1
|
Lee JH, Shin SJ, Lee JH, Knowles JC, Lee HH, Kim HW. Adaptive immunity of materials: Implications for tissue healing and regeneration. Bioact Mater 2024; 41:499-522. [PMID: 39206299 PMCID: PMC11350271 DOI: 10.1016/j.bioactmat.2024.07.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/16/2024] [Accepted: 07/21/2024] [Indexed: 09/04/2024] Open
Abstract
Recent cumulative findings signify the adaptive immunity of materials as a key agenda in tissue healing that can improve regenerative events and outcomes. Modulating immune responses, mainly the recruitment and functions of T and B cells and their further interplay with innate immune cells (e.g., dendritic cells, macrophages) can be orchestrated by materials. For instance, decellularized matrices have been shown to promote muscle healing by inducing T helper 2 (Th2) cell immunity, while synthetic biopolymers exhibit differential effects on B cell responses and fibrosis compared decellularized matrices. We discuss the recent findings on how implantable materials instruct the adaptive immune events and the subsequent tissue healing process. In particular, we dissect the materials' physicochemical properties (shape, size, topology, degradation, rigidity, and matrix dynamic mechanics) to demonstrate the relations of these parameters with the adaptive immune responses in vitro and the underlying biological mechanisms. Furthermore, we present evidence of recent in vivo phenomena, including tissue healing, cancer progression, and fibrosis, wherein biomaterials potentially shape adaptive immune cell functions and in vivo outcomes. Our discussion will help understand the materials-regulated immunology events more deeply, and offer the design rationale of materials with tunable matrix properties for accelerated tissue repair and regeneration.
Collapse
Affiliation(s)
- Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
| | - Seong-Jin Shin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
| | - Jun Hee Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
| | - Jonathan C. Knowles
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman Dental Institute, University College London, London NW3 2PX, United Kingdom
| | - Hae-Hyoung Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
2
|
Engel L, Zaoralová M, Zhou M, Dunn AR, Oliver SL. Extracellular filaments revealed by affinity capture cryo-electron tomography. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.05.552110. [PMID: 37577490 PMCID: PMC10418515 DOI: 10.1101/2023.08.05.552110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Cryogenic-electron tomography (cryo-ET) has provided an unprecedented glimpse into the nanoscale architecture of cells by combining cryogenic preservation of biological structures with electron tomography. Micropatterning of extracellular matrix proteins is increasingly used as a method to prepare adherent cell types for cryo-ET as it promotes optimal positioning of cells and subcellular regions of interest for vitrification, cryo-focused ion beam (cryo-FIB) milling, and data acquisition. Here we demonstrate a micropatterning workflow for capturing minimally adherent cell types, human T-cells and Jurkat cells, for cryo-FIB and cryo-ET. Our affinity capture system facilitated the nanoscale imaging of Jurkat cells, revealing extracellular filamentous structures. It improved workflow efficiency by consistently producing grids with a sufficient number of well positioned cells for an entire cryo-FIB session. Affinity capture can be extended to facilitate high resolution imaging of other adherent and non-adherent cell types with cryo-ET.
Collapse
Affiliation(s)
- Leeya Engel
- Dept. of Chemical Engineering, Stanford University, Stanford, CA, USA 94305
- Faculty of Mechanical Engineering, Technion - Israel Institute of Technology, Haifa, Israel 3200003
| | - Magda Zaoralová
- Dept. of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA 94305
| | - Momei Zhou
- Dept. of Pediatrics, Division of Infectious Diseases, Stanford School of Medicine, Biomedical Innovations 240 Pasteur Drive, Stanford, CA, USA 94305
| | - Alexander R. Dunn
- Dept. of Chemical Engineering, Stanford University, Stanford, CA, USA 94305
| | - Stefan L. Oliver
- Dept. of Pediatrics, Division of Infectious Diseases, Stanford School of Medicine, Biomedical Innovations 240 Pasteur Drive, Stanford, CA, USA 94305
| |
Collapse
|
3
|
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
|
4
|
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: 3.0] [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
|
5
|
Sun Y, Sun J, Xiao M, Lai W, Li L, Fan C, Pei H. DNA origami-based artificial antigen-presenting cells for adoptive T cell therapy. SCIENCE ADVANCES 2022; 8:eadd1106. [PMID: 36459554 PMCID: PMC10936057 DOI: 10.1126/sciadv.add1106] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 10/17/2022] [Indexed: 06/17/2023]
Abstract
Nanosized artificial antigen-presenting cells (aAPCs) with efficient signal presentation hold great promise for in vivo adoptive cell therapy. Here, we used DNA origami nanostructures as two-dimensional scaffolds to regulate the spatial presentation of activating ligands at nanoscale to construct high-effective aAPCs. The DNA origami-based aAPC comprises costimulatory ligands anti-CD28 antibody anchored at three vertices and T cell receptor (TCR) ligands peptide-major histocompatibility complex (pMHC) anchored at three edges with varying density. The DNA origami scaffold enables quantitative analysis of ligand-receptor interactions in T cell activation at the single-particle, single-molecule resolution. The pMHC-TCR-binding dwell time is increased from 9.9 to 12.1 s with increasing pMHC density, driving functional T cell responses. In addition, both in vitro and in vivo assays demonstrate that the optimized DNA origami-based aAPCs show effective tumor growth inhibiting capability in adoptive immunotherapy. These results provide important insights into the rational design of molecular vaccines for cancer immunotherapy.
Collapse
Affiliation(s)
- Yueyang Sun
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Jiajia Sun
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Mingshu Xiao
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Wei Lai
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Li Li
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, and Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hao Pei
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| |
Collapse
|
6
|
Qu Y, Shen F, Zhang Z, Wang Q, Huang H, Xu Y, Li Q, Zhu X, Sun L. Applications of Functional DNA Materials in Immunomodulatory Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:45079-45095. [PMID: 36171537 DOI: 10.1021/acsami.2c13768] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
In recent years, nanoscale or microscale functional materials derived from DNA have shown great potential for immunotherapy as superior delivery carriers. DNA nanostructures with excellent programmability and addressability enable the precise assembly of molecules or nanoparticles. DNA hydrogels have predictable structures and adjustable mechanical strength, thus being advantageous in controllable release of cargos. In addition, utilizing systematic evolution of ligands by exponential enrichment technology, a variety of DNA aptamers have been screened for specific recognition of ions, molecules, and even cells. Moreover, a wide variety of chemical modifications can further enrich the function of DNA. The unique advantages of functional DNA materials make them extremely attractive in immunomodulation. Recently, functional DNA materials-based immunotherapy has shown great potential in fighting against many diseases like cancer, viral infection, and inflammation. Therefore, in this review, we focus on discussing the progress of the applications of functional DNA materials in immunotherapy; before that, we also summarize the characteristics of the functional DNA materials descried above. Finally, we discuss the challenges and future opportunities of functional DNA materials in immunomodulatory therapy.
Collapse
Affiliation(s)
- Yanfei Qu
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Fengyun Shen
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ziyi Zhang
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Qi Wang
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Hao Huang
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Yufei Xu
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Qian Li
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaoli Zhu
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Lele Sun
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| |
Collapse
|
7
|
Zhang K, Ma Y, Wang D, Liu J, An J, Li Y, Ma C, Pei Y, Zhang Z, Liu J, Shi J. In Vivo Activation of T-Cell Proliferation by Regulating Cell Surface Receptor Clustering Using a pH-Driven Interlocked DNA Nano-Spring. NANO LETTERS 2022; 22:1937-1945. [PMID: 35225623 DOI: 10.1021/acs.nanolett.1c04562] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Activation of T-cell proliferation specifically in a tumor is crucial for reducing the autoimmune side effects of antitumor immunotherapy. Herein, we developed a pH-driven interlocked DNA nano-spring (iDNS) to stimulate T-cell activation in vivo in response to the low pH value in a tumor microenvironment. The interlocked structure of iDNS provide a more rigid scaffold in comparison to double-stranded DNA for ligand assembly, which can help to control the spatial distribution of ligands with more accuracy. We have demonstrated that the pH-driven reversible reconfiguration of iDNS provides a powerful way to regulate the nanoscale distribution of T-cell receptors (CD3) on the cell surface. The relatively low pH value (pH 6.5) in a solid tumor was able to drive the springlike shrinking of iDNS and induce significant T-cell proliferation, leading to an enhanced antitumor effect, thus providing a tool for specifically inducing an immune response in a tumor for immunotherapy.
