional functionalities. Here, we review the recent advancements in particle-based aAPCs for T cell activation in ACTs. Following a brief introduction, we first describe the manufacturing processes of ACT products. Next, the design and synthetic strategies for micro- and nanoparticle-based aAPCs are discussed separately to emphasize their features, advantages, and limitations. Then, the impact of design parameters of aAPCs, such as size, shape, ligand density/mobility, and stiffness, on their functionality and biomedical performance is explored to provide deeper insights into the design concepts and principles for more efficient and safer aAPCs. The review concludes by discussing current challenges and proposing future perspectives for the development of more advanced aAPCs.
Collapse
Affiliation(s)
- Fei Hou
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Zichao Guo
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Minh Trang Ho
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Yue Hui
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Chun-Xia Zhao
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| |
Collapse
|
14
|
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
|
15
|
Jeffreys N, Brockman JM, Zhai Y, Ingber DE, Mooney DJ. Mechanical forces amplify TCR mechanotransduction in T cell activation and function. APPLIED PHYSICS REVIEWS 2024; 11:011304. [PMID: 38434676 PMCID: PMC10848667 DOI: 10.1063/5.0166848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 12/08/2023] [Indexed: 03/05/2024]
Abstract
Adoptive T cell immunotherapies, including engineered T cell receptor (eTCR) and chimeric antigen receptor (CAR) T cell immunotherapies, have shown efficacy in treating a subset of hematologic malignancies, exhibit promise in solid tumors, and have many other potential applications, such as in fibrosis, autoimmunity, and regenerative medicine. While immunoengineering has focused on designing biomaterials to present biochemical cues to manipulate T cells ex vivo and in vivo, mechanical cues that regulate their biology have been largely underappreciated. This review highlights the contributions of mechanical force to several receptor-ligand interactions critical to T cell function, with central focus on the TCR-peptide-loaded major histocompatibility complex (pMHC). We then emphasize the role of mechanical forces in (i) allosteric strengthening of the TCR-pMHC interaction in amplifying ligand discrimination during T cell antigen recognition prior to activation and (ii) T cell interactions with the extracellular matrix. We then describe approaches to design eTCRs, CARs, and biomaterials to exploit TCR mechanosensitivity in order to potentiate T cell manufacturing and function in adoptive T cell immunotherapy.
Collapse
Affiliation(s)
| | | | - Yunhao Zhai
- Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
16
|
Dubay R, Darling EM, Fiering J. Microparticles with tunable, cell-like properties for quantitative acoustic mechanophenotyping. MICROSYSTEMS & NANOENGINEERING 2023; 9:90. [PMID: 37448969 PMCID: PMC10336031 DOI: 10.1038/s41378-023-00556-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/21/2023] [Accepted: 05/08/2023] [Indexed: 07/18/2023]
Abstract
Mechanical properties of biological cells have been shown to correlate with their biomolecular state and function, and therefore methods to measure these properties at scale are of interest. Emerging microfluidic technologies can measure the mechanical properties of cells at rates over 20,000 cells/s, which is more than four orders of magnitude faster than conventional instrumentation. However, precise and repeatable means to calibrate and test these new tools remain lacking, since cells themselves are by nature variable. Commonly, microfluidic tools use rigid polymer microspheres for calibration because they are widely available in cell-similar sizes, but conventional microspheres do not fully capture the physiological range of other mechanical properties that are equally important to device function (e.g., elastic modulus and density). Here, we present for the first time development of monodisperse polyacrylamide microparticles with both tunable elasticity and tunable density. Using these size, elasticity, and density tunable particles, we characterized a custom acoustic microfluidic device that makes single-cell measurements of mechanical properties. We then applied the approach to measure the distribution of the acoustic properties within samples of human leukocytes and showed that the system successfully discriminates lymphocytes from other leukocytes. This initial demonstration shows how the tunable microparticles with properties within the physiologically relevant range can be used in conjunction with microfluidic devices for efficient high-throughput measurements of mechanical properties at single-cell resolution.
Collapse
Affiliation(s)
- Ryan Dubay
- Center for Biomedical Engineering, Brown University, Providence, RI 02912 USA
- Biological Microsystems, Draper, Cambridge, MA 02139 USA
| | - Eric M. Darling
- Center for Biomedical Engineering, Brown University, Providence, RI 02912 USA
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02912 USA
- School of Engineering, Brown University, Providence, RI 02912 USA
- Department of Orthopaedics, Brown University, Providence, RI 02912 USA
| | - Jason Fiering
- Biological Microsystems, Draper, Cambridge, MA 02139 USA
| |
Collapse
|
17
|
Zhao C, Yang Q, Tang R, Li W, Wang J, Yang F, Zhao J, Zhu J, Pang W, Li N, Zhang X, Tian XY, Yao W, Zhou J. DNA methyltransferase 1 deficiency improves macrophage motility and wound healing by ameliorating cholesterol accumulation. NPJ Regen Med 2023; 8:29. [PMID: 37291182 DOI: 10.1038/s41536-023-00306-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 05/30/2023] [Indexed: 06/10/2023] Open
Abstract
Healing of the cutaneous wound requires macrophage recruitment at the sites of injury, where chemotactic migration of macrophages toward the wound is regulated by local inflammation. Recent studies suggest a positive contribution of DNA methyltransferase 1 (Dnmt1) to macrophage pro-informatory responses; however, its role in regulating macrophage motility remains unknown. In this study, myeloid-specific depletion of Dnmt1 in mice promoted cutaneous wound healing and de-suppressed the lipopolysaccharides (LPS)-inhibited macrophage motility. Dnmt1 inhibition in macrophages eliminated the LPS-stimulated changes in cellular mechanical properties in terms of elasticity and viscoelasticity. LPS increased the cellular accumulation of cholesterol in a Dnmt1-depedent manner; cholesterol content determined cellular stiffness and motility. Lipidomic analysis indicated that Dnmt1 inhibition altered the cellular lipid homeostasis, probably through down-regulating the expression of cluster of differentiation 36 CD36 (facilitating lipid influx) and up-regulating the expression of ATP-binding cassette transporter ABCA1 (mediating lipid efflux) and sterol O-acyltransferase 1 SOAT1 (also named ACAT1, catalyzing the esterification of cholesterol). Our study revealed a Dnmt1-dependent epigenetic mechanism in the control of macrophage mechanical properties and the related chemotactic motility, indicating Dnmt1 as both a marker of diseases and a potential target of therapeutic intervention for wound healing.
Collapse
Affiliation(s)
- Chuanrong Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Qianru Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Runze Tang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Wang Li
- School of Engineering Sciences, University of Chinese Academy of Science, Beijing, 100190, China
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jin Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Fangfang Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Jianan Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Juanjuan Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Wei Pang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Ning Li
- School of Engineering Sciences, University of Chinese Academy of Science, Beijing, 100190, China
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xu Zhang
- Tianjin Key Laboratory of Metabolic Diseases, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Center for Cardiovascular Diseases, Research Center of Basic Medical Sciences, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300070, China
| | - Xiao Yu Tian
- School of Biomedical Sciences, Heart and Vascular Institute, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Weijuan Yao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| | - Jing Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China.
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China.
| |
Collapse
|
18
|
Flé G, Houten EV, Rémillard-Labrosse G, FitzHarris G, Cloutier G. Imaging the subcellular viscoelastic properties of mouse oocytes. Proc Natl Acad Sci U S A 2023; 120:e2213836120. [PMID: 37186851 PMCID: PMC10214128 DOI: 10.1073/pnas.2213836120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 04/16/2023] [Indexed: 05/17/2023] Open
Abstract
In recent years, cellular biomechanical properties have been investigated as an alternative to morphological assessments for oocyte selection in reproductive science. Despite the high relevance of cell viscoelasticity characterization, the reconstruction of spatially distributed viscoelastic parameter images in such materials remains a major challenge. Here, a framework for mapping viscoelasticity at the subcellular scale is proposed and applied to live mouse oocytes. The strategy relies on the principles of optical microelastography for imaging in combination with the overlapping subzone nonlinear inversion technique for complex-valued shear modulus reconstruction. The three-dimensional nature of the viscoelasticity equations was accommodated by applying an oocyte geometry-based 3D mechanical motion model to the measured wave field. Five domains-nucleolus, nucleus, cytoplasm, perivitelline space, and zona pellucida-could be visually differentiated in both oocyte storage and loss modulus maps, and statistically significant differences were observed between most of these domains in either property reconstruction. The method proposed herein presents excellent potential for biomechanical-based monitoring of oocyte health and complex transformations across lifespan. It also shows appreciable latitude for generalization to cells of arbitrary shape using conventional microscopy equipment.
Collapse
Affiliation(s)
- Guillaume Flé
- Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montreal, QCH2X 0A9, Canada
| | - Elijah Van Houten
- Mechanical Engineering Department, University of Sherbrooke, Sherbrooke, QCJ1K 2R1, Canada
| | - Gaudeline Rémillard-Labrosse
- Oocyte and Embryo Research Laboratory, University of Montreal Hospital Research Center, Montreal, QCH2X 0A9, Canada
| | - Greg FitzHarris
- Oocyte and Embryo Research Laboratory, University of Montreal Hospital Research Center, Montreal, QCH2X 0A9, Canada
- Department of Obstetrics and Gynecology, University of Montreal, Montreal, QCH3T 1J4, Canada
| | - Guy Cloutier
- Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montreal, QCH2X 0A9, Canada
- Department of Radiology, Radio-Oncology and Nuclear Medicine, and Institute of Biomedical Engineering, University of Montreal, Montreal, QCH3T 1J4, Canada
| |
Collapse
|
19
|
Waugh RE, Lomakina E, Amitrano A, Kim M. Activation effects on the physical characteristics of T lymphocytes. Front Bioeng Biotechnol 2023; 11:1175570. [PMID: 37256117 PMCID: PMC10225623 DOI: 10.3389/fbioe.2023.1175570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/18/2023] [Indexed: 06/01/2023] Open
Abstract
The deformability of leukocytes is relevant to a wide array of physiological and pathophysiological behaviors. The goal of this study is to provide a detailed, quantitative characterization of the mechanical properties of T cells and how those properties change with activation. We tested T cells and CD8+ cells isolated from peripheral blood samples of healthy donors either immediately (naïve population) or after 7 days of activation in vitro. Single-cell micropipette aspiration was used to test the mechanical properties. T cells exhibit the general characteristics of a highly viscous liquid drop with a cortical "surface" tension, T cort. The time course of each cell entry into the micropipette was measured at two different aspiration pressures to test for shear thinning behavior. The data were analyzed in the framework of an approximate mechanical model of the cell deformation to determine the cortical tension, the cell volume, the magnitude of the initial cell entry, the characteristic viscosity μ o, and the shear thinning coefficient, b. Activation generally caused increases in cellular resistance to deformation and a broadening of the distribution of cell properties. The cell volume increased substantially upon cell activation from ∼200 μm3 to ∼650 μm3. Naive and activated T cells had similar mean cortical tension (∼150 pN/μm). However, compared to naïve CD8+ cells, the cortical tension of activated CD8+ cells increased significantly to ∼250 pN/μm. Dynamic resistance of naive CD8+ T cells, as reflected in their characteristic viscosity, was ∼870 Pa and significantly increased to 1,180 Pa after in vitro activation. The magnitude of the instantaneous projection length as the cell enters the pipette (L init) was more than doubled for activated vs. naive cells. All cell types exhibited shear thinning behavior with coefficients b in the range 0.5-0.65. Increased cell size, cortical tension, and characteristic viscosity all point to increased resistance of activated T cells to passage through the microvasculature, likely contributing to cell trapping. The increased initial elastic response of cells after activation was unexpected and could point to instability in the cell that might contribute to spontaneous cell motility.
Collapse
Affiliation(s)
- Richard E. Waugh
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Elena Lomakina
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Andrea Amitrano
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, United States
| | - Minsoo Kim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, United States
| |
Collapse
|
20
|
Hyun J, Kim SJ, Cho SD, Kim HW. Mechano-modulation of T cells for cancer immunotherapy. Biomaterials 2023; 297:122101. [PMID: 37023528 DOI: 10.1016/j.biomaterials.2023.122101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 03/12/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023]
Abstract
Immunotherapy, despite its promise for future anti-cancer approach, faces significant challenges, such as off-tumor side effects, innate or acquired resistance, and limited infiltration of immune cells into stiffened extracellular matrix (ECM). Recent studies have highlighted the importance of mechano-modulation/-activation of immune cells (mainly T cells) for effective caner immunotherapy. Immune cells are highly sensitive to the applied physical forces and matrix mechanics, and reciprocally shape the tumor microenvironment. Engineering T cells with tuned properties of materials (e.g., chemistry, topography, and stiffness) can improve their expansion and activation ex vivo, and their ability to mechano-sensing the tumor specific ECM in vivo where they perform cytotoxic effects. T cells can also be exploited to secrete enzymes that soften ECM, thus increasing tumor infiltration and cellular therapies. Furthermore, T cells, such as chimeric antigen receptor (CAR)-T cells, genomic engineered to be spatiotemporally controllable by physical stimuli (e.g., ultrasound, heat, or light), can mitigate adverse off-tumor effects. In this review, we communicate these recent cutting-edge endeavors devoted to mechano-modulating/-activating T cells for effective cancer immunotherapy, and discuss future prospects and challenges in this field.
Collapse
|
21
|
Jiang H, Wang S. Immune cells use active tugging forces to distinguish affinity and accelerate evolution. Proc Natl Acad Sci U S A 2023; 120:e2213067120. [PMID: 36897986 PMCID: PMC10089171 DOI: 10.1073/pnas.2213067120] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 01/11/2023] [Indexed: 03/12/2023] Open
Abstract
Cells are known to exert forces to sense their physical surroundings for guidance of motion and fate decisions. Here, we propose that cells might do mechanical work to drive their own evolution, taking inspiration from the adaptive immune system. Growing evidence indicates that immune B cells-capable of rapid Darwinian evolution-use cytoskeletal forces to actively extract antigens from other cells' surfaces. To elucidate the evolutionary significance of force usage, we develop a theory of tug-of-war antigen extraction that maps receptor binding characteristics to clonal reproductive fitness, revealing physical determinants of selection strength. This framework unifies mechanosensing and affinity-discrimination capabilities of evolving cells: Pulling against stiff antigen tethers enhances discrimination stringency at the expense of absolute extraction. As a consequence, active force usage can accelerate adaptation but may also cause extinction of cell populations, resulting in an optimal range of pulling strength that matches molecular rupture forces observed in cells. Our work suggests that nonequilibrium, physical extraction of environmental signals can make biological systems more evolvable at a moderate energy cost.
Collapse
Affiliation(s)
- Hongda Jiang
- Department of Physics and Astronomy, University of California Los Angeles, Los Angeles, CA90095
| | - Shenshen Wang
- Department of Physics and Astronomy, University of California Los Angeles, Los Angeles, CA90095
| |
Collapse
|
22
|
Immature and mature bone marrow-derived dendritic cells exhibit distinct intracellular mechanical properties. Sci Rep 2023; 13:1967. [PMID: 36737470 PMCID: PMC9898242 DOI: 10.1038/s41598-023-28625-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/20/2023] [Indexed: 02/05/2023] Open
Abstract
Dendritic cells (DCs) patrol the organism at an immature stage to detect the presence of pathogens. Once activated, these mature DCs reach the lymph nodes to activate antigen-specific T lymphocytes and thus initiate an adaptative immune response to control the pathogen. The migration of both immature and mature DCs is a key process for their optimal function. DC migration requires transit through narrow constrictions that is allowed by their high local and global deformation capabilities. In addition to cytoplasmic changes, the nucleus mechanical properties also have a major impact for cellular migration and motility. Yet, nucleus intracellular mobility of dendritic cells or its variation upon maturation have not been investigated. Our study defines the biophysical phenotypic variations of dendritic cells upon maturation using interferometric deformability cytometry. This method characterizes different cellular mechanical properties, such as elongation and nucleus offset, by assessing the refractive index spatial distribution of shear-induced deformed cells. By using these parameters, our data suggest that in vitro bone marrow derived dendritic cell (BMDC) maturation induces cell stiffening and reduces nucleus mobility, allowing to distinguish immature and mature dendritic cells. Overall, our method provides insights on intracellular mechanical properties of two dendritic cell states.
Collapse
|
23
|
Pineau J, Moreau H, Duménil AML, Pierobon P. Polarity in immune cells. Curr Top Dev Biol 2023; 154:197-222. [PMID: 37100518 DOI: 10.1016/bs.ctdb.2023.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Immune cells are responsible for pathogen detection and elimination, as well as for signaling to other cells the presence of potential danger. In order to mount an efficient immune response, they need to move and search for a pathogen, interact with other cells, and diversify the population by asymmetric cell division. All these actions are regulated by cell polarity: cell polarity controls cell motility, which is crucial for scanning peripheral tissues to detect pathogens, and recruiting immune cells to sites of infection; immune cells, in particular lymphocytes, communicate with each other by a direct contact called immunological synapse, which entails a global polarization of the cell and plays a role in activating lymphocyte response; finally, immune cells divide asymmetrically from a precursor, generating a diversity of phenotypes and cell types among daughter cells, such as memory and effector cells. This review aims at providing an overview from both biology and physics perspectives of how cell polarity shapes the main immune cell functions.