Collapse
Affiliation(s)
- Kaixiang Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| | - Yanrui Ma
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| | - Danyu Wang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| | - Jingwen Liu
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| | - Jingyi An
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| | - Yanan Li
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| | - Chihong Ma
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| | - Yiran Pei
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| | - Junjie Liu
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| |
Collapse
|
8
|
Schluck M, Eggermont LJ, Weiden J, Popelier C, Weiss L, Pilzecker B, Kolder S, Heinemans A, Rodriguez Mogeda C, Verdoes M, Figdor CG, Hammink R. Dictating Phenotype, Function, and Fate of Human T Cells with Co‐Stimulatory Antibodies Presented by Filamentous Immune Cell Mimics. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200019] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Marjolein Schluck
- Department of Tumor Immunology Radboud Institute for Molecular Life Sciences Radboud University Medical Center Geert Grooteplein 26 Nijmegen GA 6525 The Netherlands
| | - Loek J. Eggermont
- Department of Tumor Immunology Radboud Institute for Molecular Life Sciences Radboud University Medical Center Geert Grooteplein 26 Nijmegen GA 6525 The Netherlands
| | - Jorieke Weiden
- Department of Tumor Immunology Radboud Institute for Molecular Life Sciences Radboud University Medical Center Geert Grooteplein 26 Nijmegen GA 6525 The Netherlands
| | - Carlijn Popelier
- Department of Tumor Immunology Radboud Institute for Molecular Life Sciences Radboud University Medical Center Geert Grooteplein 26 Nijmegen GA 6525 The Netherlands
| | - Lea Weiss
- Department of Tumor Immunology Radboud Institute for Molecular Life Sciences Radboud University Medical Center Geert Grooteplein 26 Nijmegen GA 6525 The Netherlands
| | - Bas Pilzecker
- Department of Tumor Immunology Radboud Institute for Molecular Life Sciences Radboud University Medical Center Geert Grooteplein 26 Nijmegen GA 6525 The Netherlands
| | - Sigrid Kolder
- Department of Tumor Immunology Radboud Institute for Molecular Life Sciences Radboud University Medical Center Geert Grooteplein 26 Nijmegen GA 6525 The Netherlands
| | - Anne Heinemans
- Department of Tumor Immunology Radboud Institute for Molecular Life Sciences Radboud University Medical Center Geert Grooteplein 26 Nijmegen GA 6525 The Netherlands
| | - Carla Rodriguez Mogeda
- Department of Tumor Immunology Radboud Institute for Molecular Life Sciences Radboud University Medical Center Geert Grooteplein 26 Nijmegen GA 6525 The Netherlands
| | - Martijn Verdoes
- Department of Tumor Immunology Radboud Institute for Molecular Life Sciences Radboud University Medical Center Geert Grooteplein 26 Nijmegen GA 6525 The Netherlands
| | - Carl G. Figdor
- Department of Tumor Immunology Radboud Institute for Molecular Life Sciences Radboud University Medical Center Geert Grooteplein 26 Nijmegen GA 6525 The Netherlands
| | - Roel Hammink
- Department of Tumor Immunology Radboud Institute for Molecular Life Sciences Radboud University Medical Center Geert Grooteplein 26 Nijmegen GA 6525 The Netherlands
| |
Collapse
|
9
|
Sachar C, Kam LC. Probing T Cell 3D Mechanosensing With Magnetically-Actuated Structures. Front Immunol 2021; 12:704693. [PMID: 34566962 PMCID: PMC8458571 DOI: 10.3389/fimmu.2021.704693] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/23/2021] [Indexed: 12/02/2022] Open
Abstract
The ability of cells to recognize and respond to the mechanical properties of their environment is of increasing importance in T cell physiology. However, initial studies in this direction focused on planar hydrogel and elastomer surfaces, presenting several challenges in interpretation including difficulties in separating mechanical stiffness from changes in chemistry needed to modulate this property. We introduce here the use of magnetic fields to change the structural rigidity of microscale elastomer pillars loaded with superparamagnetic nanoparticles, independent of substrate chemistry. This magnetic modulation of rigidity, embodied as the pillar spring constant, changed the interaction of mouse naïve CD4+ T cells from a contractile morphology to one involving deep embedding into the array. Furthermore, increasing spring constant was associated with higher IL-2 secretion, showing a functional impact on mechanosensing. The system introduced here thus separates local substrate stiffness and long-range structural rigidity, revealing new facets of T cell interaction with their environment.
Collapse
Affiliation(s)
- Chirag Sachar
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Lance C Kam
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| |
Collapse
|
10
|
Leithner A, Altenburger LM, Hauschild R, Assen FP, Rottner K, Stradal TEB, Diz-Muñoz A, Stein JV, Sixt M. Dendritic cell actin dynamics control contact duration and priming efficiency at the immunological synapse. J Cell Biol 2021; 220:211749. [PMID: 33533935 PMCID: PMC7863705 DOI: 10.1083/jcb.202006081] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 11/25/2020] [Accepted: 01/12/2021] [Indexed: 01/22/2023] Open
Abstract
Dendritic cells (DCs) are crucial for the priming of naive T cells and the initiation of adaptive immunity. Priming is initiated at a heterologous cell–cell contact, the immunological synapse (IS). While it is established that F-actin dynamics regulates signaling at the T cell side of the contact, little is known about the cytoskeletal contribution on the DC side. Here, we show that the DC actin cytoskeleton is decisive for the formation of a multifocal synaptic structure, which correlates with T cell priming efficiency. DC actin at the IS appears in transient foci that are dynamized by the WAVE regulatory complex (WRC). The absence of the WRC in DCs leads to stabilized contacts with T cells, caused by an increase in ICAM1-integrin–mediated cell–cell adhesion. This results in lower numbers of activated and proliferating T cells, demonstrating an important role for DC actin in the regulation of immune synapse functionality.
Collapse
Affiliation(s)
- Alexander Leithner
- Institute of Science and Technology Austria, Klosterneuburg, Austria.,Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Lukas M Altenburger
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Robert Hauschild
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Frank P Assen
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Klemens Rottner
- Zoological Institute, Technical University Braunschweig, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Theresia E B Stradal
- Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Alba Diz-Muñoz
- Cell Biology and Biophysics Units, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Michael Sixt
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| |
Collapse
|
11
|
Rodríguez-Fernández JL, Criado-García O. The Actin Cytoskeleton at the Immunological Synapse of Dendritic Cells. Front Cell Dev Biol 2021; 9:679500. [PMID: 34409027 PMCID: PMC8366227 DOI: 10.3389/fcell.2021.679500] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 07/05/2021] [Indexed: 01/02/2023] Open
Abstract
Dendritic cells (DCs) are considered the most potent antigen-presenting cells. DCs control the activation of T cells (TCs) in the lymph nodes. This process involves forming a specialized superstructure at the DC-TC contact zone called the immunological synapse (IS). For the sake of clarity, we call IS(DC) and IS(TC) the DC and TC sides of the IS, respectively. The IS(DC) and IS(TC) seem to organize as multicentric signaling hubs consisting of surface proteins, including adhesion and costimulatory molecules, associated with cytoplasmic components, which comprise cytoskeletal proteins and signaling molecules. Most of the studies on the IS have focused on the IS(TC), and the information on the IS(DC) is still sparse. However, the data available suggest that both IS sides are involved in the control of TC activation. The IS(DC) may govern activities of DCs that confer them the ability to activate the TCs. One key component of the IS(DC) is the actin cytoskeleton. Herein, we discuss experimental data that support the concept that actin polarized at the IS(DC) is essential to maintaining IS stability necessary to induce TC activation.
Collapse
Affiliation(s)
- José Luis Rodríguez-Fernández
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | | |
Collapse
|
12
|
Zhovmer AS, Chandler M, Manning A, Afonin KA, Tabdanov ED. Programmable DNA-augmented hydrogels for controlled activation of human lymphocytes. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 37:102442. [PMID: 34284132 DOI: 10.1016/j.nano.2021.102442] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/25/2021] [Accepted: 07/07/2021] [Indexed: 12/25/2022]
Abstract
Contractile forces within the planar interface between T cell and antigen-presenting surface mechanically stimulate T cell receptors (TCR) in the mature immune synapses. However, the origin of mechanical stimulation during the initial, i.e., presynaptic, microvilli-based TCR activation in the course of immune surveillance remains unknown and new tools to help address this problem are needed. In this work, we develop nucleic acid nanoassembly (NAN)-based technology for functionalization of hydrogels using isothermal toehold-mediated reassociation of RNA/DNA heteroduplexes. Resulting platform allows for regulation with NAN linkers of 3D force momentum along the TCR mechanical axis, whereas hydrogels contribute to modulation of 2D shear modulus. By utilizing different lengths of NAN linkers conjugated to polyacrylamide gels of different shear moduli, we demonstrate an efficient capture of human T lymphocytes and tunable activation of TCR, as confirmed by T-cell spreading and pY foci.
Collapse
Affiliation(s)
- Alexander S Zhovmer
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA.
| | - Morgan Chandler
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Alexis Manning
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Kirill A Afonin
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, USA.
| | - Erdem D Tabdanov
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA.
| |
Collapse
|
13
|
Nassereddine A, Abdelrahman A, Benard E, Bedu F, Ozerov I, Limozin L, Sengupta K. Ligand Nanocluster Array Enables Artificial-Intelligence-Based Detection of Hidden Features in T-Cell Architecture. NANO LETTERS 2021; 21:5606-5613. [PMID: 34170136 DOI: 10.1021/acs.nanolett.1c01073] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Protein patterning has emerged as a powerful means to interrogate adhering cells. However, the tools to apply a sub-micrometer periodic stimulus and the analysis of the response are still being standardized. We propose a technique combining electron beam lithography and surface functionalization to fabricate nanopatterns compatible with advanced imaging. The repetitive pattern enables a deep-learning algorithm to reveal that T cells organize their membrane and actin network differently depending upon whether the ligands are clustered or homogeneously distributed, an effect invisible to the unassisted human eye even after extensive image analysis. This fabrication and analysis toolbox should be useful, both together and separately, for exploring general correlation between a spatially structured subcellular stimulation and a subtle cellular response.
Collapse
Affiliation(s)
- Aya Nassereddine
- Aix Marseille Univ, CNRS, CINAM, 13009 Marseille, France
- Aix Marseille Univ, CNRS, INSERM, LAI, Turing Centre for Living Systems, 13009 Marseille, France
| | - Ahmed Abdelrahman
- Aix Marseille Univ, CNRS, CINAM, 13009 Marseille, France
- Aix Marseille Univ, CNRS, INSERM, LAI, Turing Centre for Living Systems, 13009 Marseille, France
| | | | - Frederic Bedu
- Aix Marseille Univ, CNRS, CINAM, 13009 Marseille, France
| | - Igor Ozerov
- Aix Marseille Univ, CNRS, CINAM, 13009 Marseille, France
| | - Laurent Limozin
- Aix Marseille Univ, CNRS, INSERM, LAI, Turing Centre for Living Systems, 13009 Marseille, France
| | - Kheya Sengupta
- Aix Marseille Univ, CNRS, CINAM, 13009 Marseille, France
| |
Collapse
|
14
|
Yuan DJ, Shi L, Kam LC. Biphasic response of T cell activation to substrate stiffness. Biomaterials 2021; 273:120797. [PMID: 33878536 DOI: 10.1016/j.biomaterials.2021.120797] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/28/2021] [Accepted: 03/29/2021] [Indexed: 01/01/2023]
Abstract
T cell activation is sensitive to the mechanical properties of an activating substrate. However, there are also contrasting results on how substrate stiffness affects T cell activation, including differences between T cells of mouse and human origin. Towards reconciling these differences, this report examines the response of primary human T cells to polyacrylamide gels with stiffness between 5 and 110 kPa presenting activating antibodies to CD3 and CD28. T cell proliferation and IL-2 secretion exhibited a biphasic functional response to substrate stiffness, which can be shifted by changing density of activating antibodies and abrogated by inhibition of cellular contractility. T cell morphology was modulated by stiffness at early time points. RNA-seq indicates that T cells show differing monotonic trends in upregulated genes and pathways towards both ends of the stiffness spectrum. These studies provide a framework of T cell mechanosensing and suggest an effect of ligand density that may reconcile different, contrasting patterns of stiffness sensing seen in previous studies.