Collapse
Affiliation(s)
- Judith Pineau
- Institut Curie, PSL Research University, INSERM U932, Paris, Cedex, France; Université Paris Cité, Paris, France
| | - Hélène Moreau
- Institut Curie, PSL Research University, INSERM U932, Paris, Cedex, France
| | | | - Paolo Pierobon
- Institut Curie, PSL Research University, INSERM U932, Paris, Cedex, France.
| |
Collapse
|
24
|
Alatoom A, ElGindi M, Sapudom J, Teo JCM. The T Cell Journey: A Tour de Force. Adv Biol (Weinh) 2023; 7:e2200173. [PMID: 36190140 DOI: 10.1002/adbi.202200173] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/30/2022] [Indexed: 11/07/2022]
Abstract
T cells act as the puppeteers in the adaptive immune response, and their dysfunction leads to the initiation and progression of pathological conditions. During their lifetime, T cells experience myriad forces that modulate their effector functions. These forces are imposed by interacting cells, surrounding tissues, and shear forces from fluid movement. In this review, a journey with T cells is made, from their development to their unique characteristics, including the early studies that uncovered their mechanosensitivity. Then the studies pertaining to the responses of T cell activation to changes in antigen-presenting cells' physical properties, to their immediate surrounding extracellular matrix microenvironment, and flow conditions are highlighted. In addition, it is explored how pathological conditions like the tumor microenvironment can hinder T cells and allow cancer cells to escape elimination.
Collapse
Affiliation(s)
- Aseel Alatoom
- Laboratory for Immuno Bioengineering Research and Applications Division of Engineering, New York University Abu Dhabi, Saadiyat Campus, P.O. Box 127788, Abu Dhabi, UAE.,Department of Mechanical Engineering Tandon School of Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA
| | - Mei ElGindi
- Laboratory for Immuno Bioengineering Research and Applications Division of Engineering, New York University Abu Dhabi, Saadiyat Campus, P.O. Box 127788, Abu Dhabi, UAE
| | - Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications Division of Engineering, New York University Abu Dhabi, Saadiyat Campus, P.O. Box 127788, Abu Dhabi, UAE
| | - Jeremy C M Teo
- Laboratory for Immuno Bioengineering Research and Applications Division of Engineering, New York University Abu Dhabi, Saadiyat Campus, P.O. Box 127788, Abu Dhabi, UAE.,Department of Mechanical Engineering Tandon School of Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA.,Department of Biomedical Engineering Tandon School of Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA
| |
Collapse
|
25
|
Pinon L, Ruyssen N, Pineau J, Mesdjian O, Cuvelier D, Chipont A, Allena R, Guerin CL, Asnacios S, Asnacios A, Pierobon P, Fattaccioli J. Phenotyping polarization dynamics of immune cells using a lipid droplet-cell pairing microfluidic platform. CELL REPORTS METHODS 2022; 2:100335. [PMID: 36452873 PMCID: PMC9701611 DOI: 10.1016/j.crmeth.2022.100335] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 09/20/2022] [Accepted: 10/19/2022] [Indexed: 05/12/2023]
Abstract
The immune synapse is the tight contact zone between a lymphocyte and a cell presenting its cognate antigen. This structure serves as a signaling platform and entails a polarization of intracellular components necessary to the immunological function of the cell. While the surface properties of the presenting cell are known to control the formation of the synapse, their impact on polarization has not yet been studied. Using functional lipid droplets as tunable artificial presenting cells combined with a microfluidic pairing device, we simultaneously observe synchronized synapses and dynamically quantify polarization patterns of individual B cells. By assessing how ligand concentration, surface fluidity, and substrate rigidity impact lysosome polarization, we show that its onset and kinetics depend on the local antigen concentration at the synapse and on substrate rigidity. Our experimental system enables a fine phenotyping of monoclonal cell populations based on their synaptic readout.
Collapse
Affiliation(s)
- Léa Pinon
- École Normale Supérieure, UMR 8640, Laboratoire PASTEUR, Département de Chimie, PSL Research University, Sorbonne Université, CNRS, 75005 Paris, France
- Institut Curie, U932, Immunology and Cancer, INSERM, 75005 Paris, France
- Institut Pierre-Gilles de Gennes pour la Microfluidique, 75005 Paris, France
| | - Nicolas Ruyssen
- Arts et Métiers Institute of Technology, Université Paris 13, Sorbonne Paris Cité, IBHGC, HESAM Université, 75013 Paris, France
| | - Judith Pineau
- Institut Curie, U932, Immunology and Cancer, INSERM, 75005 Paris, France
| | - Olivier Mesdjian
- École Normale Supérieure, UMR 8640, Laboratoire PASTEUR, Département de Chimie, PSL Research University, Sorbonne Université, CNRS, 75005 Paris, France
- Institut Pierre-Gilles de Gennes pour la Microfluidique, 75005 Paris, France
| | - Damien Cuvelier
- Institut Pierre-Gilles de Gennes pour la Microfluidique, 75005 Paris, France
- Institut Curie, UMR 144, PSL Research University, CNRS, Paris, France
- Sorbonne Université, Faculté des Sciences et Ingénierie, UFR 926 Chemistry, 75005 Paris, France
| | - Anna Chipont
- Institut Curie, Cytometry Platform, 75005 Paris, France
| | - Rachele Allena
- Arts et Métiers Institute of Technology, Université Paris 13, Sorbonne Paris Cité, IBHGC, HESAM Université, 75013 Paris, France
- LJAD, UMR 7351, Université Côte d’Azur, 06100 Nice, France
| | - Coralie L. Guerin
- Institut Curie, Cytometry Platform, 75005 Paris, France
- Université Paris Cité, INSERM, Innovative Therapies in Haemostasis, 75006 Paris, France
| | - Sophie Asnacios
- Université de Paris, CNRS, Laboratoire Matière et Systèmes Complexes, UMR 7057, 75013 Paris, France
- Sorbonne Université, Faculté des Sciences et Ingénierie, UFR 925 Physics, 75005 Paris, France
| | - Atef Asnacios
- Université de Paris, CNRS, Laboratoire Matière et Systèmes Complexes, UMR 7057, 75013 Paris, France
| | - Paolo Pierobon
- Institut Curie, U932, Immunology and Cancer, INSERM, 75005 Paris, France
| | - Jacques Fattaccioli
- École Normale Supérieure, UMR 8640, Laboratoire PASTEUR, Département de Chimie, PSL Research University, Sorbonne Université, CNRS, 75005 Paris, France
- Institut Pierre-Gilles de Gennes pour la Microfluidique, 75005 Paris, France
| |
Collapse
|
26
|
Anikeeva N, Steblyanko M, Kuri-Cervantes L, Buggert M, Betts MR, Sykulev Y. The immune synapses reveal aberrant functions of CD8 T cells during chronic HIV infection. Nat Commun 2022; 13:6436. [PMID: 36307445 PMCID: PMC9616955 DOI: 10.1038/s41467-022-34157-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 10/14/2022] [Indexed: 02/05/2023] Open
Abstract
Chronic HIV infection causes persistent low-grade inflammation that induces premature aging of the immune system including senescence of memory and effector CD8 T cells. To uncover the reasons of gradually diminished potency of CD8 T cells from people living with HIV, here we expose the T cells to planar lipid bilayers containing ligands for T-cell receptor and a T-cell integrins and analyze the cellular morphology, dynamics of synaptic interface formation and patterns of the cellular degranulation. We find a large fraction of phenotypically naive T cells from chronically infected people are capable to form mature synapse with focused degranulation, a signature of a differentiated T cells. Further, differentiation of aberrant naive T cells may lead to the development of anomalous effector T cells undermining their capacity to control HIV and other pathogens that could be contained otherwise.
Collapse
Affiliation(s)
- Nadia Anikeeva
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Maria Steblyanko
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Leticia Kuri-Cervantes
- Department of Microbiology and Institute of Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marcus Buggert
- Department of Microbiology and Institute of Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Michael R Betts
- Department of Microbiology and Institute of Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuri Sykulev
- Departments of Immunology and Medical Oncology, Thomas Jefferson University, Philadelphia, PA, USA.
- Sydney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
27
|
Matei AE, Kubánková M, Xu L, Györfi AH, Boxberger E, Soteriou D, Papava M, Prater J, Hong X, Bergmann C, Kräter M, Schett G, Guck J, Distler JHW. Identification of a Distinct Monocyte-Driven Signature in Systemic Sclerosis Using Biophysical Phenotyping of Circulating Immune Cells. Arthritis Rheumatol 2022; 75:768-781. [PMID: 36281753 DOI: 10.1002/art.42394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 09/08/2022] [Accepted: 10/18/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Pathologically activated circulating immune cells, including monocytes, play major roles in systemic sclerosis (SSc). Their functional characterization can provide crucial information with direct clinical relevance. However, tools for the evaluation of pathologic immune cell activation and, in general, of clinical outcomes in SSc are scarce. Biophysical phenotyping (including characterization of cell mechanics and morphology) provides access to a novel, mostly unexplored layer of information regarding pathophysiologic immune cell activation. We hypothesized that the biophysical phenotyping of circulating immune cells, reflecting their pathologic activation, can be used as a clinical tool for the evaluation and risk stratification of patients with SSc. METHODS We performed biophysical phenotyping of circulating immune cells by real-time fluorescence and deformability cytometry (RT-FDC) in 63 SSc patients, 59 rheumatoid arthritis (RA) patients, 28 antineutrophil cytoplasmic antibody-associated vasculitis (AAV) patients, and 22 age- and sex-matched healthy donors. RESULTS We identified a specific signature of biophysical properties of circulating immune cells in SSc patients that was mainly driven by monocytes. Since it is absent in RA and AAV, this signature reflects an SSc-specific monocyte activation rather than general inflammation. The biophysical properties of monocytes indicate current disease activity, the extent of skin or lung fibrosis, and the severity of manifestations of microvascular damage, as well as the risk of disease progression in SSc patients. CONCLUSION Changes in the biophysical properties of circulating immune cells reflect their pathologic activation in SSc patients and are associated with clinical outcomes. As a high-throughput approach that requires minimal preparations, RT-FDC-based biophysical phenotyping of monocytes can serve as a tool for the evaluation and risk stratification of patients with SSc.
Collapse
Affiliation(s)
- Alexandru-Emil Matei
- Department of Rheumatology and Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Markéta Kubánková
- Max Planck Institute for the Science of Light & Max-Planck-Center für Physik und Medizin, Erlangen, Germany, and Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Liyan Xu
- Department of Rheumatology and Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Andrea-Hermina Györfi
- Department of Rheumatology and Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Evgenia Boxberger
- Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Despina Soteriou
- Max Planck Institute for the Science of Light & Max-Planck-Center für Physik und Medizin, Erlangen, Germany
| | - Maria Papava
- Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Julia Prater
- Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Xuezhi Hong
- Department of Rheumatology and Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christina Bergmann
- Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Martin Kräter
- Max Planck Institute for the Science of Light & Max-Planck-Center für Physik und Medizin, Erlangen, Germany, and Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Georg Schett
- Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jochen Guck
- Max Planck Institute for the Science of Light & Max-Planck-Center für Physik und Medizin, Erlangen, Germany, and Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Jörg H W Distler
- Department of Rheumatology and Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
28
|
Kalashnikov N, Moraes C. Engineering physical microenvironments to study innate immune cell biophysics. APL Bioeng 2022; 6:031504. [PMID: 36156981 PMCID: PMC9492295 DOI: 10.1063/5.0098578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/22/2022] [Indexed: 12/04/2022] Open
Abstract
Innate immunity forms the core of the human body's defense system against infection, injury, and foreign objects. It aims to maintain homeostasis by promoting inflammation and then initiating tissue repair, but it can also lead to disease when dysregulated. Although innate immune cells respond to their physical microenvironment and carry out intrinsically mechanical actions such as migration and phagocytosis, we still do not have a complete biophysical description of innate immunity. Here, we review how engineering tools can be used to study innate immune cell biophysics. We first provide an overview of innate immunity from a biophysical perspective, review the biophysical factors that affect the innate immune system, and then explore innate immune cell biophysics in the context of migration, phagocytosis, and phenotype polarization. Throughout the review, we highlight how physical microenvironments can be designed to probe the innate immune system, discuss how biophysical insight gained from these studies can be used to generate a more comprehensive description of innate immunity, and briefly comment on how this insight could be used to develop mechanical immune biomarkers and immunomodulatory therapies.
Collapse
Affiliation(s)
- Nikita Kalashnikov
- Department of Chemical Engineering, McGill University, Montreal, Quebec H3A 0G4, Canada
| | | |
Collapse
|
29
|
Zhao L, Fu X, Zhang L, Ye Z. Effect of mechanical properties of Jurkat cell on adhesion properties of Jurkat integrin and VCAM-1: An AFM study. Colloids Surf B Biointerfaces 2022; 218:112784. [PMID: 36030725 DOI: 10.1016/j.colsurfb.2022.112784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 08/07/2022] [Accepted: 08/20/2022] [Indexed: 10/15/2022]
Abstract
Mechanical properties play key roles in the immune system, especially the activation, transformation and subsequent effector responses of immune cells. As transmembrane adhesion receptors, integrins mediate the adhesion events of both cells and cell-extracellular matrix (ECM). Integrin affinity would influence the crosslinking of cytoskeleton, leading to the change of elastic properties of cells. In this study, the cells were treated with F-actin destabilizing agent Cytochalasin-D (Cyt-D), fixed by Glutaraldehyde, and cultivated in hypotonic solution respectively. We used Atomic force microscopy (AFM) to quantitatively measure the elasticity of Jurkat cells and adhesion properties between integrins and vascular cell adhesion molecule-1 (VCAM-1), and immunofluorescence to study the alteration of cytoskeleton. Glutaraldehyde had a positive effect on the adhesion force and Young's modulus. However, these mechanical properties decreased in a hypotonic environment, confirming the findings of cellular physiological structure. There was no significant difference in the bond strength and elasticity of Jurkat cells treated with Cytochalasin-D, probably because of lower importance of actin in suspension cells. All the treatments in this study pose a negative effect on the adhesion probability between integrins and VCAM-1, which demonstrates the effect of structural alteration of the cytoskeleton on the conformation of integrin. Clear consistency between adhesion force of integrin/VCAM-1 bond and Young's modulus of Jurkat cells was shown. Our results further demonstrated the relationship between cytoskeleton and integrin-ligand by mechanical characteristics.
Collapse
Affiliation(s)
- Leqian Zhao
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing, China
| | - Xingliang Fu
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing, China
| | - Liyuan Zhang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing, China
| | - Zhiyi Ye
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing, China.
| |
Collapse
|
30
|
Mustapha F, Sengupta K, Puech PH. May the force be with your (immune) cells: an introduction to traction force microscopy in Immunology. Front Immunol 2022; 13:898558. [PMID: 35990636 PMCID: PMC9389945 DOI: 10.3389/fimmu.2022.898558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/28/2022] [Indexed: 11/21/2022] Open
Abstract
For more than a couple of decades now, "force" has been recognized as an important physical parameter that cells employ to adapt to their microenvironment. Whether it is externally applied, or internally generated, cells use force to modulate their various actions, from adhesion and migration to differentiation and immune function. T lymphocytes use such mechano-sensitivity to decipher signals when recognizing cognate antigens presented on the surface of antigen presenting cells (APCs), a critical process in the adaptive immune response. As such, many techniques have been developed and used to measure the forces felt/exerted by these small, solitary and extremely reactive cells to decipher their influence on diverse T cell functions, primarily activation. Here, we focus on traction force microscopy (TFM), in which a deformable substrate, coated with the appropriate molecules, acts as a force sensor on the cellular scale. This technique has recently become a center of interest for many groups in the "ImmunoBiophysics" community and, as a consequence, has been subjected to refinements for its application to immune cells. Here, we present an overview of TFM, the precautions and pitfalls, and the most recent developments in the context of T cell immunology.
Collapse
Affiliation(s)
- Farah Mustapha
- Laboratory Adhesion Inflammation (LAI), INSERM, CNRS, Aix Marseille University, Marseille, France
- Centre Interdisciplinaire de Nanoscience de Marseille (CINaM), CNRS, Aix Marseille University, Marseille, France
- Turing Center for Living Systems (CENTURI), Marseille, France
| | - Kheya Sengupta
- Centre Interdisciplinaire de Nanoscience de Marseille (CINaM), CNRS, Aix Marseille University, Marseille, France
- Turing Center for Living Systems (CENTURI), Marseille, France
| | - Pierre-Henri Puech
- Laboratory Adhesion Inflammation (LAI), INSERM, CNRS, Aix Marseille University, Marseille, France
- Turing Center for Living Systems (CENTURI), Marseille, France
| |
Collapse
|
31
|
Zhang T, Jia Y, Yu Y, Zhang B, Xu F, Guo H. Targeting the tumor biophysical microenvironment to reduce resistance to immunotherapy. Adv Drug Deliv Rev 2022; 186:114319. [PMID: 35545136 DOI: 10.1016/j.addr.2022.114319] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 04/28/2022] [Accepted: 04/30/2022] [Indexed: 02/06/2023]
Abstract
Immunotherapy based on immune checkpoint inhibitors has evolved into a new pillar of cancer treatment in clinics, but dealing with treatment resistance (either primary or acquired) is a major challenge. The tumor microenvironment (TME) has a substantial impact on the pathological behaviors and treatment response of many cancers. The biophysical clues in TME have recently been considered as important characteristics of cancer. Furthermore, there is mounting evidence that biophysical cues in TME play important roles in each step of the cascade of cancer immunotherapy that synergistically contribute to immunotherapy resistance. In this review, we summarize five main biophysical cues in TME that affect resistance to immunotherapy: extracellular matrix (ECM) structure, ECM stiffness, tumor interstitial fluid pressure (IFP), solid stress, and vascular shear stress. First, the biophysical factors involved in anti-tumor immunity and therapeutic antibody delivery processes are reviewed. Then, the causes of these five biophysical cues and how they contribute to immunotherapy resistance are discussed. Finally, the latest treatment strategies that aim to improve immunotherapy efficacy by targeting these biophysical cues are shared. This review highlights the biophysical cues that lead to immunotherapy resistance, also supplements their importance in related technologies for studying TME biophysical cues in vitro and therapeutic strategies targeting biophysical cues to improve the effects of immunotherapy.