Collapse
Affiliation(s)
- Dennis J Yuan
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Lingting Shi
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Lance C Kam
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA.
| |
Collapse
|
15
|
Lee JH, Shao S, Kim M, Fernandes SM, Brown JR, Kam LC. Multi-Factor Clustering Incorporating Cell Motility Predicts T Cell Expansion Potential. Front Cell Dev Biol 2021; 9:648925. [PMID: 33898440 PMCID: PMC8063612 DOI: 10.3389/fcell.2021.648925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 03/18/2021] [Indexed: 11/19/2022] Open
Abstract
Expansion of an initial population of T cells is essential for cellular immunotherapy. In Chronic Lymphocytic Leukemia (CLL), expansion is often complicated by lack of T cell proliferation, as these cells frequently show signs of exhaustion. This report seeks to identify specific biomarkers or measures of cell function that capture the proliferative potential of a starting population of cells. Mixed CD4+/CD8+ T cells from healthy donors and individuals previously treated for CLL were characterized on the basis of proliferative potential and in vitro cellular functions. Single-factor analysis found little correlation between the number of populations doublings reached during expansion and either Rai stage (a clinical measure of CLL spread) or PD-1 expression. However, inclusion of in vitro IL-2 secretion and the propensity of cells to align onto micropatterned features of activating proteins as factors identified three distinct groups of donors. Notably, these group assignments provided an elegant separation of donors with regards to proliferative potential. Furthermore, these groups exhibited different motility characteristics, suggesting a mechanism that underlies changes in proliferative potential. This study describes a new set of functional readouts that augment surface marker panels to better predict expansion outcomes and clinical prognosis.
Collapse
Affiliation(s)
- Joanne H Lee
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Shuai Shao
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Michelle Kim
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Stacey M Fernandes
- Department of Medical Oncology, Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Jennifer R Brown
- Department of Medical Oncology, Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Lance C Kam
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| |
Collapse
|
16
|
Lei Q, Guo J, Kong F, Cao J, Wang L, Zhu W, Brinker CJ. Bioinspired Cell Silicification: From Extracellular to Intracellular. J Am Chem Soc 2021; 143:6305-6322. [PMID: 33826324 DOI: 10.1021/jacs.1c00814] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In nature, biosilicification directs the formation of elaborate amorphous silica exoskeletons that provide diatoms mechanically strong, chemically inert, non-decomposable silica armor conferring chemical and thermal stability as well as resistance to microbial attack, without changing the optical transparency or adversely effecting nutrient and waste exchange required for growth. These extraordinary silica/cell biocomposites have inspired decades of biomimetic research aimed at replication of diatoms' hierarchically organized exoskeletons, immobilization of cells or living organisms within silica matrices and coatings to protect them against harmful external stresses, genetic re-programming of cellular functions by virtue of physico-chemical confinement within silica, cellular integration into devices, and endowment of cells with non-native, abiotic properties through facile silica functionalization. In this Perspective, we focus our discussions on the development and concomitant challenges of bioinspired cell silicification ranging from "cells encapsulated within 3D silica matrices" and "cells encapsulated within 2D silica shells" to extra- and intracellular silica replication, wherein all biomolecular interfaces are encased within nanoscopic layers of amorphous silica. We highlight notable examples of advances in the science and technology of biosilicification and consider challenges to advancing the field, where we propose cellular "mineralization" with arbitrary nanoparticle exoskeletons as a generalizable means to impart limitless abiotic properties and functions to cells, and, based on the interchangeability of water and silicic acid and analogies between amorphous ice and amorphous silica, we consider "freezing" cells within amorphous silica as an alternative to cryo-preservation.
Collapse
Affiliation(s)
- Qi Lei
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Jimin Guo
- Center for Micro-Engineered Materials, Department of Chemical and Biological Engineering, The University of New Mexico, Albuquerque, New Mexico 87131, United States.,Department of Internal Medicine, Molecular Medicine, The University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Fanhui Kong
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Jiangfan Cao
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Lu Wang
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Wei Zhu
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - C Jeffrey Brinker
- Center for Micro-Engineered Materials, Department of Chemical and Biological Engineering, The University of New Mexico, Albuquerque, New Mexico 87131, United States
| |
Collapse
|
17
|
Esrafili A, Wagner A, Inamdar S, Acharya AP. Covalent Organic Frameworks for Biomedical Applications. Adv Healthc Mater 2021; 10:e2002090. [PMID: 33475260 DOI: 10.1002/adhm.202002090] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/23/2020] [Indexed: 12/15/2022]
Abstract
Covalent organic frameworks (COFs) are porous organic polymeric materials that are composed of organic elements and linked together by the thermodynamically stable covalent bonds. The applications of COFs in energy sector and drug delivery are afforded because of the desirable properties of COFs, such as high stability, low density, large surface area, multidimensionality, porosity, and high-ordered crystalline structure expanded. In this review COFs are reviewed, from the perspective of different types of reported COFs, different methods for their synthesis, and their potential applications in the biomedical field. The main goal of this review is to introduce COFs as a biomaterial and to identify specific advantages of different types of COFs that can be exploited for specialized biomedical applications, such as immune engineering.
Collapse
Affiliation(s)
- Arezoo Esrafili
- Chemical Engineering School for the Engineering of Matter, Transport, and Energy Arizona State University Tempe AZ 85281 USA
| | - Avery Wagner
- Chemical Engineering School for the Engineering of Matter, Transport, and Energy Arizona State University Tempe AZ 85281 USA
| | - Sahil Inamdar
- Chemical Engineering School for the Engineering of Matter, Transport, and Energy Arizona State University Tempe AZ 85281 USA
| | - Abhinav P. Acharya
- Chemical Engineering School for the Engineering of Matter, Transport, and Energy Arizona State University Tempe AZ 85281 USA
- Biological Design Graduate Program School for Biological and Health Systems Engineering Arizona State University Tempe AZ 85281 USA
- Materials Science and Engineering School for the Engineering of Matter Transport and Energy Arizona State University Tempe AZ 85281 USA
- Biodesign Center for Immunotherapy Vaccines and Virotherapy Arizona State University Tempe AZ 85281 USA
| |
Collapse
|
18
|
Li M, Yu Y. Innate immune receptor clustering and its role in immune regulation. J Cell Sci 2021; 134:134/4/jcs249318. [PMID: 33597156 DOI: 10.1242/jcs.249318] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The discovery of receptor clustering in the activation of adaptive immune cells has revolutionized our understanding of the physical basis of immune signal transduction. In contrast to the extensive studies of adaptive immune cells, particularly T cells, there is a lesser, but emerging, recognition that the formation of receptor clusters is also a key regulatory mechanism in host-pathogen interactions. Many kinds of innate immune receptors have been found to assemble into nano- or micro-sized domains on the surfaces of cells. The clusters formed between diverse categories of innate immune receptors function as a multi-component apparatus for pathogen detection and immune response regulation. Here, we highlight these pioneering efforts and the outstanding questions that remain to be answered regarding this largely under-explored research topic. We provide a critical analysis of the current literature on the clustering of innate immune receptors. Our emphasis is on studies that draw connections between the phenomenon of receptor clustering and its functional role in innate immune regulation.
Collapse
Affiliation(s)
- Miao Li
- Department of Chemistry, Indiana University, Bloomington, IN 47401, USA
| | - Yan Yu
- Department of Chemistry, Indiana University, Bloomington, IN 47401, USA
| |
Collapse
|
19
|
Isser A, Livingston NK, Schneck JP. Biomaterials to enhance antigen-specific T cell expansion for cancer immunotherapy. Biomaterials 2021; 268:120584. [PMID: 33338931 PMCID: PMC7856270 DOI: 10.1016/j.biomaterials.2020.120584] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/22/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023]
Abstract
T cells are often referred to as the 'guided missiles' of our immune system because of their capacity to traffic to and accumulate at sites of infection or disease, destroy infected or mutated cells with high specificity and sensitivity, initiate systemic immune responses, sterilize infections, and produce long-lasting memory. As a result, they are a common target for a range of cancer immunotherapies. However, the myriad of challenges of expanding large numbers of T cells specific to each patient's unique tumor antigens has led researchers to develop alternative, more scalable approaches. Biomaterial platforms for expansion of antigen-specific T cells offer a path forward towards broadscale translation of personalized immunotherapies by providing "off-the-shelf", yet modular approaches to customize the phenotype, function, and specificity of T cell responses. In this review, we discuss design considerations and progress made in the development of ex vivo and in vivo technologies for activating antigen-specific T cells, including artificial antigen presenting cells, T cell stimulating scaffolds, biomaterials-based vaccines, and artificial lymphoid organs. Ultimate translation of these platforms as a part of cancer immunotherapy regimens hinges on an in-depth understanding of T cell biology and cell-material interactions.
Collapse
Affiliation(s)
- Ariel Isser
- Department of Biomedical Engineering, School of Medicine, USA; Institute for Cell Engineering, School of Medicine, USA
| | - Natalie K Livingston
- Department of Biomedical Engineering, School of Medicine, USA; Institute for Cell Engineering, School of Medicine, USA; Translational Tissue Engineering Center, USA; Institute for Nanobiotechnology, USA
| | - Jonathan P Schneck
- Institute for Cell Engineering, School of Medicine, USA; Department of Pathology, School of Medicine, USA; Institute for Nanobiotechnology, USA; Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
20
|
Zhang J, Zhao R, Li B, Farrukh A, Hoth M, Qu B, Del Campo A. Micropatterned soft hydrogels to study the interplay of receptors and forces in T cell activation. Acta Biomater 2021; 119:234-246. [PMID: 33099024 DOI: 10.1016/j.actbio.2020.10.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 09/28/2020] [Accepted: 10/15/2020] [Indexed: 12/30/2022]
Abstract
The analysis of T cell responses to mechanical properties of antigen presenting cells (APC) is experimentally challenging at T cell-APC interfaces. Soft hydrogels with adjustable mechanical properties and biofunctionalization are useful reductionist models to address this problem. Here, we report a methodology to fabricate micropatterned soft hydrogels with defined stiffness to form spatially confined T cell/hydrogel contact interfaces at micrometer scale. Using automatized microcontact printing we prepared arrays of anti-CD3 microdots on poly(acrylamide) hydrogels with Young's Modulus in the range of 2 to 50 kPa. We optimized the printing process to obtain anti-CD3 microdots with constant area (50 µm2, corresponding to 8 µm diameter) and comparable anti-CD3 density on hydrogels of different stiffness. The anti-CD3 arrays were recognized by T cells and restricted cell attachment to the printed areas. To test functionality of the hydrogel-T cell contact, we analyzed several key events downstream of T cell receptor (TCR) activation. Anti-CD3 arrays on hydrogels activated calcium influx, induced rearrangement of the actin cytoskeleton, and led to Zeta-chain-associated protein kinase 70 (ZAP70) phosphorylation. Interestingly, upon increase in the stiffness, ZAP70 phosphorylation was enhanced, whereas the rearrangements of F-actin (F-actin clearance) and phosphorylated ZAP70 (ZAP70/pY centralization) were unaffected. Our results show that micropatterned hydrogels allow tuning of stiffness and receptor presentation to analyze TCR mediated T cell activation as function of mechanical, biochemical, and geometrical parameters.