Collapse
Affiliation(s)
- Tian Zhang
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710061, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yuanbo Jia
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yang Yu
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710061, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710049, PR China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China.
| | - Hui Guo
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710061, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| |
Collapse
|
32
|
Sudarsanam H, Buhmann R, Henschler R. Influence of Culture Conditions on Ex Vivo Expansion of T Lymphocytes and Their Function for Therapy: Current Insights and Open Questions. Front Bioeng Biotechnol 2022; 10:886637. [PMID: 35845425 PMCID: PMC9277485 DOI: 10.3389/fbioe.2022.886637] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/16/2022] [Indexed: 01/03/2023] Open
Abstract
Ex vivo expansion of T lymphocytes is a central process in the generation of cellular therapies targeted at tumors and other disease-relevant structures, which currently cannot be reached by established pharmaceuticals. The influence of culture conditions on T cell functions is, however, incompletely understood. In clinical applications of ex vivo expanded T cells, so far, a relatively classical standard cell culture methodology has been established. The expanded cells have been characterized in both preclinical models and clinical studies mainly using a therapeutic endpoint, for example antitumor response and cytotoxic function against cellular targets, whereas the influence of manipulations of T cells ex vivo including transduction and culture expansion has been studied to a much lesser detail, or in many contexts remains unknown. This includes the circulation behavior of expanded T cells after intravenous application, their intracellular metabolism and signal transduction, and their cytoskeletal (re)organization or their adhesion, migration, and subsequent intra-tissue differentiation. This review aims to provide an overview of established T cell expansion methodologies and address unanswered questions relating in vivo interaction of ex vivo expanded T cells for cellular therapy.
Collapse
Affiliation(s)
| | | | - Reinhard Henschler
- Institute of Transfusion Medicine, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
33
|
Nguyen DT, Ogando-Rivas E, Liu R, Wang T, Rubin J, Jin L, Tao H, Sawyer WW, Mendez-Gomez HR, Cascio M, Mitchell DA, Huang J, Sawyer WG, Sayour EJ, Castillo P. CAR T Cell Locomotion in Solid Tumor Microenvironment. Cells 2022; 11:1974. [PMID: 35741103 PMCID: PMC9221866 DOI: 10.3390/cells11121974] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 01/25/2023] Open
Abstract
The promising outcomes of chimeric antigen receptor (CAR) T cell therapy in hematologic malignancies potentiates its capability in the fight against many cancers. Nevertheless, this immunotherapy modality needs significant improvements for the treatment of solid tumors. Researchers have incrementally identified limitations and constantly pursued better CAR designs. However, even if CAR T cells are armed with optimal killer functions, they must overcome and survive suppressive barriers imposed by the tumor microenvironment (TME). In this review, we will discuss in detail the important role of TME in CAR T cell trafficking and how the intrinsic barriers contribute to an immunosuppressive phenotype and cancer progression. It is of critical importance that preclinical models can closely recapitulate the in vivo TME to better predict CAR T activity. Animal models have contributed immensely to our understanding of human diseases, but the intensive care for the animals and unreliable representation of human biology suggest in vivo models cannot be the sole approach to CAR T cell therapy. On the other hand, in vitro models for CAR T cytotoxic assessment offer valuable insights to mechanistic studies at the single cell level, but they often lack in vivo complexities, inter-individual heterogeneity, or physiologically relevant spatial dimension. Understanding the advantages and limitations of preclinical models and their applications would enable more reliable prediction of better clinical outcomes.
Collapse
Affiliation(s)
- Duy T. Nguyen
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (D.T.N.); (W.W.S.); (W.G.S.)
| | - Elizabeth Ogando-Rivas
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - Ruixuan Liu
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - Theodore Wang
- College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Jacob Rubin
- Warrington College of Business, University of Florida, Gainesville, FL 32610, USA;
| | - Linchun Jin
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - Haipeng Tao
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - William W. Sawyer
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (D.T.N.); (W.W.S.); (W.G.S.)
- Department of Pediatrics, Division of Pediatric Hematology Oncology, University of Florida, Gainesville, FL 32610, USA;
| | - Hector R. Mendez-Gomez
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - Matthew Cascio
- Department of Pediatrics, Division of Pediatric Hematology Oncology, University of Florida, Gainesville, FL 32610, USA;
| | - Duane A. Mitchell
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - Jianping Huang
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
| | - W. Gregory Sawyer
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (D.T.N.); (W.W.S.); (W.G.S.)
| | - Elias J. Sayour
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (E.O.-R.); (R.L.); (L.J.); (H.T.); (H.R.M.-G.); (D.A.M.); (J.H.); (E.J.S.)
- Department of Pediatrics, Division of Pediatric Hematology Oncology, University of Florida, Gainesville, FL 32610, USA;
| | - Paul Castillo
- Department of Pediatrics, Division of Pediatric Hematology Oncology, University of Florida, Gainesville, FL 32610, USA;
| |
Collapse
|
34
|
Mechanical Forces Govern Interactions of Host Cells with Intracellular Bacterial Pathogens. Microbiol Mol Biol Rev 2022; 86:e0009420. [PMID: 35285720 PMCID: PMC9199418 DOI: 10.1128/mmbr.00094-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
To combat infectious diseases, it is important to understand how host cells interact with bacterial pathogens. Signals conveyed from pathogen to host, and vice versa, may be either chemical or mechanical. While the molecular and biochemical basis of host-pathogen interactions has been extensively explored, relatively less is known about mechanical signals and responses in the context of those interactions. Nevertheless, a wide variety of bacterial pathogens appear to have developed mechanisms to alter the cellular biomechanics of their hosts in order to promote their survival and dissemination, and in turn many host responses to infection rely on mechanical alterations in host cells and tissues to limit the spread of infection. In this review, we present recent findings on how mechanical forces generated by host cells can promote or obstruct the dissemination of intracellular bacterial pathogens. In addition, we discuss how in vivo extracellular mechanical signals influence interactions between host cells and intracellular bacterial pathogens. Examples of such signals include shear stresses caused by fluid flow over the surface of cells and variable stiffness of the extracellular matrix on which cells are anchored. We highlight bioengineering-inspired tools and techniques that can be used to measure host cell mechanics during infection. These allow for the interrogation of how mechanical signals can modulate infection alongside biochemical signals. We hope that this review will inspire the microbiology community to embrace those tools in future studies so that host cell biomechanics can be more readily explored in the context of infection studies.
Collapse
|
35
|
Göhring J, Schrangl L, Schütz GJ, Huppa JB. Mechanosurveillance: Tiptoeing T Cells. Front Immunol 2022; 13:886328. [PMID: 35693808 PMCID: PMC9178122 DOI: 10.3389/fimmu.2022.886328] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/19/2022] [Indexed: 11/28/2022] Open
Abstract
Efficient scanning of tissue that T cells encounter during their migratory life is pivotal to protective adaptive immunity. In fact, T cells can detect even a single antigenic peptide/MHC complex (pMHC) among thousands of structurally similar yet non-stimulatory endogenous pMHCs on the surface of antigen-presenting cells (APCs) or target cells. Of note, the glycocalyx of target cells, being composed of proteoglycans and bulky proteins, is bound to affect and even modulate antigen recognition by posing as a physical barrier. T cell-resident microvilli are actin-rich membrane protrusions that puncture through such barriers and thereby actively place the considerably smaller T-cell antigen receptors (TCRs) in close enough proximity to APC-presented pMHCs so that productive interactions may occur efficiently yet under force. We here review our current understanding of how the plasticity of T-cell microvilli and physicochemical properties of the glycocalyx may affect early events in T-cell activation. We assess insights gained from studies on T-cell plasma membrane ultrastructure and provide an update on current efforts to integrate biophysical aspects such as the amplitude and directionality of TCR-imposed mechanical forces and the distribution and lateral mobility of plasma membrane-resident signaling molecules into a more comprehensive view on sensitized T-cell antigen recognition.