Collapse
Affiliation(s)
- Jingnan Zhang
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123 Saarbrücken, Germany; Chemistry Department, Saarland University, 66123 Saarbrücken, Germany
| | - Renping Zhao
- Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Homburg, 66421 Germany
| | - Bin Li
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123 Saarbrücken, Germany
| | - Aleeza Farrukh
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123 Saarbrücken, Germany
| | - Markus Hoth
- Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Homburg, 66421 Germany
| | - Bin Qu
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123 Saarbrücken, Germany; Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Homburg, 66421 Germany
| | - Aránzazu Del Campo
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123 Saarbrücken, Germany; Chemistry Department, Saarland University, 66123 Saarbrücken, Germany.
| |
Collapse
|
21
|
Lei K, Kurum A, Tang L. Mechanical Immunoengineering of T cells for Therapeutic Applications. Acc Chem Res 2020; 53:2777-2790. [PMID: 33258577 DOI: 10.1021/acs.accounts.0c00486] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
T cells, a key component in adaptive immunity, are central to many immunotherapeutic modalities aimed at treating various diseases including cancer, infectious diseases, and autoimmune disorders. The past decade has witnessed tremendous progress in immunotherapy, which aims at activation or suppression of the immune responses for disease treatments. Most strikingly, cancer immunotherapy has led to curative responses in a fraction of patients with relapsed or refractory cancers. However, extending those clinical benefits to a majority of cancer patients remains challenging. In order to improve both efficacy and safety of T cell-based immunotherapies, significant effort has been devoted to modulating biochemical signals to enhance T cell proliferation, effector functions, and longevity. Such strategies include discovery of new immune checkpoints, design of armored chimeric antigen receptor (CAR) T cells, and targeted delivery of stimulatory cytokines and so on.Despite the intense global research effort in developing novel cancer immunotherapies, a major dimension of the interactions between cancer and the immune system, its biomechanical aspect, has been largely underappreciated. Throughout their lifecycle, T cells constantly survey a multitude of organs and tissues and experience diverse biomechanical environments, such as shear force in the blood flow and a broad range of tissue stiffness. Furthermore, biomechanical properties of tissues or cells may be altered in disease and inflammation. Biomechanical cues, including both passive mechanical cues and active mechanical forces, have been shown to govern T cell development, activation, migration, differentiation, and effector functions. In other words, T cells can sense, respond to, and adapt to both passive mechanical cues and active mechanical forces.Biomechanical cues have been intensively studied at a fundamental level but are yet to be extensively incorporated in the design of immunotherapies. Nonetheless, the growing knowledge of T cell mechanobiology has formed the basis for the development of novel engineering strategies to mechanically modulate T cell immunity, a nascent field that we termed "mechanical immunoengineering". Mechanical immunoengineering exploits biomechanical cues (e.g., stiffness and external forces) to modulate T cell differentiation, proliferation, effector functions, etc., for diagnostic or therapeutic applications. It provides an additional dimension, complementary to traditional modulation of biochemical cues (e.g., antigen density and co-stimulatory signals), to tailor T cell immune responses and enhance therapeutic outcomes. For example, stiff antigen-presenting matrices have been shown to enhance T cell proliferation independently of the intensity of biochemical stimulatory signals. Current strategies of mechanical immunoengineering of T cells can be categorized into two major fields including passive mechanical cue-oriented and active force-oriented strategies. In this Account, we first present a brief overview of T cell mechanobiology. Next, we summarize recent advances in mechanical immunoengineering, discuss the roles of chemistry and material science in the development of these engineering strategies, and highlight potential therapeutic applications. Finally, we present our perspective on the future directions in mechanical immunoengineering and critical steps to translate mechanical immunoengineering strategies into therapeutic applications in the clinic.
Collapse
|
22
|
Maynard SA, Winter CW, Cunnane EM, Stevens MM. Advancing Cell-Instructive Biomaterials Through Increased Understanding of Cell Receptor Spacing and Material Surface Functionalization. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2020; 7:553-547. [PMID: 34805482 PMCID: PMC8594271 DOI: 10.1007/s40883-020-00180-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Abstract Regenerative medicine is aimed at restoring normal tissue function and can benefit from the application of tissue engineering and nano-therapeutics. In order for regenerative therapies to be effective, the spatiotemporal integration of tissue-engineered scaffolds by the native tissue, and the binding/release of therapeutic payloads by nano-materials, must be tightly controlled at the nanoscale in order to direct cell fate. However, due to a lack of insight regarding cell–material interactions at the nanoscale and subsequent downstream signaling, the clinical translation of regenerative therapies is limited due to poor material integration, rapid clearance, and complications such as graft-versus-host disease. This review paper is intended to outline our current understanding of cell–material interactions with the aim of highlighting potential areas for knowledge advancement or application in the field of regenerative medicine. This is achieved by reviewing the nanoscale organization of key cell surface receptors, the current techniques used to control the presentation of cell-interactive molecules on material surfaces, and the most advanced techniques for characterizing the interactions that occur between cell surface receptors and materials intended for use in regenerative medicine. Lay Summary The combination of biology, chemistry, materials science, and imaging technology affords exciting opportunities to better diagnose and treat a wide range of diseases. Recent advances in imaging technologies have enabled better understanding of the specific interactions that occur between human cells and their immediate surroundings in both health and disease. This biological understanding can be used to design smart therapies and tissue replacements that better mimic native tissue. Here, we discuss the advances in molecular biology and technologies that can be employed to functionalize materials and characterize their interaction with biological entities to facilitate the design of more sophisticated medical therapies.
Collapse
Affiliation(s)
- Stephanie A. Maynard
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ UK
| | - Charles W. Winter
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ UK
| | - Eoghan M. Cunnane
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ UK
| | - Molly M. Stevens
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ UK
| |
Collapse
|
23
|
Pérez del Río E, Santos F, Rodriguez Rodriguez X, Martínez-Miguel M, Roca-Pinilla R, Arís A, Garcia-Fruitós E, Veciana J, Spatz JP, Ratera I, Guasch J. CCL21-loaded 3D hydrogels for T cell expansion and differentiation. Biomaterials 2020; 259:120313. [DOI: 10.1016/j.biomaterials.2020.120313] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 08/08/2020] [Accepted: 08/08/2020] [Indexed: 01/21/2023]
|
24
|
Zhang X, Mariano CF, Ando Y, Shen K. Bioengineering tools for probing intracellular events in T lymphocytes. WIREs Mech Dis 2020; 13:e1510. [PMID: 33073545 DOI: 10.1002/wsbm.1510] [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: 07/16/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 11/11/2022]
Abstract
T lymphocytes are the central coordinator and executor of many immune functions. The activation and function of T lymphocytes are mediated through the engagement of cell surface receptors and regulated by a myriad of intracellular signaling network. Bioengineering tools, including imaging modalities and fluorescent probes, have been developed and employed to elucidate the cellular events throughout the functional lifespan of T cells. A better understanding of these events can broaden our knowledge in the immune systems biology, as well as accelerate the development of effective diagnostics and immunotherapies. Here we review the commonly used and recently developed techniques and probes for monitoring T lymphocyte intracellular events, following the order of intracellular events in T cells from activation, signaling, metabolism to apoptosis. The techniques introduced here can be broadly applied to other immune cells and cell systems. This article is categorized under: Immune System Diseases > Molecular and Cellular Physiology Immune System Diseases > Biomedical Engineering Infectious Diseases > Biomedical Engineering.
Collapse
Affiliation(s)
- Xinyuan Zhang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Chelsea F Mariano
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Yuta Ando
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Keyue Shen
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA.,USC Stem Cell, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
25
|
Demetriou P, Abu-Shah E, Valvo S, McCuaig S, Mayya V, Kvalvaag A, Starkey T, Korobchevskaya K, Lee LYW, Friedrich M, Mann E, Kutuzov MA, Morotti M, Wietek N, Rada H, Yusuf S, Afrose J, Siokis A, Meyer-Hermann M, Ahmed AA, Depoil D, Dustin ML. A dynamic CD2-rich compartment at the outer edge of the immunological synapse boosts and integrates signals. Nat Immunol 2020; 21:1232-1243. [PMID: 32929275 PMCID: PMC7611174 DOI: 10.1038/s41590-020-0770-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 07/28/2020] [Indexed: 12/18/2022]
Abstract
The CD2-CD58 recognition system promotes adhesion and signaling and counters exhaustion in human T cells. We found that CD2 localized to the outer edge of the mature immunological synapse, with cellular or artificial APC, in a pattern we refer to as a 'CD2 corolla'. The corolla captured engaged CD28, ICOS, CD226 and SLAM-F1 co-stimulators. The corolla amplified active phosphorylated Src-family kinases (pSFK), LAT and PLC-γ over T cell receptor (TCR) alone. CD2-CD58 interactions in the corolla boosted signaling by 77% as compared with central CD2-CD58 interactions. Engaged PD-1 invaded the CD2 corolla and buffered CD2-mediated amplification of TCR signaling. CD2 numbers and motifs in its cytoplasmic tail controlled corolla formation. CD8+ tumor-infiltrating lymphocytes displayed low expression of CD2 in the majority of people with colorectal, endometrial or ovarian cancer. CD2 downregulation may attenuate antitumor T cell responses, with implications for checkpoint immunotherapies.