Collapse
Affiliation(s)
- Janett Göhring
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Institute of Applied Physics, TU Wien, Vienna, Austria
- *Correspondence: Janett Göhring,
| | | | | | - Johannes B. Huppa
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
36
|
The interface between biochemical signaling and cell mechanics shapes T lymphocyte migration and activation. Eur J Cell Biol 2022; 101:151236. [DOI: 10.1016/j.ejcb.2022.151236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/06/2022] [Accepted: 05/07/2022] [Indexed: 11/18/2022] Open
|
37
|
Brameshuber M, Klotzsch E, Ponjavic A, Sezgin E. Understanding immune signaling using advanced imaging techniques. Biochem Soc Trans 2022; 50:853-866. [PMID: 35343569 PMCID: PMC9162467 DOI: 10.1042/bst20210479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 12/13/2022]
Abstract
Advanced imaging is key for visualizing the spatiotemporal regulation of immune signaling which is a complex process involving multiple players tightly regulated in space and time. Imaging techniques vary in their spatial resolution, spanning from nanometers to micrometers, and in their temporal resolution, ranging from microseconds to hours. In this review, we summarize state-of-the-art imaging methodologies and provide recent examples on how they helped to unravel the mysteries of immune signaling. Finally, we discuss the limitations of current technologies and share our insights on how to overcome these limitations to visualize immune signaling with unprecedented fidelity.
Collapse
Affiliation(s)
- Mario Brameshuber
- Institute of Applied Physics – Biophysics, TU Wien, 1040 Vienna, Austria
| | - Enrico Klotzsch
- Humboldt-Universität zu Berlin, Institut für Biophysik, Experimentelle Biophysik Mechanobiologie, Sitz Invalidenstrasse 42, 10115 Berlin, Germany
| | - Aleks Ponjavic
- School of Physics and Astronomy, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, U.K
- School of Food Science and Nutrition, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, U.K
| | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17165 Solna, Sweden
| |
Collapse
|
38
|
Vicar T, Chmelik J, Navratil J, Kolar R, Chmelikova L, Cmiel V, Jagos J, Provaznik I, Masarik M, Gumulec J. Cancer Cells Viscoelasticity Measurement by Quantitative Phase and Flow Stress Induction. Biophys J 2022; 121:1632-1642. [PMID: 35390297 PMCID: PMC9117928 DOI: 10.1016/j.bpj.2022.04.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 03/02/2022] [Accepted: 03/31/2022] [Indexed: 11/02/2022] Open
Abstract
Cell viscoelastic properties are affected by the cell cycle, differentiation, pathological processes such as malignant transformation. Therefore, evaluation of the mechanical properties of the cells proved to be an approach to obtaining information on the functional state of the cells. Most of the currently used methods for cell mechanophenotypisation are limited by low robustness or the need for highly expert operation. In this paper, the system and method for viscoelasticity measurement using shear stress induction by fluid flow is described and tested. Quantitative Phase Imaging (QPI) is used for image acquisition because this technique enables to quantify optical path length delays introduced by the sample, thus providing a label-free objective measure of morphology and dynamics. Viscosity and elasticity determination were refined using a new approach based on the linear system model and parametric deconvolution. The proposed method allows high-throughput measurements during live cell experiments and even through a time-lapse, where we demonstrated the possibility of simultaneous extraction of shear modulus, viscosity, cell morphology, and QPI-derived cell parameters like circularity or cell mass. Additionally, the proposed method provides a simple approach to measure cell refractive index with the same setup, which is required for reliable cell height measurement with QPI, an essential parameter for viscoelasticity calculation. Reliability of the proposed viscoelasticity measurement system was tested in several experiments including cell types of different Young/shear modulus and treatment with cytochalasin D or docetaxel, and an agreement with atomic force microscopy was observed. The applicability of the proposed approach was also confirmed by a time-lapse experiment with cytochalasin D washout, where an increase of stiffness corresponded to actin repolymerisation in time.
Collapse
Affiliation(s)
- Tomas Vicar
- Department of Biomedical Engineering, Faculty of Electrical Engineering and Communications, Brno University of Technology, Brno, Czech Republic; Department of Pathological Physiology, Masaryk University, Brno, Czech Republic
| | - Jiri Chmelik
- Department of Biomedical Engineering, Faculty of Electrical Engineering and Communications, Brno University of Technology, Brno, Czech Republic
| | - Jiri Navratil
- Department of Pathological Physiology, Masaryk University, Brno, Czech Republic
| | - Radim Kolar
- Department of Biomedical Engineering, Faculty of Electrical Engineering and Communications, Brno University of Technology, Brno, Czech Republic
| | - Larisa Chmelikova
- Department of Biomedical Engineering, Faculty of Electrical Engineering and Communications, Brno University of Technology, Brno, Czech Republic
| | - Vratislav Cmiel
- Department of Biomedical Engineering, Faculty of Electrical Engineering and Communications, Brno University of Technology, Brno, Czech Republic
| | - Jiri Jagos
- Dept. of Tissue Biomechanics and Numerical Modelling in Medicine, Faculty of Mechanical Engineering, Brno University of Technology, Brno, Czech Republic
| | - Ivo Provaznik
- Department of Biomedical Engineering, Faculty of Electrical Engineering and Communications, Brno University of Technology, Brno, Czech Republic
| | - Michal Masarik
- Department of Pathological Physiology, Masaryk University, Brno, Czech Republic
| | - Jaromir Gumulec
- Department of Pathological Physiology, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
39
|
Zak A, Dupré-Crochet S, Hudik E, Babataheri A, Barakat AI, Nüsse O, Husson J. Distinct timing of neutrophil spreading and stiffening during phagocytosis. Biophys J 2022; 121:1381-1394. [PMID: 35318004 PMCID: PMC9072703 DOI: 10.1016/j.bpj.2022.03.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 11/29/2021] [Accepted: 03/17/2022] [Indexed: 11/17/2022] Open
Abstract
Phagocytic cells form the first line of defense in an organism, engulfing microbial pathogens. Phagocytosis involves cell mechanical changes that are not yet well understood. Understanding these mechanical modifications promises to shed light on the immune processes that trigger pathological complications. Previous studies showed that phagocytes undergo a sequence of spreading events around their target followed by an increase in cell tension. Seemingly in contradiction, other studies observed an increase in cell tension concomitant with membrane expansion. Even though phagocytes are viscoelastic, few studies have quantified viscous changes during phagocytosis. It is also unclear whether cell lines behave mechanically similarly to primary neutrophils. We addressed the question of simultaneous versus sequential spreading and mechanical changes during phagocytosis by using immunoglobulin-G-coated 8- and 20-μm-diameter beads as targets. We used a micropipette-based single-cell rheometer to monitor viscoelastic properties during phagocytosis by both neutrophil-like PLB cells and primary human neutrophils. We show that the faster expansion of PLB cells on larger beads is a geometrical effect reflecting a constant advancing speed of the phagocytic cup. Cells become stiffer on 20- than on 8-μm beads, and the relative timing of spreading and stiffening of PLB cells depends on target size: on larger beads, stiffening starts before maximal spreading area is reached but ends after reaching maximal area. On smaller beads, the stiffness begins to increase after cells have engulfed the bead. Similar to PLB cells, primary cells become stiffer on larger beads but start spreading and stiffen faster, and the stiffening begins before the end of spreading on both bead sizes. Our results show that mechanical changes in phagocytes are not a direct consequence of cell spreading and that models of phagocytosis should be amended to account for causes of cell stiffening other than membrane expansion.
Collapse
Affiliation(s)
- Alexandra Zak
- LadHyX, CNRS, École polytechnique, Institut Polytechnique de Paris, Palaiseau, France; Institut de Chimie Physique, CNRS UMR 8000, Université Paris-Saclay, Orsay, France
| | - Sophie Dupré-Crochet
- Institut de Chimie Physique, CNRS UMR 8000, Université Paris-Saclay, Orsay, France
| | - Elodie Hudik
- Institut de Chimie Physique, CNRS UMR 8000, Université Paris-Saclay, Orsay, France
| | - Avin Babataheri
- LadHyX, CNRS, École polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Abdul I Barakat
- LadHyX, CNRS, École polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Oliver Nüsse
- Institut de Chimie Physique, CNRS UMR 8000, Université Paris-Saclay, Orsay, France
| | - Julien Husson
- LadHyX, CNRS, École polytechnique, Institut Polytechnique de Paris, Palaiseau, France.
| |
Collapse
|
40
|
Terada M, Ide S, Naito T, Kimura N, Matsusaki M, Kaji N. Label-Free Cancer Stem-like Cell Assay Conducted at a Single Cell Level Using Microfluidic Mechanotyping Devices. Anal Chem 2021; 93:14409-14416. [PMID: 34628861 DOI: 10.1021/acs.analchem.1c02316] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The mechanical phenotype of cells is an intrinsic property of individual cells. In fact, this property could serve as a label-free, non-destructive, diagnostic marker of the state of cells owing to its remarkable translational potential. A microfluidic device is a strong candidate for meeting the demand of this translational research as it can be used to diagnose a large population of cells at a single cell level in a high-throughput manner, without the need for off-line pretreatment operations. In this study, we investigated the mechanical phenotype of the human colon adenocarcinoma cell, HT29, which is known to be a heterogeneous cell line with both multipotency and self-renewal abilities. This type of cancer stem-like cell (CSC) is believed to be the unique originators of all tumor cells and may serve as the leading cause of cancer metastasis and drug resistance. By combining consecutive constrictions and microchannels with an ionic current sensing system, we found a high heterogeneity of cell deformability in the population of HT29 cells. Moreover, based on the level of aldehyde dehydrogenase (ALDH) activity and the expression level of CD44s, which are biochemical markers that suggest the multipotency of cells, the high heterogeneity of cell deformability was concluded to be a potential mechanical marker of CSCs. The development of label-free and non-destructive identification and collection techniques for CSCs has remarkable potential not only for cancer diagnosis and prognosis but also for the discovery of a new treatment for cancer.