Collapse
Affiliation(s)
| | - Enas Abu-Shah
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Salvatore Valvo
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Sarah McCuaig
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Viveka Mayya
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Skirball Institute of Biomolecular Medicine, New York University of School of Medicine, New York, NY, USA
| | - Audun Kvalvaag
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Thomas Starkey
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | | | - Lennard Y W Lee
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | | | - Elizabeth Mann
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Mikhail A Kutuzov
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Matteo Morotti
- Ovarian Cancer Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Nina Wietek
- Ovarian Cancer Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Heather Rada
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Shamsideen Yusuf
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Jehan Afrose
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Skirball Institute of Biomolecular Medicine, New York University of School of Medicine, New York, NY, USA
- Department of Medical Laboratory Sciences, CUNY Hunter College, New York, NY, USA
| | - Anastasios Siokis
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology (BRICS), Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology (BRICS), Helmholtz Centre for Infection Research, Braunschweig, Germany
- Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Ahmed Ashour Ahmed
- Ovarian Cancer Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - David Depoil
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Skirball Institute of Biomolecular Medicine, New York University of School of Medicine, New York, NY, USA
- Immunocore Ltd, Abingdon, Oxford, UK
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
- Skirball Institute of Biomolecular Medicine, New York University of School of Medicine, New York, NY, USA.
| |
Collapse
|
26
|
Activation and degranulation of CAR-T cells using engineered antigen-presenting cell surfaces. PLoS One 2020; 15:e0238819. [PMID: 32976541 PMCID: PMC7518621 DOI: 10.1371/journal.pone.0238819] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 08/24/2020] [Indexed: 11/19/2022] Open
Abstract
Adoptive cell transfer of Chimeric Antigen Receptor (CAR)-T cells showed promising results in patients with B cell malignancies. However, the detailed mechanism of CAR-T cell interaction with the target tumor cells is still not well understood. This work provides a systematic method for analyzing the activation and degranulation of second-generation CAR-T cells utilizing antigen-presenting cell surfaces. Antigen-presenting cell surfaces composed of circular micropatterns of CAR-specific anti-idiotype antibodies have been developed to mimic the interaction of CAR-T cells with target tumor cells using micro-contact printing. The levels of activation and degranulation of fixed non-transduced T cells (NT), CD19.CAR-T cells, and GD2.CAR-T cells on the antigen-presenting cell surfaces were quantified and compared by measuring the intensity of the CD3ζ chain phosphorylation and the Lysosome-Associated Membrane Protein 1 (LAMP-1), respectively. The size and morphology of the cells were also measured. The intracellular Ca2+ flux of NT and CAR-T cells upon engagement with the antigen-presenting cell surface was reported. Results suggest that NT and CD19.CAR-T cells have comparable activation levels, while NT have higher degranulation levels than CD19.CAR-T cells and GD2.CAR-T cells. The findings show that antigen-presenting cell surfaces allow a quantitative analysis of the molecules involved in synapse formation in different CAR-T cells in a systematic, reproducible manner.
Collapse
|
27
|
Sun L, Shen F, Xu J, Han X, Fan C, Liu Z. DNA‐Edited Ligand Positioning on Red Blood Cells to Enable Optimized T Cell Activation for Adoptive Immunotherapy. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202003367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Lele Sun
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices Soochow University Suzhou 215123 Jiangsu China
| | - Fengyun Shen
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices Soochow University Suzhou 215123 Jiangsu China
| | - Jun Xu
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices Soochow University Suzhou 215123 Jiangsu China
| | - Xiao Han
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices Soochow University Suzhou 215123 Jiangsu China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering Shanghai Jiao Tong University Shanghai 201240 China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices Soochow University Suzhou 215123 Jiangsu China
| |
Collapse
|
28
|
DNA‐Edited Ligand Positioning on Red Blood Cells to Enable Optimized T Cell Activation for Adoptive Immunotherapy. Angew Chem Int Ed Engl 2020; 59:14842-14853. [DOI: 10.1002/anie.202003367] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/25/2020] [Indexed: 12/21/2022]
|
29
|
Abstract
We present a reproducible protocol for fabrication of polyacrylamide (PAA) hydrogel-based nano-patterns and nano-textures with a wide range of elastic rigidities to study fundamental cell behaviors, such as mechanosensitivity and motility. We explore the benefits of this protocol by successfully testing the compatibility of the PAA platforms with super-resolution microscopy, which is largely unavailable with platforms of nano-scale textures made from different polymers. We also utilized soft and rigid nano-textures to study the mechanosensing basis of T cell behavior and phenotype. For complete information on the generation and use of this protocol, please refer to Tabdanov et al. (2018b). Contact guidance cues are critical for directed cell migration in health and disease Manufacturing nano- and micro- scale patterns and textures with a wide range of rigidities The engineered platforms can be used to study motility in a wide range of cell types Deformable nano-textures are compatible with super-resolution imaging
Collapse
|
30
|
T cell activation and immune synapse organization respond to the microscale mechanics of structured surfaces. Proc Natl Acad Sci U S A 2019; 116:19835-19840. [PMID: 31527238 DOI: 10.1073/pnas.1906986116] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cells have the remarkable ability to sense the mechanical stiffness of their surroundings. This has been studied extensively in the context of cells interacting with planar surfaces, a conceptually elegant model that also has application in biomaterial design. However, physiological interfaces are spatially complex, exhibiting topographical features that are described over multiple scales. This report explores mechanosensing of microstructured elastomer surfaces by CD4+ T cells, key mediators of the adaptive immune response. We show that T cells form complex interactions with elastomer micropillar arrays, extending processes into spaces between structures and forming local areas of contraction and expansion dictated by the layout of microtubules within this interface. Conversely, cytoskeletal reorganization and intracellular signaling are sensitive to the pillar dimensions and flexibility. Unexpectedly, these measures show different responses to substrate rigidity, suggesting competing processes in overall T cell mechanosensing. The results of this study demonstrate that T cells sense the local rigidity of their environment, leading to strategies for biomaterial design.
Collapse
|
31
|
Mayya V, Judokusumo E, Abu Shah E, Peel CG, Neiswanger W, Depoil D, Blair DA, Wiggins CH, Kam LC, Dustin ML. Durable Interactions of T Cells with T Cell Receptor Stimuli in the Absence of a Stable Immunological Synapse. Cell Rep 2019; 22:340-349. [PMID: 29320731 PMCID: PMC5775504 DOI: 10.1016/j.celrep.2017.12.052] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 10/02/2017] [Accepted: 12/14/2017] [Indexed: 12/03/2022] Open
Abstract
T cells engage in two modes of interaction with antigen-presenting surfaces: stable synapses and motile kinapses. Although it is surmised that durable interactions of T cells with antigen-presenting cells involve synapses, in situ 3D imaging cannot resolve the mode of interaction. We have established in vitro 2D platforms and quantitative metrics to determine cell-intrinsic modes of interaction when T cells are faced with spatially continuous or restricted stimulation. All major resting human T cell subsets, except memory CD8 T cells, spend more time in the kinapse mode on continuous stimulatory surfaces. Surprisingly, we did not observe any concordant relationship between the mode and durability of interaction on cell-sized stimulatory spots. Naive CD8 T cells maintain kinapses for more than 3 hr before leaving stimulatory spots, whereas their memory counterparts maintain synapses for only an hour before leaving. Thus, durable interactions do not require stable synapses. Naive T cells spend more time in the motile kinapse state Only human memory CD8 T cells spend more time in the stable synapse state Kinapses do not reduce durability of interaction with cell-sized stimulatory spots Spatial restriction of TCR stimulation does not force formation of a synapse
Collapse
Affiliation(s)
- Viveka Mayya
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK; Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Edward Judokusumo
- Department of Biological Engineering, Columbia University, New York, NY 10027, USA
| | - Enas Abu Shah
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Christopher G Peel
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Willie Neiswanger
- Machine Learning Department, Carnegie Mellon University, Pittsburgh, PA 15213, USA; Department of Applied Physics and Applied Mathematics, Columbia University, New York, NY 10027, USA
| | - David Depoil
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - David A Blair
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Chris H Wiggins
- Department of Applied Physics and Applied Mathematics, Columbia University, New York, NY 10027, USA
| | - Lance C Kam
- Department of Biological Engineering, Columbia University, New York, NY 10027, USA
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK; Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
32
|
Kim S, Shah SB, Graney PL, Singh A. Multiscale engineering of immune cells and lymphoid organs. NATURE REVIEWS. MATERIALS 2019; 4:355-378. [PMID: 31903226 PMCID: PMC6941786 DOI: 10.1038/s41578-019-0100-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Immunoengineering applies quantitative and materials-based approaches for the investigation of the immune system and for the development of therapeutic solutions for various diseases, such as infection, cancer, inflammatory diseases and age-related malfunctions. The design of immunomodulatory and cell therapies requires the precise understanding of immune cell formation and activation in primary, secondary and ectopic tertiary immune organs. However, the study of the immune system has long been limited to in vivo approaches, which often do not allow multidimensional control of intracellular and extracellular processes, and to 2D in vitro models, which lack physiological relevance. 3D models built with synthetic and natural materials enable the structural and functional recreation of immune tissues. These models are being explored for the investigation of immune function and dysfunction at the cell, tissue and organ levels. In this Review, we discuss 2D and 3D approaches for the engineering of primary, secondary and tertiary immune structures at multiple scales. We highlight important insights gained using these models and examine multiscale engineering strategies for the design and development of immunotherapies. Finally, dynamic 4D materials are investigated for their potential to provide stimuli-dependent and context-dependent scaffolds for the generation of immune organ models.
Collapse
Affiliation(s)
- Sungwoong Kim
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, USA
- These authors contributed equally: Sungwoong Kim, Shivem B. Shah, Pamela L. Graney
| | - Shivem B. Shah
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- These authors contributed equally: Sungwoong Kim, Shivem B. Shah, Pamela L. Graney
| | - Pamela L. Graney
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
- These authors contributed equally: Sungwoong Kim, Shivem B. Shah, Pamela L. Graney
| | - Ankur Singh
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
- Englander Institute for Precision Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
33
|
Motsch V, Brameshuber M, Baumgart F, Schütz GJ, Sevcsik E. A micropatterning platform for quantifying interaction kinetics between the T cell receptor and an intracellular binding protein. Sci Rep 2019; 9:3288. [PMID: 30824760 PMCID: PMC6397226 DOI: 10.1038/s41598-019-39865-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 02/01/2019] [Indexed: 12/14/2022] Open
Abstract
A complete understanding of signaling processes at the plasma membrane depends on a quantitative characterization of the interactions of the involved proteins. Fluorescence recovery after photobleaching (FRAP) is a widely used and convenient technique to obtain kinetic parameters on protein interactions in living cells. FRAP experiments to determine unbinding time constants for proteins at the plasma membrane, however, are often hampered by non-specific contributions to the fluorescence recovery signal. On the example of the interaction between the T cell receptor (TCR) and the Syk kinase ZAP70, we present here an approach based on protein micropatterning that allows the elimination of such non-specific contributions and considerably simplifies analysis of FRAP data. Specifically, detection and reference areas are created within single cells, each being either enriched or depleted in TCR, which permits the isolation of ZAP70-TCR binding in a straight-forward manner. We demonstrate the applicability of our method by comparing it to a conventional FRAP approach.