Collapse
Affiliation(s)
- Miyu Terada
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Sachiko Ide
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Toyohiro Naito
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Niko Kimura
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Noritada Kaji
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.,Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Nagoya 464-8603, Japan
| |
Collapse
|
41
|
Fowell DJ, Kim M. The spatio-temporal control of effector T cell migration. Nat Rev Immunol 2021; 21:582-596. [PMID: 33627851 PMCID: PMC9380693 DOI: 10.1038/s41577-021-00507-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2021] [Indexed: 02/08/2023]
Abstract
Effector T cells leave the lymph nodes armed with specialized functional attributes. Their antigenic targets may be located anywhere in the body, posing the ultimate challenge: how to efficiently identify the target tissue, navigate through a complex tissue matrix and, ultimately, locate the immunological insult. Recent advances in real-time in situ imaging of effector T cell migratory behaviour have revealed a great degree of mechanistic plasticity that enables effector T cells to push and squeeze their way through inflamed tissues. This process is shaped by an array of 'stop' and 'go' guidance signals including target antigens, chemokines, integrin ligands and the mechanical cues of the inflamed microenvironment. Effector T cells must sense and interpret these competing signals to correctly position themselves to mediate their effector functions for complete and durable responses in infectious disease and malignancy. Tuning T cell migration therapeutically will require a new understanding of this complex decision-making process.
Collapse
Affiliation(s)
- Deborah J. Fowell
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute for Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY.,Department of Microbiology and Immunology, Cornell University, Ithaca, NY
| | - Minsoo Kim
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute for Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
42
|
Est-Witte SE, Livingston NK, Omotoso MO, Green JJ, Schneck JP. Nanoparticles for generating antigen-specific T cells for immunotherapy. Semin Immunol 2021; 56:101541. [PMID: 34922816 PMCID: PMC8900015 DOI: 10.1016/j.smim.2021.101541] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 12/25/2022]
Abstract
T cell therapy shows promise as an immunotherapy in both immunostimulatory and immunosuppressive applications. However, the forms of T cell-based therapy that are currently in the clinic, such as adoptive cell transfer and vaccines, are limited by cost, time-to-treatment, and patient variability. Nanoparticles offer a modular, universal platform to improve the efficacy of various T cell therapies as nanoparticle properties can be easily modified for enhanced cell targeting, organ targeting, and cell internalization. Nanoparticles can enhance or even replace endogenous cells during each step of generating an antigen-specific T cell response - from antigen presentation and T cell activation to T cell maintenance. In this review, we discuss the unique applications of nanoparticles for antigen-specific T cell therapy, focusing on nanoparticles as vaccines (to activate endogenous antigen presenting cells (APCs)), as artificial Antigen Presenting Cells (aAPCs, to directly activate T cells), and as drug delivery vehicles (to support activated T cells).
Collapse
Affiliation(s)
- Savannah E Est-Witte
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, USA, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Natalie K Livingston
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, USA, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD 21218, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Mary O Omotoso
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD 21218, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jordan J Green
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, USA, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD 21218, USA; Departments of Ophthalmology, Oncology, Neurosurgery, Materials Science & Engineering, and Chemical & Biomolecular Engineering, and The Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - Jonathan P Schneck
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD 21218, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Departments of Pathology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
43
|
Jung P, Zhou X, Iden S, Bischoff M, Qu B. T cell stiffness is enhanced upon formation of immunological synapse. eLife 2021; 10:66643. [PMID: 34313220 PMCID: PMC8360652 DOI: 10.7554/elife.66643] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 07/26/2021] [Indexed: 12/19/2022] Open
Abstract
T cells are activated by target cells via an intimate contact, termed immunological synapse (IS). Cellular mechanical properties, especially stiffness, are essential to regulate cell functions. However, T cell stiffness at a subcellular level at the IS still remains largely elusive. In this work, we established an atomic force microscopy (AFM)-based elasticity mapping method on whole T cells to obtain an overview of the stiffness with a resolution of ~60 nm. Using primary human CD4+ T cells, we show that when T cells form IS with stimulating antibody-coated surfaces, the lamellipodia are stiffer than the cell body. Upon IS formation, T cell stiffness is enhanced both at the lamellipodia and on the cell body. Chelation of intracellular Ca2+ abolishes IS-induced stiffening at the lamellipodia but has no influence on cell-body-stiffening, suggesting different regulatory mechanisms of IS-induced stiffening at the lamellipodia and the cell body.
Collapse
Affiliation(s)
- Philipp Jung
- Institute for Medical Microbiology and Hygiene, Saarland University, Homburg, Germany
| | - Xiangda Zhou
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Sandra Iden
- Cell and Developmental Biology, School of Medicine, Center of Human and Molecular Biology (ZHMB), Saarland University, Homburg, Germany
| | - Markus Bischoff
- Institute for Medical Microbiology and Hygiene, Saarland University, Homburg, Germany
| | - Bin Qu
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany.,Leibniz Institute for New Materials, Saarbrücken, Germany
| |
Collapse
|
44
|
Kim SHJ, Hammer DA. Integrin cross-talk modulates stiffness-independent motility of CD4+ T lymphocytes. Mol Biol Cell 2021; 32:1749-1757. [PMID: 34232700 PMCID: PMC8684734 DOI: 10.1091/mbc.e21-03-0131] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
To carry out their physiological responsibilities, CD4+ T lymphocytes interact with various tissues of different mechanical properties. Recent studies suggest that T cells migrate upstream on surfaces expressing intracellular adhesion molecule-1 (ICAM-1) through interaction with leukocyte function-associated antigen-1 (αLβ2) (LFA-1) integrins. LFA-1 likely behaves as a mechanosensor, and thus we hypothesized that substrate mechanics might affect the ability of LFA-1 to support upstream migration of T cells under flow. Here we measured motility of CD4+ T lymphocytes on polyacrylamide gels with predetermined stiffnesses containing ICAM-1, vascular cell adhesion molecule-1 (VCAM-1), or a 1:1 mixture of VCAM-1/ICAM-1. Under static conditions, we found that CD4+ T cells exhibit an increase in motility on ICAM-1, but not on VCAM-1 or VCAM-1/ICAM-1 mixed, surfaces as a function of matrix stiffness. The mechanosensitivity of T-cell motility on ICAM-1 is overcome when VLA-4 (very late antigen-4 [α4β1]) is ligated with soluble VCAM-1. Last, we observed that CD4+ T cells migrate upstream under flow on ICAM-1-functionalized hydrogels, independent of substrate stiffness. In summary, we show that CD4+ T cells under no flow respond to matrix stiffness through LFA-1, and that the cross-talk of VLA-4 and LFA-1 can compensate for deformable substrates. Interestingly, CD4+ T lymphocytes migrated upstream on ICAM-1 regardless of the substrate stiffness, suggesting that flow can compensate for substrate stiffness.
Collapse
Affiliation(s)
- Sarah Hyun Ji Kim
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104
| | - Daniel A Hammer
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
45
|
Abstract
T cell activation is a critical event in the adaptive immune response, indispensable for cell-mediated and humoral immunity as well as for immune regulation. Recent years have witnessed an emerging trend emphasizing the essential role that physical force and mechanical properties play at the T cell interface. In this review, we integrate current knowledge of T cell antigen recognition and the different models of T cell activation from the perspective of mechanobiology, focusing on the interaction between the T cell receptor (TCR) and the peptide-major histocompatibility complex (pMHC) antigen. We address the shortcomings of TCR affinity alone in explaining T cell functional outcomes and the rising status of force-regulated TCR bond lifetimes, most notably the TCR catch bond. Ultimately, T cell activation and the ensuing physiological responses result from mechanical interaction between TCRs and the pMHC. Expected final online publication date for the Annual Review of Cell and Developmental Biology, Volume 37 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Baoyu Liu
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112, USA; , ,
| | - Elizabeth M Kolawole
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112, USA; , ,
| | - Brian D Evavold
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112, USA; , ,
| |
Collapse
|
46
|
Porro C, Pennella A, Panaro MA, Trotta T. Functional Role of Non-Muscle Myosin II in Microglia: An Updated Review. Int J Mol Sci 2021; 22:ijms22136687. [PMID: 34206505 PMCID: PMC8267657 DOI: 10.3390/ijms22136687] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 02/08/2023] Open
Abstract
Myosins are a remarkable superfamily of actin-based motor proteins that use the energy derived from ATP hydrolysis to translocate actin filaments and to produce force. Myosins are abundant in different types of tissues and involved in a large variety of cellular functions. Several classes of the myosin superfamily are expressed in the nervous system; among them, non-muscle myosin II (NM II) is expressed in both neurons and non-neuronal brain cells, such as astrocytes, oligodendrocytes, endothelial cells, and microglia. In the nervous system, NM II modulates a variety of functions, such as vesicle transport, phagocytosis, cell migration, cell adhesion and morphology, secretion, transcription, and cytokinesis, as well as playing key roles during brain development, inflammation, repair, and myelination functions. In this review, we will provide a brief overview of recent emerging roles of NM II in resting and activated microglia cells, the principal regulators of immune processes in the central nervous system (CNS) in both physiological and pathological conditions. When stimulated, microglial cells react and produce a number of mediators, such as pro-inflammatory cytokines, free radicals, and nitric oxide, that enhance inflammation and contribute to neurodegenerative diseases. Inhibition of NM II could be a new therapeutic target to treat or to prevent CNS diseases.
Collapse
Affiliation(s)
- Chiara Porro
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (C.P.); (A.P.)
| | - Antonio Pennella
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (C.P.); (A.P.)
| | - Maria Antonietta Panaro
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125 Bari, Italy;
| | - Teresa Trotta
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (C.P.); (A.P.)
- Correspondence:
| |
Collapse
|
47
|
Abdollahiyan P, Oroojalian F, Baradaran B, de la Guardia M, Mokhtarzadeh A. Advanced mechanotherapy: Biotensegrity for governing metastatic tumor cell fate via modulating the extracellular matrix. J Control Release 2021; 335:596-618. [PMID: 34097925 DOI: 10.1016/j.jconrel.2021.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/19/2022]
Abstract
Mechano-transduction is the procedure of mechanical stimulus translation via cells, among substrate shear flow, topography, and stiffness into a biochemical answer. TAZ and YAP are transcriptional coactivators which are recognized as relay proteins that promote mechano-transduction within the Hippo pathway. With regard to healthy cells in homeostasis, mechano-transduction regularly restricts proliferation, and TAZ and YAP are totally inactive. During cancer development a YAP/TAZ - stimulating positive response loop is formed between the growing tumor and the stiffening ECM. As tumor developments, local stromal and cancerous cells take advantage of mechanotransduction to enhance proliferation, induce their migratory into remote tissues, and promote chemotherapeutic resistance. As a newly progresses paradigm, nanoparticle-conjunctions (such as magnetic nanoparticles, and graphene derivatives nanoparticles) hold significant promises for remote regulation of cells and their relevant events at molecular scale. Despite outstanding developments in employing nanoparticles for drug targeting studies, the role of nanoparticles on cellular behaviors (proliferation, migration, and differentiation) has still required more evaluations in the field of mechanotherapy. In this paper, the in-depth contribution of mechano-transduction is discussed during tumor progression, and how these consequences can be evaluated in vitro.
Collapse
Affiliation(s)
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Miguel de la Guardia
- Department of Analytical Chemistry, University of Valencia, Dr. Moliner 50, 46100 Burjassot, Valencia, Spain
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
48
|
Li Y, Tang W, Guo M. The Cell as Matter: Connecting Molecular Biology to Cellular Functions. MATTER 2021; 4:1863-1891. [PMID: 35495565 PMCID: PMC9053450 DOI: 10.1016/j.matt.2021.03.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Viewing cell as matter to understand the intracellular biomolecular processes and multicellular tissue behavior represents an emerging research area at the interface of physics and biology. Cellular material displays various physical and mechanical properties, which can strongly affect both intracellular and multicellular biological events. This review provides a summary of how cells, as matter, connect molecular biology to cellular and multicellular scale functions. As an impact in molecular biology, we review recent progresses in utilizing cellular material properties to direct cell fate decisions in the communities of immune cells, neurons, stem cells, and cancer cells. Finally, we provide an outlook on how to integrate cellular material properties in developing biophysical methods for engineered living systems, regenerative medicine, and disease treatments.
Collapse
Affiliation(s)
- Yiwei Li
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Wenhui Tang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ming Guo
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
49
|
Bhingardive V, Kossover A, Iraqi M, Khand B, Le Saux G, Porgador A, Schvartzman M. Antibody-Functionalized Nanowires: A Tuner for the Activation of T Cells. NANO LETTERS 2021; 21:4241-4248. [PMID: 33989498 DOI: 10.1021/acs.nanolett.1c00245] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
T cells sense both chemical cues delivered by antigen molecules and physical cues delivered by the environmental elasticity and topography; yet, it is still largely unclear how these cues cumulatively regulate the immune activity of T cells. Here, we engineered a nanoscale platform for ex vivo stimulation of T cells based on antigen-functionalized nanowires. The nanowire topography and elasticity, as well as the immobilized antigens, deliver the physical and chemical cues, respectively, enabling the systematic study of the integrated effect of these cues on a T cell's immune response. We found that T cells sense both the topography and bending modulus of the nanowires and modulate their signaling, degranulation, and cytotoxicity with the variation in these physical features. Our study provides an important insight into the physical mechanism of T cell activation and paves the way to novel nanomaterials for the controlled ex vivo activation of T cells in immunotherapy.
Collapse
|
50
|
Alibert C, Pereira D, Lardier N, Etienne-Manneville S, Goud B, Asnacios A, Manneville JB. Multiscale rheology of glioma cells. Biomaterials 2021; 275:120903. [PMID: 34102526 DOI: 10.1016/j.biomaterials.2021.120903] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 04/08/2021] [Accepted: 05/19/2021] [Indexed: 12/30/2022]
Abstract
Cells tend to soften during cancer progression, suggesting that mechanical phenotyping could be used as a diagnostic or prognostic method. Here we investigate the cell mechanics of gliomas, brain tumors that originate from glial cells or glial progenitors. Using two microrheology techniques, a single-cell parallel plates rheometer to probe whole-cell mechanics and optical tweezers to probe intracellular rheology, we show that cell mechanics discriminates human glioma cells of different grades. When probed globally, grade IV glioblastoma cells are softer than grade III astrocytoma cells, while they are surprisingly stiffer at the intracellular level. We explain this difference between global and local intracellular behaviours by changes in the composition and spatial organization of the cytoskeleton, and by changes in nuclear mechanics. Our study highlights the need to combine rheology techniques for potential diagnostic or prognostic methods based on cancer cell mechanophenotyping.
Collapse
Affiliation(s)
- Charlotte Alibert
- Institut Curie, PSL Research University, CNRS, UMR 144, 26 Rue D'Ulm, F-75005, Paris, France; Sorbonne Universités, UPMC University Paris 06, CNRS, UMR 144, 26 Rue D'Ulm, F-75005, Paris, France
| | - David Pereira
- Institut Curie, PSL Research University, CNRS, UMR 144, 26 Rue D'Ulm, F-75005, Paris, France; Laboratoire Matières et Systèmes Complexes, Université de Paris, CNRS, UMR7057, Université Paris-Diderot, 10 Rue Alice Domon et Léonie Duquet, F-75013, Paris, France
| | - Nathan Lardier
- Institut Curie, PSL Research University, CNRS, UMR 144, 26 Rue D'Ulm, F-75005, Paris, France; Sorbonne Universités, UPMC University Paris 06, CNRS, UMR 144, 26 Rue D'Ulm, F-75005, Paris, France
| | - Sandrine Etienne-Manneville
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, F-75015, Paris, France
| | - Bruno Goud
- Institut Curie, PSL Research University, CNRS, UMR 144, 26 Rue D'Ulm, F-75005, Paris, France; Sorbonne Universités, UPMC University Paris 06, CNRS, UMR 144, 26 Rue D'Ulm, F-75005, Paris, France
| | - Atef Asnacios
- Laboratoire Matières et Systèmes Complexes, Université de Paris, CNRS, UMR7057, Université Paris-Diderot, 10 Rue Alice Domon et Léonie Duquet, F-75013, Paris, France
| | - Jean-Baptiste Manneville
- Institut Curie, PSL Research University, CNRS, UMR 144, 26 Rue D'Ulm, F-75005, Paris, France; Sorbonne Universités, UPMC University Paris 06, CNRS, UMR 144, 26 Rue D'Ulm, F-75005, Paris, France.
| |
Collapse
|