Collapse
Affiliation(s)
- Viktoria Motsch
- Institute of Applied Physics, TU Wien, Wiedner Hauptstrasse 8-10, 1040, Vienna, Austria
| | - Mario Brameshuber
- Institute of Applied Physics, TU Wien, Wiedner Hauptstrasse 8-10, 1040, Vienna, Austria
| | - Florian Baumgart
- Institute of Applied Physics, TU Wien, Wiedner Hauptstrasse 8-10, 1040, Vienna, Austria
| | - Gerhard J Schütz
- Institute of Applied Physics, TU Wien, Wiedner Hauptstrasse 8-10, 1040, Vienna, Austria
| | - Eva Sevcsik
- Institute of Applied Physics, TU Wien, Wiedner Hauptstrasse 8-10, 1040, Vienna, Austria.
| |
Collapse
|
34
|
Olden BR, Perez CR, Wilson AL, Cardle II, Lin YS, Kaehr B, Gustafson JA, Jensen MC, Pun SH. Cell-Templated Silica Microparticles with Supported Lipid Bilayers as Artificial Antigen-Presenting Cells for T Cell Activation. Adv Healthc Mater 2019; 8:e1801188. [PMID: 30549244 PMCID: PMC6394850 DOI: 10.1002/adhm.201801188] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 11/28/2018] [Indexed: 01/18/2023]
Abstract
Biomaterial properties that modulate T cell activation, growth, and differentiation are of significant interest in the field of cellular immunotherapy manufacturing. In this work, a new platform technology that allows for the modulation of various activation particle design parameters important for polyclonal T cell activation is presented. Artificial antigen presenting cells (aAPCs) are successfully created using supported lipid bilayers on various cell-templated silica microparticles with defined membrane fluidity and stimulating antibody density. This panel of aAPCs is used to probe the importance of activation particle shape, size, membrane fluidity, and stimulation antibody density on T cell outgrowth and differentiation. All aAPC formulations are able to stimulate T cell growth, and preferentially promote CD8+ T cell growth over CD4+ T cell growth when compared to commercially available pendant antibody-conjugated particles. T cells cultured with HeLa- and red blood cell-templated aAPCs have a less-differentiated and less-exhausted phenotype than those cultured with spherical aAPCs with matched membrane coatings when cultured for 14 days. These results support continued exploration of silica-supported lipid bilayers as an aAPC platform.
Collapse
Affiliation(s)
- Brynn R. Olden
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, USA,
| | - Caleb R. Perez
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, USA,
| | - Ashley L. Wilson
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Ian I. Cardle
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, USA,
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Yu-Shen Lin
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, USA,
| | - Bryan Kaehr
- Advanced Materials Laboratory, Sandia National Laboratories, Albuquerque, NM 87185, USA
| | - Joshua A. Gustafson
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Michael C. Jensen
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Suzie H. Pun
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, USA,
| |
Collapse
|
35
|
Hager R, Arnold A, Sevcsik E, Schütz GJ, Howorka S. Tunable DNA Hybridization Enables Spatially and Temporally Controlled Surface-Anchoring of Biomolecular Cargo. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:15021-15027. [PMID: 30160973 PMCID: PMC6291803 DOI: 10.1021/acs.langmuir.8b01942] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 07/30/2018] [Indexed: 05/04/2023]
Abstract
The controlled immobilization of biomolecules onto surfaces is relevant in biosensing and cell biological research. Spatial control is achieved by surface-tethering molecules in micro- or nanoscale patterns. Yet, there is an increasing demand for temporal control over how long biomolecular cargo stays immobilized until released into the medium. Here, we present a DNA hybridization-based approach to reversibly anchor biomolecular cargo onto micropatterned surfaces. Cargo is linked to a DNA oligonucleotide that hybridizes to a sequence-complementary, surface-tethered strand. The cargo is released from the substrate by the addition of an oligonucleotide that disrupts the duplex interaction via toehold-mediated strand displacement. The unbound tether strand can be reloaded. The generic strategy is implemented with small-molecule or protein cargo, varying DNA sequences, and multiple surface patterning routes. The approach may be used as a tool in biological research to switch membrane proteins from a locally fixed to a free state, or in biosensing to shed biomolecular receptors to regenerate the sensor surface.
Collapse
Affiliation(s)
- Roland Hager
- Center
for Advanced Bioanalysis GmbH. Linz, 4020, Austria
| | - Andreas Arnold
- Institute
of Applied Physics, TU Wien, Wien, 1040, Austria
| | - Eva Sevcsik
- Institute
of Applied Physics, TU Wien, Wien, 1040, Austria
| | | | - Stefan Howorka
- Center
for Advanced Bioanalysis GmbH. Linz, 4020, Austria
- Department
of Chemistry, Institute for Structural and Molecular Biology, University College London (UCL), London, WC1E 6BT, U.K.
| |
Collapse
|
36
|
Hickey JW, Kosmides AK, Schneck JP. Engineering Platforms for T Cell Modulation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 341:277-362. [PMID: 30262034 DOI: 10.1016/bs.ircmb.2018.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
T cells are crucial contributors to mounting an effective immune response and increasingly the focus of therapeutic interventions in cancer, infectious disease, and autoimmunity. Translation of current T cell immunotherapies has been hindered by off-target toxicities, limited efficacy, biological variability, and high costs. As T cell therapeutics continue to develop, the application of engineering concepts to control their delivery and presentation will be critical for their success. Here, we outline the engineer's toolbox and contextualize it with the biology of T cells. We focus on the design principles of T cell modulation platforms regarding size, shape, material, and ligand choice. Furthermore, we review how application of these design principles has already impacted T cell immunotherapies and our understanding of T cell biology. Recent, salient examples from protein engineering, synthetic particles, cellular and genetic engineering, and scaffolds and surfaces are provided to reinforce the importance of design considerations. Our aim is to provide a guide for immunologists, engineers, clinicians, and the pharmaceutical sector for the design of T cell-targeting platforms.
Collapse
Affiliation(s)
- John W Hickey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Institute for NanoBiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Alyssa K Kosmides
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Institute for NanoBiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jonathan P Schneck
- Institute for NanoBiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
37
|
de la Zerda A, Kratochvil MJ, Suhar NA, Heilshorn SC. Review: Bioengineering strategies to probe T cell mechanobiology. APL Bioeng 2018; 2:021501. [PMID: 31069295 PMCID: PMC6324202 DOI: 10.1063/1.5006599] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 01/29/2018] [Indexed: 01/08/2023] Open
Abstract
T cells play a major role in adaptive immune response, and T cell dysfunction can lead to the progression of several diseases that are often associated with changes in the mechanical properties of tissues. However, the concept that mechanical forces play a vital role in T cell activation and signaling is relatively new. The endogenous T cell microenvironment is highly complex and dynamic, involving multiple, simultaneous cell-cell and cell-matrix interactions. This native complexity has made it a challenge to isolate the effects of mechanical stimuli on T cell activation. In response, researchers have begun developing engineered platforms that recapitulate key aspects of the native microenvironment to dissect these complex interactions in order to gain a better understanding of T cell mechanotransduction. In this review, we first describe some of the unique characteristics of T cells and the mounting research that has shown they are mechanosensitive. We then detail the specific bioengineering strategies that have been used to date to measure and perturb the mechanical forces at play during T cell activation. In addition, we look at engineering strategies that have been used successfully in mechanotransduction studies for other cell types and describe adaptations that may make them suitable for use with T cells. These engineering strategies can be classified as 2D, so-called 2.5D, or 3D culture systems. In the future, findings from this emerging field will lead to an optimization of culture environments for T cell expansion and the development of new T cell immunotherapies for cancer and other immune diseases.
Collapse
Affiliation(s)
- Adi de la Zerda
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA
| | | | - Nicholas A Suhar
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
38
|
Piscopo NJ, Mueller KP, Das A, Hematti P, Murphy WL, Palecek SP, Capitini CM, Saha K. Bioengineering Solutions for Manufacturing Challenges in CAR T Cells. Biotechnol J 2018; 13:10.1002/biot.201700095. [PMID: 28840981 PMCID: PMC5796845 DOI: 10.1002/biot.201700095] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/26/2017] [Indexed: 12/13/2022]
Abstract
The next generation of therapeutic products to be approved for the clinic is anticipated to be cell therapies, termed "living drugs" for their capacity to dynamically and temporally respond to changes during their production ex vivo and after their administration in vivo. Genetically engineered chimeric antigen receptor (CAR) T cells have rapidly developed into powerful tools to harness the power of immune system manipulation against cancer. Regulatory agencies are beginning to approve CAR T cell therapies due to their striking efficacy in treating some hematological malignancies. However, the engineering and manufacturing of such cells remains a challenge for widespread adoption of this technology. Bioengineering approaches including biomaterials, synthetic biology, metabolic engineering, process control and automation, and in vitro disease modeling could offer promising methods to overcome some of these challenges. Here, we describe the manufacturing process of CAR T cells, highlighting potential roles for bioengineers to partner with biologists and clinicians to advance the manufacture of these complex cellular products under rigorous regulatory and quality control.
Collapse
Affiliation(s)
- Nicole J Piscopo
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, WI, USA
| | - Katherine P Mueller
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, WI, USA
| | - Amritava Das
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, WI, USA
| | - Peiman Hematti
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Department of Orthopedics and Rehabilitation, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin, Madison, WI, USA
| | - Christian M Capitini
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Krishanu Saha
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| |
Collapse
|
39
|
Foncy J, Estève A, Degache A, Colin C, Cau JC, Malaquin L, Vieu C, Trévisiol E. Fabrication of Biomolecule Microarrays for Cell Immobilization Using Automated Microcontact Printing. Methods Mol Biol 2018; 1771:83-95. [PMID: 29633206 DOI: 10.1007/978-1-4939-7792-5_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Biomolecule microarrays are generally produced by conventional microarrayer, i.e., by contact or inkjet printing. Microcontact printing represents an alternative way of deposition of biomolecules on solid supports but even if various biomolecules have been successfully microcontact printed, the production of biomolecule microarrays in routine by microcontact printing remains a challenging task and needs an effective, fast, robust, and low-cost automation process. Here, we describe the production of biomolecule microarrays composed of extracellular matrix protein for the fabrication of cell microarrays by using an automated microcontact printing device. Large scale cell microarrays can be reproducibly obtained by this method.
Collapse
Affiliation(s)
- Julie Foncy
- Laboratory for Analysis and Architecture of Systems (LAAS-CNRS), Université de Toulouse, CNRS, INSA, Toulouse, France
| | - Aurore Estève
- Laboratory for Analysis and Architecture of Systems (LAAS-CNRS), Université de Toulouse, CNRS, INSA, Toulouse, France
| | | | - Camille Colin
- Laboratory for Analysis and Architecture of Systems (LAAS-CNRS), Université de Toulouse, CNRS, INSA, Toulouse, France
| | | | - Laurent Malaquin
- Laboratory for Analysis and Architecture of Systems (LAAS-CNRS), Université de Toulouse, CNRS, INSA, Toulouse, France
| | - Christophe Vieu
- Laboratory for Analysis and Architecture of Systems (LAAS-CNRS), Université de Toulouse, CNRS, INSA, Toulouse, France
| | - Emmanuelle Trévisiol
- Laboratory for Analysis and Architecture of Systems (LAAS-CNRS), Université de Toulouse, CNRS, INSA, Toulouse, France.
| |
Collapse
|
40
|
Abstract
Background Immunotherapy consists of activating the patient’s immune system to fight cancer and has the great potential of preventing future relapses thanks to immunological memory. A great variety of strategies have emerged to harness the immune system against tumors, from the administration of immunomodulatory agents that activate immune cells, to therapeutic vaccines or infusion of previously activated cancer-specific T cells. However, despite great recent progress many difficulties still remain, which prevent the widespread use of immunotherapy. Some of these limitations include: systemic toxicity, weak immune cellular responses or persistence over time and most ultimately costly and time-consuming procedures. Main body Synthetic and natural biomaterials hold great potential to address these hurdles providing biocompatible systems capable of targeted local delivery, co-delivery, and controlled and/or sustained release. In this review we discuss some of the bioengineered solutions and approaches developed so far and how biomaterials can be further implemented to help and shape the future of cancer immunotherapy. Conclusion The bioengineering strategies here presented constitute a powerful toolkit to develop safe and successful novel cancer immunotherapies.
Collapse
|
41
|
Ben-Akiva E, Meyer RA, Wilson DR, Green JJ. Surface engineering for lymphocyte programming. Adv Drug Deliv Rev 2017; 114:102-115. [PMID: 28501510 PMCID: PMC5688954 DOI: 10.1016/j.addr.2017.05.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 05/01/2017] [Accepted: 05/08/2017] [Indexed: 12/11/2022]
Abstract
The once nascent field of immunoengineering has recently blossomed to include approaches to deliver and present biomolecules to program diverse populations of lymphocytes to fight disease. Building upon improved understanding of the molecular and physical mechanics of lymphocyte activation, varied strategies for engineering surfaces to activate and deactivate T-Cells, B-Cells and natural killer cells are in preclinical and clinical development. Surfaces have been engineered at the molecular level in terms of the presence of specific biological factors, their arrangement on a surface, and their diffusivity to elicit specific lymphocyte fates. In addition, the physical and mechanical characteristics of the surface including shape, anisotropy, and rigidity of particles for lymphocyte activation have been fine-tuned. Utilizing these strategies, acellular systems have been engineered for the expansion of T-Cells and natural killer cells to clinically relevant levels for cancer therapies as well as engineered to program B-Cells to better combat infectious diseases.
Collapse
Affiliation(s)
- Elana Ben-Akiva
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Johns Hopkins Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Randall A Meyer
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - David R Wilson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jordan J Green
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Johns Hopkins Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Materials Science and Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
42
|
Delcassian D, Sattler S, Dunlop IE. T cell immunoengineering with advanced biomaterials. Integr Biol (Camb) 2017; 9:211-222. [PMID: 28252135 PMCID: PMC6034443 DOI: 10.1039/c6ib00233a] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 02/15/2017] [Indexed: 12/25/2022]
Abstract
Recent advances in biomaterials design offer the potential to actively control immune cell activation and behaviour. Many human diseases, such as infections, cancer, and autoimmune disorders, are partly mediated by inappropriate or insufficient activation of the immune system. T cells play a central role in the host immune response to these diseases, and so constitute a promising cell type for manipulation. In vivo, T cells are stimulated by antigen presenting cells (APC), therefore to design immunoengineering biomaterials that control T cell behaviour, artificial interfaces that mimic the natural APC-T cell interaction are required. This review draws together research in the design and fabrication of such biomaterial interfaces, and highlights efforts to elucidate key parameters in T cell activation, such as substrate mechanical properties and spatial organization of receptors, illustrating how they can be manipulated by bioengineering approaches to alter T cell function.
Collapse
Affiliation(s)
- Derfogail Delcassian
- School of Pharmacy, University of Nottingham, NG7 2RD, UK. and Koch Institute for Integrative Cancer Research, MIT, Massachusetts, 02139, USA
| | - Susanne Sattler
- Imperial College London National Heart and Lung Institute, Du Cane Road, W12 0NN, London, UK
| | - Iain E Dunlop
- Department of Materials, Imperial College London, SW7 2AZ, UK.
| |
Collapse
|
43
|
Yi Y, Sanchez L, Gao Y, Lee K, Yu Y. Interrogating Cellular Functions with Designer Janus Particles. CHEMISTRY OF MATERIALS : A PUBLICATION OF THE AMERICAN CHEMICAL SOCIETY 2017; 29:1448-1460. [PMID: 31530969 PMCID: PMC6748339 DOI: 10.1021/acs.chemmater.6b05322] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Janus particles have two distinct surfaces or compartments. This enables novel applications that are impossible with homogeneous particles, ranging from the engineering of active colloidal metastructures to creating multimodal therapeutic materials. Recent years have witnessed a rapid development of novel Janus structures and exploration of their applications, particularly in the biomedical arena. It, therefore, becomes crucial to understand how Janus particles with surface or structural anisotropy might interact with biological systems and how such interactions may be exploited to manipulate biological responses. This perspective highlights recent studies that have employed Janus particles as novel toolsets to manipulate, measure, and understand cellular functions. Janus particles have been shown to have biological interactions different from uniform particles. Their surface anisotropy has been used to control the cell entry of synthetic particles, to spatially organize stimuli for the activation of immune cells, and to enable direct visualization and measurement of rotational dynamics of particles in living systems. The work included in this perspective showcases the significance of understanding the biological interactions of Janus particles and the tremendous potential of harnessing such interactions to advance the development of Janus structure-based biomaterials.
Collapse
Affiliation(s)
| | | | | | | | - Yan Yu
- Corresponding Author (Y.Yu)
| |
Collapse
|
44
|
Lee JH, Kam LC. Revealing the Role of Microscale Architecture in Immune Synapse Function Through Surface Micropatterning. Methods Mol Biol 2017; 1584:291-306. [PMID: 28255708 DOI: 10.1007/978-1-4939-6881-7_17] [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: 06/06/2023]
Abstract
The immune synapse has emerged as a compelling example of structural complexity within cell-cell interfaces. This chapter focuses on the use of microcontact printing to isolate and investigate how spatial organization of signaling molecules drives the function of immune cells. In the process detailed here, multiple rounds of microcontact printing are combined to create patterned surfaces that control the relative spatial localization of CD3 and CD28 signaling in T cells, effectively replacing an antigen presenting cell with an engineered surface. A set of approaches used to address key issues of T cell activation are described and discussed.
Collapse
Affiliation(s)
- Joung-Hyun Lee
- Department of Biomedical Engineering, Columbia University, 120 Amsterdam Ave Mail Code 8904, New York, NY, 10027, USA
| | - Lance C Kam
- Department of Biomedical Engineering, Columbia University, 120 Amsterdam Ave Mail Code 8904, New York, NY, 10027, USA.
| |
Collapse
|
45
|
Dillard P, Pi F, Lellouch AC, Limozin L, Sengupta K. Nano-clustering of ligands on surrogate antigen presenting cells modulates T cell membrane adhesion and organization. Integr Biol (Camb) 2016; 8:287-301. [PMID: 26887857 DOI: 10.1039/c5ib00293a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We investigate the adhesion and molecular organization of the plasma membrane of T lymphocytes interacting with a surrogate antigen presenting cell comprising glass supported ordered arrays of antibody (α-CD3) nano-dots dispersed in a non-adhesive matrix of polyethylene glycol (PEG). The local membrane adhesion and topography, as well as the distribution of the T cell receptors (TCRs) and the kinase ZAP-70, are influenced by dot-geometry, whereas the cell spreading area is determined by the overall average density of the ligands rather than specific characteristics of the dots. TCR clusters are recruited preferentially to the nano-dots and the TCR cluster size distribution has a weak dot-size dependence. On the patterns, the clusters are larger, more numerous, and more enriched in TCRs, as compared to the homogeneously distributed ligands at comparable concentrations. These observations support the idea that non-ligated TCRs residing in the non-adhered parts of the proximal membrane are able to diffuse and enrich the existing clusters at the ligand dots. However, long distance transport is impaired and cluster centralization in the form of a central supramolecular cluster (cSMAC) is not observed. Time-lapse imaging of early cell-surface contacts indicates that the ZAP-70 microclusters are directly recruited to the site of the antibody dots and this process is concomitant with membrane adhesion. These results together point to a complex interplay of adhesion, molecular organization and activation in response to spatially modulated stimulation.
Collapse
Affiliation(s)
- Pierre Dillard
- Aix-Marseille Université, CNRS, CINaM-UMR 7325, Marseille, 13288, France. and Laboratoire Adhésion & Inflammation Aix-Marseille Université\Inserm U1067\CNRS-UMR7333, Marseille 13288, France.
| | - Fuwei Pi
- Aix-Marseille Université, CNRS, CINaM-UMR 7325, Marseille, 13288, France.
| | - Annemarie C Lellouch
- Laboratoire Adhésion & Inflammation Aix-Marseille Université\Inserm U1067\CNRS-UMR7333, Marseille 13288, France.
| | - Laurent Limozin
- Laboratoire Adhésion & Inflammation Aix-Marseille Université\Inserm U1067\CNRS-UMR7333, Marseille 13288, France.
| | - Kheya Sengupta
- Aix-Marseille Université, CNRS, CINaM-UMR 7325, Marseille, 13288, France.
| |
Collapse
|
46
|
Sarkar S, Sabhachandani P, Stroopinsky D, Palmer K, Cohen N, Rosenblatt J, Avigan D, Konry T. Dynamic analysis of immune and cancer cell interactions at single cell level in microfluidic droplets. BIOMICROFLUIDICS 2016; 10:054115. [PMID: 27795747 PMCID: PMC5065572 DOI: 10.1063/1.4964716] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 09/29/2016] [Indexed: 05/06/2023]
Abstract
Cell-cell communication mediates immune responses to physiological stimuli at local and systemic levels. Intercellular communication occurs via a direct contact between cells as well as by secretory contact-independent mechanisms. However, there are few existing methods that allow quantitative resolution of contact-dependent and independent cellular processes in a rapid, precisely controlled, and dynamic format. This study utilizes a high-throughput microfluidic droplet array platform to analyze cell-cell interaction and effector functions at single cell level. Controlled encapsulation of distinct heterotypic cell pairs was achieved in a single-step cell loading process. Dynamic analysis of dendritic cell (DC)-T cell interactions demonstrated marked heterogeneity in the type of contact and duration. Non-stimulated DCs and T cells interacted less frequently and more transiently while antigen and chemokine-loaded DCs and T cells depicted highly stable interactions in addition to transient and sequential contact. The effector function of CD8+ T cells was assessed via cytolysis of multiple myeloma cell line. Variable cell conjugation periods and killing time were detected irrespective of the activation of T cells, although activated T cells delivered significantly higher cytotoxicity. T cell alloreactivity against the target cells was partially mediated by secretion of interferon gamma, which was abrogated by the addition of a neutralizing antibody. These results suggest that the droplet array-based microfluidic platform is a powerful technique for dynamic phenotypic screening and potentially applicable for evaluation of novel cell-based immunotherapeutic agents.
Collapse
Affiliation(s)
- S Sarkar
- Department of Pharmaceutical Sciences, Northeastern University , 360 Huntington Avenue, Boston, Massachusetts 02115, USA
| | - P Sabhachandani
- Department of Pharmaceutical Sciences, Northeastern University , 360 Huntington Avenue, Boston, Massachusetts 02115, USA
| | - D Stroopinsky
- Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, Massachusetts 02115, USA
| | - K Palmer
- Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, Massachusetts 02115, USA
| | - N Cohen
- Department of Pharmaceutical Sciences, Northeastern University , 360 Huntington Avenue, Boston, Massachusetts 02115, USA
| | - J Rosenblatt
- Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, Massachusetts 02115, USA
| | - D Avigan
- Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, Massachusetts 02115, USA
| | - T Konry
- Department of Pharmaceutical Sciences, Northeastern University , 360 Huntington Avenue, Boston, Massachusetts 02115, USA
| |
Collapse
|
47
|
Meacci G, Wolfenson H, Liu S, Stachowiak MR, Iskratsch T, Mathur A, Ghassemi S, Gauthier N, Tabdanov E, Lohner J, Gondarenko A, Chander AC, Roca-Cusachs P, O'Shaughnessy B, Hone J, Sheetz MP. α-Actinin links extracellular matrix rigidity-sensing contractile units with periodic cell-edge retractions. Mol Biol Cell 2016; 27:3471-3479. [PMID: 27122603 PMCID: PMC5221581 DOI: 10.1091/mbc.e16-02-0107] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 04/20/2016] [Indexed: 11/11/2022] Open
Abstract
During spreading and migration, the leading edges of cells undergo periodic protrusion-retraction cycles. The functional purpose of these cycles is unclear. Here, using submicrometer polydimethylsiloxane pillars as substrates for cell spreading, we show that periodic edge retractions coincide with peak forces produced by local contractile units (CUs) that assemble and disassemble along the cell edge to test matrix rigidity. We find that, whereas actin rearward flow produces a relatively constant force inward, the peak of local contractile forces by CUs scales with rigidity. The cytoskeletal protein α-actinin is shared between these two force-producing systems. It initially localizes to the CUs and subsequently moves inward with the actin flow. Knockdown of α-actinin causes aberrant rigidity sensing, loss of CUs, loss of protrusion-retraction cycles, and, surprisingly, enables the cells to proliferate on soft matrices. We present a model based on these results in which local CUs drive rigidity sensing and adhesion formation.
Collapse
Affiliation(s)
- Giovanni Meacci
- Department of Biological Sciences, Columbia University, New York, NY 10027 .,Department of Chemical Engineering, Columbia University, New York, NY 10027
| | - Haguy Wolfenson
- Department of Biological Sciences, Columbia University, New York, NY 10027
| | - Shuaimin Liu
- Department of Mechanical Engineering, Columbia University, New York, NY 10027
| | | | - Thomas Iskratsch
- Department of Biological Sciences, Columbia University, New York, NY 10027.,Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Anurag Mathur
- Department of Mechanical Engineering, Columbia University, New York, NY 10027
| | - Saba Ghassemi
- Department of Mechanical Engineering, Columbia University, New York, NY 10027
| | - Nils Gauthier
- Department of Biological Sciences, Columbia University, New York, NY 10027.,Mechanobiology Institute, National University of Singapore, Singapore, 117411
| | - Erdem Tabdanov
- Department of Biomedical Engineering, Columbia University, New York, NY 10027
| | - James Lohner
- Department of Biological Sciences, Columbia University, New York, NY 10027
| | | | - Ashok C Chander
- Department of Biological Sciences, Columbia University, New York, NY 10027
| | - Pere Roca-Cusachs
- Department of Biological Sciences, Columbia University, New York, NY 10027.,Institute for Bioengineering of Catalonia and University of Barcelona, 08028 Barcelona, Spain
| | - Ben O'Shaughnessy
- Department of Chemical Engineering, Columbia University, New York, NY 10027
| | - James Hone
- Department of Mechanical Engineering, Columbia University, New York, NY 10027
| | - Michael P Sheetz
- Department of Biological Sciences, Columbia University, New York, NY 10027 .,Mechanobiology Institute, National University of Singapore, Singapore, 117411
| |
Collapse
|
48
|
Hu J, Gondarenko AA, Dang AP, Bashour KT, O’Connor RS, Lee S, Liapis A, Ghassemi S, Milone MC, Sheetz MP, Dustin ML, Kam LC, Hone JC. High-Throughput Mechanobiology Screening Platform Using Micro- and Nanotopography. NANO LETTERS 2016; 16:2198-204. [PMID: 26990380 PMCID: PMC5403373 DOI: 10.1021/acs.nanolett.5b04364] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
We herein demonstrate the first 96-well plate platform to screen effects of micro- and nanotopographies on cell growth and proliferation. Existing high-throughput platforms test a limited number of factors and are not fully compatible with multiple types of testing and assays. This platform is compatible with high-throughput liquid handling, high-resolution imaging, and all multiwell plate-based instrumentation. We use the platform to screen for topographies and drug-topography combinations that have short- and long-term effects on T cell activation and proliferation. We coated nanofabricated "trench-grid" surfaces with anti-CD3 and anti-CD28 antibodies to activate T cells and assayed for interleukin 2 (IL-2) cytokine production. IL-2 secretion was enhanced at 200 nm trench width and >2.3 μm grating pitch; however, the secretion was suppressed at 100 nm width and <0.5 μm pitch. The enhancement on 200 nm grid trench was further amplified with the addition of blebbistatin to reduce contractility. The 200 nm grid pattern was found to triple the number of T cells in long-term expansion, a result with direct clinical applicability in adoptive immunotherapy.
Collapse
Affiliation(s)
- Junqiang Hu
- Department of Mechanical Engineering, Columbia University, New York, New York 10027, United States
| | - Alexander A. Gondarenko
- Department of Mechanical Engineering, Columbia University, New York, New York 10027, United States
| | - Alex P. Dang
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - Keenan T. Bashour
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - Roddy S. O’Connor
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Sunwoo Lee
- Department of Electrical Engineering, Columbia University, New York, New York 10027, United States
| | - Anastasia Liapis
- Department of Pathology, New York University School of Medicine, New York, New York 10016, United States
| | - Saba Ghassemi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Michael C. Milone
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Michael P. Sheetz
- Department of Biological Sciences, Columbia University, New York, New York 10027, United States
| | - Michael L. Dustin
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, The University of Oxford, Oxford, OX3 7FY, U.K
| | - Lance C. Kam
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - James C. Hone
- Department of Mechanical Engineering, Columbia University, New York, New York 10027, United States
| |
Collapse
|
49
|
Andersen AS, Aslan H, Dong M, Jiang X, Sutherland DS. Podosome Formation and Development in Monocytes Restricted by the Nanoscale Spatial Distribution of ICAM1. NANO LETTERS 2016; 16:2114-21. [PMID: 26861163 DOI: 10.1021/acs.nanolett.6b00519] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
We studied podosome formation and development in activated monocytes (THP1) at ICAM1 (intercellular adhesion molecule 1) nanopatterns of circular and ring-shaped domains and show that cellular binding to a preclustered ICAM1 nanopattern requires ligand patches of at least 200 nm (corresponding to 14 or more integrins). Podosome-like adhesion formation depends on the structure of the ligand pattern under the developing podosome with larger single domains promoting adhesion in a single patch and multiple smaller domains allowing podosome formation by integration of at least 2 smaller domains on either side of the podosome core. Maturation to rosette structures and recruitment of proteases were only observed with macroscopic ICAM1 presentation.
Collapse
Affiliation(s)
- Andreas S Andersen
- Interdisciplinary Nanoscience Center (iNANO), Århus University , Århus 8000, Denmark
| | - Hüsnü Aslan
- Interdisciplinary Nanoscience Center (iNANO), Århus University , Århus 8000, Denmark
| | - Mingdong Dong
- Interdisciplinary Nanoscience Center (iNANO), Århus University , Århus 8000, Denmark
| | - Xingyu Jiang
- National Center for Nanoscience and Technology (NCNST), Chinese Academy of Sciences (CAS) , Beijing, China
| | - Duncan S Sutherland
- Interdisciplinary Nanoscience Center (iNANO), Århus University , Århus 8000, Denmark
| |
Collapse
|
50
|
Konry T, Sarkar S, Sabhachandani P, Cohen N. Innovative Tools and Technology for Analysis of Single Cells and Cell-Cell Interaction. Annu Rev Biomed Eng 2016; 18:259-84. [PMID: 26928209 DOI: 10.1146/annurev-bioeng-090215-112735] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Heterogeneity in single-cell responses and intercellular interactions results from complex regulation of cell-intrinsic and environmental factors. Single-cell analysis allows not only detection of individual cellular characteristics but also correlation of genetic content with phenotypic traits in the same cell. Technological advances in micro- and nanofabrication have benefited single-cell analysis by allowing precise control of the localized microenvironment, cell manipulation, and sensitive detection capabilities. Additionally, microscale techniques permit rapid, high-throughput, multiparametric screening that has become essential for -omics research. This review highlights innovative applications of microscale platforms in genetic, proteomic, and metabolic detection in single cells; cell sorting strategies; and heterotypic cell-cell interaction. We discuss key design aspects of single-cell localization and isolation in microfluidic systems, dynamic and endpoint analyses, and approaches that integrate highly multiplexed detection of various intracellular species.
Collapse
Affiliation(s)
- Tania Konry
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts 02115; , , ,
| | - Saheli Sarkar
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts 02115; , , ,
| | - Pooja Sabhachandani
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts 02115; , , ,
| | - Noa Cohen
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts 02115; , , ,
| |
Collapse
|